1
|
Ogawara R, Misaka T, Ohashi N, Ichimura S, Tomita Y, Tani T, Anzai F, Sato Y, Yokokawa T, Sato A, Shimizu T, Sato T, Oikawa M, Kobayashi A, Yoshihisa A, Takeishi Y. Very short-term blood pressure variability by pulse transit time-based measurements during night-time predicts future cardiovascular events in patients with ischemic heart disease. Clin Res Cardiol 2024:10.1007/s00392-024-02539-7. [PMID: 39297937 DOI: 10.1007/s00392-024-02539-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 08/30/2024] [Indexed: 09/21/2024]
Abstract
Blood pressure (BP) variability (BPV) is associated with an increased risk of cardiovascular events, independent of absolute BP values. However, the predictive significance of very short-term BPV, occurring within seconds or minutes, in patients with ischemic heart disease (IHD) has yet to be established. This prospective study involved 206 consecutive hospitalized patients with IHD (mean age 67.6 years, 78.2% male) who underwent pulse transit time (PTT)-based continuous BP recording during the night-time. Very short-term BPV was assessed by standard deviation (SD), coefficient of variation (CV), and variation independent of mean (VIM) of PTT-BP. Clinical outcome data were collected. When the patients were categorized into two groups according to the median value of very short-term BPV, Kaplan-Meier analysis revealed that patients with elevated SD, CV, and VIM of systolic and diastolic PTT-BP were associated with lower event-free survival rates from the composite cardiovascular events including cardiac deaths, worsening heart failure cases, nonfatal myocardial infarctions, unplanned revascularizations, and strokes over a median follow-up of 797 days. In a multivariate Cox proportional hazards analysis adjusting for confounding variables, each parameter as a continuous variable was independently associated with adverse events. Incorporating very short-term BPV into basic models had a significant impact on risk reclassification and integrated discrimination for cardiovascular outcomes. In conclusion, the identification of patients with elevated very short-term BPV during the night-time through a PTT-driven approach helps stratify the future risk in IHD patients.
Collapse
Affiliation(s)
- Ryo Ogawara
- Department of Cardiovascular Medicine, Fukushima Medical University, 1 Hikarigaoka, Fukushima, 960-1295, Japan
| | - Tomofumi Misaka
- Department of Cardiovascular Medicine, Fukushima Medical University, 1 Hikarigaoka, Fukushima, 960-1295, Japan.
- Department of Community Cardiovascular Medicine, Fukushima Medical University, Fukushima, Japan.
| | - Naoto Ohashi
- Department of Cardiovascular Medicine, Fukushima Medical University, 1 Hikarigaoka, Fukushima, 960-1295, Japan
| | - Shohei Ichimura
- Department of Cardiovascular Medicine, Fukushima Medical University, 1 Hikarigaoka, Fukushima, 960-1295, Japan
| | - Yusuke Tomita
- Department of Cardiovascular Medicine, Fukushima Medical University, 1 Hikarigaoka, Fukushima, 960-1295, Japan
| | - Tetsuya Tani
- Department of Cardiovascular Medicine, Fukushima Medical University, 1 Hikarigaoka, Fukushima, 960-1295, Japan
| | - Fumiya Anzai
- Department of Cardiovascular Medicine, Fukushima Medical University, 1 Hikarigaoka, Fukushima, 960-1295, Japan
| | - Yu Sato
- Department of Cardiovascular Medicine, Fukushima Medical University, 1 Hikarigaoka, Fukushima, 960-1295, Japan
| | - Tetsuro Yokokawa
- Department of Cardiovascular Medicine, Fukushima Medical University, 1 Hikarigaoka, Fukushima, 960-1295, Japan
| | - Akihiko Sato
- Department of Cardiovascular Medicine, Fukushima Medical University, 1 Hikarigaoka, Fukushima, 960-1295, Japan
| | - Takeshi Shimizu
- Department of Cardiovascular Medicine, Fukushima Medical University, 1 Hikarigaoka, Fukushima, 960-1295, Japan
| | - Takamasa Sato
- Department of Cardiovascular Medicine, Fukushima Medical University, 1 Hikarigaoka, Fukushima, 960-1295, Japan
- Department of Community Cardiovascular Medicine, Fukushima Medical University, Fukushima, Japan
| | - Masayoshi Oikawa
- Department of Cardiovascular Medicine, Fukushima Medical University, 1 Hikarigaoka, Fukushima, 960-1295, Japan
| | - Atsushi Kobayashi
- Department of Cardiovascular Medicine, Fukushima Medical University, 1 Hikarigaoka, Fukushima, 960-1295, Japan
| | - Akiomi Yoshihisa
- Department of Cardiovascular Medicine, Fukushima Medical University, 1 Hikarigaoka, Fukushima, 960-1295, Japan
- Department of Clinical Laboratory Sciences, Fukushima Medical University School of Health Sciences, Fukushima, Japan
| | - Yasuchika Takeishi
- Department of Cardiovascular Medicine, Fukushima Medical University, 1 Hikarigaoka, Fukushima, 960-1295, Japan
| |
Collapse
|
2
|
Gallo G, Volterrani M, Fini M, Sposato B, Autore C, Tocci G, Volpe M. Position Statement of the Italian Society of Cardiovascular Prevention (SIPREC) and Italian Heart Failure Association (ITAHFA) on Cardiac Rehabilitation and Protection Programs as a Cornerstone of Secondary Prevention after Myocardial Infarction or Revascularization. High Blood Press Cardiovasc Prev 2024; 31:417-423. [PMID: 39060868 DOI: 10.1007/s40292-024-00663-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 07/12/2024] [Indexed: 07/28/2024] Open
Abstract
Despite the remarkable and progressive advances made in the prevention and management of cardiovascular diseases, the recurrence of cardiovascular events remains unacceptably elevated with a notable size of the residual risk. Indeed, in patients who suffered from myocardial infarction or who underwent percutaneous or surgical myocardial revascularization, life-style changes and optimized pharmacological therapy with antiplatelet drugs, lipid lowering agents, beta-blockers, renin angiotensin system inhibitors and antidiabetic drugs, when appropriate, are systematically prescribed but they might be insufficient to protect from further events. In such a context, an increasing body of evidence supports the benefits of cardiac rehabilitation (CR) in the setting of secondary cardiovascular prevention, consisting in the reduction of myocardial oxygen demands, in the inhibition of atherosclerotic plaque progression and in an improvement of exercise performance, quality of life and survival. However, prescription and implementation of CR programs is still not sufficiently considered.The aim of this position paper of the Italian Society of Cardiovascular Prevention (SIPREC) and of the Italian Heart Failure Association (ITAHFA) is to examine the reasons of the insufficient use of this strategy in clinical practice and to propose some feasible solutions to overcome this clinical gap.
Collapse
Affiliation(s)
- Giovanna Gallo
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, Via di Grottarossa 1035-1039, Rome, 00189, Italy
| | | | | | | | - Camillo Autore
- Cardio-Pulmonary Department, San Raffaele Cassino, Cassino (FR), 03043, Italy
| | - Giuliano Tocci
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, Via di Grottarossa 1035-1039, Rome, 00189, Italy
| | | |
Collapse
|
3
|
Toprak K, Kaplangöray M, Karataş M, Dursun A, Arğa Y, Tascanov MB, Biçer A, Demirbağ R. Atherogenic Combined Index: Validation of a Novel Predictive Lipid Biomarker for the Presence and Severity of Coronary Artery Disease. Arch Med Res 2024; 55:103065. [PMID: 39098112 DOI: 10.1016/j.arcmed.2024.103065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 07/06/2024] [Accepted: 07/24/2024] [Indexed: 08/06/2024]
Abstract
BACKGROUND/AIM The balance between atherogenic and antiatherogenic lipid particles significantly influences coronary artery disease (CAD), as an imbalance may contribute to the development and progression of atherosclerosis, which affects the risk and severity of CAD. This study aims to introduce and validate the atherogenic combined index (ACI) as a novel lipid biomarker that, comprehensively assesses the balance between atherogenic and antiatherogenic particles in the blood to effectively reflect the cumulative atherogenic effect and its association with the presence and severity of CAD. MATERIAL AND METHODS In this cross-sectional study, 1,830 patients diagnosed with CAD and a total of 650 patients without CAD were included in the study cohort for comprehensive analysis and comparison. Based on the tertiles of the SYNTAX score (SS), three subgroups of patients with CAD were identified. ACI and other atherogenic indices were compared to predict the presence and severity of CAD. RESULTS The levels of ACI and other non-traditional lipid markers levels were higher in the CAD group compared to the non-CAD group (p <0.05, for all). ACI showed a good linear association with the SYNTAX score (r = 0.527; p <0.001). The multivariate logistic regression model showed that ACI was an independent predictor of the presence (OR: 1.602, 95% CI: 1.509-1.701, p <0.001) and severity (OR: 1.296, 95% CI: 1.243-1.351, p <0.001) of CAD after adjustment for various confounders. CONCLUSION The results suggest that ACI may serve as a promising and stronger tool for predicting the presence and severity of CAD.
Collapse
Affiliation(s)
- Kenan Toprak
- Harran University, Faculty of Medicine, Department of Cardiology, Sanliurfa, Turkey.
| | - Mustafa Kaplangöray
- Şeyh Edebali University, Medical Faculty, Department of Cardiology, Bilecik, Turkey
| | - Mesut Karataş
- Kartal Koşuyolu High Specialization Training and Research Hospital, Istanbul, Turkey
| | - Ayten Dursun
- Şanlıurfa Provincial Health Directorate, Nursing Department, Sanliurfa, Turkey
| | - Yakup Arğa
- Viranşehir State Hospital, Department of Cardiology, Sanliurfa, Turkey
| | | | - Asuman Biçer
- Harran University, Faculty of Medicine, Department of Cardiology, Sanliurfa, Turkey
| | - Recep Demirbağ
- Harran University, Faculty of Medicine, Department of Cardiology, Sanliurfa, Turkey
| |
Collapse
|
4
|
Therre M, Wenzel P, Haring B. Atherosclerosis is an inflammatory disease: treat it as such! Clin Res Cardiol 2024; 113:1274-1275. [PMID: 38806822 PMCID: PMC11269445 DOI: 10.1007/s00392-024-02470-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/21/2024] [Indexed: 05/30/2024]
Affiliation(s)
- Markus Therre
- Department of Internal Medicine III, Cardiology, Angiology and Intensive Care Medicine, University Hospital Saarland, Saarland University, Kirrberger Str. 100, Homburg, 66421, Saar, Germany
| | - Philip Wenzel
- Department of Cardiology, University Medical Center of the Johannes Gutenberg University Mainz and German Center for Cardiovascular Research - Partner Site Rhine-Main, Mainz, Germany
| | - Bernhard Haring
- Department of Internal Medicine III, Cardiology, Angiology and Intensive Care Medicine, University Hospital Saarland, Saarland University, Kirrberger Str. 100, Homburg, 66421, Saar, Germany.
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
5
|
Wang W, Liu L, Qiu W, Chen C, Huang Y, Cai A, Nie Z, Ou Y, Zhu Y, Feng Y. The Non-Targeted Lipidomic-Based Classifier Reveals Two Candidate Biomarkers for Ischemic Stroke in Hypertensive Individuals. Risk Manag Healthc Policy 2024; 17:1889-1901. [PMID: 39100548 PMCID: PMC11297523 DOI: 10.2147/rmhp.s465135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 07/11/2024] [Indexed: 08/06/2024] Open
Abstract
Introduction Traditional clinical risk factors are insufficient to estimate the residual risk of large-vessel ischemic stroke. Non-targeted lipidomic techniques provide an opportunity to evaluate these risks. Methods Plasma samples were collected from 113 hypertensive individuals, including 55 individuals at high risk of ischemic stroke and 58 matched individuals, in a prospective nested case-control cohort. To identify dysregulated lipid metabolites, we conducted multivariate and univariate analyses. A classifier based on a cross-validated procedure was employed to select the optimal combination of lipid species and their ratios. Results We identified 23 dysregulated lipid species in patients with and without ischemic stroke, including 16 (69.6%) up-regulated and 7 (30.4%) down-regulated lipid species. Through internal cross-validation, the optimal combination of two lipid features (phosphatidylcholine 34:2 and triglyceride 18:1/18:1/22:1 / phosphatidylcholine 34:2, referred to as ischemic stroke-related 2 lipid features - IS2LP) was selected, leading to a more precise prediction probability for ischemic stroke within 3.9 years. In the comparison of different risk factors, the traditional risk score, the IS2LP risk score, and the combination of the traditional risk score with IS2LP yield AUC values of 0.613(95% CI:0.509-0.717), 0.833(95% CI:0.755-0.911), and 0.843(95% CI:0.777-0.916), respectively. The combination of the traditional risk score and IS2LP exhibited significantly improved discriminative performance, with an integrated discrimination improvement (IDI) of 0.31 (p<0.001) and a continuous net reclassification improvement (NRI) of 1.06 (p < 0.001) compared to the traditional risk score. Conclusion We identified new lipidomic biomarkers associated with the futural event of large-vessel ischemic stroke. These lipid species could serve as potential blood biomarkers for assessing the residual risk of ischemic stroke in hypertensive individuals.
Collapse
Affiliation(s)
- Wenbin Wang
- Department of Cardiology, Hypertension Research Laboratory, Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, People’s Republic of China
| | - Lin Liu
- Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Weida Qiu
- Department of Cardiology, Hypertension Research Laboratory, Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, People’s Republic of China
| | - Chaolei Chen
- Department of Cardiology, Hypertension Research Laboratory, Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, People’s Republic of China
| | - Yuqing Huang
- Department of Cardiology, Hypertension Research Laboratory, Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, People’s Republic of China
| | - Anping Cai
- Department of Cardiology, Hypertension Research Laboratory, Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, People’s Republic of China
| | - Zhiqiang Nie
- Department of Cardiology, Hypertension Research Laboratory, Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, People’s Republic of China
| | - Yanqiu Ou
- Department of Cardiology, Hypertension Research Laboratory, Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, People’s Republic of China
| | - Yicheng Zhu
- Department of Cardiology, Hypertension Research Laboratory, Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, People’s Republic of China
| | - Yingqing Feng
- Department of Cardiology, Hypertension Research Laboratory, Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, People’s Republic of China
| |
Collapse
|
6
|
Okan T, Doruk M, Ozturk A, Topaloglu C, Dogdus M, Yilmaz MB. Evaluation of Plasma Atherogenic Index, Triglyceride-Glucose Index and Other Lipid Ratios as Predictive Biomarkers of Coronary Artery Disease in Different Age Groups. Diagnostics (Basel) 2024; 14:1495. [PMID: 39061632 PMCID: PMC11276137 DOI: 10.3390/diagnostics14141495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 07/03/2024] [Accepted: 07/09/2024] [Indexed: 07/28/2024] Open
Abstract
(1) Background: Dyslipidaemia and insulin resistance are major risk factors for coronary artery disease (CAD). This study investigated the relationship between plasma atherogenic index (PA-I), triglyceride-glucose index (TGI) and other lipid ratios with the presence and prediction of CAD among different age categories. (2) Methods: The study included 223 participants diagnosed with CAD and those with normal coronary arteries (normal group) by coronary computed tomography angiography (CCTA). Participants were categorised by age and sex: premature CAD (PCAD) for men under 55 and women under 65, and older groups as elderly. (3) Results: PA-I, Lipid Combined Index, Castelli Risk Indices, and TGI were significantly higher in the PCAD group compared to the control group (p < 0.05). ROC analysis showed that a PA-I cut-off of 0.41 had a sensitivity of 62% and a specificity of 58% for predicting PCAD, while a TGI cut-off of 8.74 had a sensitivity of 68% and a specificity of 62%. In the elderly, no significant differences in these indices were found between the CAD and normal groups. (4) Conclusions: Traditional lipid profiles and non-traditional lipid indices such as PA-I and TGI show significant differences in predicting CAD in younger populations but not in older groups. TGI and PA-I may be promising biomarkers for the prediction of PAD, although further validation is needed.
