1
|
La X, He X, Liang J, Zhang Z, Li Z. Investigating the separation and purification of flavonoids extracted from foxtail millet, with an in-depth study on its functions in alleviating metabolic syndrome through suppressing lipid absorption. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2025. [PMID: 40285665 DOI: 10.1002/jsfa.14295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Revised: 03/01/2025] [Accepted: 03/19/2025] [Indexed: 04/29/2025]
Abstract
BACKGROUND Rising living standards alongside high-fat diets (HFDs) have fueled the prevalence of metabolic syndrome (MetS), posing a significant health challenge with limited effective therapies. Foxtail millet is recognized as a health food with rich flavonoids. The objective of this work is to isolate and purify flavonoids from foxtail millet, which have the potential to alleviate MetS and to clarify their mechanism. RESULTS Our research combined single-factor experiments and a Box-Behnken design to optimize the extraction processes for millet whole-grain flavonoids (MWGFs) followed by the purification using D101 resin. Components were eluted using varied alcohol concentrations. Additionally, the investigation revealed that MWGF30 notably decreased triglyceride (TG) levels, mitigated lipid uptake in Caco-2 cells induced by free fatty acids, and suppressed lipid absorption in HFD mice while modulating TG, high‑ and low-density lipoprotein cholesterol and total cholesterol levels. Further investigation uncovered that MWGF30 achieved this lipid uptake reduction ability by downregulating the CD36 protein level. CONCLUSION Collectively, MWGF30 demonstrated a potent ability to regulate blood lipids and curb lipid absorption, enhancing our comprehension of millet's benefits and furnishing a scientifically grounded dietary approach for MetS. © 2025 Society of Chemical Industry.
Collapse
Affiliation(s)
- Xiaoqin La
- Institutes of Biomedical Sciences, Shanxi University, Taiyuan, China
- Shanxi Provincial Key Laboratory of Medical Molecular Cell Biology, Shanxi University, Taiyuan, China
| | - Xiaoting He
- School of Life Science, Shanxi University, Taiyuan, China
| | - Jingyi Liang
- Institute of Biotechnology, Shanxi University, Taiyuan, China
| | - Zhaoyan Zhang
- Institutes of Biomedical Sciences, Shanxi University, Taiyuan, China
- Shanxi Provincial Key Laboratory of Medical Molecular Cell Biology, Shanxi University, Taiyuan, China
| | - Zhuoyu Li
- Institute of Biotechnology, Shanxi University, Taiyuan, China
| |
Collapse
|
2
|
Xu P, Fan C, Yan M, Liu J, Zhang X. Remnant cholesterol and suicide attempts in untreated first-episode major depressive disorder. Front Psychiatry 2025; 16:1493509. [PMID: 40104336 PMCID: PMC11913861 DOI: 10.3389/fpsyt.2025.1493509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 02/18/2025] [Indexed: 03/20/2025] Open
Abstract
Objective The objective of this research was to investigate the relationship between remnant cholesterol (RC) levels and suicide attempts (SA) made by Chinese patients with untreated first-episode major depressive disorder (UFE MDD). Methods This study included 1718 patients with UFE MDD. Demographic, clinical characteristics, and blood lipid parameters were collected. The 17-item Hamilton Depression Rating Scale (HAMD), the 14-item Hamilton Anxiety Rating Scale (HAMA), and the positive subscale of the Positive and Negative Syndrome Scale (PANSS) were used to assess their depression, anxiety, and psychotic symptoms, respectively. Multivariable binary logistic regression analysis was used to estimate the association between RC and the risk of SA. A two-piecewise linear regression model was used to investigate the threshold effects if non-linear associations existed. Results Univariate logistic regression analysis showed a significant positive correlation between RC and SA, but after controlling for confounding factors, the association between them was not statistically significant. After dividing the RC into quartiles, only the RC in the Q4 group was significantly positively correlated with suicide attempts (OR = 1.73, 95% CI: 1.13-2.65, P = 0.012, vs. Q1) in a fully adjusted model. Curve fitting analysis also showed a nonlinear relationship between RC and suicide attempts with an inflection point at 1.99 mmol/L in RC. On the left of the inflection point, a significant positive correlation was observed between RC and SA (OR: 1.36, 95% CI: 1.09-1.69, p=0.006). However, on the right of the inflection point, no significant correlation was found (OR: 0.79, 95% CI: 0.55-1.14, p=0.214). Conclusion This study demonstrates a non-linear association between RC levels and SA in patients with untreated first-episode major depressive disorder. When RC was less than 1.99 mmol/L, they showed a significant positive correlation.
