2101
|
Impact of Cholesterol on Ischemic Stroke in Different Human-Like Hamster Models: A New Animal Model for Ischemic Stroke Study. Cells 2019; 8:cells8091028. [PMID: 31487778 PMCID: PMC6770656 DOI: 10.3390/cells8091028] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 08/28/2019] [Accepted: 08/29/2019] [Indexed: 12/03/2022] Open
Abstract
Rationale: While high low-density lipoprotein cholesterol (LDL-C) and low high-density lipoprotein cholesterol (HDL-C) levels are positively associated with cardiovascular events, it is still unclear whether familial hypercholesterolemia (FH) and Tangier’s disease (TD), caused by mutations in LDLR and ABCA1, respectively, influence ischemic stroke (IS) in humans. Objective: We sought to establish an easier, more effective, and time-saving method to induce IS, then studied the precise effects of different types of lipoproteins on IS. Methods and Results: A new technique termed contralateral middle cerebral artery occlusion (c-MCAO) was introduced to human-like hamster models to induce IS. Compared to traditional distal MCAO (d-MCAO) induced by electrocoagulation, c-MCAO resulted in a more severe IS with larger infarct sizes and more blood–brain barrier (BBB) disruption after 24 h. It was shown that c-MCAO markedly elicited an increase in brain infarct volume and BBB leakage in both homozygous LDLR (LDLR–/–) and ABCA1 knockout (ABCA1–/–) hamsters, but not in heterozygous LDLR knockout (LDLR+/–) hamsters when compared to wild-type (WT) controls. Conclusions: Using human-like genetically engineered hamsters, our findings demonstrated that both high LDL-C level caused by homozygous LDLR deficiency and severe low HDL-C level caused by deleting ABCA1 were risk factors of IS. As such, we believe the development of this novel IS hamster model is suitable for future ischemic/reperfusion studies.
Collapse
|
2102
|
Du R, Wu X, Peng K, Lin L, Li M, Xu Y, Xu M, Chen Y, Li D, Lu J, Bi Y, Wang W, Ning G. Serum apolipoprotein B is associated with increased risk of metabolic syndrome among middle-aged and elderly Chinese: A cross-sectional and prospective cohort study. J Diabetes 2019; 11:752-760. [PMID: 30652427 DOI: 10.1111/1753-0407.12904] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 01/02/2019] [Accepted: 01/12/2019] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Serum apolipoprotein (apo) B has been associated with an increased risk of atherosclerotic cardiovascular diseases. However, findings on the association between apoB and the risk of metabolic syndrome (MetS) are inconsistent. The purpose of this study was to investigate the association between serum apoB and MetS risk in Chinese population. METHODS A baseline survey was conducted in a population-based cohort of 10 340 adults aged ≥40 years in Shanghai, China, in 2010. A follow-up visit was conducted to assess incident diabetes in 2015. RESULTS At baseline, 2794 of 10 340 participants (27.02%) had MetS. Serum apoB was significantly associated with an increased risk of prevalent MetS. Multivariable-adjusted odds ratios and 95% confidence intervals (CIs) for quintiles 2-5 compared with quintile 1 (reference) were 1.29 (1.02-1.63), 1.47 (1.18-1.84), 1.32 (1.06-1.65), and 2.02 (1.61-2.51), respectively (Ptrend < 0.05). During an average of 5.1 years follow-up, 4627 individuals without MetS at baseline showed a significant association between apoB and the risk of incident MetS. Multivariable-adjusted risk ratios (95% CIs) for subjects in apoB quintiles 2-5 compared with the reference were 1.43 (1.13-1.82), 1.57 (1.25-1.98), 1.74 (1.38-2.18), and 2.07 (1.66-2.58), respectively (Ptrend < 0.05). Stratified analysis suggested that the above association was much stronger among normal weight individuals than in those who were overweight or obese. CONCLUSION These cross-sectional and prospective studies provide evidence that serum apoB is associated with existing MetS and is a possible predictor of the risk of MetS, especially among normal weight individuals.
Collapse
Affiliation(s)
- Rui Du
- National Clinical Research Center for Metabolic Diseases, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xueyan Wu
- National Clinical Research Center for Metabolic Diseases, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kui Peng
- National Clinical Research Center for Metabolic Diseases, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lin Lin
- National Clinical Research Center for Metabolic Diseases, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mian Li
- Shanghai Institute of Endocrine and Metabolic Diseases, Shanghai, China
| | - Yu Xu
- Shanghai Institute of Endocrine and Metabolic Diseases, Shanghai, China
| | - Min Xu
- Shanghai Institute of Endocrine and Metabolic Diseases, Shanghai, China
| | - Yuhong Chen
- National Clinical Research Center for Metabolic Diseases, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Donghui Li
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jieli Lu
- National Clinical Research Center for Metabolic Diseases, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Institute of Endocrine and Metabolic Diseases, Shanghai, China
| | - Yufang Bi
- National Clinical Research Center for Metabolic Diseases, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Institute of Endocrine and Metabolic Diseases, Shanghai, China
| | - Weiqing Wang
- National Clinical Research Center for Metabolic Diseases, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guang Ning
- National Clinical Research Center for Metabolic Diseases, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Institute of Endocrine and Metabolic Diseases, Shanghai, China
| |
Collapse
|
2103
|
Wierzbicki AS. Treating lipids in secondary prevention in a developing country: Lies, damn lies and the internet. Int J Clin Pract 2019; 73:1-3. [PMID: 31168908 DOI: 10.1111/ijcp.13381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Affiliation(s)
- Anthony S Wierzbicki
- Department of Metabolic Medicine/Chemical Pathology, Guy's & St Thomas' Hospitals, London, UK
| |
Collapse
|
2104
|
Annemans L, Stock JK, Chapman MJ. PCSK9 inhibition, atherosclerotic cardiovascular disease, and health economics: Challenges at the crossroads. J Clin Lipidol 2019; 13:714-720. [DOI: 10.1016/j.jacl.2019.07.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 07/14/2019] [Accepted: 07/16/2019] [Indexed: 12/22/2022]
|
2105
|
Emanuelsson F, Nordestgaard BG, Tybjærg-Hansen A, Benn M. Impact of LDL Cholesterol on Microvascular Versus Macrovascular Disease. J Am Coll Cardiol 2019; 74:1465-1476. [DOI: 10.1016/j.jacc.2019.07.037] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 06/24/2019] [Accepted: 07/02/2019] [Indexed: 01/22/2023]
|
2106
|
Kapellos TS, Bonaguro L, Gemünd I, Reusch N, Saglam A, Hinkley ER, Schultze JL. Human Monocyte Subsets and Phenotypes in Major Chronic Inflammatory Diseases. Front Immunol 2019; 10:2035. [PMID: 31543877 PMCID: PMC6728754 DOI: 10.3389/fimmu.2019.02035] [Citation(s) in RCA: 576] [Impact Index Per Article: 96.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 08/12/2019] [Indexed: 12/14/2022] Open
Abstract
Human monocytes are divided in three major populations; classical (CD14+CD16−), non-classical (CD14dimCD16+), and intermediate (CD14+CD16+). Each of these subsets is distinguished from each other by the expression of distinct surface markers and by their functions in homeostasis and disease. In this review, we discuss the most up-to-date phenotypic classification of human monocytes that has been greatly aided by the application of novel single-cell transcriptomic and mass cytometry technologies. Furthermore, we shed light on the role of these plastic immune cells in already recognized and emerging human chronic diseases, such as obesity, atherosclerosis, chronic obstructive pulmonary disease, lung fibrosis, lung cancer, and Alzheimer's disease. Our aim is to provide an insight into the contribution of human monocytes to the progression of these diseases and highlight their candidacy as potential therapeutic cell targets.
Collapse
Affiliation(s)
- Theodore S Kapellos
- Genomics and Immunoregulation, Life and Medical Sciences Institute (LIMES), Bonn, Germany
| | - Lorenzo Bonaguro
- Genomics and Immunoregulation, Life and Medical Sciences Institute (LIMES), Bonn, Germany
| | - Ioanna Gemünd
- Genomics and Immunoregulation, Life and Medical Sciences Institute (LIMES), Bonn, Germany
| | - Nico Reusch
- Genomics and Immunoregulation, Life and Medical Sciences Institute (LIMES), Bonn, Germany
| | - Adem Saglam
- Platform for Single Cell Genomics and Epigenomics, German Center for Neurodegenerative Diseases and University of Bonn, Bonn, Germany
| | - Emily R Hinkley
- Platform for Single Cell Genomics and Epigenomics, German Center for Neurodegenerative Diseases and University of Bonn, Bonn, Germany
| | - Joachim L Schultze
- Genomics and Immunoregulation, Life and Medical Sciences Institute (LIMES), Bonn, Germany.,Platform for Single Cell Genomics and Epigenomics, German Center for Neurodegenerative Diseases and University of Bonn, Bonn, Germany
| |
Collapse
|
2107
|
Drexel H, Rosano GMC, Lewis BS, Huber K, Vonbank A, Dopheide JF, Mader A, Niessner A, Savarese G, Wassmann S, Agewall S. The age of randomized clinical trials: three important aspects of randomized clinical trials in cardiovascular pharmacotherapy with examples from lipid and diabetes trials. EUROPEAN HEART JOURNAL. CARDIOVASCULAR PHARMACOTHERAPY 2019; 6:97-103. [DOI: 10.1093/ehjcvp/pvz029] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 05/24/2019] [Accepted: 07/10/2019] [Indexed: 01/17/2023]
Abstract
Abstract
Randomized clinical trials (RCTs) are important and the Gold Standard for drugs in modern cardiovascular (CV) therapy. The cornerstone of RCTs is the recording of hard clinical endpoints instead of surrogates. It is important to select an appropriate endpoint. Efficacy endpoints must be clinically relevant and can be hierarchically divided. A very interesting innovation in endpoint acquisition is the total event paradigm.
Collapse
Affiliation(s)
- Heinz Drexel
- Vorarlberg Institute for Vascular Investigation and Treatment (VIVIT), Academic Teaching Hospital Feldkirch, Carinagasse 47, 6800 Feldkirch, Austria
- Division of Angiology, Swiss Cardiovascular Center, Freiburgstrasse 18, University Hospital Bern, 3010 Bern, Switzerland
- Doctorate Studies Medical Science, Private University of the Principality of Liechtenstein, Dorfstrasse 24, 9495 Triesen, Principality of Liechtenstein
- Drexel University College of Medicine, 2900 W Queen Ln, Philadelphia, PA 19129, USA
| | - Giuseppe M C Rosano
- Department of Medical Sciences, IRCCS San Raffaele Hospital, Via della Pisana 235, 00163 Rome, Italy
| | - Basil S Lewis
- Technion-Israel Institute of Technology, Ruth and Bruce Rappaport School of Medicine, Efron St 1, Bat Galim, 3525433 Haifa, Israel
| | - Kurt Huber
- 3rd Medical Department, Cardiology and Intensive Care Medicine, Wilhelminen Hospital, Montleartstraße 37, 1160 Vienna, Austria
- Medical School, Cardiology, Sigmund Freud University, Campus Prater, Freudplatz 1, 1020 Vienna, Austria
| | - Alexander Vonbank
- Vorarlberg Institute for Vascular Investigation and Treatment (VIVIT), Academic Teaching Hospital Feldkirch, Carinagasse 47, 6800 Feldkirch, Austria
- Doctorate Studies Medical Science, Private University of the Principality of Liechtenstein, Dorfstrasse 24, 9495 Triesen, Principality of Liechtenstein
- Department of Medicine I, Academic Teaching Hospital Feldkirch, Carinagasse 47, 6800 Feldkirch, Austria
| | - Jörn F Dopheide
- Division of Angiology, Swiss Cardiovascular Center, Freiburgstrasse 18, University Hospital Bern, 3010 Bern, Switzerland
| | - Arthur Mader
- Vorarlberg Institute for Vascular Investigation and Treatment (VIVIT), Academic Teaching Hospital Feldkirch, Carinagasse 47, 6800 Feldkirch, Austria
- Doctorate Studies Medical Science, Private University of the Principality of Liechtenstein, Dorfstrasse 24, 9495 Triesen, Principality of Liechtenstein
- Department of Medicine I, Academic Teaching Hospital Feldkirch, Carinagasse 47, 6800 Feldkirch, Austria
| | - Alexander Niessner
- Department of Internal Medicine II, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria
| | - Gianluigi Savarese
- Cardiology Unit, Department of Medicine, Karolinska Institute, Solnavägen 1, 171 77 Solna, Sweden
| | - Sven Wassmann
- Cardiology Practice Pasing, Institutstraße 14, 81241 Munich, Germany
- Medical Faculty, Clinical Medicine, University of the Saarland, Kirrberger Straße, 66421 Homburg, Germany
| | - Stefan Agewall
- Department of Cardiology, Oslo University Hospital Ulleval, Kirkeveien 166, 0450 Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Søsterhjemmet, Kirkeveien 166, 0450 Oslo, Norway
| |
Collapse
|
2108
|
Postprandial changes in gene expression of cholesterol influx and efflux mediators after intake of SFA compared with n-6 PUFA in subjects with and without familial hypercholesterolaemia: secondary outcomes of a randomised controlled trial. J Nutr Sci 2019; 8:e27. [PMID: 31448116 PMCID: PMC6692810 DOI: 10.1017/jns.2019.25] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Revised: 07/04/2019] [Accepted: 07/09/2019] [Indexed: 12/24/2022] Open
Abstract
The long-term cholesterol-lowering effect of replacing intake of SFA with PUFA is well established, but has not been fully explained mechanistically. We examined the postprandial response of meals with different fat quality on expression of lipid genes in peripheral blood mononuclear cells (PBMC) in subjects with and without familial hypercholesterolaemia (FH). Thirteen subjects with FH (who had discontinued lipid-lowering treatment ≥4 weeks prior to both test days) and fourteen normolipidaemic controls were included in a randomised controlled double-blind crossover study with two meals, each with 60 g of fat either mainly SFA (about 40% energy) or n-6 PUFA (about 40% energy). PBMC were isolated in fasting, and 4 and 6 h postprandial blood samples. Expression of thirty-three lipid genes was analysed by reverse transcription quantitative PCR. A linear mixed model was used to assess postprandial effects between meals and groups. There was a significant interaction between meal and group for MSR1 (P = 0·03), where intake of SFA compared with n-6 PUFA induced a larger reduction in gene expression in controls only (P = 0·01). Intake of SFA compared with n-6 PUFA induced larger reductions in gene expression levels of LDLR and FADS1/2, smaller increases of INSIG1 and FASN, and larger increases of ABCA1 and ABCG1 (P = 0·01 for all, no group interaction). Intake of SFA compared with n-6 PUFA induced changes in gene expression of cholesterol influx and efflux mediators in PBMC including lower LDLR and higher ABCA1/G1, potentially explaining the long-term cholesterol-raising effect of a high SFA intake.