Collapse
Affiliation(s)
- Taha Okan
- Kardiya Medical Center, 35000 Izmir, Turkey
| | - Mehmet Doruk
- Izmir Endocrinology Clinic, 35500 Izmir, Turkey;
| | - Ali Ozturk
- Department of Cardiology, Ozel Saglik International Hospital, 35000 Izmir, Turkey;
| | - Caner Topaloglu
- Department of Cardiology, Faculty of Medicine, Izmir Economy University, 35550 Izmir, Turkey; (C.T.); (M.D.)
| | - Mustafa Dogdus
- Department of Cardiology, Faculty of Medicine, Izmir Economy University, 35550 Izmir, Turkey; (C.T.); (M.D.)
| | - Mehmet Birhan Yilmaz
- Department of Cardiology, Faculty of Medicine, Dokuz Eylul University, 35220 Izmir, Turkey;
| |
Collapse
|
7
|
Sarraju A, Nissen SE. Atherosclerotic plaque stabilization and regression: a review of clinical evidence. Nat Rev Cardiol 2024; 21:487-497. [PMID: 38177454 DOI: 10.1038/s41569-023-00979-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/01/2023] [Indexed: 01/06/2024]
Abstract
Atherosclerotic plaque results from a complex interplay between lipid deposition, inflammatory changes, cell migration and arterial wall injury. Over the past two decades, clinical trials utilizing invasive arterial imaging modalities, such as intravascular ultrasonography, have shown that reducing levels of atherogenic lipoproteins, mainly serum LDL-cholesterol (LDL-C), to very low levels can safely reduce overall atherosclerotic plaque burden and favourably modify plaque composition. Classically, this outcome has been achieved with intensive statin therapy. Since 2016, newer and potent lipid-lowering strategies, such as proprotein convertase subtilisin-kexin type 9 inhibition, have shown incremental effects on plaque regression and risk of clinical events. Despite maximal reduction in plasma LDL-C levels, considerable residual cardiovascular risk remains in some patients. Therefore, there is a need to study therapeutic approaches that address residual risk beyond LDL-C reduction to promote plaque stabilization or regression. Contemporary imaging modalities, such as coronary computed tomography angiography, enable non-invasive assessment of the overall atherosclerotic plaque burden as well as of certain local plaque characteristics. This technology could allow further study of plaque stabilization and regression using novel therapeutic approaches. Non-invasive plaque assessment might also offer the potential to guide personalized management strategies if validated for this purpose.
Collapse
Affiliation(s)
- Ashish Sarraju
- Department of Cardiovascular Medicine, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Steven E Nissen
- Department of Cardiovascular Medicine, Cleveland Clinic Foundation, Cleveland, OH, USA.
| |
Collapse
|
8
|
Kanda D, Tokushige A, Ohishi M. Association between Estimated Small Dense Low-Density Lipoprotein Cholesterol and Occurrence of New Lesions after Percutaneous Coronary Intervention in Japanese Patients with Stable Angina and Receiving Statin Therapy. Rev Cardiovasc Med 2024; 25:218. [PMID: 39076334 PMCID: PMC11270076 DOI: 10.31083/j.rcm2506218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 03/27/2024] [Accepted: 03/29/2024] [Indexed: 07/31/2024] Open
Abstract
Background Low-density lipoprotein cholesterol (LDL-C) is considered the most important risk factor for coronary artery disease (CAD). Although lipid-lowering therapy using high-intensity statins for patients with stable CAD is one of the cornerstones of medication therapy, there is still a risk of residual cardiovascular events, even after controlling for LDL-C. Recently, attention has focused on the association between small dense LDL-C as a residual risk factor for CAD, and it has been reported that a formula can be used to calculate the small LDL-C. Methods We investigated the association between estimated small dense LDL-C (Esd LDL-C) and the occurrence of new lesions with myocardial ischemia ≤ 2 years after percutaneous coronary intervention (PCI) in 537 patients with stable angina who underwent PCI. In this study, all patients had been prescribed statins. This study was based on previously reported data regarding the relationship between non-high-density lipoprotein cholesterol levels and stable angina pectoris after PCI. Results Revascularization, including new lesions and in-stent restenosis, and new lesions appeared in 130 and 90 patients, respectively, ≤ 2 years after PCI. Age, diabetes mellitus (DM), LDL-C, and Esd LDL-C were associated with the occurrence of revascularization and new lesions ≤ 2 years after PCI. Multivariate logistic regression analysis models revealed that Esd LDL-C [odds ratio (OR) 1.03, 95% confidence interval (CI) 1.004-1.048, p = 0.020; and OR 1.03, 95% CI 1.009-1.057, p = 0.007, respectively] were associated with the revascularization and occurrence of new lesions ≤ 2 years after PCI. Conclusions As well as total cholesterol and LDL-C, Esd LDL-C was an independent risk factor for the revascularization and occurrence of new lesions ≤ 2 years after PCI for stable angina in Japanese patients receiving statin therapy. In patients with stable angina who are on lipid-lowering therapy with statins, calculating the Esd LDL-C may provide useful information for predicting revascularization and the occurrence of new lesions.
Collapse
Affiliation(s)
- Daisuke Kanda
- Department of Cardiovascular Medicine and Hypertension, Graduate School of Medical and Dental Sciences, Kagoshima University, 890‑8520 Kagoshima, Japan
| | - Akihiro Tokushige
- Department of Cardiovascular Medicine and Hypertension, Graduate School of Medical and Dental Sciences, Kagoshima University, 890‑8520 Kagoshima, Japan
| | - Mitsuru Ohishi
- Department of Cardiovascular Medicine and Hypertension, Graduate School of Medical and Dental Sciences, Kagoshima University, 890‑8520 Kagoshima, Japan
| |
Collapse
|
9
|
Singh A, Bruemmer D. Cardiometabolic Risk: Shifting the Paradigm Toward Comprehensive Assessment. JACC. ADVANCES 2024; 3:100867. [PMID: 38939673 PMCID: PMC11198385 DOI: 10.1016/j.jacadv.2024.100867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/29/2024]
Affiliation(s)
- Abhayjit Singh
- Robert and Suzanne Tomsich Department of Cardiovascular Medicine, Section of Preventive Cardiology and Rehabilitation, Heart, Vascular and Thoracic Institute, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| | - Dennis Bruemmer
- Robert and Suzanne Tomsich Department of Cardiovascular Medicine, Section of Preventive Cardiology and Rehabilitation, Heart, Vascular and Thoracic Institute, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| |
Collapse
|
10
|
Nazari I, Feinstein MJ. Evolving mechanisms and presentations of cardiovascular disease in people with HIV: implications for management. Clin Microbiol Rev 2024; 37:e0009822. [PMID: 38299802 PMCID: PMC10938901 DOI: 10.1128/cmr.00098-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2024] Open
Abstract
People with HIV (PWH) are at elevated risk for cardiovascular diseases (CVDs), including myocardial infarction, heart failure, and sudden cardiac death, among other CVD manifestations. Chronic immune dysregulation resulting in persistent inflammation is common among PWH, particularly those with sustained viremia and impaired CD4+ T cell recovery. This inflammatory milieu is a major contributor to CVDs among PWH, in concert with common comorbidities (such as dyslipidemia and smoking) and, to a lesser extent, off-target effects of antiretroviral therapy. In this review, we discuss the clinical and mechanistic evidence surrounding heightened CVD risks among PWH, implications for specific CVD manifestations, and practical guidance for management in the setting of evolving data.
Collapse
Affiliation(s)
- Ilana Nazari
- Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Matthew J. Feinstein
- Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Division of Cardiology in the Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| |
Collapse
|
11
|
Xue Y, Ju J, Zhao W, Ma X, Li S, Zhao Y, Wang X, Liu X, Li J, Yan P, Sun Q. Association between atherogenic index of plasma and asymptomatic intracranial arterial stenosis in middle-aged and elderly women: A cross-sectional study in Shandong, China. Nutr Metab Cardiovasc Dis 2024; 34:598-605. [PMID: 38000995 DOI: 10.1016/j.numecd.2023.09.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 08/31/2023] [Accepted: 09/26/2023] [Indexed: 11/26/2023]
Abstract
BACKGROUND AND AIMS The atherogenic index of plasma (AIP) is associated with progression of atherosclerosis and used to describe how pro- or anti-atherogenic components are balanced. However, the association of AIP with asymptomatic intracranial arterial stenosis (aICAS) is uncertain. The purpose of this study is to investigate the association between AIP and aICAS in rural China. METHODS AND RESULTS A total of 1990 participants aged ≥40 years free of stroke or transient ischemic attack were enrolled in this study. The presence of aICAS was examined by Transcranial Doppler ultrasound and confirmed by magnetic resonance angiography. The adjusted AIP (aAIP) was calculated according to the ratio of TG and HDL-C and further separated into 4 quartiles. Multiple logistic regression was used to investigate the association between aAIP and aICAS, and the dose-response relationship was explored by restricted cubic spline. After adjusting for conventional confounders, aAIP was significantly higher in the aICAS group than that in the non-aICAS group. Furthermore, the common odds ratios for aICAS risk increased with increasing aAIP quartiles. Multivariate logistic regression revealed that aAIP was independently associated with aICAS in female or middle-aged and elderly (age ≥50 years), and superior to other lipid profiles. Multiple-adjusted spline regression showed the dose-response association between aAIP levels and aICAS prevalence. CONCLUSIONS AIP might be independently and positively associated with the prevalence of aICAS in middle-aged and elderly women, which might be superior to traditional and nontraditional lipid profiles in rural China.
Collapse
Affiliation(s)
- Yuan Xue
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Jiachen Ju
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Weihua Zhao
- Department of Neurology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
| | - Xiaotong Ma
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Shan Li
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Yuanyuan Zhao
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Xiang Wang
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Xiaohui Liu
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Jifeng Li
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Peng Yan
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China.
| | - Qinjian Sun
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China.
| |
Collapse
|
12
|
Sánchez-Cabo F, Fuster V, Lara-Pezzi E. Inflammation contributes to the pathogenic effects of subclinical atherosclerosis. Eur Heart J 2024; 45:313. [PMID: 37936251 DOI: 10.1093/eurheartj/ehad701] [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: 11/09/2023] Open
Affiliation(s)
- Fátima Sánchez-Cabo
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernandez Almagro, 3, 28029 Madrid, Spain
- Centro de Investigacion Biomedica en Red en Enfermedades Cardiovasculares (CIBERCV), Spain
| | - Valentín Fuster
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernandez Almagro, 3, 28029 Madrid, Spain
- The Zena and Michael A. Wiener Cardiovascular Institute/Marie-Josée and Henry R. Kravis Center for Cardiovascular Health, Mount Sinai School of Medicine, One Gustave L. Levy. Place, New York, NY 10029, USA
| | - Enrique Lara-Pezzi
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernandez Almagro, 3, 28029 Madrid, Spain
- Centro de Investigacion Biomedica en Red en Enfermedades Cardiovasculares (CIBERCV), Spain
| |
Collapse
|
13
|
Boutagy NE, Gamez-Mendez A, Fowler JW, Zhang H, Chaube BK, Esplugues E, Kuo A, Lee S, Horikami D, Zhang J, Citrin KM, Singh AK, Coon BG, Lee MY, Suarez Y, Fernandez-Hernando C, Sessa WC. Dynamic metabolism of endothelial triglycerides protects against atherosclerosis in mice. J Clin Invest 2024; 134:e170453. [PMID: 38175710 PMCID: PMC10866653 DOI: 10.1172/jci170453] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 12/20/2023] [Indexed: 01/05/2024] Open
Abstract
Blood vessels are continually exposed to circulating lipids, and elevation of ApoB-containing lipoproteins causes atherosclerosis. Lipoprotein metabolism is highly regulated by lipolysis, largely at the level of the capillary endothelium lining metabolically active tissues. How large blood vessels, the site of atherosclerotic vascular disease, regulate the flux of fatty acids (FAs) into triglyceride-rich (TG-rich) lipid droplets (LDs) is not known. In this study, we showed that deletion of the enzyme adipose TG lipase (ATGL) in the endothelium led to neutral lipid accumulation in vessels and impaired endothelial-dependent vascular tone and nitric oxide synthesis to promote endothelial dysfunction. Mechanistically, the loss of ATGL led to endoplasmic reticulum stress-induced inflammation in the endothelium. Consistent with this mechanism, deletion of endothelial ATGL markedly increased lesion size in a model of atherosclerosis. Together, these data demonstrate that the dynamics of FA flux through LD affects endothelial cell homeostasis and consequently large vessel function during normal physiology and in a chronic disease state.
Collapse
Affiliation(s)
- Nabil E. Boutagy
- Department of Pharmacology
- Vascular Biology and Therapeutics Program, and
| | - Ana Gamez-Mendez
- Department of Pharmacology
- Vascular Biology and Therapeutics Program, and
| | - Joseph W.M. Fowler
- Department of Pharmacology
- Vascular Biology and Therapeutics Program, and
| | - Hanming Zhang
- Vascular Biology and Therapeutics Program, and
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Bal K. Chaube
- Vascular Biology and Therapeutics Program, and
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Enric Esplugues
- Vascular Biology and Therapeutics Program, and
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Andrew Kuo
- Vascular Biology Program, Department of Surgery, Boston Children’s Hospital, Boston, Massachusetts, USA
| | - Sungwoon Lee
- Department of Pharmacology
- Vascular Biology and Therapeutics Program, and
| | - Daiki Horikami
- Department of Pharmacology
- Vascular Biology and Therapeutics Program, and
| | - Jiasheng Zhang
- Department of Cardiology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Kathryn M. Citrin
- Vascular Biology and Therapeutics Program, and
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Abhishek K. Singh
- Department of Pharmacology
- Vascular Biology and Therapeutics Program, and
| | - Brian G. Coon
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Monica Y. Lee
- Department of Physiology and Biophysics, Center for Cardiovascular Research, University of Illinois at Chicago School of Medicine, Chicago, Illinois, USA
| | - Yajaira Suarez
- Vascular Biology and Therapeutics Program, and
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Carlos Fernandez-Hernando
- Vascular Biology and Therapeutics Program, and
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - William C. Sessa
- Department of Pharmacology
- Vascular Biology and Therapeutics Program, and
- Department of Cardiology, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
14
|
Norata GD, Tokgözoğlu L. Oral strategies to target proprotein convertase subtilisin/kexin type 9 and lipoprotein(a): the new frontier of lipid lowering. Eur Heart J 2023; 44:5018-5020. [PMID: 37947270 DOI: 10.1093/eurheartj/ehad682] [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: 11/12/2023] Open
Affiliation(s)
- Giuseppe Danilo Norata
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Lale Tokgözoğlu
- Department of Cardiology, Hacettepe University School of Medicine, 06100 Ankara, Turkey
| |
Collapse
|
15
|
Delialis D, Georgiopoulos G, Aivalioti E, Konstantaki C, Oikonomou E, Bampatsias D, Mavraganis G, Vardavas C, Liberopoulos E, Stellos K, Stamatelopoulos K. Remnant cholesterol in atherosclerotic cardiovascular disease: A systematic review and meta-analysis. Hellenic J Cardiol 2023; 74:48-57. [PMID: 37116829 DOI: 10.1016/j.hjc.2023.04.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 04/20/2023] [Accepted: 04/24/2023] [Indexed: 04/30/2023] Open
Abstract
BACKGROUND Accumulating evidence suggests a substantial contribution of remnant cholesterol (RC) to residual risk for the development or relapse of atherosclerotic cardiovascular disease (ASCVD). We aimed to evaluate the association of RC levels with ASCVD risk by different risk categories and methods of RC assessment. We also assessed available evidence of the effects of lipid-lowering therapies (LLTs) on RC levels. METHODS English-language searches of Medline, PubMed, and Embase (inception to 31 January 2023); ClinicalTrials.gov (October 2022); and reference lists of studies and reviews. Studies reporting on the risk of the composite endpoint [all-cause mortality, cardiovascular mortality, and major adverse cardiac events (MACE)] by RC levels were included. Moreover, we searched for studies reporting differences in RC levels after the administration of LLT(s). RESULTS Among n = 29 studies with 257,387 participants, we found a pooled linear (pooled HR: 1.27 per 1-SD increase, 95% CI: 1.12-1.43, P < 0.001, I2 = 95%, n = 15 studies) and non-linear association (pooled HR: 1.59 per quartile increase, 95% CI: 1.35-1.85, P < 0.001, I2 = 87.9%, n = 15 studies) of RC levels and the risk of M ACE both in patients with and without established ASCVD. Interestingly, the risk of MACE was higher in studies with directly measured vs. calculated RC levels. In a limited number of studies and participants, LLTs reduced RC levels. CONCLUSION RC levels are associated with ASCVD risk both in primary and secondary prevention. Directly measured RC levels are associated with ASCVD risk more evidently. Available LLTs tend to decrease RC levels, although the clinical relevance of RC decrease merits further investigation. PROSPERO REGISTRATION CRD42022371346.