Collapse
Affiliation(s)
- Ping Xu
- Department of Psychiatry, Nanjing Lishui District Psychiatric Hospital, Lishui, China
- Nanjing Department of Psychiatry, The Third People's Hospital of Lishui District, Lishui, China
| | - Cheng Fan
- Department of Psychiatry, Nanjing Lishui District Psychiatric Hospital, Lishui, China
- Nanjing Department of Psychiatry, The Third People's Hospital of Lishui District, Lishui, China
| | - Mingxing Yan
- Department of Psychiatry, Nanjing Lishui District Psychiatric Hospital, Lishui, China
- Nanjing Department of Psychiatry, The Third People's Hospital of Lishui District, Lishui, China
| | - Junjun Liu
- Department of Psychiatry, Nanjing Meishan Hospital, Nanjing, China
- Medical College of Soochow University, Suzhou, China
- Suzhou Guangji Hospital, The Affiliated Guangji Hospital of Soochow University, Suzhou, China
| | - Xiangyang Zhang
- Chinese Academy of Sciences (CAS) Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
3
|
Gong S, Jin J, Mao J, Li H, Mo Y, Zhou Q, Gan S. Plasma atherogenicity index is a powerful indicator for identifying metabolic syndrome in adults with type 2 diabetes mellitus: A cross-sectional study. Medicine (Baltimore) 2024; 103:e39792. [PMID: 39331941 PMCID: PMC11441968 DOI: 10.1097/md.0000000000039792] [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: 01/16/2024] [Accepted: 08/30/2024] [Indexed: 09/29/2024] Open
Abstract
Metabolic syndrome (MetS) is an important risk factor for atherosclerotic cardiovascular disease (ASCVD). Elevated triglyceride (TG) levels and decreased high-density lipoprotein levels (HDL-C) are predisposing factors for the development of ASCVD. Evidence on the association between atherosclerotic index of plasma [AIP = log (TG/HDL-C)] and MetS is limited. Our study aimed to investigate the association between AIP and MetS. This is a cross-sectional study that determines the presence of MetS by assessing anthropometric and biochemical parameters. Multivariate log-binomial regression models were used to analyze the relationship between AIP and MetS risk. To further test the stability of the results, we performed sensitivity analyses in young, non-obese, and normal lipid population. Smoothing plots explored the potential nonlinear relationship between the AIP index for MetS and the estimated potential risk threshold. Predictive power of AIP for MetS using respondent operating characteristic (ROC) curves. The prevalence of MetS was 67.35%. Multivariate logistic regression analysis showed an independent and positive association between AIP and MetS (Per 1 SD increase, PR = 1.31, 95% CI: 1.15-1.47). Sensitivity analysis demonstrated the stability of the results. Smoothing plot showed a nonlinear relationship between AIP and MetS, with an inflection point of 0.66. ROC curve analysis, AIP was an accurate indicator for assessing MetS in type 2 diabetics (AUC = 0.840, 95% CI: 0.819-0.862). AIP is a stable and independently powerful predictor of MetS in T2DM patients. AIP can be used as a simple assessment tool for the early detection of MetS and disease management for the prevention of cardiovascular disease.
Collapse
Affiliation(s)
- Shijun Gong
- Department of Ultrasound, Changde Hospital, Xiangya School of Medicine, Central South University, Changde, China
| | - Jing Jin
- Department of Endocrinology, Changde Hospital, Xiangya School of Medicine, Central South University, Changde, China
| | - Jing Mao
- Department of Science and Education, Changde Hospital, Xiangya School of Medicine, Central South University, Changde, China
| | - Heng Li
- Department of Ultrasound, Changde Hospital, Xiangya School of Medicine, Central South University, Changde, China
| | - YePing Mo
- Department of Ultrasound, Changde Hospital, Xiangya School of Medicine, Central South University, Changde, China
| | - Quan Zhou
- Department of Science and Education, Changde Hospital, Xiangya School of Medicine, Central South University, Changde, China
| | - Shenglian Gan
- Department of Endocrinology, Changde Hospital, Xiangya School of Medicine, Central South University, Changde, China
| |
Collapse
|
4
|
Zhang Y, Chen S, Tian X, Xu Q, Xia X, Zhang X, Li J, Wu S, Wang A. Elevated atherogenic index of plasma associated with stroke risk in general Chinese. Endocrine 2024; 84:934-942. [PMID: 38197990 DOI: 10.1007/s12020-023-03677-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 12/26/2023] [Indexed: 01/11/2024]
Abstract
BACKGROUND AND AIMS The atherogenic index of plasma (AIP) is supposed to be associated with cardiovascular disease (CVD), but there is limited evidence on its longitudinal effect. Our study aimed to explore the associations between baseline and long-term AIP with the risk of stroke and its subtypes. METHODS AND RESULTS A total of 97,959 participants free of stroke at baseline were included in the Kailuan study. The AIP was calculated as the logarithm of the ratio of triglyceride to high-density lipoprotein cholesterol. The updated mean AIP was calculated as the average of the AIP from baseline to the first occurrence of outcome or the end of follow-up. The outcome was the first occurrence of stroke, including ischemic stroke (IS) and intracerebral hemorrhage (ICH). We used univariable and multivariable Cox proportional hazard models to explore the association between AIP and the risk of stroke. During a median follow-up of 12.79 years, a total of 6307 participants developed stroke, including 5482 IS and 1024 ICH. Compared with the 1st quartile of baseline AIP, the multivariate-adjusted HR in the 4th quartile was 1.12 (95% CI, 1.03-1.22, p for trend <0.001) for stroke risk. Same results were found in IS, but no significant association was found for ICH. The associations between updated mean AIP and stroke and its subtypes showed similar results. CONCLUSIONS Elevated levels of both baseline and long-term updated mean AIP were associated with the risk of stroke and IS but not ICH in the general population.