Collapse
|
2109
|
Gupta R, Wood DA. Primary prevention of ischaemic heart disease: populations, individuals, and health professionals. Lancet 2019; 394:685-696. [PMID: 31448740 DOI: 10.1016/s0140-6736(19)31893-8] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2019] [Revised: 07/03/2019] [Accepted: 07/11/2019] [Indexed: 12/24/2022]
Abstract
Ischaemic heart disease has a multifactorial aetiology and can be prevented from developing in populations primordially, and in individuals at high risk by primary prevention. The primordial approach focuses on social determinants of health in populations: political, economic, and social factors, principally unplanned urbanisation, illiteracy, poverty, and working and living conditions. Implementation of the UN Sustainable Development Goals can lead to major improvements in cardiovascular health, and adequate health-care financing and universal health care are important for achieving these goals. Population-level interventions should focus on tobacco control, promotion of healthy foods (fruits, vegetables, legumes, and nuts), curbing unhealthy foods (saturated fats, trans fats, refined carbohydrates, excessive salt, and alcohol), promotion of physical activity in everyday living, and control of ambient and indoor pollution. At the individual level, identification of people at high multifactorial risk and guideline-driven management of hypertension, LDL cholesterol, and diabetes is required. Strategies to improve adherence to healthy lifestyles and drug therapies are essential and can be implemented at health system, health care, and patient levels with use of education, technology, and personalised approaches. Improving quality of medical education with a focus on ischaemic heart disease prevention for physicians, nurses, allied health workers, and the public is required.
Collapse
Affiliation(s)
- Rajeev Gupta
- Academic Research Development Unit, Rajasthan University of Health Sciences, Jaipur, Rajasthan, India; Department of Preventive Cardiology and Internal Medicine, Eternal Heart Care Centre and Research Institute, Jaipur, Rajasthan, India.
| | - David A Wood
- National Institute for Prevention and Cardiovascular Health, National University of Ireland Galway, Galway, Ireland; National Heart and Lung Institute, Imperial College, London, UK
| |
Collapse
|
2110
|
Ray KK, Corral P, Morales E, Nicholls SJ. Pharmacological lipid-modification therapies for prevention of ischaemic heart disease: current and future options. Lancet 2019; 394:697-708. [PMID: 31448741 DOI: 10.1016/s0140-6736(19)31950-6] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 07/24/2019] [Accepted: 07/31/2019] [Indexed: 12/16/2022]
Abstract
Atherosclerosis and its clinical manifestation as ischaemic heart disease remains a considerable health burden. Given that many factors contribute to ischaemic heart disease, a multifactorial approach to prevention is recommended, starting with lifestyle advice, smoking cessation, and control of known cardiovascular risk factors, such as blood pressure and lipids. Within the lipid profile, the principal target is lowering LDL cholesterol, firstly with lifestyle interventions and subsequently with pharmacological therapy. Statins are the recommended first-line pharmacological treatment. Some individuals might require further lowering of LDL cholesterol or be unable to tolerate statins. Additional therapies targeting different pathways in cholesterol metabolism are now available, ranging from small molecules taken orally, to injectable therapies. Examples include ezetimibe, which targets Niemann-Pick C1-like protein, and monoclonal antibodies that target PCSK9. Phase 3 trials have also been completed for bempedoic acid (targeting ATP-citrate lyase) and inclisiran (an interference RNA-based therapeutic targeting hepatic PCSK9 synthesis). In addition to LDL cholesterol, mendelian randomisation studies support a causal role for lipoprotein(a) and triglycerides in ischaemic heart disease. In this Series paper, we appraise currently available and emerging therapies for lowering LDL cholesterol, lipoprotein(a), and triglycerides for prevention of ischaemic heart disease.
Collapse
Affiliation(s)
- Kausik K Ray
- Imperial Centre for Cardiovascular Disease Prevention, School of Public Health, Imperial College London, London, UK.
| | - Pablo Corral
- Pharmacology Department, School of Medicine, FASTA University, Mar del Plata, Argentina
| | - Enrique Morales
- Cardiometabolic Research Center, MAC Hospital, Aguascalientes, Mexico
| | - Stephen J Nicholls
- Monash Cardiovascular Research Centre, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
2111
|
Abstract
CVD remains the greatest cause of death globally, and with the escalating prevalence of metabolic diseases, including type-2 diabetes, CVD mortality is predicted to rise. While the replacement of SFA has been the cornerstone of effective dietary recommendations to decrease CVD risk since the 1980s, the validity of these recommendations have been recently challenged. A review of evidence for the impact of SFA reduction revealed no effect on CVD mortality, but a significant reduction in risk of CVD events (7-17%). The greatest effect was found when SFA were substituted with PUFA, resulting in 27% risk reduction in CVD events, with no effect of substitution with carbohydrate or protein. There was insufficient evidence from randomised controlled trials to conclude upon the impact of SFA replacement with MUFA on CVD and metabolic outcomes. However, there was high-quality evidence that reducing SFA lowered serum total, and specifically LDL-cholesterol, a key risk factor for CVD, with greatest benefits achieved by replacing SFA with unsaturated fats. The exchange of SFA with either PUFA or MUFA, also produced favourable effects on markers of glycaemia, reducing HbA1c, a long-term marker of glycaemic control. In conclusion, the totality of evidence supports lowering SFA intake and replacement with unsaturated fats to reduce the risk of CVD events, and to a lesser extent, cardiometabolic risk factors, which is consistent with current dietary guidelines.
Collapse
|
2112
|
Oh M, Kim H, Shin EW, Sung C, Kim DH, Moon DH, Lee CW. Effects of ezetimibe/simvastatin 10/10 mg versus Rosuvastatin 10 mg on carotid atherosclerotic plaque inflammation. BMC Cardiovasc Disord 2019; 19:201. [PMID: 31426749 PMCID: PMC6700958 DOI: 10.1186/s12872-019-1184-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Accepted: 08/12/2019] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND Using 18F-fluorodeoxyglucose (18FDG) positron emission tomography-computed tomography (PET/CT) imaging, we examined the effects of ezetimibe/simvastatin 10/10 mg versus rosuvastatin 10 mg on carotid atherosclerotic plaque inflammation. Whether the combination therapy of ezetimibe with low-dose statin is as effective as potent statin monotherapy in attenuating carotid atherosclerotic plaque inflammation remains unclear. METHODS In this 2-by-2 factorial trial, 50 patients with 18FDG uptake (target-to-background ratio [TBR] ≥1.6) in the carotid artery and acute coronary syndrome were randomized to receive either simvastatin/ezetimibe 10/10 mg or rosuvastatin 10 mg. 18FDG PET/CT examinations were performed at baseline and at 6 months. The percent change in the TBR of the index vessel at the most diseased segment (MDS) was the primary endpoint. RESULTS Baseline characteristics of the two groups were largely similar. At 6-month follow-up, the MDS TBR of the index vessel and aorta significantly decreased in ezetimibe/simvastatin group and tended to decrease in rosuvastatin group. However, the percent change in the MDS TBR of the index vessel was similar between the 2 groups (- 10.22 ± 17.49% vs. -5.84 ± 15.78%, respectively, p = 0.357), as was the percent change in the whole vessel TBR of the index vessel. Likewise, the changes in the MDS TBR or whole vessel TBR of the aorta were similar in both groups. Total cholesterol and low-density lipoprotein cholesterol levels improved to a similar degree in both groups. CONCLUSION Treatment with ezetimibe/simvastatin versus rosuvastatin resulted in a similar improvement of carotid atherosclerotic plaque inflammation, suggesting their equivalent anti-inflammatory effects. TRIAL REGISTRATION The trial is registered at ClinicalTrials.gov : NCT02378064, 3-4-2015. /IRB No. 2015-0194.
Collapse
Affiliation(s)
- Minyoung Oh
- Department of Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Hyunji Kim
- Department of Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Eon Woo Shin
- Department of Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Changhwan Sung
- Department of Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Do-Hoon Kim
- Department of Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Dae Hyuk Moon
- Department of Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Cheol Whan Lee
- Division of Cardiology, Heart Institute, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea.
| |
Collapse
|
2113
|
Calabrò P, Gragnano F. Event recurrence after myocardial infarction: Prediction is very difficult, especially about the future. Int J Cardiol 2019; 296:30-31. [PMID: 31447227 DOI: 10.1016/j.ijcard.2019.08.032] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 08/07/2019] [Accepted: 08/14/2019] [Indexed: 10/26/2022]
Affiliation(s)
- Paolo Calabrò
- Division of Cardiology, A.O.R.N. "Sant'Anna e San Sebastiano", Caserta, Italy; Department of Translational Medical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy.
| | - Felice Gragnano
- Division of Cardiology, A.O.R.N. "Sant'Anna e San Sebastiano", Caserta, Italy; Department of Translational Medical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| |
Collapse
|
2114
|
Libby P, Buring JE, Badimon L, Hansson GK, Deanfield J, Bittencourt MS, Tokgözoğlu L, Lewis EF. Atherosclerosis. Nat Rev Dis Primers 2019; 5:56. [PMID: 31420554 DOI: 10.1038/s41572-019-0106-z] [Citation(s) in RCA: 1858] [Impact Index Per Article: 309.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/04/2019] [Indexed: 12/12/2022]
Abstract
Atherosclerosis, the formation of fibrofatty lesions in the artery wall, causes much morbidity and mortality worldwide, including most myocardial infarctions and many strokes, as well as disabling peripheral artery disease. Development of atherosclerotic lesions probably requires low-density lipoprotein, a particle that carries cholesterol through the blood. Other risk factors for atherosclerosis and its thrombotic complications include hypertension, cigarette smoking and diabetes mellitus. Increasing evidence also points to a role of the immune system, as emerging risk factors include inflammation and clonal haematopoiesis. Studies of the cell and molecular biology of atherogenesis have provided considerable insight into the mechanisms that link all these risk factors to atheroma development and the clinical manifestations of this disease. An array of diagnostic techniques, both invasive (such as selective coronary arteriography) and noninvasive (such as blood biomarkers, stress testing, CT and nuclear scanning), permit assessment of cardiovascular disease risk and targeting of therapies. An expanding armamentarium of therapies that can modify risk factors and confer clinical benefit is available; however, we face considerable challenge in providing equitable access to these treatments and in maximizing adherence. Yet, the clinical application of the fruits of research has advanced preventive strategies, enhanced clinical outcomes in affected individuals, and improved their quality of life. Rapidly accelerating knowledge and continued research promise to provide further progress in combating this common chronic disease.
Collapse
Affiliation(s)
- Peter Libby
- Department of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Julie E Buring
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Lina Badimon
- Centre d'Investigació Cardiovascular CSIC-ICCC, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Göran K Hansson
- Center for Molecular Medicine, Karolinska Universitetssjukhuset, Stockholm, Sweden
| | - John Deanfield
- Institute of Cardiovascular Sciences, University College London, London, UK
| | - Márcio Sommer Bittencourt
- Center for Clinical and Epidemiological Research, University Hospital, University of São Paulo, São Paulo, Brazil.,Faculdade Israelita de Ciencias da Saude Albert Einstein, São Paulo, Brazil.,DASA, São Paulo, Brazil
| | | | - Eldrin F Lewis
- Department of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
2115
|
Abstract
Atherosclerotic cardiovascular disease is a leading cause of death and morbidity globally. Over the past several years, arterial inflammation has been implicated in the pathophysiology of athero-thrombosis, substantially confirming what pathologist Rudolf Virchow had observed in the 19th century. Lipid lowering, lifestyle changes, and modification of other risk factors have reduced cardiovascular complications of athero-thrombosis, but a substantial residual risk remains. In view of the pathogenic role of inflammation in athero-thrombosis, directly targeting inflammation has emerged as an additional potential therapeutic option; and some early promising results have been suggested by the Canakinumab Anti-inflammatory Thrombosis Outcome Study (CANTOS), in which canakinumab, a fully human monoclonal antibody targeting the pro-inflammatory and pro-atherogenic cytokine interleukin 1 beta, was shown to reduce cardiovascular events.