Collapse
Affiliation(s)
- Dimitrios Delialis
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Georgios Georgiopoulos
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens Medical School, Athens, Greece; School of Biomedical Engineering and Imaging Sciences, King's College, London, UK
| | - Evmorfia Aivalioti
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Christina Konstantaki
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Ermioni Oikonomou
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Dimitrios Bampatsias
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Georgios Mavraganis
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Constantine Vardavas
- Department of Social Medicine, Faculty of Medicine, University of Crete, University Campus of Voutes, 700 13, Heraklion, Crete, Greece; Center for Global Tobacco Control, Department of Society, Human Development and Health, Harvard School of Public Health, 677 Huntington Avenue, Boston, MA 02115, USA
| | - Evangelos Liberopoulos
- 1(st) Department of Propedeutic Medicine, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Konstantinos Stellos
- Department of Cardiovascular Research, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Kimon Stamatelopoulos
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens Medical School, Athens, Greece; Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK.
| |
Collapse
|
16
|
Reijnders E, van der Laarse A, Jukema JW, Cobbaert CM. High residual cardiovascular risk after lipid-lowering: prime time for Predictive, Preventive, Personalized, Participatory, and Psycho-cognitive medicine. Front Cardiovasc Med 2023; 10:1264319. [PMID: 37908502 PMCID: PMC10613690 DOI: 10.3389/fcvm.2023.1264319] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 10/03/2023] [Indexed: 11/02/2023] Open
Abstract
As time has come to translate trial results into individualized medical diagnosis and therapy, we analyzed how to minimize residual risk of cardiovascular disease (CVD) by reviewing papers on "residual cardiovascular disease risk". During this review process we found 989 papers that started off with residual CVD risk after initiating statin therapy, continued with papers on residual CVD risk after initiating therapy to increase high-density lipoprotein-cholesterol (HDL-C), followed by papers on residual CVD risk after initiating therapy to decrease triglyceride (TG) levels. Later on, papers dealing with elevated levels of lipoprotein remnants and lipoprotein(a) [Lp(a)] reported new risk factors of residual CVD risk. And as new risk factors are being discovered and new therapies are being tested, residual CVD risk will be reduced further. As we move from CVD risk reduction to improvement of patient management, a paradigm shift from a reductionistic approach towards a holistic approach is required. To that purpose, a personalized treatment dependent on the individual's CVD risk factors including lipid profile abnormalities should be configured, along the line of P5 medicine for each individual patient, i.e., with Predictive, Preventive, Personalized, Participatory, and Psycho-cognitive approaches.
Collapse
Affiliation(s)
- E. Reijnders
- Department of Clinical Chemistry and Laboratory Medicine, Leiden University Medical Center, Leiden, Netherlands
| | - A. van der Laarse
- Department of Clinical Chemistry and Laboratory Medicine, Leiden University Medical Center, Leiden, Netherlands
| | - J. W. Jukema
- Department of Cardiology, Leiden University Medical Center, Leiden, Netherlands
- Netherlands Heart Institute, Utrecht, Netherlands
| | - C. M. Cobbaert
- Department of Clinical Chemistry and Laboratory Medicine, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
17
|
Wei D, Melgarejo JD, Van Aelst L, Vanassche T, Verhamme P, Janssens S, Peter K, Zhang ZY. Prediction of coronary artery disease using urinary proteomics. Eur J Prev Cardiol 2023; 30:1537-1546. [PMID: 36943304 DOI: 10.1093/eurjpc/zwad087] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 03/13/2023] [Accepted: 03/20/2023] [Indexed: 03/23/2023]
Abstract
AIMS Coronary artery disease (CAD) is multifactorial, caused by complex pathophysiology, and contributes to a high burden of mortality worldwide. Urinary proteomic analyses may help to identify predictive biomarkers and provide insights into the pathogenesis of CAD. METHODS AND RESULTS Urinary proteome was analysed in 965 participants using capillary electrophoresis coupled with mass spectrometry. A proteomic classifier was developed in a discovery cohort with 36 individuals with CAD and 36 matched controls using the support vector machine. The classifier was tested in a validation cohort with 115 individuals who progressed to CAD and 778 controls and compared with two previously developed CAD-associated classifiers, CAD238 and ACSP75. The Framingham and SCORE2 risk scores were available in 737 participants. Bioinformatic analysis was performed based on the CAD-associated peptides. The novel proteomic classifier was comprised of 160 urinary peptides, mainly related to collagen turnover, lipid metabolism, and inflammation. In the validation cohort, the classifier provided an area under the receiver operating characteristic curve (AUC) of 0.82 [95% confidence interval (CI): 0.78-0.87] for the CAD prediction in 8 years, superior to CAD238 (AUC: 0.71, 95% CI: 0.66-0.77) and ACSP75 (AUC: 0.53 and 95% CI: 0.47-0.60). On top of CAD238 and ACSP75, the addition of the novel classifier improved the AUC to 0.84 (95% CI: 0.80-0.89). In a multivariable Cox model, a 1-SD increment in the novel classifier was associated with a higher risk of CAD (HR: 1.54, 95% CI: 1.26-1.89, P < 0.0001). The new classifier further improved the risk reclassification of CAD on top of the Framingham or SCORE2 risk scores (net reclassification index: 0.61, 95% CI: 0.25-0.95, P = 0.001; 0.64, 95% CI: 0.28-0.98, P = 0.001, correspondingly). CONCLUSION A novel urinary proteomic classifier related to collagen metabolism, lipids, and inflammation showed potential for the risk prediction of CAD. Urinary proteome provides an alternative approach to personalized prevention.
Collapse
Affiliation(s)
- Dongmei Wei
- Studies Coordinating Centre, Research Unit Hypertension and Cardiovascular Epidemiology, KU Leuven Department of Cardiovascular Sciences, University of Leuven, Campus Sint Rafaël, Kapucijnenvoer 7, Box 7001, BE-3000 Leuven, Belgium
| | - Jesus D Melgarejo
- Studies Coordinating Centre, Research Unit Hypertension and Cardiovascular Epidemiology, KU Leuven Department of Cardiovascular Sciences, University of Leuven, Campus Sint Rafaël, Kapucijnenvoer 7, Box 7001, BE-3000 Leuven, Belgium
| | - Lucas Van Aelst
- Division of Cardiology, University Hospitals Leuven, University of Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Thomas Vanassche
- Division of Cardiology, University Hospitals Leuven, University of Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Peter Verhamme
- Division of Cardiology, University Hospitals Leuven, University of Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Stefan Janssens
- Division of Cardiology, University Hospitals Leuven, University of Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Karlheinz Peter
- Baker Heart and Diabetes Institute, 75 Commercial Rd, Melbourne VIC 3004, Australia
- Department of Cardiology, The Alfred Hospital, 55 Commercial Rd, Melbourne VIC 3004, Australia
| | - Zhen-Yu Zhang
- Studies Coordinating Centre, Research Unit Hypertension and Cardiovascular Epidemiology, KU Leuven Department of Cardiovascular Sciences, University of Leuven, Campus Sint Rafaël, Kapucijnenvoer 7, Box 7001, BE-3000 Leuven, Belgium
| |
Collapse
|
18
|
Sakamoto A, Suwa K, Kawakami R, Finn AV, Maekawa Y, Virmani R, Finn AV. Significance of Intra-plaque Hemorrhage for the Development of High-Risk Vulnerable Plaque: Current Understanding from Basic to Clinical Points of View. Int J Mol Sci 2023; 24:13298. [PMID: 37686106 PMCID: PMC10487895 DOI: 10.3390/ijms241713298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 08/18/2023] [Accepted: 08/22/2023] [Indexed: 09/10/2023] Open
Abstract
Acute coronary syndromes due to atherosclerotic coronary artery disease are a leading cause of morbidity and mortality worldwide. Intra-plaque hemorrhage (IPH), caused by disruption of intra-plaque leaky microvessels, is one of the major contributors of plaque progression, causing a sudden increase in plaque volume and eventually plaque destabilization. IPH and its healing processes are highly complex biological events that involve interactions between multiple types of cells in the plaque, including erythrocyte, macrophages, vascular endothelial cells and vascular smooth muscle cells. Recent investigations have unveiled detailed molecular mechanisms by which IPH leads the development of high-risk "vulnerable" plaque. Current advances in clinical diagnostic imaging modalities, such as magnetic resonance image and intra-coronary optical coherence tomography, increasingly allow us to identify IPH in vivo. To date, retrospective and prospective clinical trials have revealed the significance of IPH as detected by various imaging modalities as a reliable prognostic indicator of high-risk plaque. In this review article, we discuss recent advances in our understanding for the significance of IPH on the development of high-risk plaque from basic to clinical points of view.
Collapse
Affiliation(s)
- Atsushi Sakamoto
- CVPath Institute, Inc., Gaithersburg, MD 20878, USA; (A.S.); (R.K.); (A.V.F.); (R.V.)
- Division of Cardiology, Internal Medicine III, Hamamatsu University School of Medicine, Hamamatsu 431-3125, Japan; (K.S.); (Y.M.)
| | - Kenichiro Suwa
- Division of Cardiology, Internal Medicine III, Hamamatsu University School of Medicine, Hamamatsu 431-3125, Japan; (K.S.); (Y.M.)
| | - Rika Kawakami
- CVPath Institute, Inc., Gaithersburg, MD 20878, USA; (A.S.); (R.K.); (A.V.F.); (R.V.)
| | - Alexandra V. Finn
- CVPath Institute, Inc., Gaithersburg, MD 20878, USA; (A.S.); (R.K.); (A.V.F.); (R.V.)
| | - Yuichiro Maekawa
- Division of Cardiology, Internal Medicine III, Hamamatsu University School of Medicine, Hamamatsu 431-3125, Japan; (K.S.); (Y.M.)
| | - Renu Virmani
- CVPath Institute, Inc., Gaithersburg, MD 20878, USA; (A.S.); (R.K.); (A.V.F.); (R.V.)
| | - Aloke V. Finn
- CVPath Institute, Inc., Gaithersburg, MD 20878, USA; (A.S.); (R.K.); (A.V.F.); (R.V.)
| |
Collapse
|
19
|
Skoumas I, Andrikou I, Grigoriou K, Dima I, Lazarou E, Vlachopoulos C, Tsioufis K. Lipoprotein(a), metabolic profile and new-onset type 2 diabetes in patients with familial combined hyperlipidemia: A 9 year follow-up study. J Clin Lipidol 2023; 17:512-518. [PMID: 37321915 DOI: 10.1016/j.jacl.2023.05.103] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 05/17/2023] [Accepted: 05/29/2023] [Indexed: 06/17/2023]
Abstract
BACKGROUND Lipoprotein(a) [Lp(a)] appears to have an inverse association with the risk of type 2 diabetes mellitus in the general population. OBJECTIVE This study aimed to investigate the prognostic role of Lp(a) regarding the development of type 2 diabetes in the special population of subjects with familial combined hyperlipidemia (FCH). METHODS This cohort study included 474 patients (mean age 49.7±11.3 years, 64% males) with FCH, without diabetes at baseline who were followed for a mean period of 8.2±6.8 years. At baseline evaluation venous blood samples were obtained for the determination of lipid profile and Lp(a) levels. The endpoint of interest was the development of diabetes. RESULTS Patients with increased Lp(a) levels ≥30 mg/dl compared to those with low Lp(a) levels <30 mg/dl had lower levels of triglycerides (238±113 vs 268±129 mg/dl, p = 0.01), greater levels of high-density lipoprotein (HDL) cholesterol (44±10 vs 41±10 mg/dl, p = 0.01) and hypertension in a greater percentage (42% vs 32%, p = 0.03). The incidence of new-onset diabetes during the follow-up period was 10.1% (n = 48). Multiple Cox regression analysis revealed that increased Lp(a) is an independent predictor of lower diabetes incidence (HR 0.39, 95% CI 0.17-0.90, p = 0.02) after adjustment for confounders. CONCLUSION Among subjects with FCH those with higher Lp(a) levels have lower risk for the development of type 2 diabetes. Moreover, the presence of increased Lp(a) seems to differentiate the expression of metabolic syndrome characteristics in patients with FCH, as increased Lp(a) is related to lower levels of triglycerides, greater prevalence of hypertension and higher levels of HDL cholesterol.
Collapse
Affiliation(s)
- Ioannis Skoumas
- First Department of Cardiology, Hippokration Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Ioannis Andrikou
- First Department of Cardiology, Hippokration Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece.
| | - Kalliopi Grigoriou
- First Department of Cardiology, Hippokration Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Ioanna Dima
- First Department of Cardiology, Hippokration Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Emilia Lazarou
- First Department of Cardiology, Hippokration Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Charalambos Vlachopoulos
- First Department of Cardiology, Hippokration Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Konstantinos Tsioufis
- First Department of Cardiology, Hippokration Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
20
|
Li Y, Feng Y, Li S, Ma Y, Lin J, Wan J, Zhao M. The atherogenic index of plasma (AIP) is a predictor for the severity of coronary artery disease. Front Cardiovasc Med 2023; 10:1140215. [PMID: 37441702 PMCID: PMC10333749 DOI: 10.3389/fcvm.2023.1140215] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 06/14/2023] [Indexed: 07/15/2023] Open
Abstract
Objective Dyslipidemia is a key risk factor for coronary artery disease (CAD). This study aimed to investigate the correlation between the atherogenic index of plasma (AIP) and the severity of CAD. Methods 2,491 patients were enrolled in this study and analyzed retrospectively, including 665 non-CAD patients as the control group and 1,826 CAD patients. The CAD patients were classified into three subgroups according to tertiles of SYNTAX score (SS). Non-high-density lipoprotein cholesterol (Non-HDL-C) was defined as serum total cholesterol (TC) minus serum high-density lipoprotein cholesterol (Non-HDL-C), atherogenic index (AI) was defined as the ratio of non-HDL-C to HDL-C; AIP was defined as the logarithm of the ratio of the concentration of triglyceride (TG) to HDL-C; lipoprotein combine index (LCI) was defined as the ratio of TC∗TG∗ low-density lipoprotein cholesterol (LDL)to HDL-C; Castelli Risk Index I (CRI I) was defined as the ratio of TC to HDL-C; Castelli Risk Index II (CRI II) was defined as the ratio of LDL-C to HDL-C. Results The levels of AIP (P < 0.001), AI (P < 0.001), and LCI (P = 0.013) were higher in the CAD group compared with the non-CAD group. The Spearman correlation analysis showed that AIP (r = 0.075, P < 0.001), AI (r = 0.132, P < 0.001), and LCI (r = 0.072, P = 0.001) were positively correlated with SS. The multivariate logistic regression model showed CRI I (OR: 1.11, 95% CI: 1.03-1.19, P = 0.005), CRI II (OR: 1.26, 95% CI: 1.15-1.39, P < 0.001), AI (OR: 1.28, 95% CI: 1.17-1.40, P < 0.001), AIP (OR: 2.06, 95% CI: 1.38-3.07, P < 0.001), and LCI (OR: 1.01, 95% CI: 1.01-1.02, P < 0.001) were independent predictors of severity of CAD After adjusting various confounders. Conclusion CRI I, CRI II, AIP, AI, and LCI were independent predictors of the severity of CAD, which could be used as a biomarker for the evaluation of the severity of CAD.