Collapse
Affiliation(s)
- Yijun Zhang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Department of Clinical Epidemiology and Clinical Trial, Capital Medical University, Beijing, China
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, Beijing, China
- Beijing Municipal Key Laboratory of Clinical Epidemiology, Beijing, China
| | - Shuohua Chen
- Department of Cardiology, Kailuan Hospital, North China University of Science and Technology, Tangshan, China
| | - Xue Tian
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Department of Clinical Epidemiology and Clinical Trial, Capital Medical University, Beijing, China
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, Beijing, China
- Beijing Municipal Key Laboratory of Clinical Epidemiology, Beijing, China
| | - Qin Xu
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Department of Clinical Epidemiology and Clinical Trial, Capital Medical University, Beijing, China
| | - Xue Xia
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Department of Clinical Epidemiology and Clinical Trial, Capital Medical University, Beijing, China
| | - Xiaoli Zhang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Department of Clinical Epidemiology and Clinical Trial, Capital Medical University, Beijing, China
| | - Jing Li
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Department of Clinical Epidemiology and Clinical Trial, Capital Medical University, Beijing, China
| | - Shouling Wu
- Department of Cardiology, Kailuan Hospital, North China University of Science and Technology, Tangshan, China.
| | - Anxin Wang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
- Department of Clinical Epidemiology and Clinical Trial, Capital Medical University, Beijing, China.
| |
Collapse
|
5
|
Ahmad M, Kennedy BA, Son S, McIntyre AD, Lazarte J, Wang J, Hegele RA. Carotid intima-medial thickness in patients with severe hypertriglyceridemia. ATHEROSCLEROSIS PLUS 2024; 56:7-11. [PMID: 38694144 PMCID: PMC11060956 DOI: 10.1016/j.athplu.2024.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 04/16/2024] [Accepted: 04/19/2024] [Indexed: 05/04/2024]
Abstract
Background and aims Severe hypertriglyceridemia (HTG), defined as plasma triglyceride (TG) concentration > 10 mmol/L, is relatively uncommon, and its implications for atherosclerotic cardiovascular disease (ASCVD) risk remain somewhat unclear. We evaluated the association between severe HTG and carotid intima-media thickness (IMT), a marker for ASCVD. Methods We studied three clinical cohorts: 88 patients with severe HTG (mean TG level 20.6 mmol/L), 271 patients with familial hypercholesterolemia (FH) as a contrast group, and 70 normolipidemic controls. Carotid IMT was measured using standardized ultrasound imaging. Statistical analysis was conducted using one-way analysis of variance (ANOVA) to compare mean IMT values, analysis of covariance (ANCOVA) to adjust for confounding variables, specifically age and sex, as well as Spearman pairwise correlation analysis between variables. Results Unadjusted mean carotid IMT was greater in severe HTG and FH groups compared to controls, however, this was no longer significant for severe HTG after adjustment for age and sex. In contrast, adjusted carotid IMT remained significantly different between the FH and control groups. Conclusions Our findings suggest that extreme TG elevations in severe HTG patients are not significantly associated with carotid IMT, in contrast to the increased IMT seen in FH patients. These findings add perspective to the complex relationship between severe HTG and ASCVD risk.
Collapse
Affiliation(s)
- Maud Ahmad
- Department of Medicine, Schulich School of Medicine and Dentistry, Western University, London, Ontario, N6A 5B7, Canada
| | - Brooke A. Kennedy
- Robarts Research Institute, Western University, London, Ontario, N6A 5B7, Canada
| | - Surim Son
- Department of Epidemiology and Biostatistics, Western University, London, ON, N6A 5B7, Canada
| | - Adam D. McIntyre
- Robarts Research Institute, Western University, London, Ontario, N6A 5B7, Canada
| | - Julieta Lazarte
- Department of Medicine, Schulich School of Medicine and Dentistry, Western University, London, Ontario, N6A 5B7, Canada
| | - Jian Wang
- Robarts Research Institute, Western University, London, Ontario, N6A 5B7, Canada
| | - Robert A. Hegele
- Department of Medicine, Schulich School of Medicine and Dentistry, Western University, London, Ontario, N6A 5B7, Canada
- Robarts Research Institute, Western University, London, Ontario, N6A 5B7, Canada
| |
Collapse
|
6
|
Solnica B, Sygitowicz G, Sitkiewicz D, Jóźwiak J, Kasperczyk S, Broncel M, Wolska A, Odrowąż-Sypniewska G, Banach M. 2024 Guidelines of the Polish Society of Laboratory Diagnostics and the Polish Lipid Association on laboratory diagnostics of lipid metabolism disorders. Arch Med Sci 2024; 20:357-374. [PMID: 38757022 PMCID: PMC11094830 DOI: 10.5114/aoms/186191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 03/18/2024] [Indexed: 05/18/2024] Open
Abstract
Lipid disorders are the most common (even 70%) and worst monitored cardiovascular risk factor (only 1/4 of patients in Poland and in CEE countries are on the low-density lipoprotein cholesterol (LDL-C) goal). To improve this, clear and simple diagnostic criteria should be introduced for all components of the lipid profile. These are the updated guidelines of the two main scientific societies in Poland in the area - the Polish Society of Laboratory Diagnostics (PSLD) and the Polish Lipid Association (PoLA), which, in comparison to those from 2020, introduce few important changes in recommendations (two main lipid targets, new recommendations on LDL-C measurements, calculations new goals for triglycerides, new recommendations on remnants and small dense LDL) that should help the practitioners to be early with the diagnosis of lipid disorders and in the effective monitoring (after therapy initiation), and in the consequence to avoid the first and recurrent cardiovascular events.