Collapse
Affiliation(s)
- Prediman K Shah
- Helga and Walter Oppenheimer Atherosclerosis Research Center, Smidt Heart Institute, Cedars Sinai Medical Center, Los Angeles, CA, USA
| | - Dalgisio Lecis
- Helga and Walter Oppenheimer Atherosclerosis Research Center, Smidt Heart Institute, Cedars Sinai Medical Center, Los Angeles, CA, USA.,Department of Cardiovascular Medicine, "Tor Vergata" University of Rome, Rome, Italy
| |
Collapse
|
2116
|
Abstract
PURPOSE OF REVIEW A number of novel trials have assessed the efficacy of new lipid-lowering therapies in cardiovascular disease (CVD). RECENT FINDINGS Proprotein convertase subtilisin kexin-9 inhibitors reduce low-density lipoprotein cholesterol (LDL-C) by 50-55%. A CVD outcome trial in patients with acute coronary syndromes with evolocumab achieved a LDL-C of 0.8 mmol/l (31 mg/dl) and a 20% relative risk reduction in CVD events in 2.2 years. Cholesterol ester transfer protein inhibitors raise high-density lipoprotein cholesterol and can lower LDL-C. Anacetrapib reduced coronary artery disease events by 7%, but not wider composite CVD outcomes, in a population with chronic CVD with pretreatment LDL-C of 1.6 mmol/l (62 mg/dl). The conflicting outcomes of cholesterol ester transfer protein inhibitor trials means these compounds are not being developed further. Trials using lipid drugs targeting inflammation have previously been generally unsuccessful, but recent data on the interleukin-1B receptor antagonist canakinumab has proven the concept of intervention on inflammation in atherosclerosis by showing a reduction in acute coronary interventions, but at the predictable cost of increased infections. SUMMARY Despite the success of proprotein convertase subtilisin kexin-9 inhibition, the ability to achieve low LDL-C with off-patent medications and the costs of novel therapies will limit their use even in high-risk patients and confine them to the highest-risk sub-groups of patients.
Collapse
|
2117
|
Banerjee S, Andrew RJ, Duff CJ, Fisher K, Jackson CD, Lawrence CB, Maeda N, Greenspan DS, Kellett KAB, Hooper NM. Proteolysis of the low density lipoprotein receptor by bone morphogenetic protein-1 regulates cellular cholesterol uptake. Sci Rep 2019; 9:11416. [PMID: 31388055 PMCID: PMC6684651 DOI: 10.1038/s41598-019-47814-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 07/22/2019] [Indexed: 12/14/2022] Open
Abstract
The development of cardiovascular disease is intimately linked to elevated levels of low-density lipoprotein (LDL) cholesterol in the blood. Hepatic LDL receptor (LDLR) levels regulate the amount of plasma LDL. We identified the secreted zinc metalloproteinase, bone morphogenetic protein 1 (BMP1), as responsible for the cleavage of human LDLR within its extracellular ligand-binding repeats at Gly171↓Asp172. The resulting 120 kDa membrane-bound C-terminal fragment (CTF) of LDLR had reduced capacity to bind LDL and when expressed in LDLR null cells had compromised LDL uptake as compared to the full length receptor. Pharmacological inhibition of BMP1 or siRNA-mediated knockdown prevented the generation of the 120 kDa CTF and resulted in an increase in LDL uptake into cells. The 120 kDa CTF was detected in the livers from humans and mice expressing human LDLR. Collectively, these results identify that BMP1 regulates cellular LDL uptake and may provide a target to modulate plasma LDL cholesterol.
Collapse
Affiliation(s)
- Sreemoti Banerjee
- School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Sciences, Manchester, M13 9PT, UK.,Jack Birch Unit for Molecular Carcinogenesis, Department of Biology, University of York, York, YO10 5DD, UK
| | - Robert J Andrew
- School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Sciences, Manchester, M13 9PT, UK.,Department of Neurobiology, The University of Chicago, Chicago, IL, 60637, USA
| | - Christopher J Duff
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, LS2 9JT, UK.,Department of Clinical Biochemistry, University Hospitals of North Midlands NHS Trust, Stoke-on-Trent, ST4 6QG, UK
| | - Kate Fisher
- School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Sciences, Manchester, M13 9PT, UK
| | - Carolyn D Jackson
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, LS2 9JT, UK
| | - Catherine B Lawrence
- School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Sciences, Manchester, M13 9PT, UK
| | - Nobuyo Maeda
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Daniel S Greenspan
- Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | - Katherine A B Kellett
- School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Sciences, Manchester, M13 9PT, UK.
| | - Nigel M Hooper
- School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Sciences, Manchester, M13 9PT, UK.
| |
Collapse
|
2118
|
Berman AN, Blankstein R. Optimizing Dyslipidemia Management for the Prevention of Cardiovascular Disease: a Focus on Risk Assessment and Therapeutic Options. Curr Cardiol Rep 2019; 21:110. [PMID: 31378838 DOI: 10.1007/s11886-019-1175-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Primary prevention of incident atherosclerotic cardiovascular disease (ASCVD) as well as decreasing the risk of future events in those with established atherosclerosis is critical from a public health perspective. Management of dyslipidemias constitutes a key target in decreasing the risk of developing ASCVD events. While there have been great strides in the treatment of dyslipidemia over the last three decades, there are important recent developments and ongoing research that will expand the available therapeutic options and enable further cardiovascular risk reduction. PURPOSE OF REVIEW: The purpose of this paper is to review new developments relating to the primary prevention and management of ASCVD with a specific focus on optimizing the treatment of dyslipidemias. RECENT FINDINGS: In the realm of ASCVD risk prediction, mounting evidence over the last decade has demonstrated that coronary artery calcium testing is superior to any serum biomarker in the prediction of future ASCVD events and in discriminating future cardiovascular risk. As such, it has been incorporated into the most recent ACC/AHA primary prevention guideline to help guide management decisions in select patients. In terms of the management of dyslipidemias, PCSK9 inhibitors lower LDL-C by 50-70% and provide an additional 15% reduction in key cardiovascular events in high-risk patients with known ASCVD, as demonstrated in the ODYSSEY and FOURIER trials. Cholesteryl ester transfer protein (CETP) inhibitors, which significantly increase HDL-C levels, demonstrated mixed results in large clinical trials and have helped reframe HDL-C as a risk marker rather than a modifiable risk factor. In regard to the management of triglycerides, the REDUCE-IT trial demonstrated a nearly 5% absolute reduction in key cardiovascular events with a highly purified fish-oil derivative named icosapent ethyl in high-risk patients already on statin therapy. Finally, in regard to lipoprotein(a)-which is a strong risk factor for ASCVD-there are exciting developments in the therapeutic pipeline which reduce circulating lipoprotein(a) levels by nearly 90%. The management of dyslipidemias continues to be an exciting field with several ongoing cardiovascular outcomes trials, improvement in risk prediction models, and new therapeutic agents in the pipeline that will further mitigate residual cardiovascular risk in both primary and secondary prevention patients.
Collapse
Affiliation(s)
- Adam N Berman
- Departments of Medicine (Cardiovascular Division) and Radiology, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA, 02115, USA
| | - Ron Blankstein
- Departments of Medicine (Cardiovascular Division) and Radiology, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA, 02115, USA.
| |
Collapse
|
2119
|
Paul A, Lydic TA, Hogan R, Goo YH. Cholesterol Acceptors Regulate the Lipidome of Macrophage Foam Cells. Int J Mol Sci 2019; 20:E3784. [PMID: 31382484 PMCID: PMC6695943 DOI: 10.3390/ijms20153784] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 07/29/2019] [Accepted: 07/30/2019] [Indexed: 12/17/2022] Open
Abstract
Arterial foam cells are central players of atherogenesis. Cholesterol acceptors, apolipoprotein A-I (apoA-I) and high-density lipoprotein (HDL), take up cholesterol and phospholipids effluxed from foam cells into the circulation. Due to the high abundance of cholesterol in foam cells, most previous studies focused on apoA-I/HDL-mediated free cholesterol (FC) transport. However, recent lipidomics of human atherosclerotic plaques also identified that oxidized sterols (oxysterols) and non-sterol lipid species accumulate as atherogenesis progresses. While it is known that these lipids regulate expression of pro-inflammatory genes linked to plaque instability, how cholesterol acceptors impact the foam cell lipidome, particularly oxysterols and non-sterol lipids, remains unexplored. Using lipidomics analyses, we found cholesterol acceptors remodel foam cell lipidomes. Lipid subclass analyses revealed various oxysterols, sphingomyelins, and ceramides, species uniquely enriched in human plaques were significantly reduced by cholesterol acceptors, especially by apoA-I. These results indicate that the function of lipid-poor apoA-I is not limited to the efflux of cholesterol and phospholipids but suggest that apoA-I serves as a major regulator of the foam cell lipidome and might play an important role in reducing multiple lipid species involved in the pathogenesis of atherosclerosis.
Collapse
Affiliation(s)
- Antoni Paul
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA
| | - Todd A Lydic
- Department of Physiology, Michigan State University, East Lansing, MI 48824, USA
| | - Ryan Hogan
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA
| | - Young-Hwa Goo
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA.
| |
Collapse
|
2120
|
da Silva PM, Aguiar C, Morais J. Suboptimal lipid levels in clinical practice among Portuguese adults with dyslipidemia under lipid-lowering therapy: Data from the DISGEN-LIPID study. Rev Port Cardiol 2019; 38:559-569. [DOI: 10.1016/j.repc.2019.02.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 01/24/2019] [Accepted: 02/03/2019] [Indexed: 12/28/2022] Open
|
2121
|
Wibaek R, Vistisen D, Girma T, Admassu B, Abera M, Abdissa A, Jørgensen ME, Kæstel P, Michaelsen KF, Friis H, Wells JCK, Andersen GS. Associations of fat mass and fat-free mass accretion in infancy with body composition and cardiometabolic risk markers at 5 years: The Ethiopian iABC birth cohort study. PLoS Med 2019; 16:e1002888. [PMID: 31430287 PMCID: PMC6701744 DOI: 10.1371/journal.pmed.1002888] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 07/16/2019] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Accelerated growth in early childhood is an established risk factor for later obesity and cardiometabolic disease, but the relative importance of fat mass (FM) and fat-free mass (FFM) accretion is not well understood. We aimed to study how FM and FFM at birth and their accretion during infancy were associated with body composition and cardiometabolic risk markers at 5 years. METHODS AND FINDINGS Healthy children born at term were enrolled in the Infant Anthropometry and Body Composition (iABC) birth cohort between December 2008 and October 2012 at Jimma University Specialized Hospital in the city of Jimma, Ethiopia. FM and FFM were assessed using air displacement plethysmography a median of 6 times between birth and 6 months of age. In 507 children, we estimated individual FM and FFM at birth and their accretion over 0-3 and 3-6 months of age using linear-spline mixed-effects modelling. We analysed associations of FM and FFM at birth and their accretion in infancy with height, waist circumference, FM, FFM, and cardiometabolic risk markers at 5 years using multiple linear regression analysis. A total of 340 children were studied at the 5-year follow-up (mean age: 60.0 months; girls: 50.3%; mean wealth index: 45.5 out of 100; breastfeeding status at 4.5 to 6 months post-partum: 12.5% exclusive, 21.4% almost exclusive, 60.6% predominant, 5.5% partial/none). Higher FM accretion in infancy was associated with higher FM and waist circumference at 5 years. For instance, 100-g/month higher FM accretion in the periods 0-3 and 3-6 months was associated with 339 g (95% CI: 243-435 g, p < 0.001) and 367 g (95% CI: 250-484 g, p < 0.001) greater FM at 5 years, respectively. Higher FM at birth and FM accretion from 0 to 3 months were associated with higher FFM and cholesterol concentrations at 5 years. Associations for cholesterol were strongest for low-density lipoprotein (LDL)-cholesterol, and remained significant after adjusting for current FM. A 100-g higher FM at birth and 100-g/month higher FM accretion from 0 to 3 months were associated with 0.16 mmol/l (95% CI: 0.05-0.26 mmol/l, p = 0.005) and 0.06 mmol/l (95% CI: 0.01-0.12 mmol/l, p = 0.016) higher LDL-cholesterol at 5 years, respectively. Higher FFM at birth and FFM accretion in infancy were associated with higher FM, FFM, waist circumference, and height at 5 years. For instance, 100-g/month higher FFM accretion in the periods 0-3 and 3-6 months was associated with 1,002 g (95% CI: 815-1,189 g, p < 0.001) and 624 g (95% CI: 419-829 g, p < 0.001) greater FFM at 5 years, respectively. We found no associations of FM and FFM growth with any of the other studied cardiometabolic markers including glucose, HbA1c, insulin, C-peptide, HOMA-IR, triglycerides, and blood pressure. Non-attendance at the 5-year follow-up visit was the main limitation of this study, which may have introduced selection bias and limited the power of the regression analyses. CONCLUSIONS FM accretion in early life was positively associated with markers of adiposity and lipid metabolism, but not with blood pressure and cardiometabolic markers related to glucose homeostasis. FFM accretion was primarily related to linear growth and FFM at 5 years.