Collapse
Affiliation(s)
- Ya Li
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yujia Feng
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Shu Li
- Department of Intensive Care Unit, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yulin Ma
- Department of Cardiology, Hubei Jianghan Oilfield General Hospital, Qianjiang, China
| | - Jiesheng Lin
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
- Institute for Medical Information Processing, Biometry, and Epidemiology (IBE), Faculty of Medicine, Pettenkofer School of Public Health, LMU Munich, Munich, Germany
| | - Jing Wan
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Min Zhao
- Demonstration Center for Experimental Basic Medicine Education, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| |
Collapse
|
21
|
Sánchez-Cabo F, Fuster V, Silla-Castro JC, González G, Lorenzo-Vivas E, Alvarez R, Callejas S, Benguría A, Gil E, Núñez E, Oliva B, Mendiguren JM, Cortes-Canteli M, Bueno H, Andrés V, Ordovás JM, Fernández-Friera L, Quesada AJ, Garcia JM, Rossello X, Vázquez J, Dopazo A, Fernández-Ortiz A, Ibáñez B, Fuster JJ, Lara-Pezzi E. Subclinical atherosclerosis and accelerated epigenetic age mediated by inflammation: a multi-omics study. Eur Heart J 2023:ehad361. [PMID: 37339167 PMCID: PMC10393076 DOI: 10.1093/eurheartj/ehad361] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 04/28/2023] [Accepted: 05/23/2023] [Indexed: 06/22/2023] Open
Abstract
AIMS Epigenetic age is emerging as a personalized and accurate predictor of biological age. The aim of this article is to assess the association of subclinical atherosclerosis with accelerated epigenetic age and to investigate the underlying mechanisms mediating this association. METHODS AND RESULTS Whole blood methylomics, transcriptomics, and plasma proteomics were obtained for 391 participants of the Progression of Early Subclinical Atherosclerosis study. Epigenetic age was calculated from methylomics data for each participant. Its divergence from chronological age is termed epigenetic age acceleration. Subclinical atherosclerosis burden was estimated by multi-territory 2D/3D vascular ultrasound and by coronary artery calcification. In healthy individuals, the presence, extension, and progression of subclinical atherosclerosis were associated with a significant acceleration of the Grim epigenetic age, a predictor of health and lifespan, regardless of traditional cardiovascular risk factors. Individuals with an accelerated Grim epigenetic age were characterized by an increased systemic inflammation and associated with a score of low-grade, chronic inflammation. Mediation analysis using transcriptomics and proteomics data revealed key pro-inflammatory pathways (IL6, Inflammasome, and IL10) and genes (IL1B, OSM, TLR5, and CD14) mediating the association between subclinical atherosclerosis and epigenetic age acceleration. CONCLUSION The presence, extension, and progression of subclinical atherosclerosis in middle-aged asymptomatic individuals are associated with an acceleration in the Grim epigenetic age. Mediation analysis using transcriptomics and proteomics data suggests a key role of systemic inflammation in this association, reinforcing the relevance of interventions on inflammation to prevent cardiovascular disease.
Collapse
Affiliation(s)
- Fátima Sánchez-Cabo
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3, 28029 Madrid, Spain
- Centro de Investigacion Biomedica en Red en Enfermedades Cardiovasculares (CIBERCV), Spain
| | - Valentín Fuster
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3, 28029 Madrid, Spain
- The Zena and Michael A. Wiener Cardiovascular Institute/Marie-Josée and Henry R. Kravis Center for Cardiovascular Health, Mount Sinai School of Medicine, One Gustave L. Levy. Place, New York, NY 10029, USA
| | - Juan Carlos Silla-Castro
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3, 28029 Madrid, Spain
| | - Gema González
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3, 28029 Madrid, Spain
| | - Erika Lorenzo-Vivas
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3, 28029 Madrid, Spain
| | - Rebeca Alvarez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3, 28029 Madrid, Spain
| | - Sergio Callejas
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3, 28029 Madrid, Spain
| | - Alberto Benguría
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3, 28029 Madrid, Spain
| | - Eduardo Gil
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3, 28029 Madrid, Spain
| | - Estefanía Núñez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3, 28029 Madrid, Spain
| | - Belén Oliva
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3, 28029 Madrid, Spain
| | | | - Marta Cortes-Canteli
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3, 28029 Madrid, Spain
- Cardiology, IIS-Fundación Jiménez Díaz Hospital, Av. de los Reyes Católicos, 2, 28040 Madrid, Spain
- Centro de Investigacion Biomedica en Red en Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Héctor Bueno
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3, 28029 Madrid, Spain
- Cardiology Department, Hospital Universitario 12 de Octubre and Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Avda. de Córdoba, s/n 28041 Madrid, Spain
| | - Vicente Andrés
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3, 28029 Madrid, Spain
- Centro de Investigacion Biomedica en Red en Enfermedades Cardiovasculares (CIBERCV), Spain
| | - Jose María Ordovás
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3, 28029 Madrid, Spain
- Precision Nutrition and Obesity Research Program, IMDEA Food Institute, CEI UAM + CSIC, Carr. de Canto Blanco, nº 8 E, 28049 Madrid, Spain
- U.S. Department of Agriculture Human Nutrition Research Center on Aging, Tufts University, 711 Washington Street, Boston, MA 02111, USA
| | - Leticia Fernández-Friera
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3, 28029 Madrid, Spain
- Centro de Investigacion Biomedica en Red en Enfermedades Cardiovasculares (CIBERCV), Spain
- HM Hospitales-Centro Integral de Enfermedades Cardiovasculares HM CIEC, Av. de Montepríncipe, 25, 28660 Boadilla del Monte, Madrid, Spain
| | - Antonio J Quesada
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3, 28029 Madrid, Spain
| | - Jose Manuel Garcia
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3, 28029 Madrid, Spain
- Hospital Universitario Central de Oviedo, Av. Roma, s/n, 33011 Asturias, Spain
| | - Xavier Rossello
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3, 28029 Madrid, Spain
- Centro de Investigacion Biomedica en Red en Enfermedades Cardiovasculares (CIBERCV), Spain
- Hospital Universitari Son Espases-IDISBA, Carretera de Valldemossa, 79, 07120 Palma de Mallorca, Mallorca, Islas Baleares (Balearic Islands), Spain
| | - Jesús Vázquez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3, 28029 Madrid, Spain
- Centro de Investigacion Biomedica en Red en Enfermedades Cardiovasculares (CIBERCV), Spain
| | - Ana Dopazo
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3, 28029 Madrid, Spain
- Centro de Investigacion Biomedica en Red en Enfermedades Cardiovasculares (CIBERCV), Spain
| | - Antonio Fernández-Ortiz
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3, 28029 Madrid, Spain
- Centro de Investigacion Biomedica en Red en Enfermedades Cardiovasculares (CIBERCV), Spain
- Hospital Clínico San Carlos, Calle del Prof Martín Lagos, S/N, 28040 Madrid, Spain
| | - Borja Ibáñez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3, 28029 Madrid, Spain
- Centro de Investigacion Biomedica en Red en Enfermedades Cardiovasculares (CIBERCV), Spain
- Cardiology, IIS-Fundación Jiménez Díaz Hospital, Av. de los Reyes Católicos, 2, 28040 Madrid, Spain
| | - Jose Javier Fuster
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3, 28029 Madrid, Spain
- Centro de Investigacion Biomedica en Red en Enfermedades Cardiovasculares (CIBERCV), Spain
| | - Enrique Lara-Pezzi
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3, 28029 Madrid, Spain
- Centro de Investigacion Biomedica en Red en Enfermedades Cardiovasculares (CIBERCV), Spain
| |
Collapse
|
22
|
Georgoulis M, Chrysohoou C, Georgousopoulou E, Damigou E, Skoumas I, Pitsavos C, Panagiotakos D. Long-term prognostic value of LDL-C, HDL-C, lp(a) and TG levels on cardiovascular disease incidence, by body weight status, dietary habits and lipid-lowering treatment: the ATTICA epidemiological cohort study (2002-2012). Lipids Health Dis 2022; 21:141. [PMID: 36529737 PMCID: PMC9762061 DOI: 10.1186/s12944-022-01747-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 12/01/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND The link between blood lipids and cardiovascular disease (CVD) is complex. Our aim was to assess the differential effect of blood lipids on CVD risk according to age, sex, body weight, diet quality, use of lipid-lowering drugs and presence of hypercholesterolemia. METHODS In this secondary analysis of the ATTICA prospective cohort study, serum blood lipids, i.e., total cholesterol (TC), low-density lipoprotein cholesterol (LDL-C), high-density lipoprotein cholesterol (HDL-C), triglycerides (TG) and liproprotein(a) [Lp(a)], and sociodemographic, anthropometric, lifestyle and clinical parameters were evaluated at baseline (2001/2002) in 2020 CVD-free men and women. CVD incidence was recorded at the 10-year follow-up (2011/2012). RESULTS All blood lipids assessed were univariately related to CVD risk; however, associations remained significant only for HDL-C and TG in multivariate models adjusted for age, sex, body mass index, smoking, Mediterranean Diet Score, physical activity, presence of hypercholesterolemia, hypertension and diabetes mellitus, use of lipid-lowering drugs, and family history of CVD [RR per 1 mg/dL (95% CI): 0.983 (0.967, 1.000) and 1.002 (1.001, 1.003), respectively]. In stratified analyses, TC and LDL-C predicted CVD risk in younger subjects, normal-weight subjects, and those not on lipid-lowering drugs, while HDL-C and TG were significant predictors in older subjects, those with low adherence to the Mediterranean diet, and hypercholesterolemic subjects; a significant effect on CVD risk was also observed for TG in males, overweight participants and lipid-lowering medication users and for Lp(a) in older subjects and females (all p ≤ 0.050). CONCLUSIONS The impact of blood lipids on CVD risk differs according to several biological, lifestyle and clinical parameters.
Collapse
Affiliation(s)
- Michael Georgoulis
- grid.15823.3d0000 0004 0622 2843Department of Nutrition and Dietetics, School of Health Sciences and Education, Harokopio University of Athens, 17676 Athens, Greece
| | - Christina Chrysohoou
- grid.5216.00000 0001 2155 0800First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Ekavi Georgousopoulou
- grid.15823.3d0000 0004 0622 2843Department of Nutrition and Dietetics, School of Health Sciences and Education, Harokopio University of Athens, 17676 Athens, Greece
| | - Evangelia Damigou
- grid.15823.3d0000 0004 0622 2843Department of Nutrition and Dietetics, School of Health Sciences and Education, Harokopio University of Athens, 17676 Athens, Greece
| | - Ioannis Skoumas
- grid.5216.00000 0001 2155 0800First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Christos Pitsavos
- grid.5216.00000 0001 2155 0800First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Demosthenes Panagiotakos
- grid.15823.3d0000 0004 0622 2843Department of Nutrition and Dietetics, School of Health Sciences and Education, Harokopio University of Athens, 17676 Athens, Greece
| |
Collapse
|
23
|
May L, Bartolo B, Harrison D, Guzik T, Drummond G, Figtree G, Ritchie R, Rye KA, de Haan J. Translating atherosclerosis research from bench to bedside: navigating the barriers for effective preclinical drug discovery. Clin Sci (Lond) 2022; 136:1731-1758. [PMID: 36459456 PMCID: PMC9727216 DOI: 10.1042/cs20210862] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 10/21/2022] [Accepted: 11/04/2022] [Indexed: 08/10/2023]
Abstract
Cardiovascular disease (CVD) remains the leading cause of death worldwide. An ongoing challenge remains the development of novel pharmacotherapies to treat CVD, particularly atherosclerosis. Effective mechanism-informed development and translation of new drugs requires a deep understanding of the known and currently unknown biological mechanisms underpinning atherosclerosis, accompanied by optimization of traditional drug discovery approaches. Current animal models do not precisely recapitulate the pathobiology underpinning human CVD. Accordingly, a fundamental limitation in early-stage drug discovery has been the lack of consensus regarding an appropriate experimental in vivo model that can mimic human atherosclerosis. However, when coupled with a clear understanding of the specific advantages and limitations of the model employed, preclinical animal models remain a crucial component for evaluating pharmacological interventions. Within this perspective, we will provide an overview of the mechanisms and modalities of atherosclerotic drugs, including those in the preclinical and early clinical development stage. Additionally, we highlight recent preclinical models that have improved our understanding of atherosclerosis and associated clinical consequences and propose model adaptations to facilitate the development of new and effective treatments.
Collapse
Affiliation(s)
- Lauren T. May
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | | | - David G. Harrison
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville TN, U.S.A
| | - Tomasz Guzik
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, U.K
- Department of Medicine, Jagiellonian University Medical College, Krakow, Poland
| | - Grant R. Drummond
- Centre for Cardiovascular Biology and Disease Research, Department of Microbiology, Anatomy, Physiology and Pharmacology, La Trobe University, Melbourne, Victoria, Australia
| | - Gemma A. Figtree
- Kolling Research Institute, University of Sydney, Sydney, Australia
- Imaging and Phenotyping Laboratory, Charles Perkins Centre and Faculty of Medicine and Health, University of Sydney, Sydney, Australia
| | - Rebecca H. Ritchie
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Kerry-Anne Rye
- Lipid Research Group, School of Medical Sciences, Faculty of Medicine, University of New South Wales, Sydney 2052, Australia
| | - Judy B. de Haan
- Cardiovascular Inflammation and Redox Biology Lab, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Department of Chemistry and Biotechnology, Swinburne University of Technology, Hawthorn, Victoria 3122, Australia
- Department Cardiometabolic Health, University of Melbourne, Parkville, Victoria 3010, Australia
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, Victoria 3086, Australia
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, Victoria 3004, Australia
| |
Collapse
|
24
|
Kolossváry M, Mayrhofer T, Ferencik M, Karády J, Pagidipati NJ, Shah SH, Nanna MG, Foldyna B, Douglas PS, Hoffmann U, Lu MT. Are risk factors necessary for pretest probability assessment of coronary artery disease? A patient similarity network analysis of the PROMISE trial. J Cardiovasc Comput Tomogr 2022; 16:397-403. [PMID: 35393245 PMCID: PMC9452442 DOI: 10.1016/j.jcct.2022.03.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 03/05/2022] [Accepted: 03/22/2022] [Indexed: 11/26/2022]
Abstract
BACKGROUND Pretest probability (PTP) calculators utilize epidemiological-level findings to provide patient-level risk assessment of obstructive coronary artery disease (CAD). However, their limited accuracies question whether dissimilarities in risk factors necessarily result in differences in CAD. Using patient similarity network (PSN) analyses, we wished to assess the accuracy of risk factors and imaging markers to identify ≥50% luminal narrowing on coronary CT angiography (CCTA) in stable chest-pain patients. METHODS We created four PSNs representing: patient characteristics, risk factors, non-coronary imaging markers and calcium score. We used spectral clustering to group individuals with similar risk profiles. We compared PSNs to a contemporary PTP score incorporating calcium score and risk factors to identify ≥50% luminal narrowing on CCTA in the CT-arm of the PROMISE trial. We also conducted subanalyses in different age and sex groups. RESULTS In 3556 individuals, the calcium score PSN significantly outperformed patient characteristic, risk factor, and non-coronary imaging marker PSNs (AUC: 0.81 vs. 0.57, 0.55, 0.54; respectively, p < 0.001 for all). The calcium score PSN significantly outperformed the contemporary PTP score (AUC: 0.81 vs. 0.78, p < 0.001), and using 0, 1-100 and > 100 cut-offs provided comparable results (AUC: 0.81 vs. 0.81, p = 0.06). Similar results were found in all subanalyses. CONCLUSION Calcium score on its own provides better individualized obstructive CAD prediction than contemporary PTP scores incorporating calcium score and risk factors. Risk factors may not be able to improve the diagnostic accuracy of calcium score to predict ≥50% luminal narrowing on CCTA.