Collapse
Affiliation(s)
- Bogdan Solnica
- Polish Society for Laboratory Diagnostics
- Department of Clinical Biochemistry, Jagiellonian University Medical College, Krakow, Poland
| | - Grażyna Sygitowicz
- Polish Society for Laboratory Diagnostics
- Department of Clinical Chemistry and Laboratory Diagnostics, Medical University of Warsaw, Warsaw, Poland
| | - Dariusz Sitkiewicz
- Polish Society for Laboratory Diagnostics
- Department of Clinical Chemistry and Laboratory Diagnostics, Medical University of Warsaw, Warsaw, Poland
| | - Jacek Jóźwiak
- Polish Lipid Association
- Department of Family Medicine and Public Health, Faculty of Medicine, University of Opole, Opole, Poland
| | - Sławomir Kasperczyk
- Polish Lipid Association
- Department of Biochemistry, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, Poland
| | - Marlena Broncel
- Polish Lipid Association
- Department of Internal Diseases and Clinical Pharmacology, Medical University of Lodz, Lodz, Poland
| | - Anna Wolska
- Polish Lipid Association
- Lipoprotein Metabolism Laboratory, Translational Vascular Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Grażyna Odrowąż-Sypniewska
- Polish Society for Laboratory Diagnostics
- Collegium Medicum, Nicolaus Copernicus University, Torun, Poland
| | - Maciej Banach
- Polish Lipid Association
- Department of Preventive Cardiology and Lipidology, Medical University of Lodz (MUL), Lodz, Poland
- Ciccarone Center for the Prevention of Cardiovascular Disease, Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
7
|
Zhang Y, Wu S, Tian X, Xu Q, Xia X, Zhang X, Li J, Chen S, Liu F, Wang A. Elevated atherogenic index of plasma increased the risk of myocardial infarction in a general population. Ann Epidemiol 2024; 90:1-8. [PMID: 37979893 DOI: 10.1016/j.annepidem.2023.11.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 10/11/2023] [Accepted: 11/02/2023] [Indexed: 11/20/2023]
Abstract
PURPOSE The atherogenic index of plasma (AIP) has been shown to be related to cardiovascular disease risk, but evidence on the longitudinal pattern of AIP during follow-up is limited. We aimed to explore the associations of baseline and long-term AIP with the risk of myocardial infarction (MI). METHODS We included 98,861 participants in the Kailuan Study who were free of MI at baseline. AIP was calculated as log (triglyceride/high-density lipoprotein cholesterol). Long-term AIP included the long-term mean AIP (mean AIP from baseline to the first occurrence of MI or the end of follow-up) and number of visits with high AIP (above the cutoff value at the first three visits). The baseline and updated mean AIP were in operationalized as quartiles. Cox proportional hazard models were used to determine the associations between AIP and risk of MI. We excluding all deaths during the follow-up visits in the sensitivity analysis. RESULTS During a median follow-up of 12.80 years, 1804 participants developed MI. Compared with quartile 1, the adjusted hazard ratios in baseline and updated mean AIP quartile 4 were 1.63 (95% CI, 1.41-1.88) and 1.59 (95% CI, 1.37-1.83), respectively. Compared with participants who did not have a high AIP, the HR among individuals with AIP elevated by three times was 1.94 (95% CI,1.55-2.45). The sensitivity analysis showed similar associations. CONCLUSIONS Elevated levels of both baseline and long-term AIP increased the risk of MI.
Collapse
Affiliation(s)
- Yijun Zhang
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, Beijing, China; Beijing Municipal Key Laboratory of Clinical Epidemiology, Beijing, China; Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China; China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Shouling Wu
- Department of Cardiology, Kailuan Hospital, North China University of Science and Technology, Tangshan, China
| | - Xue Tian
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, Beijing, China; Beijing Municipal Key Laboratory of Clinical Epidemiology, Beijing, China; Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China; China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Qin Xu
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China; China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Xue Xia
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China; China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Xiaoli Zhang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China; China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Jing Li
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China; China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Shuohua Chen
- Department of Cardiology, Kailuan Hospital, North China University of Science and Technology, Tangshan, China
| | - Fen Liu
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, Beijing, China; Beijing Municipal Key Laboratory of Clinical Epidemiology, Beijing, China.
| | - Anxin Wang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China; China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China; Department of Clinical Epidemiology and Clinical Trial, Capital Medical University, Beijing, China.