Collapse
Affiliation(s)
- Rasmus Wibaek
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
- Clinical Epidemiology, Steno Diabetes Center Copenhagen, Gentofte, Denmark
- * E-mail:
| | - Dorte Vistisen
- Clinical Epidemiology, Steno Diabetes Center Copenhagen, Gentofte, Denmark
| | - Tsinuel Girma
- Department of Paediatrics and Child Health, Jimma University, Jimma, Ethiopia
- Jimma University Clinical and Nutrition Research Partnership (JUCAN), Jimma University, Jimma, Ethiopia
| | - Bitiya Admassu
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
- Jimma University Clinical and Nutrition Research Partnership (JUCAN), Jimma University, Jimma, Ethiopia
- Department of Population and Family Health, Jimma University, Jimma, Ethiopia
| | - Mubarek Abera
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
- Jimma University Clinical and Nutrition Research Partnership (JUCAN), Jimma University, Jimma, Ethiopia
- Department of Psychiatry, Jimma University, Jimma, Ethiopia
| | - Alemseged Abdissa
- Jimma University Clinical and Nutrition Research Partnership (JUCAN), Jimma University, Jimma, Ethiopia
- Department of Laboratory Sciences and Pathology, Jimma University, Jimma, Ethiopia
| | - Marit E. Jørgensen
- Clinical Epidemiology, Steno Diabetes Center Copenhagen, Gentofte, Denmark
- National Institute of Public Health, Southern Denmark University, Copenhagen, Denmark
| | - Pernille Kæstel
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Kim F. Michaelsen
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Henrik Friis
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Jonathan C. K. Wells
- Childhood Nutrition Research Centre, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| | | |
Collapse
|
2122
|
Raal FJ, Mohamed F. More aggressive lipid lowering in people with diabetes? Lancet Diabetes Endocrinol 2019; 7:587-589. [PMID: 31272929 DOI: 10.1016/s2213-8587(19)30190-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 05/02/2019] [Indexed: 11/30/2022]
Affiliation(s)
- Frederick J Raal
- Division of Endocrinology and Metabolism, Department of Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2193, South Africa.
| | - Farzahna Mohamed
- Division of Endocrinology and Metabolism, Department of Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2193, South Africa
| |
Collapse
|
2123
|
da Silva PM, Aguiar C, Morais J. Suboptimal lipid levels in clinical practice among Portuguese adults with dyslipidemia under lipid-lowering therapy: Data from the DISGEN-LIPID study. REVISTA PORTUGUESA DE CARDIOLOGIA (ENGLISH EDITION) 2019. [DOI: 10.1016/j.repce.2019.11.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
|
2124
|
Clinical significance of non-culprit plaque regression following acute coronary syndrome: A serial intravascular ultrasound study. J Cardiol 2019; 74:102-108. [DOI: 10.1016/j.jjcc.2018.12.023] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 12/15/2018] [Accepted: 12/30/2018] [Indexed: 11/19/2022]
|
2125
|
Zwol WV, Rimbert A, Kuivenhoven JA. The Future of Lipid-lowering Therapy. J Clin Med 2019; 8:E1085. [PMID: 31340607 PMCID: PMC6678580 DOI: 10.3390/jcm8071085] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 07/19/2019] [Accepted: 07/22/2019] [Indexed: 12/13/2022] Open
Abstract
The recent introduction of inhibitors of proprotein convertase subtilisin/kexin 9 to lower low-density lipoprotein (LDL) cholesterol on top of statins or as monotherapy is rapidly changing the landscape of treatment of atherosclerotic cardiovascular disease (ASCVD). However, existing lipid-lowering drugs have little impact on lipoprotein(a) (Lp(a)) or plasma triglycerides, two other risk factors for ASCVD. This review summarizes the evidence and the rationale to target Lp(a) and triglycerides and provides an overview of currently tested strategies to lower Lp(a), apolipoprotein C-III and angiopoietin-like protein 3. In addition, it summarizes new findings on the use of omega-3 fatty acids (OM3FA) to fight ASCVD. With the exception of OM3FA supplementation, the promise of the experimental drugs discussed here depends on the long-term safety and efficacy of monoclonal antibodies and/or antisense oligonucleotides Clinical outcome trials will ultimately prove whether these new therapeutic modalities will reduce ASCVD risk.
Collapse
Affiliation(s)
- Willemien van Zwol
- Department of Pediatrics, Section Molecular Genetics, University of Groningen, University Medical Centre Groningen, 9713 Groningen, The Netherlands
| | - Antoine Rimbert
- Department of Pediatrics, Section Molecular Genetics, University of Groningen, University Medical Centre Groningen, 9713 Groningen, The Netherlands
| | - Jan Albert Kuivenhoven
- Department of Pediatrics, Section Molecular Genetics, University of Groningen, University Medical Centre Groningen, 9713 Groningen, The Netherlands.
| |
Collapse
|
2126
|
Lu R, Zheng Z, Yin Y, Jiang Z. Effect of Genistein on Cholesterol Metabolism-Related Genes in HepG2 Cell. J Food Sci 2019; 84:2330-2336. [PMID: 31313321 DOI: 10.1111/1750-3841.14725] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2018] [Revised: 05/27/2019] [Accepted: 06/09/2019] [Indexed: 11/30/2022]
Abstract
It has been reported that genistein could improve metabolic syndromes. Our study aimed to investigate the effects and potential mechanisms of genistein on improving cholesterol metabolism in HepG2 cell. HepG2 cells were cultured with 0, 0.01, 1.00, 10.00, and 50.00 µM genistein for 24 hr. The current results showed a dose-dependent manner between genistein and intracellular contents of total cholesterol (TC), high-density lipoprotein-cholesterol (HDL-C), and cellular apolipoprotein A1 (Apo-A1) secretion. TC was increased by 25.69%, meanwhile HDL-C and Apo-A1 were decreased by 56.00% and 25.93%, respectively, when the dosage of genistein was 1.00 µM. Genistein dose-dependently upregulated the protein and mRNA levels of sterol regulatory element binding proteins-2 (SREBP-2), as well as the mRNA levels of low-density lipoprotein receptor (LDLR) and 3-hydroxy-3-methyl glutaryl coenzyme A reductase (HMGCR), by 145.91%, 72.29%, 310.23%, and 123.08%, respectively, when we gave 1.00 µM genistein, indicating that intracellular cholesterol synthesis and absorption of exogenous cholesterol were increased. In addition, the mRNA levels of peroxisome proliferator-activated receptor-γ (PPARγ) and liver X receptor (LXRα), lowered by 58.23% and 34.86% at 0.01 µM genistein, were reduced in a dose-dependent manner. LXRα and ATP-binding cassette transporter A1 (ABCA1) protein levels were significantly (P < 0.05) decreased by 50.35% and 11.60% at 1.00 µM genistein, which indicated that cellular cholesterol efflux was inhibited. Taken together, our results suggested that genistein at dosage of more than 1.00 µM was able to increase the intracellular cholesterol levels by up regulating SREBP-2/LDLR/HMGCR pathway and suppressing PPARγ/LXRα/ABCA1 pathway. PRACTICAL APPLICATION: In this study, genistein appeared to be effective in reducing plasma cholesterol levels due to increase the intracellular cholesterol levels by upregulating cholesterol absorption through SREBP-2/LDLR/HMGCR pathway, and also downregulating cholesterol efflux via PPARγ/LXRα/ABCA1 pathway in vitro. In addition, plasma cholesterol is regarded as the key indicator of atherosclerosis; therefore, we believe that our findings could be used for further exploration on a possible therapeutic application of genistein for atherosclerosis.
Collapse
Affiliation(s)
- Rongrong Lu
- Nutrition, School of Public Health, Sun Yat-Sen Univ., Guangzhou, Guangdong, 510605, China
| | - Zicong Zheng
- Nutrition, School of Public Health, Sun Yat-Sen Univ., Guangzhou, Guangdong, 510605, China
| | - Yimin Yin
- Nutrition, School of Public Health, Sun Yat-Sen Univ., Guangzhou, Guangdong, 510605, China
| | - Zhuoqin Jiang
- Nutrition, School of Public Health, Sun Yat-Sen Univ., Guangzhou, Guangdong, 510605, China
| |
Collapse
|
2127
|
Affiliation(s)
- Safi U Khan
- Department of Medicine, West Virginia University, Morgantown, USA
| | - Erin D Michos
- Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins School of Medicine, Baltimore, USA
| |
Collapse
|
2128
|
Fontes-Carvalho R, Marques Silva P, Rodrigues E, Araújo F, Gavina C, Ferreira J, Morais J. Practical guide for the use of PCSK9 inhibitors in Portugal. Rev Port Cardiol 2019; 38:391-405. [PMID: 31324407 DOI: 10.1016/j.repc.2019.05.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Abstract
Reducing low-density lipoprotein cholesterol (LDL-C) levels is one of the most important strategies for reducing the risk of cardiovascular events. However, in clinical practice, a high proportion of patients do not achieve recommended LDL-C levels through lifestyle and lipid-lowering therapy with statins and ezetimibe. PCSK9 inhibitors (PCSK9i) are a new therapeutic option that significantly (50-60%) reduces LDL-C levels, which in clinical trials translates into an additional reduction in risk for cardiovascular events, and has a good safety profile. However, it is a high-cost therapy, and therefore its use in clinical practice should take its cost-effectiveness into account. Priority should be given to use in patients at higher cardiovascular risk and those in whom high LDL-C levels persist despite optimal lipid-lowering therapy. This consensus document aims to summarize the main data on the clinical use of PCSK9i and to make recommendations for Portugal on the profile of patients who may benefit most from this therapy.
Collapse
Affiliation(s)
- Ricardo Fontes-Carvalho
- Departamento de Cardiologia, Centro Hospitalar de Vila Nova de Gaia/Espinho, Portugal; Departamento de Cirurgia e Fisiologia, Faculdade de Medicina da Universidade do Porto, Porto, Portugal.
| | - Pedro Marques Silva
- Núcleo de Investigação Arterial, Hospital de Santa Marta, Centro Hospitalar de Lisboa Central, Lisboa, Portugal
| | - Elisabete Rodrigues
- Departamento de Endocrinologia, Diabetes e Metabolismo, Centro Hospitalar S. João, Porto, Portugal; Departamento de Medicina, Faculdade de Medicina da Universidade do Porto, Porto, Portugal
| | - Francisco Araújo
- Serviço de Medicina Interna, Hospital Beatriz Ângelo, Loures, Portugal
| | - Cristina Gavina
- Departamento de Medicina, Faculdade de Medicina da Universidade do Porto, Porto, Portugal; Serviço de Cardiologia, Hospital Pedro Hispano - Unidade Local de Saúde de Matosinhos, Senhora da Hora, Portugal
| | - Jorge Ferreira
- Serviço de Cardiologia, Hospital de Santa Cruz, Centro Hospitalar de Lisboa Ocidental, Lisboa, Portugal
| | - João Morais
- Serviço de Cardiologia, Centro Hospitalar de Leiria, Leiria, Portugal
| |
Collapse
|
2129
|
Gut Microbiome: Profound Implications for Diet and Disease. Nutrients 2019; 11:nu11071613. [PMID: 31315227 PMCID: PMC6682904 DOI: 10.3390/nu11071613] [Citation(s) in RCA: 660] [Impact Index Per Article: 110.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 07/05/2019] [Accepted: 07/11/2019] [Indexed: 02/07/2023] Open
Abstract
The gut microbiome plays an important role in human health and influences the development of chronic diseases ranging from metabolic disease to gastrointestinal disorders and colorectal cancer. Of increasing prevalence in Western societies, these conditions carry a high burden of care. Dietary patterns and environmental factors have a profound effect on shaping gut microbiota in real time. Diverse populations of intestinal bacteria mediate their beneficial effects through the fermentation of dietary fiber to produce short-chain fatty acids, endogenous signals with important roles in lipid homeostasis and reducing inflammation. Recent progress shows that an individual’s starting microbial profile is a key determinant in predicting their response to intervention with live probiotics. The gut microbiota is complex and challenging to characterize. Enterotypes have been proposed using metrics such as alpha species diversity, the ratio of Firmicutes to Bacteroidetes phyla, and the relative abundance of beneficial genera (e.g., Bifidobacterium, Akkermansia) versus facultative anaerobes (E. coli), pro-inflammatory Ruminococcus, or nonbacterial microbes. Microbiota composition and relative populations of bacterial species are linked to physiologic health along different axes. We review the role of diet quality, carbohydrate intake, fermentable FODMAPs, and prebiotic fiber in maintaining healthy gut flora. The implications are discussed for various conditions including obesity, diabetes, irritable bowel syndrome, inflammatory bowel disease, depression, and cardiovascular disease.
Collapse
|
2130
|
Blauw LL, Li-Gao R, Noordam R, de Mutsert R, Trompet S, Berbée JFP, Wang Y, van Klinken JB, Christen T, van Heemst D, Mook-Kanamori DO, Rosendaal FR, Jukema JW, Rensen PCN, Willems van Dijk K. CETP (Cholesteryl Ester Transfer Protein) Concentration: A Genome-Wide Association Study Followed by Mendelian Randomization on Coronary Artery Disease. CIRCULATION-GENOMIC AND PRECISION MEDICINE 2019; 11:e002034. [PMID: 29728394 DOI: 10.1161/circgen.117.002034] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Accepted: 02/26/2018] [Indexed: 01/22/2023]
Abstract
BACKGROUND We aimed to identify independent genetic determinants of circulating CETP (cholesteryl ester transfer protein) to assess causal effects of variation in CETP concentration on circulating lipid concentrations and cardiovascular disease risk. METHODS A genome-wide association discovery and replication study on serum CETP concentration were embedded in the NEO study (Netherlands Epidemiology of Obesity). Based on the independent identified variants, Mendelian randomization was conducted on serum lipids (NEO study) and coronary artery disease (CAD; CARDIoGRAMplusC4D consortium). RESULTS In the discovery analysis (n=4248), we identified 3 independent variants (P<5×10-8) that determine CETP concentration. These single-nucleotide polymorphisms were mapped to CETP and replicated in a separate subpopulation (n=1458). Per-allele increase (SE) in serum CETP was 0.32 (0.02) µg/mL for rs247616-C, 0.35 (0.02) µg/mL for rs12720922-A, and 0.12 (0.02) µg/mL for rs1968905-G. Combined, these 3 variants explained 16.4% of the total variation in CETP concentration. One microgram per milliliter increase in genetically determined CETP concentration strongly decreased high-density lipoprotein cholesterol (-0.23 mmol/L; 95% confidence interval, -0.26 to -0.20), moderately increased low-density lipoprotein cholesterol (0.08 mmol/L; 95% confidence interval, 0.00-0.16), and was associated with an odds ratio of 1.08 (95% confidence interval, 0.94-1.23) for CAD risk. CONCLUSIONS This is the first genome-wide association study identifying independent variants that largely determine CETP concentration. Although high-density lipoprotein cholesterol is not a causal risk factor for CAD, it has been unequivocally demonstrated that low-density lipoprotein cholesterol lowering is proportionally associated with a lower CAD risk. Therefore, the results of our study are fully consistent with the notion that CETP concentration is causally associated with CAD through low-density lipoprotein cholesterol.