Collapse
Affiliation(s)
- Márton Kolossváry
- Cardiovascular Imaging Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| | - Thomas Mayrhofer
- Cardiovascular Imaging Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; School of Business Studies, Stralsund University of Applied Sciences, Stralsund, Germany
| | - Maros Ferencik
- Cardiovascular Imaging Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Knight Cardiovascular Institute, Oregon Health and Science University, Portland, OR, USA
| | - Júlia Karády
- Cardiovascular Imaging Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Neha J Pagidipati
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC, USA
| | - Svati H Shah
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC, USA
| | - Michael G Nanna
- Section of Cardiovascular Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Borek Foldyna
- Cardiovascular Imaging Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Pamela S Douglas
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC, USA
| | - Udo Hoffmann
- Cardiovascular Imaging Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Michael T Lu
- Cardiovascular Imaging Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
25
|
Pan X, Hussain MM. Bmal1 regulates production of larger lipoproteins by modulating cAMP-responsive element-binding protein H and apolipoprotein AIV. Hepatology 2022; 76:78-93. [PMID: 34626126 PMCID: PMC8993942 DOI: 10.1002/hep.32196] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 09/22/2021] [Accepted: 10/04/2021] [Indexed: 01/05/2023]
Abstract
BACKGROUND AND AIMS High plasma lipid/lipoprotein levels are risk factors for various metabolic diseases. We previously showed that circadian rhythms regulate plasma lipids and deregulation of these rhythms causes hyperlipidemia and atherosclerosis in mice. Here, we show that global and liver-specific brain and muscle aryl hydrocarbon receptor nuclear translocator-like 1 (Bmal1)-deficient mice maintained on a chow or Western diet developed hyperlipidemia, denoted by the presence of higher amounts of triglyceride-rich and apolipoprotein AIV (ApoAIV)-rich larger chylomicron and VLDL due to overproduction. APPROACH AND RESULTS Bmal1 deficiency decreased small heterodimer partner (Shp) and increased microsomal triglyceride transfer protein (MTP), a key protein that facilitates primordial lipoprotein assembly and secretion. Moreover, we show that Bmal1 regulates cAMP-responsive element-binding protein H (Crebh) to modulate ApoAIV expression and the assembly of larger lipoproteins. This is supported by the observation that Crebh-deficient and ApoAIV-deficient mice, along with Bmal1-deficient mice with knockdown of Crebh, had smaller lipoproteins. Further, overexpression of Bmal1 in Crebh-deficient mice had no effect on ApoAIV expression and lipoprotein size. CONCLUSIONS These studies indicate that regulation of ApoAIV and assembly of larger lipoproteins by Bmal1 requires Crebh. Mechanistic studies showed that Bmal1 regulates Crebh expression by two mechanisms. First, Bmal1 interacts with the Crebh promoter to control circadian regulation. Second, Bmal1 increases Rev-erbα expression, and nuclear receptor subfamily 1 group D member 1 (Nr1D1, Rev-erbα) interacts with the Crebh promoter to repress expression. In short, Bmal1 modulates both the synthesis of primordial lipoproteins and their subsequent expansion into larger lipoproteins by regulating two different proteins, MTP and ApoAIV, through two different transcription factors, Shp and Crebh. It is likely that disruptions in circadian mechanisms contribute to hyperlipidemia and that avoiding disruptions in circadian rhythms may limit/prevent hyperlipidemia and atherosclerosis.
Collapse
Affiliation(s)
- Xiaoyue Pan
- Department of Foundations of Medicine, New York University Long Island School of Medicine, Mineola, NY, USA
- Department of Cell Biology, SUNY Downstate Medical Center, Brooklyn, NY, USA
| | - M Mahmood Hussain
- Department of Foundations of Medicine, New York University Long Island School of Medicine, Mineola, NY, USA
- Department of Cell Biology, SUNY Downstate Medical Center, Brooklyn, NY, USA
- VA New York Harbor Healthcare System, Brooklyn, NY, USA
| |
Collapse
|
26
|
Bornfeldt KE. The Remnant Lipoprotein Hypothesis of Diabetes-Associated Cardiovascular Disease. Arterioscler Thromb Vasc Biol 2022; 42:819-830. [PMID: 35616031 DOI: 10.1161/atvbaha.122.317163] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Both type 1 and type 2 diabetes are associated with an increased risk of atherosclerotic cardiovascular disease (CVD). Research based on human-first or bedside-to-bench approaches has provided new insights into likely mechanisms behind this increased risk. Although both forms of diabetes are associated with hyperglycemia, it is becoming increasingly clear that altered lipoprotein metabolism also plays a critical role in predicting CVD risk in people with diabetes. This review examines recent findings indicating that increased levels of circulating remnant lipoproteins could be a missing link between diabetes and CVD. Although CVD risk associated with diabetes is clearly multifactorial in nature, these findings suggest that we should increase efforts in evaluating whether remnant lipoproteins or the proteins that govern their metabolism are biomarkers of incident CVD in people living with diabetes and whether reducing remnant lipoproteins will prevent the increased CVD risk associated with diabetes.
Collapse
Affiliation(s)
- Karin E Bornfeldt
- Department of Medicine, Division of Metabolism, Endocrinology and Nutrition and Department of Laboratory Medicine and Pathology, University of Washington Medicine Diabetes Institute, Seattle
| |
Collapse
|
27
|
Decoding microRNA drivers in Atherosclerosis. Biosci Rep 2022; 42:231479. [PMID: 35758143 PMCID: PMC9289798 DOI: 10.1042/bsr20212355] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 05/17/2022] [Accepted: 06/26/2022] [Indexed: 11/17/2022] Open
Abstract
An estimated 97% of the human genome consists of non-protein-coding sequences. As our understanding of genome regulation improves, this has led to the characterization of a diverse array of non-coding RNAs (ncRNA). Among these, micro-RNAs (miRNAs) belong to the short ncRNA class (22–25 nucleotides in length), with approximately 2500 miRNA genes encoded within the human genome. From a therapeutic perspective, there is interest in exploiting miRNA as biomarkers of disease progression and response to treatments, as well as miRNA mimics/repressors as novel medicines. miRNA have emerged as an important class of RNA master regulators with important roles identified in the pathogenesis of atherosclerotic cardiovascular disease. Atherosclerosis is characterized by a chronic inflammatory build-up, driven largely by low-density lipoprotein cholesterol accumulation within the artery wall and vascular injury, including endothelial dysfunction, leukocyte recruitment and vascular remodelling. Conventional therapy focuses on lifestyle interventions, blood pressure-lowering medications, high-intensity statin therapy and antiplatelet agents. However, a significant proportion of patients remain at increased risk of cardiovascular disease. This continued cardiovascular risk is referred to as residual risk. Hence, a new drug class targeting atherosclerosis could synergise with existing therapies to optimise outcomes. Here, we review our current understanding of the role of ncRNA, with a focus on miRNA, in the development and progression of atherosclerosis, highlighting novel biological mechanisms and therapeutic avenues.
Collapse
|
28
|
de la Cruz-Ares S, Leon-Acuña A, Yubero-Serrano EM, Torres-Peña JD, Arenas-de Larriva AP, Cardelo MP, Rangel-Zuñiga OA, Luque RM, Alcala-Diaz JF, Ordovas JM, Perez-Martinez P, Lopez-Miranda J, Delgado-Lista J. High density lipoprotein subfractions and extent of coronary atherosclerotic lesions: From the cordioprev study. Clin Chim Acta 2022; 533:89-95. [PMID: 35700819 DOI: 10.1016/j.cca.2022.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 06/02/2022] [Accepted: 06/03/2022] [Indexed: 11/03/2022]
Abstract
BACKGROUND AND AIMS The extent of atherosclerotic coronary heart disease (CHD) is associated with its prognosis, thus discovering potential biomarkers related to worse outcomes could prove valuable. The present work aims to investigate whether lipoprotein subfractions are associated with angiographic CHD severity. MATERIALS AND METHODS Patients from the CORDIOPREV study exhibiting coronary lesions in angiography were classified into two groups (single-vessel coronary disease (SVD) or multivessel coronary disease (MVD)). High-throughput nuclear magnetic resonance (NMR) spectroscopy determined lipoprotein subfractions concentration and composition. RESULTS SVD patients showed a higher concentration of medium and small HDL particles compared with MVD patients. For medium HDL, total lipids, phospholipids, total cholesterol, cholesteryl esters and free cholesterol reflected HDL particle concentration, whereas, for small HDL, total lipids, phospholipids, and free cholesterol mirrored lipoprotein particle concentration. Among traditional cardiovascular risk factors, age, hypertension and T2D were independently associated with angiography severity. In multivariate logistic regression models, medium and small HDL particles remained inversely associated with angiography severity (OR 0.77 (95% CI: 0.64-0.91); OR 0.78 (95% CI: 0.67-0.91), respectively) after adjusting with covariates. CONCLUSION In CHD patients mostly on statin treatment, angiography severity is inversely related to small and medium HDL subclasses concentration measured by NMR. These particles are also independent predictors of the presence of MVD, and its use increased the prediction of this entity over traditional risk factors.
Collapse
Affiliation(s)
- Silvia de la Cruz-Ares
- Lipids and Atherosclerosis Unit, Internal Medicine Unit, Reina Sofia University Hospital, 14004 Cordoba, Spain; Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004 Cordoba, Spain; Department of Medical and Surgical Sciences, University of Cordoba, 14004 Cordoba, Spain; CIBER Fisiopatologia de la Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Ana Leon-Acuña
- Lipids and Atherosclerosis Unit, Internal Medicine Unit, Reina Sofia University Hospital, 14004 Cordoba, Spain; Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004 Cordoba, Spain; Department of Medical and Surgical Sciences, University of Cordoba, 14004 Cordoba, Spain; CIBER Fisiopatologia de la Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Elena M Yubero-Serrano
- Lipids and Atherosclerosis Unit, Internal Medicine Unit, Reina Sofia University Hospital, 14004 Cordoba, Spain; Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004 Cordoba, Spain; Department of Medical and Surgical Sciences, University of Cordoba, 14004 Cordoba, Spain; CIBER Fisiopatologia de la Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Jose D Torres-Peña
- Lipids and Atherosclerosis Unit, Internal Medicine Unit, Reina Sofia University Hospital, 14004 Cordoba, Spain; Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004 Cordoba, Spain; Department of Medical and Surgical Sciences, University of Cordoba, 14004 Cordoba, Spain; CIBER Fisiopatologia de la Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Antonio P Arenas-de Larriva
- Lipids and Atherosclerosis Unit, Internal Medicine Unit, Reina Sofia University Hospital, 14004 Cordoba, Spain; Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004 Cordoba, Spain
| | - Magdalena P Cardelo
- Lipids and Atherosclerosis Unit, Internal Medicine Unit, Reina Sofia University Hospital, 14004 Cordoba, Spain; Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004 Cordoba, Spain; Department of Medical and Surgical Sciences, University of Cordoba, 14004 Cordoba, Spain; CIBER Fisiopatologia de la Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Oriol A Rangel-Zuñiga
- Lipids and Atherosclerosis Unit, Internal Medicine Unit, Reina Sofia University Hospital, 14004 Cordoba, Spain; Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004 Cordoba, Spain; Department of Medical and Surgical Sciences, University of Cordoba, 14004 Cordoba, Spain; CIBER Fisiopatologia de la Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Raul M Luque
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004 Cordoba, Spain; CIBER Fisiopatologia de la Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain; Department of Cell Biology, Physiology, and Immunology, University of Cordoba, 14004 Cordoba, Spain
| | - Juan F Alcala-Diaz
- Lipids and Atherosclerosis Unit, Internal Medicine Unit, Reina Sofia University Hospital, 14004 Cordoba, Spain; Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004 Cordoba, Spain; Department of Medical and Surgical Sciences, University of Cordoba, 14004 Cordoba, Spain; CIBER Fisiopatologia de la Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Jose M Ordovas
- Nutrition and Genomics Laboratory, Human Nutrition Research Center of Aging, Tufts University, Boston, MA 02111, USA; IMDEA Food Institute, 28049 Madrid, Spain
| | - Pablo Perez-Martinez
- Lipids and Atherosclerosis Unit, Internal Medicine Unit, Reina Sofia University Hospital, 14004 Cordoba, Spain; Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004 Cordoba, Spain; Department of Medical and Surgical Sciences, University of Cordoba, 14004 Cordoba, Spain; CIBER Fisiopatologia de la Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Jose Lopez-Miranda
- Lipids and Atherosclerosis Unit, Internal Medicine Unit, Reina Sofia University Hospital, 14004 Cordoba, Spain; Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004 Cordoba, Spain; Department of Medical and Surgical Sciences, University of Cordoba, 14004 Cordoba, Spain; CIBER Fisiopatologia de la Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain.
| | - Javier Delgado-Lista
- Lipids and Atherosclerosis Unit, Internal Medicine Unit, Reina Sofia University Hospital, 14004 Cordoba, Spain; Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004 Cordoba, Spain; Department of Medical and Surgical Sciences, University of Cordoba, 14004 Cordoba, Spain; CIBER Fisiopatologia de la Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
29
|
Delialis D, Georgiopoulos G, Aivalioti E, Mavraganis G, Dimopoulou AM, Sianis A, Aggelidakis L, Patras R, Petropoulos I, Ioannou S, Syrigou R, Chatzidou S, Kanakakis I, Stellos K, Stamatelopoulos K. Remnant cholesterol and atherosclerotic disease in high cardiovascular risk patients. Beyond LDL cholesterol and hypolipidemic treatment. Hellenic J Cardiol 2022; 66:26-31. [PMID: 35667617 DOI: 10.1016/j.hjc.2022.05.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Revised: 05/03/2022] [Accepted: 05/29/2022] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND Remnant cholesterol (RC) is an emerging factor contributing to residual risk for the development of atherosclerotic cardiovascular disease (ASCVD). We aimed to investigate the association of RC with ASCVD in high ASCVD risk patients. METHODS RC was calculated in 906 participants (178 low/moderate-risk and 728 high-risk) consecutively recruited from a vascular registry. Subclinical carotid atherosclerosis was assessed by B-mode carotid ultrasonography. Maximal carotid wall thickness (maxWT) and carotid atherosclerotic burden (n≥2 atherosclerotic plaques) were set as the vascular outcomes. An independent cohort of 87 consecutively recruited high-risk patients who were followed for their lipid profile for 3 months, was also analyzed. RESULTS RC was increased in the high-risk group as compared to controls (26±17 vs. 21±11mg/dl, respectively, p<0.001). Increased RC levels were independently associated with increased maxWT and carotid atherosclerotic burden (p<0.05), after adjustment for traditional cardiovascular risk factors (TRF) and ASCVD. RC levels were associated with the presence of flow-limiting ASCVD and coronary artery disease (p<0.05), after adjustment for TRFs. These associations remained significant in those not receiving hypolipidemic treatment and in treated individuals achieving LDL-C<100 mg/dl. In the prospective cohort, there was no significant interaction between change in RC levels and hypolipidemic status, as contrasted to LDL-C levels (p <0.001). CONCLUSION In a high-risk population, RC was associated with subclinical and clinically overt ASCVD particularly in patients with the most adverse lipid phenotype (untreated) or in treated patients with low LDL-related risk profile. These findings support a residual pro-atherosclerotic role of RC in high-risk patients.
Collapse
Affiliation(s)
- Dimitrios Delialis
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Georgios Georgiopoulos
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Evmorfia Aivalioti
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Georgios Mavraganis
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Angeliki-Maria Dimopoulou
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Alexandros Sianis
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Lasthenis Aggelidakis
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Raphael Patras
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Ioannis Petropoulos
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Sofia Ioannou
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Rodanthi Syrigou
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Sofia Chatzidou
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Ioannis Kanakakis
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Konstantinos Stellos
- Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK; Department of Cardiovascular Research, European Center for Angioscience (ECAS), Heidelberg University, Mannheim, Germany; German Centre for Cardiovascular Research (Deutsches Zentrum für Herz-Kreislauf-Forschung, DZHK), Heidelberg/Mannheim Partner Site, Mannheim, Germany
| | - Kimon Stamatelopoulos
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens Medical School, Athens, Greece; Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK.
| |
Collapse
|
30
|
Abstract
Obesity has reached epidemic proportions and is a major contributor to insulin resistance (IR) and type 2 diabetes (T2D). Importantly, IR and T2D substantially increase the risk of cardiovascular (CV) disease. Although there are successful approaches to maintain glycemic control, there continue to be increased CV morbidity and mortality associated with metabolic disease. Therefore, there is an urgent need to understand the cellular and molecular processes that underlie cardiometabolic changes that occur during obesity so that optimal medical therapies can be designed to attenuate or prevent the sequelae of this disease. The vascular endothelium is in constant contact with the circulating milieu; thus, it is not surprising that obesity-driven elevations in lipids, glucose, and proinflammatory mediators induce endothelial dysfunction, vascular inflammation, and vascular remodeling in all segments of the vasculature. As cardiometabolic disease progresses, so do pathological changes in the entire vascular network, which can feed forward to exacerbate disease progression. Recent cellular and molecular data have implicated the vasculature as an initiating and instigating factor in the development of several cardiometabolic diseases. This Review discusses these findings in the context of atherosclerosis, IR and T2D, and heart failure with preserved ejection fraction. In addition, novel strategies to therapeutically target the vasculature to lessen cardiometabolic disease burden are introduced.