| |
Collapse
|
8
|
Domanski MJ, Wu CO, Tian X, Hasan AA, Ma X, Huang Y, Miao R, Reis JP, Bae S, Husain A, Jacobs DR, Allen NB, Lee MLT, Hong CC, Farkouh ME, Lloyd-Jones DM, Fuster V. Association of Incident Cardiovascular Disease With Time Course and Cumulative Exposure to Multiple Risk Factors. J Am Coll Cardiol 2023; 81:1151-1161. [PMID: 36948731 DOI: 10.1016/j.jacc.2023.01.024] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 01/06/2023] [Accepted: 01/17/2023] [Indexed: 03/24/2023]
Abstract
BACKGROUND The quantitative relationship of incident cardiovascular disease (CVD) to lifetime cumulative risk factor exposure is not well understood. OBJECTIVES Using CARDIA (Coronary Artery Risk Development in Young Adults) study data, we examined the quantitative associations of cumulative exposure over time to multiple, simultaneously operating risk factors with CVD incidence and the incidence of its components. METHODS Regression models were developed quantifying the influence of the time course and severity of multiple CVD risk factors, operating simultaneously, on risk of incident CVD. The outcomes were incident CVD and the incidence of its components: coronary heart disease, stroke, and congestive heart failure. RESULTS Our study included 4,958 asymptomatic adults enrolled in CARDIA from 1985 to 1986 (ages 18 to 30 years) who were followed for 30 years. Risk of incident CVD depends on the time course and severity of a series of independent risk factors, the impact of which is mediated by their effects on individual CVD components after age 40 years. Cumulative exposure (AUC vs time) to low-density lipoprotein cholesterol and triglycerides was independently associated with risk of incident CVD. Of the blood pressure variables, areas under the mean arterial pressure vs time curve and the pulse pressure vs time curve were strongly and independently associated with incident CVD risk. CONCLUSIONS The quantitative description of the link between risk factors and CVD informs the construction of individualized CVD mitigation strategies, design of primary prevention trials, and assessment of public health impact of risk factor-based interventions.
Collapse
Affiliation(s)
- Michael J Domanski
- Division of Cardiovascular Medicine and Data Science Initiative, University of Maryland School of Medicine and the VA Medical Center, Baltimore, Maryland, USA.
| | - Colin O Wu
- Office of Biostatistics Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Xin Tian
- Office of Biostatistics Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Ahmed A Hasan
- Division of Cardiovascular Medicine and Data Science Initiative, University of Maryland School of Medicine and the VA Medical Center, Baltimore, Maryland, USA; Division of Cardiovascular Sciences, National Heart, Lung, and Blood Institute, Bethesda, Maryland, USA
| | - Xiaoyang Ma
- Office of Biostatistics Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Yi Huang
- Division of Cardiovascular Medicine and Data Science Initiative, University of Maryland School of Medicine and the VA Medical Center, Baltimore, Maryland, USA; Department of Mathematics and Statistics, University of Maryland Baltimore County, Baltimore, Maryland, USA
| | - Rui Miao
- Office of Biostatistics Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Jared P Reis
- Division of Cardiovascular Sciences, National Heart, Lung, and Blood Institute, Bethesda, Maryland, USA
| | - Sejong Bae
- Division of Preventive Medicine, University of Alabama at Birmingham School of Medicine, Birmingham, Alabama, USA
| | - Anwar Husain
- Division of Cardiovascular Medicine and Data Science Initiative, University of Maryland School of Medicine and the VA Medical Center, Baltimore, Maryland, USA
| | - David R Jacobs
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, Minnesota, USA
| | - Norrina B Allen
- Department of Preventive Medicine, Northwestern Feinberg School of Medicine, Chicago, Illinois, USA
| | - Mei-Ling T Lee
- Department of Epidemiology and Biostatistics, University of Maryland, College Park, Maryland, USA
| | - Charles C Hong
- Division of Cardiovascular Medicine and Data Science Initiative, University of Maryland School of Medicine and the VA Medical Center, Baltimore, Maryland, USA
| | - Michael E Farkouh
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Donald M Lloyd-Jones
- Department of Preventive Medicine, Northwestern Feinberg School of Medicine, Chicago, Illinois, USA
| | - Valentin Fuster
- Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
9
|
Taşkin E, Bağci H, Turan MK. Investigation of associations between apolipoprotein A5 and C3 gene polymorphisms with plasma triglyceride and lipid levels. REVISTA DA ASSOCIACAO MEDICA BRASILEIRA (1992) 2023; 69:415-420. [PMID: 36921196 PMCID: PMC10004291 DOI: 10.1590/1806-9282.20221016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 12/20/2022] [Indexed: 03/12/2023]
Abstract
OBJECTIVE The aim of this study was to determine frequency and associations between APOA5 c.56C>G, -1131T>C, c.553G>T, and APOC3 -482C>T and SstI gene polymorphisms with hypertriglyceridemia. METHODS Under a case-control study model, 135 hypertriglyceridemic and 178 normotriglyceridemic control participants were recruited. Polymerase chain reaction and restriction fragment length polymorphism methods were utilized for genotyping. Statistical calculations were performed by comparing allele and genotype frequencies between groups. Clinical characteristics were compared between groups and intra-group genotypes. RESULTS APOC3 gene -482C>T and SstI polymorphic genotypes and allele frequencies were significantly higher in hypertriglyceridemic group (genotype frequencies, p=0.035, p=0.028, respectively). Regression analysis under unadjusted model confirmed that APOC3 -482C>T and SstI polymorphisms were significantly contributing to have hypertriglyceridemia (p=0.02, odds ratio [OR]=1.831 (95% confidence interval [CI] 1.095-3.060); p=0.04, OR=1.812 (1.031-3.183), respectively). APOA5 c.56C>G was in complete linkage disequilibrium with APOA5 c.553G>T polymorphism (D'=1). CONCLUSION For the first time in a population sample from Turkey, among the five polymorphisms of APOA5 and APOC3 genes investigated, APOC3 -482C>T and SstI polymorphisms were associated with elevated serum TG levels, while APOA5 c.56C>G, -1131T>C, and c.553G>T polymorphisms were not.