Collapse
Affiliation(s)
- Lisanne L Blauw
- Department of Internal Medicine, Division of Endocrinology (L.L.B., J.F.P.B., Y.W., P.C.N.R., K.W.v.D.) .,Department of Clinical Epidemiology (L.L.B., R.L.-G., R.d.M., T.C., D.O.M.-K., F.R.R.).,Einthoven Laboratory for Experimental Vascular Medicine (L.L.B., J.F.P.B., Y.W., J.B.v.K., P.C.N.R., K.W.v.D.)
| | - Ruifang Li-Gao
- Department of Clinical Epidemiology (L.L.B., R.L.-G., R.d.M., T.C., D.O.M.-K., F.R.R.)
| | - Raymond Noordam
- Department of Internal Medicine, Division of Gerontology and Geriatrics (R.N., S.T., D.v.H.)
| | - Renée de Mutsert
- Department of Clinical Epidemiology (L.L.B., R.L.-G., R.d.M., T.C., D.O.M.-K., F.R.R.)
| | - Stella Trompet
- Department of Internal Medicine, Division of Gerontology and Geriatrics (R.N., S.T., D.v.H.).,Department of Cardiology (S.T., J.W.J.)
| | - Jimmy F P Berbée
- Department of Internal Medicine, Division of Endocrinology (L.L.B., J.F.P.B., Y.W., P.C.N.R., K.W.v.D.).,Einthoven Laboratory for Experimental Vascular Medicine (L.L.B., J.F.P.B., Y.W., J.B.v.K., P.C.N.R., K.W.v.D.)
| | - Yanan Wang
- Department of Internal Medicine, Division of Endocrinology (L.L.B., J.F.P.B., Y.W., P.C.N.R., K.W.v.D.).,Einthoven Laboratory for Experimental Vascular Medicine (L.L.B., J.F.P.B., Y.W., J.B.v.K., P.C.N.R., K.W.v.D.)
| | - Jan B van Klinken
- Einthoven Laboratory for Experimental Vascular Medicine (L.L.B., J.F.P.B., Y.W., J.B.v.K., P.C.N.R., K.W.v.D.).,Department of Human Genetics (J.B.v.K., K.W.v.D.)
| | - Tim Christen
- Department of Clinical Epidemiology (L.L.B., R.L.-G., R.d.M., T.C., D.O.M.-K., F.R.R.)
| | - Diana van Heemst
- Department of Internal Medicine, Division of Gerontology and Geriatrics (R.N., S.T., D.v.H.)
| | - Dennis O Mook-Kanamori
- Department of Clinical Epidemiology (L.L.B., R.L.-G., R.d.M., T.C., D.O.M.-K., F.R.R.).,and Department of Public Health and Primary Care (D.O.M.-K.) Leiden University Medical Center, The Netherlands
| | - Frits R Rosendaal
- Department of Clinical Epidemiology (L.L.B., R.L.-G., R.d.M., T.C., D.O.M.-K., F.R.R.)
| | | | - Patrick C N Rensen
- Department of Internal Medicine, Division of Endocrinology (L.L.B., J.F.P.B., Y.W., P.C.N.R., K.W.v.D.).,Einthoven Laboratory for Experimental Vascular Medicine (L.L.B., J.F.P.B., Y.W., J.B.v.K., P.C.N.R., K.W.v.D.)
| | - Ko Willems van Dijk
- Department of Internal Medicine, Division of Endocrinology (L.L.B., J.F.P.B., Y.W., P.C.N.R., K.W.v.D.).,Einthoven Laboratory for Experimental Vascular Medicine (L.L.B., J.F.P.B., Y.W., J.B.v.K., P.C.N.R., K.W.v.D.).,Department of Human Genetics (J.B.v.K., K.W.v.D.)
| |
Collapse
|
2131
|
Wisløff T, Mundal LJ, Retterstøl K, Igland J, Kristiansen IS. Economic evaluation of lipid lowering with PCSK9 inhibitors in patients with familial hypercholesterolemia: Methodological aspects. Atherosclerosis 2019; 287:140-146. [PMID: 31280039 DOI: 10.1016/j.atherosclerosis.2019.06.900] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 06/10/2019] [Accepted: 06/12/2019] [Indexed: 12/11/2022]
Abstract
BACKGROUND AND AIMS Proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors have proved to reduce low density lipoprotein cholesterol levels in numerous clinical trials. In two large clinical trials, PCSK9 inhibitor treatment reduced the risk of cardiovascular disease. Our aim was to explore the impact of varying assumptions about clinical effectiveness on health and economic outcomes for patients with familial hypercholesterolemia. METHODS We used a previously published and validated Norwegian model for cardiovascular disease. The model was updated with recent data from the world's second largest registry of patients with genetically confirmed familial hypercholesterolemia. We performed analyses for 24 different subgroups of patients based on age, gender, statin tolerance and previous history of cardiovascular disease. RESULTS In 1 out of 24 subgroups, PCSK9 inhibitors were cost-effective when effectiveness was modelled using direct relative efficacy as reported in the FOURIER trial. When using assumptions, as suggested in a recent consensus statement from the European Atherosclerosis Society, 14 subgroups were cost-effective. CONCLUSIONS Cost-effectiveness of PCSK9 inhibitors depends highly on assumptions regarding effectiveness. Basing assumptions only on randomised controlled trials, and not taking into account varying effects based on baseline cholesterol level, results in much fewer groups being cost-effective.
Collapse
Affiliation(s)
- Torbjørn Wisløff
- Department of Health Management and Health Economics, University of Oslo, Oslo, Norway; Department of Infectious Disease Epidemiology and Modelling, Norwegian Institute of Public Health, Oslo, Norway.
| | - Liv J Mundal
- The Lipid Clinic, Department of Endocrinology, Morbid Obesity and Preventive Medicine, Oslo University Hospital, Oslo, Norway; Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Kjetil Retterstøl
- The Lipid Clinic, Department of Endocrinology, Morbid Obesity and Preventive Medicine, Oslo University Hospital, Oslo, Norway; Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Jannicke Igland
- Department of Global Public Health and Primary Care, University of Bergen, Bergen, Norway
| | | |
Collapse
|
2132
|
Baseline LDL-C levels and risk of cardiovascular events: is there any room for questions? Int J Cardiol 2019; 286:166-167. [DOI: 10.1016/j.ijcard.2019.03.058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 03/12/2019] [Accepted: 03/27/2019] [Indexed: 11/19/2022]
|
2133
|
Fiuza M. Lipoprotein(a) as a novel therapeutic target for preventing cardiovascular disease: A whiter shade of pale? Rev Port Cardiol 2019; 38:495-496. [DOI: 10.1016/j.repc.2019.08.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
|
2134
|
Fiuza M. Lipoprotein(a) as a novel therapeutic target for preventing cardiovascular disease: A whiter shade of pale? REVISTA PORTUGUESA DE CARDIOLOGIA (ENGLISH EDITION) 2019. [DOI: 10.1016/j.repce.2019.09.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
2135
|
Pérez de Isla L, Ray KK, Watts GF, Santos RD, Alonso R, Muñiz-Grijalvo O, Diaz-Diaz JL, Badimon L, Catapano AL, Mata P. Potential utility of the SAFEHEART risk equation for rationalising the use of PCSK9 monoclonal antibodies in adults with heterozygous familial hypercholesterolemia. Atherosclerosis 2019; 286:40-45. [DOI: 10.1016/j.atherosclerosis.2019.05.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Revised: 04/25/2019] [Accepted: 05/03/2019] [Indexed: 01/06/2023]
|
2136
|
Lindh M, Banefelt J, Fox KM, Hallberg S, Tai MH, Eriksson M, Villa G, Svensson MK, Qian Y. Cardiovascular event rates in a high atherosclerotic cardiovascular disease risk population: estimates from Swedish population-based register data. EUROPEAN HEART JOURNAL. QUALITY OF CARE & CLINICAL OUTCOMES 2019; 5:225-232. [PMID: 30649251 PMCID: PMC6613595 DOI: 10.1093/ehjqcco/qcy058] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 12/13/2018] [Accepted: 01/07/2019] [Indexed: 12/27/2022]
Abstract
AIMS This study aimed to estimate the rate of cardiovascular (CV) events in the real world in patients at high risk of recurrent CV events similar to the FOURIER trial population. METHODS AND RESULTS A retrospective population-based cohort study was conducted using Swedish national registers from 1 July 2001 to 31 December 2015. Patients in the atherosclerotic cardiovascular disease (ASCVD) prevalent cohort met the FOURIER-like inclusion criteria, including treatment with high/moderate-intensity statins, on 1 July 2006. Additionally, two cohorts defined by diagnosis of incident ischaemic stroke (IS) and incident myocardial infarction (MI), meeting the FOURIER-like inclusion criteria were followed from date of diagnosis. Event rates were calculated for the hard major adverse cardiovascular events (MACE) composite: MI, IS, and CV death; and the ASCVD composite: MI, IS, unstable angina, coronary revascularization, and CV death. Approximately half of patients experienced a CV event (ASCVD composite) during follow-up. The MACE composite rates/100 person-years were 6.3, 11.9, and 12.3 in the ASCVD prevalent (n = 54 992), MI incident (n = 45 895), and IS incident (n = 36 134) cohorts, respectively. The ASCVD composite rates/100 person-years were 7.0, 21.7, and 12.9 in the ASCVD prevalent, MI incident, and IS incident cohorts, respectively. The multiple-event MACE composite rates/100 person-years were 8.5 (ASCVD prevalent cohort), 15.4 (MI incident cohort), and 14.4 (IS incident cohort). CONCLUSION In this real-world setting, CV event rates were high in all studied cohorts. In particular, the MACE composite rates were two to three times higher than in the FOURIER clinical trial, indicating a substantial disease burden despite treatment with moderate or high-intensity statins.
Collapse
Affiliation(s)
- Maria Lindh
- Quantify Research, Hantverkargatan 8, Stockholm, Sweden
| | | | - Kathleen M Fox
- Strategic Healthcare Solutions LLC, 133 Cottonwood Creek Lane, Aiken, SC, USA
| | - Sara Hallberg
- Quantify Research, Hantverkargatan 8, Stockholm, Sweden
| | - Ming-Hui Tai
- Department of Global Health Economics, One Amgen center dr. Thousand Oaks, CA, USA
| | - Mats Eriksson
- Department of Medicine, Karolinska Institutet, Karolinska Universitetssjukhuset Huddinge, Stockholm, Sweden
| | - Guillermo Villa
- Department of Global Health Economics, AMGEN Europe GmbH, Suurstoffi 22, Rotkreuz, Switzerland
| | - Maria K Svensson
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Yi Qian
- Department of Global Health Economics, One Amgen center dr. Thousand Oaks, CA, USA
| |
Collapse
|
2137
|
Rämö JT, Ripatti P, Tabassum R, Söderlund S, Matikainen N, Gerl MJ, Klose C, Surma MA, Stitziel NO, Havulinna AS, Pirinen M, Salomaa V, Freimer NB, Jauhiainen M, Palotie A, Taskinen MR, Simons K, Ripatti S. Coronary Artery Disease Risk and Lipidomic Profiles Are Similar in Hyperlipidemias With Family History and Population-Ascertained Hyperlipidemias. J Am Heart Assoc 2019; 8:e012415. [PMID: 31256696 PMCID: PMC6662358 DOI: 10.1161/jaha.119.012415] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Background We asked whether, after excluding familial hypercholesterolemia, individuals with high low‐density lipoprotein cholesterol (LDL‐C) or triacylglyceride levels and a family history of the same hyperlipidemia have greater coronary artery disease risk or different lipidomic profiles compared with population‐based hyperlipidemias. Methods and Results We determined incident coronary artery disease risk for 755 members of 66 hyperlipidemic families (≥2 first‐degree relatives with similar hyperlipidemia) and 19 644 Finnish FINRISK population study participants. We quantified 151 circulating lipid species from 550 members of 73 hyperlipidemic families and 897 FINRISK participants using mass spectrometric shotgun lipidomics. Familial hypercholesterolemia was excluded using functional LDL receptor testing and genotyping. Hyperlipidemias (LDL‐C or triacylglycerides >90th population percentile) associated with increased coronary artery disease risk in meta‐analysis of the hyperlipidemic families and the population cohort (high LDL‐C: hazard ratio, 1.74 [95% CI, 1.48–2.04]; high triacylglycerides: hazard ratio, 1.38 [95% CI, 1.09–1.74]). Risk estimates were similar in the family and population cohorts also after adjusting for lipid‐lowering medication. In lipidomic profiling, high LDL‐C associated with 108 lipid species, and high triacylglycerides associated with 131 lipid species in either cohort (at 5% false discovery rate; P‐value range 0.038–2.3×10−56). Lipidomic profiles were highly similar for hyperlipidemic individuals in the families and the population (LDL‐C: r=0.80; triacylglycerides: r=0.96; no lipid species deviated between the cohorts). Conclusions Hyperlipidemias with family history conferred similar coronary artery disease risk as population‐based hyperlipidemias. We identified distinct lipidomic profiles associated with high LDL‐C and triacylglycerides. Lipidomic profiles were similar between hyperlipidemias with family history and population‐ascertained hyperlipidemias, providing evidence of similar and overlapping underlying mechanisms.