Collapse
|
31
|
Li C, Qu L, Matz AJ, Murphy PA, Liu Y, Manichaikul AW, Aguiar D, Rich SS, Herrington DM, Vu D, Johnson WC, Rotter JI, Post WS, Vella AT, Rodriguez-Oquendo A, Zhou B. AtheroSpectrum Reveals Novel Macrophage Foam Cell Gene Signatures Associated With Atherosclerotic Cardiovascular Disease Risk. Circulation 2022; 145:206-218. [PMID: 34913723 PMCID: PMC8766929 DOI: 10.1161/circulationaha.121.054285] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 11/18/2021] [Indexed: 01/21/2023]
Abstract
BACKGROUND Whereas several interventions can effectively lower lipid levels in people at risk for atherosclerotic cardiovascular disease (ASCVD), cardiovascular event risks remain, suggesting an unmet medical need to identify factors contributing to cardiovascular event risk. Monocytes and macrophages play central roles in atherosclerosis, but studies have yet to provide a detailed view of macrophage populations involved in increased ASCVD risk. METHODS A novel macrophage foaming analytics tool, AtheroSpectrum, was developed using 2 quantitative indices depicting lipid metabolism and the inflammatory status of macrophages. A machine learning algorithm was developed to analyze gene expression patterns in the peripheral monocyte transcriptome of MESA participants (Multi-Ethnic Study of Atherosclerosis; set 1; n=911). A list of 30 genes was generated and integrated with traditional risk factors to create an ASCVD risk prediction model (30-gene cardiovascular disease risk score [CR-30]), which was subsequently validated in the remaining MESA participants (set 2; n=228); performance of CR-30 was also tested in 2 independent human atherosclerotic tissue transcriptome data sets (GTEx [Genotype-Tissue Expression] and GSE43292). RESULTS Using single-cell transcriptomic profiles (GSE97310, GSE116240, GSE97941, and FR-FCM-Z23S), AtheroSpectrum detected 2 distinct programs in plaque macrophages-homeostatic foaming and inflammatory pathogenic foaming-the latter of which was positively associated with severity of atherosclerosis in multiple studies. A pool of 2209 pathogenic foaming genes was extracted and screened to select a subset of 30 genes correlated with cardiovascular event in MESA set 1. A cardiovascular disease risk score model (CR-30) was then developed by incorporating this gene set with traditional variables sensitive to cardiovascular event in MESA set 1 after cross-validation generalizability analysis. The performance of CR-30 was then tested in MESA set 2 (P=2.60×10-4; area under the receiver operating characteristic curve, 0.742) and 2 independent data sets (GTEx: P=7.32×10-17; area under the receiver operating characteristic curve, 0.664; GSE43292: P=7.04×10-2; area under the receiver operating characteristic curve, 0.633). Model sensitivity tests confirmed the contribution of the 30-gene panel to the prediction model (likelihood ratio test; df=31, P=0.03). CONCLUSIONS Our novel computational program (AtheroSpectrum) identified a specific gene expression profile associated with inflammatory macrophage foam cells. A subset of 30 genes expressed in circulating monocytes jointly contributed to prediction of symptomatic atherosclerotic vascular disease. Incorporating a pathogenic foaming gene set with known risk factors can significantly strengthen the power to predict ASCVD risk. Our programs may facilitate both mechanistic investigations and development of therapeutic and prognostic strategies for ASCVD risk.
Collapse
Affiliation(s)
- Chuan Li
- Department of Immunology, School of Medicine, University of Connecticut, Farmington, CT
| | - Lili Qu
- Department of Immunology, School of Medicine, University of Connecticut, Farmington, CT
| | - Alyssa J. Matz
- Department of Immunology, School of Medicine, University of Connecticut, Farmington, CT
| | - Patrick A. Murphy
- Center for Vascular Biology, School of Medicine, University of Connecticut, Farmington, CT
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, MD
| | - Yongmei Liu
- Department of Medicine, Divisions of Cardiology and Neurology, Duke University Medical Center, Durham, NC
| | - Ani W. Manichaikul
- Center for Public Health Genomics, Department of Public Health Sciences, University of Virginia, Charlottesville, VA
| | - Derek Aguiar
- Department of Computer Science and Engineering, University of Connecticut, Storrs, CT
| | - Stephen S. Rich
- Center for Public Health Genomics, Department of Public Health Sciences, University of Virginia, Charlottesville, VA
| | - David M Herrington
- Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC
| | - David Vu
- Department of Biostatistics, University of Washington, Seattle, WA
| | - W. Craig Johnson
- Department of Biostatistics, University of Washington, Seattle, WA
| | - Jerome I. Rotter
- Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA
| | - Wendy S. Post
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, MD
| | - Anthony T. Vella
- Department of Immunology, School of Medicine, University of Connecticut, Farmington, CT
- Institute for Systems Genomics, University of Connecticut, Farmington, CT
| | | | - Beiyan Zhou
- Department of Immunology, School of Medicine, University of Connecticut, Farmington, CT
- Institute for Systems Genomics, University of Connecticut, Farmington, CT
| |
Collapse
|
32
|
Buono MF, Slenders L, Wesseling M, Hartman RJG, Monaco C, den Ruijter HM, Pasterkamp G, Mokry M. The changing landscape of the vulnerable plaque: a call for fine-tuning of preclinical models. Vascul Pharmacol 2021; 141:106924. [PMID: 34607015 DOI: 10.1016/j.vph.2021.106924] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 09/08/2021] [Accepted: 09/28/2021] [Indexed: 11/17/2022]
Abstract
For decades, the pathological definition of the vulnerable plaque led to invaluable insights into the mechanisms that underlie myocardial infarction and stroke. Beyond plaque rupture, other mechanisms, such as erosion, may elicit thrombotic events underlining the complexity and diversity of the atherosclerotic disease. Novel insights, based on single-cell transcriptomics and other "omics" methods, provide tremendous opportunities in the ongoing search for cell-specific determinants that will fine-tune the description of the thrombosis prone lesion. It coincides with an increasing awareness that knowledge on lesion characteristics, cell plasticity and clinical presentation of ischemic cardiovascular events have shifted over the past decades. This shift correlates with an observed changes of cell composition towards phenotypical stabilizing of human plaques. These stabilization features and mechanisms are directly mediated by the cells present in plaques and can be mimicked in vitro via primary plaque cells derived from human atherosclerotic tissues. In addition, the rapidly evolving of sequencing technologies identify many candidate genes and molecular mechanisms that may influence the risk of developing an atherosclerotic thrombotic event - which bring the next challenge in sharp focus: how to translate these cell-specific insights into tangible functional and translational discoveries?
Collapse
Affiliation(s)
- Michele F Buono
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, the Netherlands
| | - Lotte Slenders
- Central Diagnostics Laboratory, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Marian Wesseling
- Central Diagnostics Laboratory, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Robin J G Hartman
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, the Netherlands
| | - Claudia Monaco
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, United Kingdom
| | - Hester M den Ruijter
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, the Netherlands
| | - Gerard Pasterkamp
- Central Diagnostics Laboratory, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Michal Mokry
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, the Netherlands; Central Diagnostics Laboratory, University Medical Center Utrecht, Utrecht, the Netherlands.
| |
Collapse
|
33
|
Fahed AC, Jang IK. Plaque erosion and acute coronary syndromes: phenotype, molecular characteristics and future directions. Nat Rev Cardiol 2021; 18:724-734. [PMID: 33953381 DOI: 10.1038/s41569-021-00542-3] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/15/2021] [Indexed: 02/03/2023]
Abstract
Although acute coronary syndromes (ACS) remain one of the leading causes of death, the clinical presentation has changed over the past three decades with a decline in the incidence of ST-segment elevation myocardial infarction (STEMI) and an increase in non-STEMI. This epidemiological shift is at least partially explained by changes in plaque biology as a result of the widespread use of statins. Historically, atherosclerotic plaque rupture of the fibrous cap was thought to be the main culprit in ACS. However, plaque erosion with an intact fibrous cap is now responsible for about one third of ACS and up to two thirds of non-STEMI. Two major research approaches have enabled a better understanding of plaque erosion. First, advanced intravascular imaging has provided opportunities for an 'optical biopsy' and extensive phenotyping of coronary plaques in living patients. Second, basic science experiments have shed light on the unique molecular characteristics of plaque erosion. At present, patients with ACS are still uniformly treated with coronary stents irrespective of the underlying pathobiology. However, pilot studies indicate that patients with plaque erosion might be treated conservatively without coronary stenting. In this Review, we discuss the patient phenotype and the molecular characteristics in atherosclerotic plaque erosion and provide our vision for a potential major shift in the management of patients with plaque erosion.
Collapse
Affiliation(s)
- Akl C Fahed
- Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.,Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Ik-Kyung Jang
- Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA. .,Kyung Hee University, Seoul, South Korea.
| |
Collapse
|
34
|
Noels H, Lehrke M, Vanholder R, Jankowski J. Lipoproteins and fatty acids in chronic kidney disease: molecular and metabolic alterations. Nat Rev Nephrol 2021; 17:528-542. [PMID: 33972752 DOI: 10.1038/s41581-021-00423-5] [Citation(s) in RCA: 78] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/22/2021] [Indexed: 02/06/2023]
Abstract
Chronic kidney disease (CKD) induces modifications in lipid and lipoprotein metabolism and homeostasis. These modifications can promote, modulate and/or accelerate CKD and secondary cardiovascular disease (CVD). Lipid and lipoprotein abnormalities - involving triglyceride-rich lipoproteins, LDL and/or HDL - not only involve changes in concentration but also changes in molecular structure, including protein composition, incorporation of small molecules and post-translational modifications. These alterations modify the function of lipoproteins and can trigger pro-inflammatory and pro-atherogenic processes, as well as oxidative stress. Serum fatty acid levels are also often altered in patients with CKD and lead to changes in fatty acid metabolism - a key process in intracellular energy production - that induce mitochondrial dysfunction and cellular damage. These fatty acid changes might not only have a negative impact on the heart, but also contribute to the progression of kidney damage. The presence of these lipoprotein alterations within a biological environment characterized by increased inflammation and oxidative stress, as well as the competing risk of non-atherosclerotic cardiovascular death as kidney function declines, has important therapeutic implications. Additional research is needed to clarify the pathophysiological link between lipid and lipoprotein modifications, and kidney dysfunction, as well as the genesis and/or progression of CVD in patients with kidney disease.
Collapse
Affiliation(s)
- Heidi Noels
- Institute for Molecular Cardiovascular Research, RWTH Aachen University, University Hospital, Aachen, Germany
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, Netherlands
| | - Michael Lehrke
- Department of Internal Medicine I, RWTH Aachen University, University Hospital, Aachen, Germany
| | - Raymond Vanholder
- Nephrology Section, Department of Internal Medicine and Pediatrics, University Hospital, Ghent, Belgium
| | - Joachim Jankowski
- Institute for Molecular Cardiovascular Research, RWTH Aachen University, University Hospital, Aachen, Germany.
- Department of Pathology, Cardiovascular Research Institute Maastricht, Maastricht University Medical Centre, Maastricht University, Maastricht, Netherlands.
| |
Collapse
|
35
|
Vulnerable atherosclerotic plaque features: findings from coronary imaging. JOURNAL OF GERIATRIC CARDIOLOGY : JGC 2021; 18:577-584. [PMID: 34404993 PMCID: PMC8352771 DOI: 10.11909/j.issn.1671-5411.2021.07.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Pathological studies have suggested that features of vulnerable atherosclerotic plaques likely to progress and lead to acute cardiovascular events have specific characteristics. Given the progress of intravascular coronary imaging technology, some large prospective studies have detected features of vulnerable atherosclerotic plaques using these imaging modalities. However, the rate of cardiovascular events, such as acute coronary syndrome, has been found to be considerably reduced in the limited follow-up period available in the statin era. Additionally, not all disrupted plaques lead to thrombus formation with clinical presentation. If sub-occlusive or occlusive thrombus formation does not occur, a thrombus on a disrupted plaque will organize without any symptoms, forming a “healed plaque”. Although vulnerable plaque detection using intracoronary imaging is focused on “thin-cap fibroatheroma” leading to plaque rupture, superficial plaque erosion is increasingly recognized; however, the underlying mechanism of thrombus formation on eroded plaques is not well understood. One of intravascular imaging, optical coherence tomography (OCT) has the highest image resolution and has enabled detailed characterization of the plaque in vivo. Here, we reviewed the status and limitations of intravascular imaging in terms of detecting vulnerable plaque through mainly OCT studies. We suggested that vulnerable plaque should be reconsidered in terms of eroded plaque and healed plaque and that both plaque and circulating blood should be assessed in greater detail accordingly.
Collapse
|
36
|
Huang H, Yu X, Li L, Shi G, Li F, Xiao J, Yun Z, Cai G. Atherogenic index of plasma is related to coronary atherosclerotic disease in elderly individuals: a cross-sectional study. Lipids Health Dis 2021; 20:68. [PMID: 34247637 PMCID: PMC8273949 DOI: 10.1186/s12944-021-01496-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 06/29/2021] [Indexed: 02/06/2023] Open
Abstract
Background Dyslipidaemia plays an important role in coronary atherosclerotic disease (CAD). The relationship between the atherogenic index of plasma (AIP) and CAD in elderly individuals was explored in this study. Methods Elderly individuals (age ≥ 65 years) who underwent coronary angiography from January 2016 to October 2020 were consecutively enrolled in the study. Results A total of 1313 individuals, including 354 controls (non-CAD) and 959 CAD patients, were enrolled. In univariate analysis of all populations, the adjusted AIP (aAIP) in the CAD group was 1.13 (0.96, 1.3), which was significantly higher than that in the controls [1.07 (0.89, 1.26)]. However, in subgroup analyses, this phenomenon was only present in males. In addition, further study showed that aAIP was positively related to CAD severity. In binary logistic regression analyses, after adjusting for sex, age, smoking status, primary hypertension (PH), type 2 diabetes mellitus (T2DM), heart rate (HR), white blood cell (WBC) and platelet (PLT), AIP remained independently related to CAD in elderly individuals and was superior to traditional and other nontraditional lipid indices. Subgroup analyses showed that AIP independently influenced CAD risk in males. Ultimately, sensitivity analyses were performed excluding all coronary emergencies, and the final results were similar. Conclusions AIP was positively related to the risk and severity of CAD in elderly individuals and was superior to traditional and other nontraditional lipid profiles. However, this association only exists in elderly males. Supplementary Information The online version contains supplementary material available at 10.1186/s12944-021-01496-8.
Collapse
Affiliation(s)
- Haomin Huang
- Department of Cardiology, Wujin Hospital Affiliated with Jiangsu University, The Wujin Clinical College of Xuzhou Medical University, Changzhou City, Jiangsu Province, China
| | - Xiaolong Yu
- Department of Ultrasound, Wujin Hospital Affiliated with Jiangsu University, The Wujin Clinical College of Xuzhou Medical University, Changzhou City, Jiangsu Province, China
| | - Lamei Li
- Department of Cardiology, Wujin Hospital Affiliated with Jiangsu University, The Wujin Clinical College of Xuzhou Medical University, Changzhou City, Jiangsu Province, China
| | - Ganwei Shi
- Department of Cardiology, Wujin Hospital Affiliated with Jiangsu University, The Wujin Clinical College of Xuzhou Medical University, Changzhou City, Jiangsu Province, China
| | - Feng Li
- Department of Cardiology, Wujin Hospital Affiliated with Jiangsu University, The Wujin Clinical College of Xuzhou Medical University, Changzhou City, Jiangsu Province, China
| | - Jianqiang Xiao
- Department of Cardiology, Wujin Hospital Affiliated with Jiangsu University, The Wujin Clinical College of Xuzhou Medical University, Changzhou City, Jiangsu Province, China
| | - Zhihua Yun
- Department of Clinical Laboratory, Wujin Hospital Affiliated with Jiangsu University, The Wujin Clinical College of Xuzhou Medical University, Changzhou City, Jiangsu Province, China
| | - Gaojun Cai
- Department of Cardiology, Wujin Hospital Affiliated with Jiangsu University, The Wujin Clinical College of Xuzhou Medical University, Changzhou City, Jiangsu Province, China.
| |
Collapse
|
37
|
Sampson M, Wolska A, Warnick R, Lucero D, Remaley AT. A New Equation Based on the Standard Lipid Panel for Calculating Small Dense Low-Density Lipoprotein-Cholesterol and Its Use as a Risk-Enhancer Test. Clin Chem 2021; 67:987-997. [PMID: 33876239 DOI: 10.1093/clinchem/hvab048] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 03/04/2021] [Indexed: 12/11/2022]
Abstract
BACKGROUND Increased small dense low-density lipoprotein-cholesterol (sdLDL-C) is a risk factor for atherosclerotic cardiovascular disease (ASCVD) but typically requires advanced lipid testing. We describe two new equations, first one for calculating large buoyant LDL-C (lbLDL-C), based only upon results from the standard lipid panel, and the second one for sdLDL-C. METHODS Equations for sdLDL-C and lbLDL-C were generated with least-squares regression analysis using the direct Denka sdLDL-C assay as reference (n = 20 171). sdLDL-C was assessed as a risk-enhancer test in the National Heart and Nutrition Examination Survey (NHANES), and for its association with ASCVD in the Multi-Ethnic Study of Atherosclerosis (MESA). RESULTS The newly derived equations depend on two terms, namely LDL-C as determined by the Sampson equation, and an interaction term between LDL-C and the natural log of triglycerides (TG). The lbLDL-C equation (lbLDLC=1.43 × LDLC-0.14 ×(ln(TG)× LDLC)- 8.99) was more accurate (R2 = 0.933, slope = 0.94) than the sdLDL-C equation (sdLDLC=LDLC- lbLDLC; R2 = 0.745, slope = 0.73). Using the 80th percentile (46 mg/dL) as a cut-point, sdLDL-C identified in NHANES additional high-risk patients not identified by other risk-enhancer tests based on TG, LDL-C, apolipoprotein B, and nonHDL-C. By univariate survival-curve analysis, estimated sdLDL-C was superior to other risk-enhancer tests in predicting ASCVD events in MESA. After multivariate adjustment for other known ASCVD risk factors, estimated sdLDL-C had the strongest association with ASCVD compared to other lipid parameters, including measured sdLDL-C. CONCLUSIONS Estimated sdLDL-C could potentially be calculated on all patients tested with a standard lipid panel to improve ASCVD risk stratification.