Collapse
Affiliation(s)
- Emre Taşkin
- Bandırma Onyedi Eylül Üniversitesi, Medical Faculty, Department of Medical Genetics – Bandırma, Turkey
- Corresponding author:
| | - Hasan Bağci
- Ondokuz Mayıs Üniversitesi, Medical Faculty, Department of Medical Biology – Samsun, Turkey
| | - Muhammed Kamil Turan
- Karabük Üniversitesi, Medical Faculty, Department of Medical Biology – Samsun, Turkey
| |
Collapse
|
10
|
Deshotels MR, Hadley TD, Roth M, Agha AM, Pulipati VP, Nugent AK, Virani SS, Nambi V, Moriarty PM, Davidson MH, Ballantyne CM. Genetic Testing for Hypertriglyceridemia in Academic Lipid Clinics: Implications for Precision Medicine-Brief Report. Arterioscler Thromb Vasc Biol 2022; 42:1461-1467. [PMID: 36325899 DOI: 10.1161/atvbaha.122.318445] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
BACKGROUND Severe hypertriglyceridemia is often caused by variants in genes of triglyceride metabolism. These variants include rare, heterozygous pathogenic variants (PVs), or multiple common, small-effect single nucleotide polymorphisms that can be quantified using a polygenic risk score (PRS). The role of genetic testing to examine PVs and PRS in predicting risk for pancreatitis and severity of hypertriglyceridemia is unknown. METHODS We examined the relationship of PVs and PRSs associated with hypertriglyceridemia with the highest recorded plasma triglyceride level and risk for acute pancreatitis in 363 patients from 3 academic lipid clinics who underwent genetic testing (GBinsight's Dyslipidemia Comprehensive Panel). Categories of hypertriglyceridemia included: normal triglyceride (<200 mg/dL), moderate (200-499 mg/dL), severe (500-999 mg/dL), or very severe (≥1000 mg/dL). RESULTS PVs and high PRSs were identified in 37 (10%) and 59 (16%) individuals, respectively. Patients with both had increased risk for very severe hypertriglyceridemia compared with those with neither genetic risk factor. Risk for acute pancreatitis was also increased in individuals with both genetic risk factors (odds ratio, 5.1 [P=0.02] after controlling for age, race, sex, body mass index, and highest triglyceride level), but not in individuals with PV or high PRS alone. CONCLUSIONS The presence of both PV and high PRS significantly increased risk for very severe hypertriglyceridemia and acute pancreatitis, whereas PV or PRS alone only modestly increased risk. Genetic testing may help identify patients with hypertriglyceridemia who have the greatest risk for developing pancreatitis and may derive the greatest benefit from novel triglyceride-lowering therapies.
Collapse
Affiliation(s)
- Matthew R Deshotels
- Sections of Cardiovascular Research and Cardiology, Department of Medicine, Baylor College of Medicine, Houston, TX (M.R.D., T.D.H., A.M.A., S.S.V., V.N., C.M.B.)
| | - Trevor D Hadley
- Sections of Cardiovascular Research and Cardiology, Department of Medicine, Baylor College of Medicine, Houston, TX (M.R.D., T.D.H., A.M.A., S.S.V., V.N., C.M.B.)
| | | | - Ali M Agha
- Sections of Cardiovascular Research and Cardiology, Department of Medicine, Baylor College of Medicine, Houston, TX (M.R.D., T.D.H., A.M.A., S.S.V., V.N., C.M.B.)
| | - Vishnu Priya Pulipati
- Section of Cardiology, Department of Medicine, University of Chicago, IL (V.P.P., M.H.D.)
| | - Anne K Nugent
- Division of Clinical Pharmacology, University of Kansas Medical Center, Kansas City (A.K.N., P.M.M.)
| | - Salim S Virani
- Section of Cardiology, Michael E. DeBakey VA Medical Center, Houston, TX (S.S.V., V.N.)
| | - Vijay Nambi
- Section of Cardiology, Michael E. DeBakey VA Medical Center, Houston, TX (S.S.V., V.N.)
| | - Patrick M Moriarty
- Division of Clinical Pharmacology, University of Kansas Medical Center, Kansas City (A.K.N., P.M.M.)
| | - Michael H Davidson
- Section of Cardiology, Department of Medicine, University of Chicago, IL (V.P.P., M.H.D.)
| | - Christie M Ballantyne
- Sections of Cardiovascular Research and Cardiology, Department of Medicine, Baylor College of Medicine, Houston, TX (M.R.D., T.D.H., A.M.A., S.S.V., V.N., C.M.B.)
| |
Collapse
|
11
|
Zhao Y, Zhang Q, Zhang X, Zhang Y, Lu Y, Ma X, Li W, Niu X, Zhang G, Chang M, Shi W, Tian Y. The roles of MMP8/MMP10 polymorphisms in ischemic stroke susceptibility. Brain Behav 2022; 12:e2797. [PMID: 36282475 PMCID: PMC9759140 DOI: 10.1002/brb3.2797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 09/30/2022] [Accepted: 10/09/2022] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Ischemic stroke (IS), a multifactorial and polygenic disease, is the most common cause of death. This study aimed to determine the roles of MMP8/MMP10 polymorphisms in IS susceptibility in the Chinese Han population. METHODS MMP8 rs1940475 and rs3765620, and MMP10 rs17860949 from 700 IS patients and 700 controls were genotyped by the MassARRAY iPLEX platform. The impact of polymorphisms on IS risk was evaluated by logistic regression analysis. RESULTS Our study indicated that rs17860949 in MMP10 was significantly associated with a reduced risk of IS (OR = 0.632, p = .002). Precisely, stratification analysis showed that rs17860949 was relate to a decreased susceptibility to IS in patients aged > 55 years (OR = 0.472, p < .001), males (OR = 0.632, p = .012), nonsmokers (OR = 0.610, p = .017), and nondrinkers (OR = 0.559, p = .006). All these significant findings were verified by false-positive report probability test. Furthermore, GG genotype and AG genotype in MMP8 rs3765620 polymorphism were related to a reduced triglycerides concentration (p = .018). CONCLUSION Our study suggests that rs17860949 in MMP10 may play a protective role in IS in the Chinese Han population.