Collapse
Affiliation(s)
- Joel T Rämö
- 1 Institute for Molecular Medicine Finland HiLIFE University of Helsinki Finland
| | - Pietari Ripatti
- 1 Institute for Molecular Medicine Finland HiLIFE University of Helsinki Finland
| | - Rubina Tabassum
- 1 Institute for Molecular Medicine Finland HiLIFE University of Helsinki Finland
| | - Sanni Söderlund
- 2 Research Programs Unit Clinical and Molecular Metabolism University of Helsinki Finland.,3 Endocrinology Abdominal Center Helsinki University Hospital Helsinki Finland
| | - Niina Matikainen
- 2 Research Programs Unit Clinical and Molecular Metabolism University of Helsinki Finland.,3 Endocrinology Abdominal Center Helsinki University Hospital Helsinki Finland
| | | | | | - Michal A Surma
- 4 Lipotype GmbH Dresden Germany.,5 Łukasiewicz Research Network-PORT Polish Center for Technology Development Wroclaw Poland
| | - Nathan O Stitziel
- 6 Cardiovascular Division Department of Medicine Washington University School of Medicine St. Louis MO.,7 Department of Genetics Washington University School of Medicine St. Louis MO.,8 McDonnell Genome Institute Washington University School of Medicine St. Louis MO
| | - Aki S Havulinna
- 1 Institute for Molecular Medicine Finland HiLIFE University of Helsinki Finland.,9 National Institute for Health and Welfare Helsinki Finland
| | - Matti Pirinen
- 1 Institute for Molecular Medicine Finland HiLIFE University of Helsinki Finland.,10 Department of Mathematics and Statistics Faculty of Science University of Helsinki Finland.,16 Department of Public Health Clinicum Faculty of Medicine University of Helsinki Finland
| | - Veikko Salomaa
- 9 National Institute for Health and Welfare Helsinki Finland
| | - Nelson B Freimer
- 11 Center for Neurobehavioral Genetics Semel Institute for Neuroscience and Human Behavior University of California Los Angeles CA
| | - Matti Jauhiainen
- 9 National Institute for Health and Welfare Helsinki Finland.,12 Minerva Foundation Institute for Medical Research Biomedicum Helsinki Finland
| | - Aarno Palotie
- 1 Institute for Molecular Medicine Finland HiLIFE University of Helsinki Finland.,13 Program in Medical and Population Genetics and The Stanley Center for Psychiatric Research The Broad Institute of MIT and Harvard Cambridge MA.,14 Psychiatric and Neurodevelopmental Genetics Unit Department of Psychiatry, Analytic and Translational Genetics Unit Department of Medicine, and the Department of Neurology Massachusetts General Hospital Boston MA
| | - Marja-Riitta Taskinen
- 2 Research Programs Unit Clinical and Molecular Metabolism University of Helsinki Finland
| | - Kai Simons
- 4 Lipotype GmbH Dresden Germany.,15 Max Planck Institute of Cell Biology and Genetics Dresden Germany
| | - Samuli Ripatti
- 1 Institute for Molecular Medicine Finland HiLIFE University of Helsinki Finland.,13 Program in Medical and Population Genetics and The Stanley Center for Psychiatric Research The Broad Institute of MIT and Harvard Cambridge MA.,16 Department of Public Health Clinicum Faculty of Medicine University of Helsinki Finland
| |
Collapse
|
2138
|
Del Pinto R, Grassi D, Properzi G, Desideri G, Ferri C. Low Density Lipoprotein (LDL) Cholesterol as a Causal Role for Atherosclerotic Disease: Potential Role of PCSK9 Inhibitors. High Blood Press Cardiovasc Prev 2019; 26:199-207. [PMID: 31236902 DOI: 10.1007/s40292-019-00323-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 06/18/2019] [Indexed: 02/07/2023] Open
Abstract
Proprotein convertase subtilisin/kexin type 9 (PCSK9)-related discoveries of the turn of the century have translated into substantial novelty in dyslipidemia treatment in the last 5 years. With chronic preventable atherosclerotic cardiovascular diseases (ASCVD) representing an epidemic of morbidity and mortality worldwide, low-density lipoprotein cholesterol (LDL-c) reduction represents a public health priority. By overcoming two major statin-related issues, namely intolerance and ineffectiveness, PCSK9 inhibitors have offered a safe and effective option in selected clinical settings where LDL-c reduction is required. Herein, we recapitulate recent findings, clinical applications, and ASCVD prevention potential of PCSK9 inhibition, with focus on anti-PCSK9 monoclonal antibodies, evolocumab and alirocumab.
Collapse
Affiliation(s)
- Rita Del Pinto
- Department of Life, Health and Environmental Sciences, San Salvatore Hospital, University of L'Aquila, Building Delta 6, L'Aquila, Italy.
| | - Davide Grassi
- Department of Life, Health and Environmental Sciences, San Salvatore Hospital, University of L'Aquila, Building Delta 6, L'Aquila, Italy
| | - Giuliana Properzi
- Department of Life, Health and Environmental Sciences, San Salvatore Hospital, University of L'Aquila, Building Delta 6, L'Aquila, Italy
| | - Giovambattista Desideri
- Department of Life, Health and Environmental Sciences, San Salvatore Hospital, University of L'Aquila, Building Delta 6, L'Aquila, Italy
| | - Claudio Ferri
- Department of Life, Health and Environmental Sciences, San Salvatore Hospital, University of L'Aquila, Building Delta 6, L'Aquila, Italy
| |
Collapse
|
2139
|
Abstract
PURPOSE OF REVIEW Low-density lipoprotein cholesterol (LDL-C) is one major cause of cardiovascular disease (CVD). In this review, we discuss current developments in the understanding of LDL-C as lifelong risk factor, treatment targets, and emerging approaches to reduce cardiovascular risk by lowering LDL-C. RECENT FINDINGS Recent evidence underscores the importance of LDL-C lowering in CVD prevention by mechanisms that increase the hepatic clearance of apolipoprotein B-containing lipoproteins from the plasma. Mendelian randomization studies provided evidence on both safety and efficacy of lower LDL-C in the long term. For young individuals, metrics other than 10-year CVD risk are required. Despite this evidence, LDL-C treatment target attainment is poor. Novel approaches are therefore needed. These include individualized strategies and new LDL-C-lowering pharmaceuticals. Early, long-term treatment with LDL-C-lowering therapies has the potential to markedly reduce CVD incidence and progression. Future research should aim to identify patient characteristics that enable physicians to tailor therapy to each individual patient.
Collapse
|
2140
|
Abstract
PURPOSE OF REVIEW The cardiovascular (CV) risk related to lipid disorders is well established and is based on a robust body of evidence from well-designed randomized clinical trials, as well as prospective observational studies. In the last two decades, significant advances have been made in understanding the genetic basis of dyslipidemias. The present review is intended as a comprehensive discussion of current knowledge about the genetics and pathophysiology of disorders that predispose to dyslipidemia. We also focus on issues related to statins and the proprotein convertase subtilisin/kexin type 9 (PCSK9) and some of its polymorphisms, as well as new cholesterol-lowering medications, including PCSK9 inhibitors. RECENT FINDING Cholesterol is essential for the proper functioning of several body systems. However, dyslipidemia-especially elevated low-density lipoprotein (LDL-c) and triglyceride levels, as well as reduced lipoprotein lipase activity-is associated with an increased risk of coronary artery disease (CAD). High-density lipoprotein (HDL-c), however, seems to play a role as a risk marker rather than as a causal factor of the disease, as suggested by Mendelian randomization studies. Several polymorphisms in the lipoprotein lipase locus have been described and are associated with variations in the activity of this enzyme, producing high concentrations of triglycerides and increased risk of CAD. Dyslipidemia, especially increased LDL-c and triglyceride levels, continues to play a significant role in CV risk. The combination of genetic testing and counseling is important in the management of patients with dyslipidemia of genetic etiology. Strategies focused on primary prevention can offer an opportunity to reduce CV events.
Collapse
Affiliation(s)
- Ricardo Stein
- Graduate Program in Cardiology and Cardiovascular Sciences, Hospital de Clínicas de Porto Alegre (HCPA), Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil. .,Exercise Cardiology Research Group (CardioEx), Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil. .,School of Medicine, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil. .,Serviço de Fisiatria e Reabilitação, Hospital de Clínicas de Porto Alegre, Rua Ramiro Barcelos, 2350, Porto Alegre, RS, 90035-903, Brazil.
| | - Filipe Ferrari
- Graduate Program in Cardiology and Cardiovascular Sciences, Hospital de Clínicas de Porto Alegre (HCPA), Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Exercise Cardiology Research Group (CardioEx), Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Fernando Scolari
- Graduate Program in Cardiology and Cardiovascular Sciences, Hospital de Clínicas de Porto Alegre (HCPA), Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| |
Collapse
|
2141
|
Kobalava JD, Gurevich VS, Galyavich AM, Kaminnyi AI, Kashtalap VV, Mareev VY, Susekov AV, Shaposhnik II. [Possibilities of clinical use of ezetimibe Otrio (JSC "AKRIKHIN", Russia) in patients with high and very high cardiovascular risk who have not reached the target values of lipid metabolism. Conclusion of the Board of experts]. ACTA ACUST UNITED AC 2019; 59:47-57. [PMID: 31221075 DOI: 10.18087/cardio.n581] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 06/20/2019] [Indexed: 11/18/2022]
Abstract
This Conclusion of the Board of experts is devoted to the analysis of the evidence base, the position in modern clinical guidelines, the efficacy and safety analysis as well as the options of combined therapy with statins and ezetimibe (Otrio, JSC "AKRIKHIN") in various categories of patients in routine clinical practice in theRussian Federation. Cardiovascular diseases (CVD) continue to lead in the structure of morbidity and mortality inRussia. Hypercholesterolemia is one of the main modifiable risk factors for CVD. Administration of HMGCo-A-reductase inhibitors (statins) remains the basis for the prevention and treatment of the main complications of atherosclerosis, but the achievement of target levels of LDL-C on of statin monotherapy in Russian practice among different categories of risk does not exceed 50%. Proportion of patients (up to 12%) does not tolerate statin therapy, which requires the search for alternative therapies. To optimize the control of the level of LDL-C, combination therapy with statins and ezetimibe is used. Ezetimibe is an effective lipid-lowering drug, an inhibitor of intestinal absorption of cholesterol, which was investigated in many international and Russian studies, the results of which have demonstrated good tolerability, safety and efficacy (reduction of LDL-C levels by 18% in monotherapy). It was noted that the combined therapy with low/medium doses of statins and ezetimibe effectively reduces the level of LDL-C by 44-53%, which is comparable to the effect of high doses of statins and reduces CV risk in patients with CKD and ACS. Otrio (INN Ezetimib) tablets 10 mg ( JSC "AKRIKHIN",Russia) has demonstrated bioequivalence to the original drug Ezetrol tablets 10 mg (Schering-plough Labo N. V,Belgium). Broad use of a new generic product Otrio in combination with different statins will significantly increase the frequency of achievement of target lipid levels in patients with high and very high CV risk, including patients with chronic renal failure, type 2 diabetes and in patients with high hypercholesterolemia (LDL-C > 5 mmol/l) and, ultimately, reduce the burden of CV disease and mortality in Russia.
Collapse
Affiliation(s)
| | - V S Gurevich
- Saint-Petersburg State University; North-Western State Medical university n.a. I.I. Mechnikov
| | | | - A I Kaminnyi
- National Medical Research Center of Cardiology, Department of atherosclerosis; Pirogov Russian National Research Medical University
| | - V V Kashtalap
- Research Institute for Complex Issues of Cardiovascular Diseases; Kemerovo State Medical University
| | - V Yu Mareev
- Medical scientific and educational center of Lomonosov Moscow state University
| | | | | |
Collapse
|
2142
|
Movva R, Greenside P, Marinov GK, Nair S, Shrikumar A, Kundaje A. Deciphering regulatory DNA sequences and noncoding genetic variants using neural network models of massively parallel reporter assays. PLoS One 2019; 14:e0218073. [PMID: 31206543 PMCID: PMC6576758 DOI: 10.1371/journal.pone.0218073] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 05/24/2019] [Indexed: 11/19/2022] Open
Abstract
The relationship between noncoding DNA sequence and gene expression is not well-understood. Massively parallel reporter assays (MPRAs), which quantify the regulatory activity of large libraries of DNA sequences in parallel, are a powerful approach to characterize this relationship. We present MPRA-DragoNN, a convolutional neural network (CNN)-based framework to predict and interpret the regulatory activity of DNA sequences as measured by MPRAs. While our method is generally applicable to a variety of MPRA designs, here we trained our model on the Sharpr-MPRA dataset that measures the activity of ∼500,000 constructs tiling 15,720 regulatory regions in human K562 and HepG2 cell lines. MPRA-DragoNN predictions were moderately correlated (Spearman ρ = 0.28) with measured activity and were within range of replicate concordance of the assay. State-of-the-art model interpretation methods revealed high-resolution predictive regulatory sequence features that overlapped transcription factor (TF) binding motifs. We used the model to investigate the cell type and chromatin state preferences of predictive TF motifs. We explored the ability of our model to predict the allelic effects of regulatory variants in an independent MPRA experiment and fine map putative functional SNPs in loci associated with lipid traits. Our results suggest that interpretable deep learning models trained on MPRA data have the potential to reveal meaningful patterns in regulatory DNA sequences and prioritize regulatory genetic variants, especially as larger, higher-quality datasets are produced.