Collapse
Affiliation(s)
- Maureen Sampson
- Department of Laboratory Medicine, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Anna Wolska
- Lipoprotein Metabolism Laboratory, Translational Vascular Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | | | - Diego Lucero
- Lipoprotein Metabolism Laboratory, Translational Vascular Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Alan T Remaley
- Lipoprotein Metabolism Laboratory, Translational Vascular Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
38
|
Bruemmer D, Cho L. Remnant Cholesterol: The Leftovers and Their Contribution to Atherosclerotic Cardiovascular Disease. Circ Cardiovasc Imaging 2021; 14:e012615. [PMID: 33877871 DOI: 10.1161/circimaging.121.012615] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Dennis Bruemmer
- Robert and Suzanne Tomsich Department of Cardiovascular Medicine, Section of Preventive Cardiology and Rehabilitation, Heart, Vascular and Thoracic Institute, Cleveland Clinic Foundation, Cleveland, OH
| | - Leslie Cho
- Robert and Suzanne Tomsich Department of Cardiovascular Medicine, Section of Preventive Cardiology and Rehabilitation, Heart, Vascular and Thoracic Institute, Cleveland Clinic Foundation, Cleveland, OH
| |
Collapse
|
39
|
Increased Circulating Malondialdehyde-Modified Low-Density Lipoprotein Level Is Associated with High-Risk Plaque in Coronary Computed Tomography Angiography in Patients Receiving Statin Therapy. J Clin Med 2021; 10:jcm10071480. [PMID: 33918383 PMCID: PMC8038255 DOI: 10.3390/jcm10071480] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 03/30/2021] [Accepted: 03/30/2021] [Indexed: 11/16/2022] Open
Abstract
Objective: To evaluate the association of serum malondialdehyde low-density lipoprotein (MDA-LDL), an oxidatively modified LDL, with the prevalence of high-risk plaques (HRP) determined with coronary computed tomography angiography (CTA) in statin-treated patients. Methods: This study was a single-center retrospective cohort comprising 268 patients (mean age 67 years, 58% men) with statin therapy and who underwent coronary CTA for suspected stable coronary artery disease. Patients were classified into two groups according to median MDA-LDL level or median LDL-C level. Coronary CTA-verified HRP was defined when two or more characteristics, including positive remodeling, low-density plaques, and spotty calcification, were present. Results: Patients with HRP had higher MDA-LDL (p = 0.011), but not LDL-C (p = 0.867) than those without HRP. High MDA-LDL was independently associated with HRP (odds ratio 1.883, 95% confidential interval 1.082–3.279) after adjustment for traditional risk factors. Regarding incremental value of MDA-LDL for predicting CTA-verified HRP, addition of serum MDA-LDL levels to the baseline model significantly increased global chi-square score from 26.1 to 32.8 (p = 0.010). Conclusions: A high serum MDA-LDL level is an independent predictor of CTA-verified HRP, which can lead to cardiovascular events in statin-treated patients.
Collapse
|
40
|
Abstract
Purpose of review Based on the recent data of the DA VINCI study, it is clear that, besides utilization of statins, there is a need to increase non-statin lipid lowering approaches to reduce the cardiovascular burden in patients at highest risk. Recent findings For hypercholesterolemia, the small synthetic molecule bempedoic acid has the added benefit of selective liver activation, whereas inclisiran, a hepatic inhibitor of the PCSK9 synthesis, has comparable effects with PCSK9 monoclonal antibodies. For hypertriglyceridemia, cardiovascular benefit has been achieved by the use of icosapent ethyl, whereas results with pemafibrate, a selective agonist of PPAR-α, are eagerly awaited. In the era of RNA-based therapies, new options are offered to dramatically reduce levels of lipoprotein(a) (APO(a)LRX) and of triglycerides (ANGPTL3LRX and APOCIII-LRx). Summary Despite the demonstrated benefits of statins, a large number of patients still remain at significant risk because of inadequate LDL-C reduction or elevated blood triglyceride-rich lipoproteins or lipoprotein(a). The area of lipid modulating agents is still ripe with ideas and major novelties are to be awaited in the next few years.
Collapse
|
41
|
Barroso WKS, Rodrigues CIS, Bortolotto LA, Mota-Gomes MA, Brandão AA, Feitosa ADDM, Machado CA, Poli-de-Figueiredo CE, Amodeo C, Mion Júnior D, Barbosa ECD, Nobre F, Guimarães ICB, Vilela-Martin JF, Yugar-Toledo JC, Magalhães MEC, Neves MFT, Jardim PCBV, Miranda RD, Póvoa RMDS, Fuchs SC, Alessi A, Lucena AJGD, Avezum A, Sousa ALL, Pio-Abreu A, Sposito AC, Pierin AMG, Paiva AMGD, Spinelli ACDS, Nogueira ADR, Dinamarco N, Eibel B, Forjaz CLDM, Zanini CRDO, Souza CBD, Souza DDSMD, Nilson EAF, Costa EFDA, Freitas EVD, Duarte EDR, Muxfeldt ES, Lima Júnior E, Campana EMG, Cesarino EJ, Marques F, Argenta F, Consolim-Colombo FM, Baptista FS, Almeida FAD, Borelli FADO, Fuchs FD, Plavnik FL, Salles GF, Feitosa GS, Silva GVD, Guerra GM, Moreno Júnior H, Finimundi HC, Back IDC, Oliveira Filho JBD, Gemelli JR, Mill JG, Ribeiro JM, Lotaif LAD, Costa LSD, Magalhães LBNC, Drager LF, Martin LC, Scala LCN, Almeida MQ, Gowdak MMG, Klein MRST, Malachias MVB, Kuschnir MCC, Pinheiro ME, Borba MHED, Moreira Filho O, Passarelli Júnior O, Coelho OR, Vitorino PVDO, Ribeiro Junior RM, Esporcatte R, Franco R, Pedrosa R, Mulinari RA, Paula RBD, Okawa RTP, Rosa RF, Amaral SLD, Ferreira-Filho SR, Kaiser SE, Jardim TDSV, Guimarães V, Koch VH, Oigman W, Nadruz W. Brazilian Guidelines of Hypertension - 2020. Arq Bras Cardiol 2021; 116:516-658. [PMID: 33909761 PMCID: PMC9949730 DOI: 10.36660/abc.20201238] [Citation(s) in RCA: 308] [Impact Index Per Article: 102.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Affiliation(s)
- Weimar Kunz Sebba Barroso
- Universidade Federal de Goiás , Goiânia , GO - Brasil
- Liga de Hipertensão Arterial , Goiânia , GO - Brasil
| | - Cibele Isaac Saad Rodrigues
- Pontifícia Universidade Católica de São Paulo , Faculdade de Ciências Médicas e da Saúde , Sorocaba , SP - Brasil
| | | | | | - Andréa Araujo Brandão
- Faculdade de Ciências Médicas da Universidade do Estado do Rio de Janeiro (FCM-UERJ), Rio de Janeiro , RJ - Brasil
| | | | | | | | - Celso Amodeo
- Universidade Federal de São Paulo (UNIFESP), São Paulo , SP - Brasil
| | - Décio Mion Júnior
- Hospital das Clínicas da Faculdade de Medicina da USP , São Paulo , SP - Brasil
| | | | - Fernando Nobre
- Hospital das Clínicas da Faculdade de Medicina de Ribeirão Preto da Universidade de São Paulo , Ribeirão Preto , SP - Brasil
- Hospital São Francisco , Ribeirão Preto , SP - Brasil
| | | | | | | | - Maria Eliane Campos Magalhães
- Hospital Universitário Pedro Ernesto da Universidade do Estado do Rio de Janeiro (UERJ), Rio de Janeiro , RJ - Brasil
| | - Mário Fritsch Toros Neves
- Faculdade de Ciências Médicas da Universidade do Estado do Rio de Janeiro (FCM-UERJ), Rio de Janeiro , RJ - Brasil
| | | | | | | | - Sandra C Fuchs
- Faculdade de Medicina da Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre , RS - Brasil
| | | | | | - Alvaro Avezum
- Hospital Alemão Oswaldo Cruz , São Paulo , SP - Brasil
| | - Ana Luiza Lima Sousa
- Universidade Federal de Goiás , Goiânia , GO - Brasil
- Liga de Hipertensão Arterial , Goiânia , GO - Brasil
| | | | | | | | | | | | | | | | - Bruna Eibel
- Instituto de Cardiologia , Fundação Universitária de Cardiologia (IC/FUC), Porto Alegre , RS - Brasil
- Centro Universitário da Serra Gaúcha (FSG), Caxias do Sul , RS - Brasil
| | | | | | | | | | | | | | - Elizabete Viana de Freitas
- Faculdade de Ciências Médicas da Universidade do Estado do Rio de Janeiro (FCM-UERJ), Rio de Janeiro , RJ - Brasil
- Departamento de Cardiogeriatria da Sociedade Brazileira de Cardiologia , Rio de Janeiro , RJ - Brasil
| | | | | | - Emilton Lima Júnior
- Hospital de Clínicas da Universidade Federal do Paraná (HC/UFPR), Curitiba , PR - Brasil
| | - Erika Maria Gonçalves Campana
- Faculdade de Ciências Médicas da Universidade do Estado do Rio de Janeiro (FCM-UERJ), Rio de Janeiro , RJ - Brasil
- Universidade Iguaçu (UNIG), Rio de Janeiro , RJ - Brasil
| | - Evandro José Cesarino
- Faculdade de Ciências Farmacêuticas de Ribeirão Preto da Universidade de São Paulo , Ribeirão Preto , SP - Brasil
- Associação Ribeirãopretana de Ensino, Pesquisa e Assistência ao Hipertenso (AREPAH), Ribeirão Preto , SP - Brasil
| | - Fabiana Marques
- Faculdade de Medicina de Ribeirão Preto da Universidade de São Paulo , Ribeirão Preto , SP - Brasil
| | | | | | | | - Fernando Antonio de Almeida
- Pontifícia Universidade Católica de São Paulo , Faculdade de Ciências Médicas e da Saúde , Sorocaba , SP - Brasil
| | | | | | - Frida Liane Plavnik
- Instituto do Coração (InCor), São Paulo , SP - Brasil
- Hospital Alemão Oswaldo Cruz , São Paulo , SP - Brasil
| | | | | | | | - Grazia Maria Guerra
- Instituto do Coração (InCor), São Paulo , SP - Brasil
- Universidade Santo Amaro (UNISA), São Paulo , SP - Brasil
| | | | | | | | | | | | - José Geraldo Mill
- Centro de Ciências da Saúde , Universidade Federal do Espírito Santo , Vitória , ES - Brasil
| | - José Marcio Ribeiro
- Faculdade Ciências Médicas de Minas Gerais , Belo Horizonte , MG - Brasil
- Hospital Felício Rocho , Belo Horizonte , MG - Brasil
| | - Leda A Daud Lotaif
- Instituto Dante Pazzanese de Cardiologia , São Paulo , SP - Brasil
- Hospital do Coração (HCor), São Paulo , SP - Brasil
| | | | | | | | | | | | - Madson Q Almeida
- Hospital das Clínicas da Faculdade de Medicina da USP , São Paulo , SP - Brasil
| | | | | | | | | | | | | | | | | | | | | | | | - Roberto Esporcatte
- Faculdade de Ciências Médicas da Universidade do Estado do Rio de Janeiro (FCM-UERJ), Rio de Janeiro , RJ - Brasil
- Hospital Pró-Cradíaco , Rio de Janeiro , RJ - Brasil
| | - Roberto Franco
- Universidade Estadual Paulista (UNESP), Bauru , SP - Brasil
| | - Rodrigo Pedrosa
- Pronto Socorro Cardiológico de Pernambuco (PROCAPE), Recife , PE - Brasil
| | | | | | | | | | | | | | - Sergio Emanuel Kaiser
- Faculdade de Ciências Médicas da Universidade do Estado do Rio de Janeiro (FCM-UERJ), Rio de Janeiro , RJ - Brasil
| | | | | | - Vera H Koch
- Universidade de São Paulo (USP), São Paulo , SP - Brasil
| | - Wille Oigman
- Faculdade de Ciências Médicas da Universidade do Estado do Rio de Janeiro (FCM-UERJ), Rio de Janeiro , RJ - Brasil
| | - Wilson Nadruz
- Universidade Estadual de Campinas (UNICAMP), Campinas , SP - Brasil
| |
Collapse
|
42
|
Shea S, Navas-Acien A, Shimbo D, Brown ER, Budoff M, Bancks MP, Graham Barr R, Kronmal R. Spatially Weighted Coronary Artery Calcium Score and Coronary Heart Disease Events in the Multi-Ethnic Study of Atherosclerosis. Circ Cardiovasc Imaging 2021; 14:e011981. [PMID: 33461306 PMCID: PMC7987729 DOI: 10.1161/circimaging.120.011981] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 11/25/2020] [Indexed: 01/07/2023]
Abstract
BACKGROUND A limitation of the Agatston coronary artery calcium (CAC) score is that it does not use all of the calcium density information in the computed tomography scan such that many individuals have a score of zero. We examined the predictive validity for incident coronary heart disease (CHD) events of the spatially weighted coronary calcium score (SWCS), an alternative scoring method for CAC that assigns scores to individuals with Agatston CAC=0. METHODS The MESA (Multi-Ethnic Study of Atherosclerosis) is a longitudinal study that conducted a baseline exam from 2000 to 2002 in 6814 participants including computed tomography scanning for CAC. Subsequent exams and systematic follow-up of the cohort for outcomes were performed. Statistical models were adjusted using the MESA risk score based on age, sex, race/ethnicity, systolic blood pressure, use of hypertension medications, diabetes, total and HDL (high-density lipoprotein) cholesterol, use of lipid-lowering medications, smoking status, and family history of heart attack. RESULTS In the 3286 participants with Agatston CAC=0 at baseline and for whom SWCS was computed, 98 incident CHD events defined as definite or probably myocardial infarction or definite CHD death occurred during a median follow-up of 15.1 years. In this group, SWCS predicted incident CHD events after multivariable adjustment (hazard ratio=1.30 per SD of natural logarithm [SWCS] [95% CI, 1.04-1.60]; P=0.005); and progression from Agatston CAC=0 at baseline to CAC>0 at subsequent exams (multivariable-adjusted incidence rate difference per SD of natural logarithm [SWCS] per 100 person-years 1.68 [95% CI, 1.03-2.33]; P<0.0001). CONCLUSIONS SWCS predicts incident CHD events in individuals with Agatston CAC score=0 as well as conversion to Agatston CAC>0 at repeat computed tomography scanning at later exams. SWCS has predictive validity as a subclinical phenotype and marker of CHD risk in individuals with Agatston CAC=0.