Collapse
Affiliation(s)
- Yong Zhao
- Department of Neurology, Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No. 3 Hospital, the Affiliated Hospital of Northwest University, Xi'an, Shaanxi, China
| | - Qi Zhang
- The College of Life Sciences, Northwest University, Xi'an, Shaanxi, China
| | - Xiaobo Zhang
- The College of Life Sciences, Northwest University, Xi'an, Shaanxi, China
| | - Yu Zhang
- The College of Life Sciences, Northwest University, Xi'an, Shaanxi, China
| | - Ying Lu
- Department of Neurology, Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No. 3 Hospital, the Affiliated Hospital of Northwest University, Xi'an, Shaanxi, China
| | - Xiaojuan Ma
- Medical Research Center, Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No. 3 Hospital, the Affiliated Hospital of Northwest University, Xi'an, Shaanxi, China
| | - Weiping Li
- The College of Life Sciences, Northwest University, Xi'an, Shaanxi, China
| | - Xiaochen Niu
- The College of Life Sciences, Northwest University, Xi'an, Shaanxi, China
| | - Gejuan Zhang
- Department of Neurology, Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No. 3 Hospital, the Affiliated Hospital of Northwest University, Xi'an, Shaanxi, China
| | - Mingze Chang
- Department of Neurology, Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No. 3 Hospital, the Affiliated Hospital of Northwest University, Xi'an, Shaanxi, China
| | - Wenzhen Shi
- Medical Research Center, Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No. 3 Hospital, the Affiliated Hospital of Northwest University, Xi'an, Shaanxi, China
| | - Ye Tian
- Department of Neurology, Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No. 3 Hospital, the Affiliated Hospital of Northwest University, Xi'an, Shaanxi, China
| |
Collapse
|
12
|
Heidemann BE, Bemelmans RHH, Marais AD, Visseren FLJ, Koopal C. Clinical heterogeneity in monogenic chylomicronaemia. BMJ Case Rep 2022; 15:15/11/e251411. [PMID: 36423940 PMCID: PMC9693862 DOI: 10.1136/bcr-2022-251411] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Chylomicronaemia accompanies hypertriglyceridaemia, usually due to a polygenic predisposition in combination with secondary risk factors. Monogenic chylomicronaemia represents a small subgroup of patients with hypertriglyceridaemia. This article describes three patients and illustrates the heterogeneity in the presentation of monogenic chylomicronaemia. The first case is a man with mild hypertriglyceridaemia who is a compound heterozygote for two variants in the LMF1 gene, without relevant medical history. The second case is a woman who is a double heterozygote of variants in the LPL and APOA5 genes. She experienced pancreatitis. The third case is a man, with recurrent pancreatitis attributed to severe hypertriglyceridaemia and homozygous for a variant in the APOC2 gene. This article highlights that in patients with hypertriglyceridaemia, the absence of pancreatitis or the presence of mild hypertriglyceridaemia does not exclude monogenic chylomicronaemia. Genetic screening should be considered in patients with unexplained or severe hypertriglyceridaemia, to determine appropriate treatment and follow-up.
Collapse
Affiliation(s)
- Britt E Heidemann
- Department of Vascular Medicine, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| | - Remy H H Bemelmans
- Department of Internal Medicine, Ziekenhuis Gelderse Vallei, Ede, The Netherlands
| | - A David Marais
- Division of Chemical Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Frank L J Visseren
- Department of Vascular Medicine, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| | - Charlotte Koopal
- Department of Vascular Medicine, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| |
Collapse
|
13
|
Goldberg IJ, Gjini J, Fisher EA. Big Fish or No Fish; Eicosapentaenoic Acid and Cardiovascular Disease. Endocrinol Metab Clin North Am 2022; 51:625-633. [PMID: 35963632 DOI: 10.1016/j.ecl.2022.02.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Benefits of omega 3 fatty acids for cardiovascular and other diseases have been touted for more than 50 years. The one clear clinical benefit of these lipids is the reduction of circulating levels of triglycerides, making them a useful approach for the prevention of pancreatitis in severely hypertriglyceridemic patients. After a series of spectacularly failed clinical trials that were criticized for the choice of subjects and doses of omega 3 fatty acids used, Reduction of Cardiovascular Events with Icosapent Ethyl-Intervention Trial (REDUCE-IT) using a high dose of icosapent ethyl (IPE) reported a reduction in cardiovascular disease (CVD) events. However, this trial has generated controversy due to the use of mineral oil in the control group and the associated side effects of the IPA. This review will focus on the following topics: What are the epidemiologic data suggesting a benefit of omega 3 fatty acids? What might be the mechanisms for these benefits? Why have the clinical trials failed to resolve whether these fatty acids provide benefit? What choices should a clinician consider?