Collapse
Affiliation(s)
- Rajiv Movva
- The Harker School, San Jose, CA, United States of America
- Department of Genetics, Stanford University, Stanford, CA, United States of America
| | - Peyton Greenside
- Biomedical Informatics Training Program, Stanford University, Stanford, CA, United States of America
| | - Georgi K. Marinov
- Department of Genetics, Stanford University, Stanford, CA, United States of America
| | - Surag Nair
- Department of Computer Science, Stanford University, Stanford, CA, United States of America
| | - Avanti Shrikumar
- Department of Computer Science, Stanford University, Stanford, CA, United States of America
| | - Anshul Kundaje
- Department of Genetics, Stanford University, Stanford, CA, United States of America
- Department of Computer Science, Stanford University, Stanford, CA, United States of America
| |
Collapse
|
2143
|
Orekhov AN, Sobenin IA. Modified and Dysfunctional Lipoproteins in Atherosclerosis: Effectors or Biomarkers? Curr Med Chem 2019; 26:1512-1524. [PMID: 29557739 DOI: 10.2174/0929867325666180320121137] [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: 12/06/2017] [Revised: 02/14/2018] [Accepted: 03/05/2018] [Indexed: 01/17/2023]
Abstract
Atherosclerotic diseases are the leading cause of mortality in industrialized countries. Correspondingly, studying the pathogenesis of atherosclerosis and developing new methods for its diagnostic and treatment remain in the focus of current medicine and health care. This review aims to discuss the mechanistic role of low-density lipoprotein (LDL) and high-density lipoprotein (HDL) in atherogenesis. In particular, the generally accepted hypothesis about the key role of oxidized LDL in atherogenesis is questioned, and an alternative concept of multiple modification of LDL is presented. The fundamental question discussed in this review is whether LDL and HDL are effectors or biomarkers, or both. This is important for understanding whether lipoproteins are a therapeutic target or just diagnostic indicators.
Collapse
Affiliation(s)
- Alexander N Orekhov
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Moscow, Russian Federation.,Institute for Atherosclerosis Research, Skolkovo Innovative Center, Moscow, Russian Federation
| | - Igor A Sobenin
- Institute of Experimental Cardiology, National Medical Research Center of Cardiology, Moscow, Russian Federation
| |
Collapse
|
2144
|
Zhu J, Wu S, Hu S, Li H, Li M, Geng X, Wang H. NLRP3 inflammasome expression in peripheral blood monocytes of coronary heart disease patients and its modulation by rosuvastatin. Mol Med Rep 2019; 20:1826-1836. [PMID: 31257469 PMCID: PMC6625427 DOI: 10.3892/mmr.2019.10382] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 05/29/2019] [Indexed: 01/11/2023] Open
Abstract
Nucleotide-binding oligomerization domain, leucine rich repeat, and pyrin domain-containing protein 3 (NLRP3) inflammasome has been implicated in a series of physiological and pathological processes. However, its correlation in coronary heart disease (CHD) still remains to be elucidated. The present study aimed to determine the expression of NLRP3 inflammasome in peripheral blood monocytes (PBMCs) of stable angina pectoris (SAP) and acute myocardial infarction (AMI) patients. In addition, the effect of rosuvastatin on their activities was analyzed in vitro. A total of 60 participants with SAP (n=20), AMI (n=20) and non-CHD controls (n=20) were enrolled. Fluorescence-activated cell sorting, real-time PCR, western blotting and enzyme-linked immunosorbent assay were performed to reveal the role of NLRP3 inflammasome. NLRP3 inflammasome was expressed in the PBMCs, and revealed an increased expression along the downstream interleukin (IL)-1β and IL-18 in both SAP and AMI groups, compared to the control group. Moreover, there was a more marked increase in the expression of these indicators in AMI patients when compared to SAP patients. Interference with rosuvastatin in vitro revealed that the expression of NLRP3 inflammasome and its downstream cytokines were significantly downregulated in both SAP and AMI groups in a time-dependent manner. The activation of NLRP3 inflammasome may be involved in the development of CHD, and rosuvastatin could attenuate the inflammatory process of atherosclerosis by downregulating NLRP3 expression and its downstream mediators. These findings indicated a potential role of NLRP3 in the pathogenesis and management of CHD, and also provided new insights into the mechanistic framework of rosuvastatin activity.
Collapse
Affiliation(s)
- Jian Zhu
- Department of Cardiology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233004, P.R. China
| | - Shili Wu
- Department of Cardiology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233004, P.R. China
| | - Sigan Hu
- Department of Cardiology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233004, P.R. China
| | - Hui Li
- Department of Cardiology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233004, P.R. China
| | - Miaonan Li
- Department of Cardiology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233004, P.R. China
| | - Xu Geng
- Department of Cardiology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233004, P.R. China
| | - Hongju Wang
- Department of Cardiology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233004, P.R. China
| |
Collapse
|
2145
|
Glenn AJ, Viguiliouk E, Seider M, Boucher BA, Khan TA, Blanco Mejia S, Jenkins DJA, Kahleová H, Rahelić D, Salas-Salvadó J, Kendall CWC, Sievenpiper JL. Relation of Vegetarian Dietary Patterns With Major Cardiovascular Outcomes: A Systematic Review and Meta-Analysis of Prospective Cohort Studies. Front Nutr 2019; 6:80. [PMID: 31263700 PMCID: PMC6585466 DOI: 10.3389/fnut.2019.00080] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 05/16/2019] [Indexed: 12/18/2022] Open
Abstract
Background: Vegetarian dietary patterns are recommended for cardiovascular disease (CVD) prevention and management due to their favorable effects on cardiometabolic risk factors, however, the role of vegetarian dietary patterns in CVD incidence and mortality remains unclear. Objective: To update the European Association for the Study of Diabetes (EASD) clinical practice guidelines for nutrition therapy, we undertook a systematic review and meta-analysis of the association of vegetarian dietary patterns with major cardiovascular outcomes in prospective cohort studies that included individuals with and without diabetes using the Grading of Recommendations, Assessment, Development, and Evaluation (GRADE) approach. Methods: MEDLINE, EMBASE, and Cochrane databases were searched through September 6th, 2018. We included prospective cohort studies ≥1 year of follow-up including individuals with or without diabetes reporting the relation of vegetarian and non-vegetarian dietary patterns with at least one cardiovascular outcome. Two independent reviewers extracted data and assessed study quality (Newcastle-Ottawa Scale). The pre-specified outcomes included CVD incidence and mortality (total CVD, coronary heart disease (CHD) and stroke). Risk ratios for associations were pooled using inverse variance random effects model and expressed as risk ratios (RRs) with 95% confidence intervals (CIs). Heterogeneity was assessed (Cochran Q-statistic) and quantified (I 2-statistic). The overall certainty of the evidence was assessed using GRADE. Results: Seven prospective cohort studies (197,737 participants, 8,430 events) were included. A vegetarian dietary pattern was associated with reduced CHD mortality [RR, 0.78 (CI, 0.69, 0.88)] and incidence [0.72 (0.61, 0.85)] but were not associated with CVD mortality [0.92 (0.84, 1.02)] and stroke mortality [0.92 (0.77, 1.10)]. The overall certainty of the evidence was graded as "very low" for all outcomes, owing to downgrades for indirectness and imprecision. Conclusions: Very low-quality evidence indicates that vegetarian dietary patterns are associated with reductions in CHD mortality and incidence but not with CVD and stroke mortality in individuals with and without diabetes. More research, particularly in different populations, is needed to improve the certainty in our estimates. Clinical Trial Registration: Clinicaltrials.gov, identifier: NCT03610828.
Collapse
Affiliation(s)
- Andrea J. Glenn
- Department of Nutritional Sciences, University of Toronto, Toronto, ON, Canada
- Clinical Nutrition and Risk Factor Modification Center, St. Michael's Hospital, Toronto, ON, Canada
- Toronto 3D Knowledge Synthesis and Clinical Trials Unit, St. Michael's Hospital, Toronto, ON, Canada
| | - Effie Viguiliouk
- Department of Nutritional Sciences, University of Toronto, Toronto, ON, Canada
- Clinical Nutrition and Risk Factor Modification Center, St. Michael's Hospital, Toronto, ON, Canada
- Toronto 3D Knowledge Synthesis and Clinical Trials Unit, St. Michael's Hospital, Toronto, ON, Canada
| | - Maxine Seider
- Department of Nutritional Sciences, University of Toronto, Toronto, ON, Canada
- Clinical Nutrition and Risk Factor Modification Center, St. Michael's Hospital, Toronto, ON, Canada
- Toronto 3D Knowledge Synthesis and Clinical Trials Unit, St. Michael's Hospital, Toronto, ON, Canada
| | - Beatrice A. Boucher
- Department of Nutritional Sciences, University of Toronto, Toronto, ON, Canada
| | - Tauseef A. Khan
- Department of Nutritional Sciences, University of Toronto, Toronto, ON, Canada
- Clinical Nutrition and Risk Factor Modification Center, St. Michael's Hospital, Toronto, ON, Canada
- Toronto 3D Knowledge Synthesis and Clinical Trials Unit, St. Michael's Hospital, Toronto, ON, Canada
| | - Sonia Blanco Mejia
- Department of Nutritional Sciences, University of Toronto, Toronto, ON, Canada
- Clinical Nutrition and Risk Factor Modification Center, St. Michael's Hospital, Toronto, ON, Canada
- Toronto 3D Knowledge Synthesis and Clinical Trials Unit, St. Michael's Hospital, Toronto, ON, Canada
| | - David J. A. Jenkins
- Department of Nutritional Sciences, University of Toronto, Toronto, ON, Canada
- Clinical Nutrition and Risk Factor Modification Center, St. Michael's Hospital, Toronto, ON, Canada
- Toronto 3D Knowledge Synthesis and Clinical Trials Unit, St. Michael's Hospital, Toronto, ON, Canada
- Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, ON, Canada
- Division of Endocrinology and Metabolism, St. Michael's Hospital, Toronto, ON, Canada
- Department of Medicine, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Hana Kahleová
- Physicians Committee for Responsible Medicine, Washington, DC, United States
- Institute for Clinical and Experimental Medicine, Prague, Czechia
| | - Dario Rahelić
- Vuk Vrhovac University Clinic for Diabetes, Endocrinology and Metabolic Diseases, Merkur University Hospital, Zagreb, Croatia
- School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Jordi Salas-Salvadó
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBER Obn), Instituto de Salud Carlos III, Madrid, Spain
- Human Nutrition Department, IISPV, Universitat Rovira i Virgili, Reus, Spain
| | - Cyril W. C. Kendall
- Department of Nutritional Sciences, University of Toronto, Toronto, ON, Canada
- Clinical Nutrition and Risk Factor Modification Center, St. Michael's Hospital, Toronto, ON, Canada
- Toronto 3D Knowledge Synthesis and Clinical Trials Unit, St. Michael's Hospital, Toronto, ON, Canada
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK, Canada
| | - John L. Sievenpiper
- Department of Nutritional Sciences, University of Toronto, Toronto, ON, Canada
- Clinical Nutrition and Risk Factor Modification Center, St. Michael's Hospital, Toronto, ON, Canada
- Toronto 3D Knowledge Synthesis and Clinical Trials Unit, St. Michael's Hospital, Toronto, ON, Canada
- Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, ON, Canada
- Division of Endocrinology and Metabolism, St. Michael's Hospital, Toronto, ON, Canada
| |
Collapse
|
2146
|
Skuratovskaia D, Vulf M, Komar A, Kirienkova E, Litvinova L. Promising Directions in Atherosclerosis Treatment Based on Epigenetic Regulation Using MicroRNAs and Long Noncoding RNAs. Biomolecules 2019; 9:E226. [PMID: 31212708 PMCID: PMC6627269 DOI: 10.3390/biom9060226] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 06/03/2019] [Accepted: 06/07/2019] [Indexed: 12/22/2022] Open
Abstract
Atherosclerosis is one of the leading causes of mortality from cardiovascular disease (CVD) and is a chronic inflammatory disease of the middle and large arteries caused by a disruption of lipid metabolism. Noncoding RNA (ncRNA), including microRNA (miRNA), small interfering RNA (siRNA) and long noncoding RNA (lncRNA), was investigated for the treatment of atherosclerosis. Regulation of the expression of noncoding RNA targets the constituent element of the pathogenesis of atherosclerosis. Currently, miRNA therapy commonly employs miRNA antagonists and mimic compounds. In this review, attention is focused on approaches to correcting molecular disorders based on the genetic regulation of the transcription of key genes responsible for the development of atherosclerosis. Promising technologies were considered for the treatment of atherosclerosis, and examples are given for technologies that have been shown to be effective in clinical trials.