Collapse
Affiliation(s)
- Steven Shea
- Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY
| | - Ana Navas-Acien
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY
| | - Daichi Shimbo
- Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY
| | - Elizabeth R. Brown
- Fred Hutchison Cancer Research Center, University of Washington, Seattle, WA
- Department of Biostatistics, University of Washington, Seattle, WA
| | - Matthew Budoff
- Lundquist Institute, University of California, Los Angeles, Torrance, CA
- Biomedical Research Institute, University of California, Los Angeles, Los Angeles, CA
| | - Michael P. Bancks
- Department of Epidemiology and Prevention, Wake Forest School of Medicine, Winston-Salem, NC
| | - R. Graham Barr
- Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY
| | - Richard Kronmal
- Department of Biostatistics, University of Washington, Seattle, WA
| |
Collapse
|
43
|
Echouffo-Tcheugui JB, Jain M, Cheng S. Breaking through the surface: more to learn about lipids and cardiovascular disease. J Clin Invest 2020; 130:1084-1086. [PMID: 31985490 DOI: 10.1172/jci134696] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
The human lipidome comprises over tens of thousands of distinct lipid species in addition to total cholesterol and the other conventional lipid traits that are routinely measurable in the peripheral circulation. Of the lipid species considered to exhibit bioactive functions, sphingolipids are a class of molecules that have shown relevance to human disease risk and cardiovascular outcomes in particular. In this issue of the JCI, Poss et al. conducted targeted lipidomics in a case-control study involving over 600 individuals and found a sphingolipid profile that predicted coronary artery disease status. In the context of emerging evidence linking sphingolipid biology with cardiovascular pathophysiology, these results suggest the potential utility of serum sphingolipids as cholesterol-independent markers of risk and even future targets for optimizing cardiovascular health.
Collapse
Affiliation(s)
- Justin B Echouffo-Tcheugui
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Mohit Jain
- Department of Medicine and.,Department of Pharmacology, School of Medicine, UCSD, La Jolla, California, USA
| | - Susan Cheng
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| |
Collapse
|
44
|
Soppert J, Lehrke M, Marx N, Jankowski J, Noels H. Lipoproteins and lipids in cardiovascular disease: from mechanistic insights to therapeutic targeting. Adv Drug Deliv Rev 2020; 159:4-33. [PMID: 32730849 DOI: 10.1016/j.addr.2020.07.019] [Citation(s) in RCA: 141] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 07/20/2020] [Accepted: 07/22/2020] [Indexed: 12/12/2022]
Abstract
With cardiovascular disease being the leading cause of morbidity and mortality worldwide, effective and cost-efficient therapies to reduce cardiovascular risk are highly needed. Lipids and lipoprotein particles crucially contribute to atherosclerosis as underlying pathology of cardiovascular disease and influence inflammatory processes as well as function of leukocytes, vascular and cardiac cells, thereby impacting on vessels and heart. Statins form the first-line therapy with the aim to block cholesterol synthesis, but additional lipid-lowering drugs are sometimes needed to achieve low-density lipoprotein (LDL) cholesterol target values. Furthermore, beyond LDL cholesterol, also other lipid mediators contribute to cardiovascular risk. This review comprehensively discusses low- and high-density lipoprotein cholesterol, lipoprotein (a), triglycerides as well as fatty acids and derivatives in the context of cardiovascular disease, providing mechanistic insights into their role in pathological processes impacting on cardiovascular disease. Also, an overview of applied as well as emerging therapeutic strategies to reduce lipid-induced cardiovascular burden is provided.
Collapse
Affiliation(s)
- Josefin Soppert
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital Aachen, Aachen, Germany
| | - Michael Lehrke
- Medical Clinic I, University Hospital Aachen, Aachen, Germany
| | - Nikolaus Marx
- Medical Clinic I, University Hospital Aachen, Aachen, Germany
| | - Joachim Jankowski
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital Aachen, Aachen, Germany; Department of Pathology, Cardiovascular Research Institute Maastricht, Maastricht University Medical Centre, Maastricht University, the Netherlands
| | - Heidi Noels
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital Aachen, Aachen, Germany; Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, the Netherlands.
| |
Collapse
|
45
|
Guo Q, Zhou S, Feng X, Yang J, Qiao J, Zhao Y, Shi D, Zhou Y. The sensibility of the new blood lipid indicator--atherogenic index of plasma (AIP) in menopausal women with coronary artery disease. Lipids Health Dis 2020; 19:27. [PMID: 32093690 PMCID: PMC7041294 DOI: 10.1186/s12944-020-01208-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 02/18/2020] [Indexed: 12/16/2022] Open
Abstract
Background Dyslipidemia is a key driver of coronary artery disease (CAD) development. This study aimed to determine whether the atherogenic index of plasma (AIP), a novel comprehensive lipid index, is an independent and reliable predictor of CAD risk in postmenopausal women. Methods A cohort of consecutive 4644 postmenopausal women (aged 50 or above) undergoing coronary angiography (CAG) in Anzhen Hospital (Beijing, China) from January–December 2014 was included in the analysis. Of them, 3039 women were CAD patients, and 1605 were non-CAD subjects. Results Relative to control subjects, TG levels in CAD patients were higher and HDL-C levels were lower. In CAD patients, non-traditional lipid profile values (TC/HDL-C, AI, and AIP) were significantly elevated relative to controls. AIP was positively correlated with TC (r = 0.157), TG (r = 0.835), LDL-C (r = 0.058), non-HDL-C (r = 0.337), TC/HDL-C (r = 0.683), LDL-C/HDL-C (r = 0.437), LCI (r = 0.662), and AI (r = 0.684), and negatively correlated with HDL-C (r = − 0.682) (all P < 0.001), but was independent of age (r = − 0.022; P = 0.130) and BMI (r = 0.020, P = 0.168). Aunivariate logistic regression analysis revealed AIP to be the measured lipid parameter most closely related to CAD, and its unadjusted odds ratio was 1.824 (95% CI: 1.467–2.267, P < 0.001). After adjusting for several CAD risk factors (age, BMI, smoking, drinking, EH, DM, hyperlipidemia, and family history of CVD, AIP was still found to represent a significant CAD risk factor (OR 1.553, 95% CI: 1.234–1.955, P < 0. 001). Conclusion AIP may be a powerful independent predictor of CAD risk in Chinese Han postmenopausal women, and may be superior to the traditional lipid indices.
Collapse
Affiliation(s)
- Qianyun Guo
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical center for coronary heart disease, Capital Medical University, Beijing, 100029, China
| | - Shu Zhou
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical center for coronary heart disease, Capital Medical University, Beijing, 100029, China
| | - Xunxun Feng
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical center for coronary heart disease, Capital Medical University, Beijing, 100029, China
| | - Jiaqi Yang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical center for coronary heart disease, Capital Medical University, Beijing, 100029, China
| | - Jiaming Qiao
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical center for coronary heart disease, Capital Medical University, Beijing, 100029, China
| | - Yingxin Zhao
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical center for coronary heart disease, Capital Medical University, Beijing, 100029, China
| | - Dongmei Shi
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical center for coronary heart disease, Capital Medical University, Beijing, 100029, China
| | - Yujie Zhou
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical center for coronary heart disease, Capital Medical University, Beijing, 100029, China.
| |
Collapse
|
46
|
Konishi T, Sunaga D, Funayama N, Yamamoto T, Murakami H, Hotta D, Nojima M, Tanaka S. Eicosapentaenoic acid therapy is associated with decreased coronary plaque instability assessed using optical frequency domain imaging. Clin Cardiol 2019; 42:618-628. [PMID: 30993750 PMCID: PMC6553360 DOI: 10.1002/clc.23185] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 04/07/2019] [Accepted: 04/16/2019] [Indexed: 01/26/2023] Open
Abstract
Background The relationship between eicosapentaenoic acid (EPA) therapy and coronary plaque stability assessed by optical frequency domain imaging (OFDI) has not been thoroughly described. Hypothesis EPA therapy is associated with decreased plaque instability in patients undergoing percutaneous coronary intervention (PCI) using OFDI. Methods Data on coronary artery plaques from 121 patients who consecutively underwent PCI between October 2015 and July 2018 were retrospectively analyzed. Of these patients, 109 were untreated (no‐EPA group), whereas 12 were treated with EPA (EPA group). Each plaque's morphological characteristics were analyzed using OFDI. Results We used 1:4 propensity score matching for patients who received or did not receive EPA therapy before PCI. Baseline characteristics were balanced between both groups (age, sex, body mass index, diabetes mellitus, hypertension, dyslipidemia, chronic kidney disease, smoking, previous PCI or coronary artery bypass grafting, previous myocardial infarction, prior statin use, acute coronary syndrome, hemoglobin A1c level, low‐density lipoprotein cholesterol concentration, triglyceride concentration, and high‐density lipoprotein cholesterol concentration). OFDI data from 60 patients were analyzed in this study. The EPA group had significantly lower mean lipid index (818 ± 806 vs 1574 ± 891) and macrophage grade (13.5 ± 5.9 vs 19.3 ± 7.4) but higher mean minimum fibrous cap thickness (109.2 ± 55.7 vs 81.6 ± 36.4 μm) than the no‐EPA group (P = 0.010, 0.019, and 0.040, respectively). Multiple logistic regression analyses showed that prior EPA use was independently associated with lower lipid index and macrophage grade (P = 0.043 and 0.024, respectively). Conclusion This OFDI analysis suggests that EPA therapy is associated with decreased plaque instability in patients undergoing PCI.
Collapse
Affiliation(s)
- Takao Konishi
- Department of Cardiology, Hokkaido Cardiovascular Hospital, Sapporo, Japan.,Department of Cancer Pathology, Hokkaido University Faculty of Medicine, Sapporo, Japan
| | - Daisuke Sunaga
- Department of Cardiology, Hokkaido Cardiovascular Hospital, Sapporo, Japan
| | - Naohiro Funayama
- Department of Cardiology, Hokkaido Cardiovascular Hospital, Sapporo, Japan
| | - Tadashi Yamamoto
- Department of Cardiology, Hokkaido Cardiovascular Hospital, Sapporo, Japan
| | - Hironori Murakami
- Department of Cardiology, Hokkaido Cardiovascular Hospital, Sapporo, Japan
| | - Daisuke Hotta
- Department of Cardiology, Hokkaido Cardiovascular Hospital, Sapporo, Japan
| | - Masanori Nojima
- Center for Translational Research, The Institute of Medical Science Hospital, The University of Tokyo, Tokyo, Japan
| | - Shinya Tanaka
- Department of Cancer Pathology, Hokkaido University Faculty of Medicine, Sapporo, Japan
| |
Collapse
|
47
|
Cai G, Liu W, Lv S, Wang X, Guo Y, Yan Z, Du Y, Zhou Y. Gender-specific associations between atherogenic index of plasma and the presence and severity of acute coronary syndrome in very young adults: a hospital-based observational study. Lipids Health Dis 2019; 18:99. [PMID: 30987629 PMCID: PMC6466804 DOI: 10.1186/s12944-019-1043-2] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 04/02/2019] [Indexed: 01/08/2023] Open
Abstract
OBJECTIVE The value of atherogenic index of plasma (AIP) as a predictive biomarker for coronary artery disease (CAD) remains controversial. In addition, whether AIP is associated with the risk of acute coronary syndrome (ACS) in very young adults has not been well established. METHODS We consecutively collected very young adults (≤35 years of age) undergoing coronary angiography (CAG) at Anzhen Hospital, between January 2008 and December 2017. Total of 1, 478 very young participants, including 1, 059 ACS patients and 419 non-CAD subjects, were enrolled in the present study. RESULTS Very young patients with ACS had higher AIP level compared with non-CAD participants (0.35 ± 0.30 vs 0.21 ± 0.33, P < 0.001). According to Gensini Score (GS) and number of lesion vessel, patients were divided into four groups, respectively. With the elevated GS score and number of lesion vessels, the AIP level increased gradually (Pfor trend all< 0.05). Multivariate logistic regression analyses suggested that AIP remained to be independently associated with the presence of ACS and was superior to traditional lipid profiles (for AIP, OR = 2.930, 95% CI = 1.855-4.627, P < 0.001; for total cholesterol, OR = 1.152, 95% CI = 1.048-1.266, P = 0.003; for triglyceride, OR = 1.078, 95% CI = 0.991-1.172, P = 0.079; for low-density lipoprotein cholesterol, OR = 1.046, 95% CI = 1.015-1.078, P < 0.001), after adjustment for other traditional confounders. Moreover, the prevalence of ACS, acute myocardial infarction, unstable angina pectoris and the value of GS were also elevated as AIP quartiles increased (Pfor trend < 0.001). Subgroup analysis based on gender revealed that AIP was only independently associated with the ACS risk in male. CONCLUSIONS AIP was independently associated with the presence and severity of ACS in very young patients in a gender-dependent manner, which might be superior to traditional lipid profiles.
Collapse
Affiliation(s)
- Gaojun Cai
- Department of Cardiology, 12th ward, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical center for coronary heart disease, Capital Medical University, Beijing, 100029, China
- Department of Cardiology, Wujin Hospital affiliated with Jiangsu University, Changzhou, 213017, Jiangsu, China
| | - Wei Liu
- Department of Cardiology, 12th ward, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical center for coronary heart disease, Capital Medical University, Beijing, 100029, China
| | - Sai Lv
- Department of Cardiology, 12th ward, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical center for coronary heart disease, Capital Medical University, Beijing, 100029, China
| | - Xu Wang
- Emergency & Critical Care Center, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, China
| | - Yonghe Guo
- Department of Cardiology, 12th ward, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical center for coronary heart disease, Capital Medical University, Beijing, 100029, China
| | - Zhenxian Yan
- Department of Cardiology, 12th ward, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical center for coronary heart disease, Capital Medical University, Beijing, 100029, China
| | - Yu Du
- Department of Cardiology, 12th ward, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical center for coronary heart disease, Capital Medical University, Beijing, 100029, China
| | - Yujie Zhou
- Department of Cardiology, 12th ward, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical center for coronary heart disease, Capital Medical University, Beijing, 100029, China.
| |
Collapse
|
48
|
Hayashi H. Enhancement of rat lymphatic lipid transport by glucose or amino acids ingestion. Physiol Rep 2019; 7:e14079. [PMID: 31016888 PMCID: PMC6478621 DOI: 10.14814/phy2.14079] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 04/02/2019] [Accepted: 04/10/2019] [Indexed: 11/24/2022] Open
Abstract
To elucidate the effect of simultaneously fed carbohydrate or protein on lipid absorption, lymphatic lipid transports in the rat intestine were observed with or without simultaneous feeding of glucose or amino acids. A lipid emulsion containing 40 μmol/h (35.4 mg/h) of triolein, 2.74 μmol/h (1.06 mg/h) of cholesterol, 7.8 μmol/h (6.08 mg/h) of egg phosphatidylcholine without any additive (the Control group), with 560 mmol/h (300 mg/h) of glucose (the Glucose group), or with 400 mmol/h (150 mg/h) of amino acids (the Amino Acids group), was infused intraduodenally at 3 mL/h for 8 h into mesenteric lymph-fistula rats. The amounts of triglyceride transported in lymph for 8 h were 185 ± 12 (mean ± SE) mg in the Amino Acids group (n = 4), 175 ± 3 mg in the Glucose group (n = 5), and 147 ± 7 mg in the Control group (n = 4), respectively, with a statistically significant difference (P < 0.05) among the groups. The amounts of cholesterol transported in lymph for 8 h of the Amino Acid group and the Glucose group seemed to be larger than that of the Control group. The amount of phosphatidylcholine transported in lymph for 8 h were 16.4 ± 1.0 mg in the Amino Acids group, 15.7 ± 0.4 mg in the Glucose group, and 12.4 ± 0.3 mg in the Control group, respectively, with a statistically significant difference (P < 0.01) among the groups. Simultaneous glucose or amino acids feeding enhanced lymphatic lipid transport in the rat intestine during lipid feeding.
Collapse
Affiliation(s)
- Hiroshi Hayashi
- Department of Internal MedicineTokyo Ariake University of Medical and Health SciencesKoto‐kuTokyoJapan
| |
Collapse
|