Collapse
Affiliation(s)
- Daria Skuratovskaia
- Laboratory of Immunology and Cell Biotechnology, Immanuel Kant Baltic Federal University, 236016 Kaliningrad, Russia.
| | - Maria Vulf
- Laboratory of Immunology and Cell Biotechnology, Immanuel Kant Baltic Federal University, 236016 Kaliningrad, Russia.
| | - Aleksandra Komar
- Laboratory of Immunology and Cell Biotechnology, Immanuel Kant Baltic Federal University, 236016 Kaliningrad, Russia.
| | - Elena Kirienkova
- Laboratory of Immunology and Cell Biotechnology, Immanuel Kant Baltic Federal University, 236016 Kaliningrad, Russia.
| | - Larisa Litvinova
- Laboratory of Immunology and Cell Biotechnology, Immanuel Kant Baltic Federal University, 236016 Kaliningrad, Russia.
| |
Collapse
|
2147
|
Affiliation(s)
- Ying Jin
- 1 Laboratory of Inflammation and Vascular Biology Institute of Clinical Medicine Hubei University of Medicine Renmin Hospital Shiyan China.,2 Center for Translational Medicine Hubei University of Medicine Renmin Hospital Shiyan China
| | - Jian Fu
- 1 Laboratory of Inflammation and Vascular Biology Institute of Clinical Medicine Hubei University of Medicine Renmin Hospital Shiyan China.,2 Center for Translational Medicine Hubei University of Medicine Renmin Hospital Shiyan China
| |
Collapse
|
2148
|
Fruchart JC, Santos RD, Aguilar-Salinas C, Aikawa M, Al Rasadi K, Amarenco P, Barter PJ, Ceska R, Corsini A, Després JP, Duriez P, Eckel RH, Ezhov MV, Farnier M, Ginsberg HN, Hermans MP, Ishibashi S, Karpe F, Kodama T, Koenig W, Krempf M, Lim S, Lorenzatti AJ, McPherson R, Nuñez-Cortes JM, Nordestgaard BG, Ogawa H, Packard CJ, Plutzky J, Ponte-Negretti CI, Pradhan A, Ray KK, Reiner Ž, Ridker PM, Ruscica M, Sadikot S, Shimano H, Sritara P, Stock JK, Su TC, Susekov AV, Tartar A, Taskinen MR, Tenenbaum A, Tokgözoğlu LS, Tomlinson B, Tybjærg-Hansen A, Valensi P, Vrablík M, Wahli W, Watts GF, Yamashita S, Yokote K, Zambon A, Libby P. The selective peroxisome proliferator-activated receptor alpha modulator (SPPARMα) paradigm: conceptual framework and therapeutic potential : A consensus statement from the International Atherosclerosis Society (IAS) and the Residual Risk Reduction Initiative (R3i) Foundation. Cardiovasc Diabetol 2019; 18:71. [PMID: 31164165 PMCID: PMC6549355 DOI: 10.1186/s12933-019-0864-7] [Citation(s) in RCA: 103] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 04/29/2019] [Indexed: 12/12/2022] Open
Abstract
In the era of precision medicine, treatments that target specific modifiable characteristics of high-risk patients have the potential to lower further the residual risk of atherosclerotic cardiovascular events. Correction of atherogenic dyslipidemia, however, remains a major unmet clinical need. Elevated plasma triglycerides, with or without low levels of high-density lipoprotein cholesterol (HDL-C), offer a key modifiable component of this common dyslipidemia, especially in insulin resistant conditions such as type 2 diabetes mellitus. The development of selective peroxisome proliferator-activated receptor alpha modulators (SPPARMα) offers an approach to address this treatment gap. This Joint Consensus Panel appraised evidence for the first SPPARMα agonist and concluded that this agent represents a novel therapeutic class, distinct from fibrates, based on pharmacological activity, and, importantly, a safe hepatic and renal profile. The ongoing PROMINENT cardiovascular outcomes trial is testing in 10,000 patients with type 2 diabetes mellitus, elevated triglycerides, and low levels of HDL-C whether treatment with this SPPARMα agonist safely reduces residual cardiovascular risk.
Collapse
Affiliation(s)
| | - Raul D. Santos
- Hospital Israelita Albert Einstein, and Lipid Clinic, Heart Institute (InCor) University of Sao Paulo Medical School Hospital, Sao Paulo, Brazil
| | - Carlos Aguilar-Salinas
- Unidad de Investigacion de Enfermedades Metabolicas, Department of Endocrinolgy and Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey, Mexico
| | - Masanori Aikawa
- Center for Interdisciplinary Cardiovascular Sciences and Center for Excellence in Vascular Biology, Division of Cardiovascular Medicine and Channing Division of Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA USA
| | - Khalid Al Rasadi
- Department of Clinical Biochemistry, Sultan Qaboos University Hospital, Muscat, Oman
| | - Pierre Amarenco
- Department of Neurology and Stroke Center, Paris-Diderot-Sorbonne University, Paris, France
| | - Philip J. Barter
- Lipid Research Group, School of Medical Sciences, University of New South Wales, Sydney, NSW Australia
| | - Richard Ceska
- IIIrd Dept Int. Med, Center for Preventive Cardiology, 3rd Internal Medicine Clinic, University General Hospital and Charles University, Prague, Czech Republic
| | - Alberto Corsini
- Department of Pharmacological and Biomolecular Sciences, Università Degli Studi di Milano, Milan, Italy
| | - Jean-Pierre Després
- Centre de recherche sur les soins et les services de première ligne-Université Laval du CIUSSS de la Capitale-Nationale, Department of Kinesiology, Faculty of Medicine, Université Laval, Québec, QC Canada
| | - Patrick Duriez
- INSERM, CHU Lille, U1171-Degenerative & Vascular Cognitive Disorders, University of Lille, Faculty of Pharmacy, University of Lille, UDSL, Lille, France
| | - Robert H. Eckel
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO USA
| | - Marat V. Ezhov
- Laboratory of Lipid Disorders, National Cardiology Research Center, Moscow, Russian Federation
| | - Michel Farnier
- Lipid Clinic, Point Médical and Department of Cardiology, CHU Dijon-Bourgogne, Dijon, France
| | - Henry N. Ginsberg
- Columbia University Vagelos College of Physicians and Surgeons, New York, USA
| | - Michel P. Hermans
- Division of Endocrinology and Nutrition, Cliniques Universitaires St-Luc and Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium
| | - Shun Ishibashi
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Jichi Medical University, Shimotsuke, Japan
| | - Fredrik Karpe
- OCDEM, University of Oxford and the NIHR Oxford Biomedical Research Centre, OUH Foundation Trust, Churchill Hospital, Oxford, UK
| | - Tatsuhiko Kodama
- Laboratory for System Biology and Medicine Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Wolfgang Koenig
- Deutsches Herzzentrum München, Technische Universitat München, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
- Institute of Epidemiology and Medical Biometry, University of Ulm, Ulm, Germany
| | - Michel Krempf
- Mass Spectrometry Core facility of West Human Nutrition Research Center (CRNHO), Hotel Dieu Hospital, Nantes, France
- Inra, UMR 1280, Physiologie des Adaptations Nutritionnelles, Nantes, France
- Department of Endocrinology, Metabolic diseases and Nutrition, G and R Laennec Hospital, Nantes, France
| | - Soo Lim
- Department of Internal Medicine, Seoul National University Bundang Hospital and Seoul National University College of Medicine, Seongnam, Republic of Korea
| | - Alberto J. Lorenzatti
- DAMIC Medical Institute/Rusculleda Foundation for Research, Córdoba, Argentina
- Cardiology Department, Córdoba Hospital, Córdoba, Argentina
| | - Ruth McPherson
- Ruddy Canadian Cardiovascular Genetics Centre, University of Ottawa Heart Institute, Ottawa, Canada
| | - Jesus Millan Nuñez-Cortes
- Internal Medicine, Lipids Unit, Gregorio Marañón University Hospital, Madrid, Spain
- Department of Medicine, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
- Instituto de Investigaciones Sanitarias Gregorio Marañón, Madrid, Spain
| | - Børge G. Nordestgaard
- Department of Clinical Biochemistry, Herlev and Gentofte Hospital, Copenhagen University Hospital, Herlev, Denmark
- The Copenhagen General Population Study, Herlev and Gentofte Hospital, Copenhagen University Hospital, Herlev, Denmark
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Hisao Ogawa
- National Cerebral and Cardiovascular Center, Suita, Osaka Japan
| | - Chris J. Packard
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Jorge Plutzky
- Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA USA
| | - Carlos I. Ponte-Negretti
- Unidad de Prevención Cardiometabólica Cardiocob. Servicio de Cardiología Hospital el Pino Santiago de Chile, Sociedad Inter Americana de Cardiología SIAC Chairman Cardiovascular Prevention Comite, Santiago de Chile, Chile
| | - Aruna Pradhan
- Division of Cardiovascular Medicine, VA Boston Medical Center, Boston, MA USA
- Division of Preventive Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA USA
| | - Kausik K. Ray
- Imperial Centre for Cardiovascular Disease Prevention, Department of Primary Care and Public Health, Imperial College London, London, UK
| | - Željko Reiner
- Department of Internal Medicine, University Hospital Centre Zagreb, School of Medicine, Zagreb University, Kispaticeva 12, Zagreb, Croatia
| | - Paul M. Ridker
- Division of Cardiovascular Medicine and Center for Cardiovascular Disease Prevention, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA USA
| | - Massimiliano Ruscica
- Department of Pharmacological and Biomolecular Sciences, Università Degli Studi di Milano, Milan, Italy
| | - Shaukat Sadikot
- Department of Endocrinology/Diabetology, Jaslok Hospital and Research Centre, Mumbai, India
| | - Hitoshi Shimano
- Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Ibaraki, 305-8575 Japan
| | - Piyamitr Sritara
- Department of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Jane K. Stock
- R3i Foundation, Picassoplatz 8, 4010 Basel, Switzerland
| | - Ta-Chen Su
- Departments of Internal Medicine and Environmental and Occupational Medicine, National Taiwan University; Institute of Occupational Medicine and Industrial Hygiene, National Taiwan University College of Public Health, Taipei, Taiwan
| | - Andrey V. Susekov
- Faculty of Clinical Pharmacology and Therapeutics, Academy for Postgraduate Continuous Medical Education, Moscow, Russian Federation
| | | | - Marja-Riitta Taskinen
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki and Clinical Research Institute, HUCH Ltd., Helsinki, Finland
| | - Alexander Tenenbaum
- Sackler Faculty of Medicine, Tel Aviv University, 6997801 Tel Aviv, Israel
- Cardiac Rehabilitation Institute, Sheba Medical Center, 5265601 Tel Hashomer, Israel
| | - Lale S. Tokgözoğlu
- Department of Cardiology, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Brian Tomlinson
- Department of Medicine & Theraputics, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Anne Tybjærg-Hansen
- Department of Clinical Biochemistry, Rigshospitalet; Copenhagen University Hospital, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- The Copenhagen General Population Study, Herlev and Gentofte Hospital, Herlev, Denmark
| | - Paul Valensi
- Department of Endocrinology, Diabetology and Nutrition, Jean-Verdier Hospital (AP-HP), Paris 13 University, Sorbonne Paris Cité, CRNH-IdF, CINFO, 93140 Bondy, France
| | - Michal Vrablík
- 3rd Department of Medicine, 1st Faculty of Medicine of Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Walter Wahli
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Clinical Sciences Building, 11 Mandalay Road, Singapore, 308232 Singapore
- Center for Integrative Genomics, Université de Lausanne, Le Génopode, CH-1015 Lausanne, Switzerland
- Institut National de La Recherche Agronomique (INRA), UMR1331 ToxAlim, Toulouse, France
| | - Gerald F. Watts
- Lipid Disorders Clinic, Department of Cardiology, Royal Perth Hospital, School of Medicine, University of Western Australia, Perth, Australia
| | - Shizuya Yamashita
- Rinku General Medical Center, Izumisano, Osaka Japan
- Department of Community Medicine, Osaka University Graduate School of Medicine, Suita, Osaka Japan
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Suita, Osaka Japan
| | - Koutaro Yokote
- Department of Endocrinology, Hematology and Gerontology, Clinical Cell Biology and Medicine, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Alberto Zambon
- Department of Medicine-DIMED, University of Padua, Padua, Italy
| | - Peter Libby
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA USA
| |
Collapse
|
2149
|
He WM, Dai T, Chen J, Wang JA. Leukocyte cell-derived chemotaxin 2 inhibits development of atherosclerosis in mice. Zool Res 2019; 40:317-323. [PMID: 31310065 PMCID: PMC6680125 DOI: 10.24272/j.issn.2095-8137.2019.030] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Leukocyte cell-derived chemotaxin 2 (LECT2), a multifunctional hepatokine, is involved in many pathological conditions. However, its role in atherosclerosis remains undefined. In this study, we administered vehicle or LECT2 to male Apoe-/- mice fed a Western diet for 15 weeks. Atherosclerotic lesions were visualized and quantified with Oil-red O and hematoxylin staining. The mRNA expression levels of MCP-1, MMP-1, IL-8, IL-1β, and TNF-α were analyzed by quantitative real-time polymerase chain reaction. Serum TNF-α, IL-1β, IL-8, MCP-1, and MMP-1 concentrations were measured by enzyme-linked immunosorbent assay. CD68, CD31, and α-SMA, markers of macrophages, endothelial cells, and smooth muscle cells, respectively, were detected by immunostaining. Results showed that LECT2 reduced total cholesterol and low-density lipoprotein concentrations in serum and inhibited the development of atherosclerotic lesions, accompanied by reductions in inflammatory cytokines and lower MCP-1, MMP-1, TNF-α, IL-8, and IL-1β mRNA abundance. Furthermore, LECT2 decreased CD68, but increased α-SMA in atherosclerotic lesions, suggesting an increase in smooth muscle cells and reduction in macrophages. In summary, LECT2 inhibited the development of atherosclerosis in mice, accompanied by reduced serum total cholesterol concentration and lower inflammatory responses.
Collapse
Affiliation(s)
- Wen-Ming He
- Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou Zhejiang 310009, China.,Department of Cardiology, the Affiliated Hospital of Medical School of Ningbo University, Ningbo Zhejiang 315010, China
| | - Ting Dai
- Department of Cardiology, the Affiliated Hospital of Medical School of Ningbo University, Ningbo Zhejiang 315010, China
| | - Jiong Chen
- Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Meishan Campus, Ningbo University, Ningbo Zhejiang 315832, China
| | - Jian-An Wang
- Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou Zhejiang 310009, China
| |
Collapse
|
2150
|
Practical guide for the use of PCSK9 inhibitors in Portugal. REVISTA PORTUGUESA DE CARDIOLOGIA (ENGLISH EDITION) 2019. [DOI: 10.1016/j.repce.2019.07.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|