Collapse
Affiliation(s)
- Ira J Goldberg
- Division of Endocrinology, Diabetes and Metabolism, New York University Grossman School of Medicine, 435 First Avenue, SB 617, New York, NY 10016, USA.
| | - Jana Gjini
- Division of Endocrinology, Diabetes and Metabolism, New York University Grossman School of Medicine, 435 First Avenue, SB 617, New York, NY 10016, USA
| | - Edward A Fisher
- Division of Cardiology and Center for the Prevention of Cardiovascular Disease, New York University Grossman School of Medicine, 435 First Avenue, SB 704, New York, NY 10016, USA
| |
Collapse
|
14
|
Alcala-Diaz JF, Arenas-de Larriva AP, Torres-Peña JD, Rodriguez-Cantalejo F, Rangel-Zuñiga OA, Yubero-Serrano EM, Gutierrez-Mariscal FM, Cardelo MP, Luque RM, Ordovas JM, Perez-Martinez P, Delgado-Lista J, Lopez-Miranda J. A Gene Variation at the ZPR1 Locus (rs964184) Interacts With the Type of Diet to Modulate Postprandial Triglycerides in Patients With Coronary Artery Disease: From the Coronary Diet Intervention With Olive Oil and Cardiovascular Prevention Study. Front Nutr 2022; 9:885256. [PMID: 35782928 PMCID: PMC9247506 DOI: 10.3389/fnut.2022.885256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Accepted: 05/09/2022] [Indexed: 11/22/2022] Open
Abstract
Background and Aims rs964184 variant in the ZPR1 gene has been associated with blood lipids levels both in fasting and postprandial state and with the risk of myocardial infarction in high-risk cardiovascular patients. However, whether this association is modulated by diet has not been studied. Objective To investigate whether the type of diet (low-fat or Mediterranean diets) interacts with genetic variability at this loci to modulate fasting and postprandial lipids in coronary patients. Materials and Methods The genotype of the rs964184 polymorphism was determined in the Cordioprev Study population (NCT00924937). Fasting and Postprandial triglycerides were assessed before and after 3 years of dietary intervention with either a Mediterranean or a low-fat diet. Postprandial lipid assessment was done by a 4-h oral fat tolerance test (OFTT). Differences in triglycerides levels were identified using repeated-measures ANCOVA. Results From 523 patients (85% males, mean age 59 years) that completed the OFTT at baseline and after 3 years of intervention and had complete genotype information, 125 of them were carriers of the risk allele G. At the start of the study, these patients showed a higher fasting and postprandial triglycerides (TG) plasma levels. After 3 years of dietary intervention, G-carriers following a Mediterranean Diet maintained higher fasting and postprandial triglycerides, while those on the low-fat diet reduced their postprandial triglycerides to similar values to the population without the G-allele. Conclusion After 3 years of dietary intervention, the altered postprandial triglyceride response induced by genetic variability in the rs964184 polymorphism of the ZPR1 gene can be modulated by a low-fat diet, better than by a Mediterranean diet, in patients with coronary artery disease.
Collapse
Affiliation(s)
- Juan F. Alcala-Diaz
- Lipids and Atherosclerosis Unit, Department of Internal Medicine, Maimonides Biomedical Research Institute of Córdoba (IMIBIC), Reina Sofía University Hospital, University of Córdoba, Córdoba, Spain
- Centro de Investigación Biomédica en Red de Fisiología de la Obesidad y Nutricion (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain
| | - Antonio P. Arenas-de Larriva
- Lipids and Atherosclerosis Unit, Department of Internal Medicine, Maimonides Biomedical Research Institute of Córdoba (IMIBIC), Reina Sofía University Hospital, University of Córdoba, Córdoba, Spain
- Centro de Investigación Biomédica en Red de Fisiología de la Obesidad y Nutricion (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain
| | - Jose D. Torres-Peña
- Lipids and Atherosclerosis Unit, Department of Internal Medicine, Maimonides Biomedical Research Institute of Córdoba (IMIBIC), Reina Sofía University Hospital, University of Córdoba, Córdoba, Spain
- Centro de Investigación Biomédica en Red de Fisiología de la Obesidad y Nutricion (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain
| | | | - Oriol A. Rangel-Zuñiga
- Lipids and Atherosclerosis Unit, Department of Internal Medicine, Maimonides Biomedical Research Institute of Córdoba (IMIBIC), Reina Sofía University Hospital, University of Córdoba, Córdoba, Spain
- Centro de Investigación Biomédica en Red de Fisiología de la Obesidad y Nutricion (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain
| | - Elena M. Yubero-Serrano
- Lipids and Atherosclerosis Unit, Department of Internal Medicine, Maimonides Biomedical Research Institute of Córdoba (IMIBIC), Reina Sofía University Hospital, University of Córdoba, Córdoba, Spain
- Centro de Investigación Biomédica en Red de Fisiología de la Obesidad y Nutricion (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain
| | - Francisco M. Gutierrez-Mariscal
- Lipids and Atherosclerosis Unit, Department of Internal Medicine, Maimonides Biomedical Research Institute of Córdoba (IMIBIC), Reina Sofía University Hospital, University of Córdoba, Córdoba, Spain
- Centro de Investigación Biomédica en Red de Fisiología de la Obesidad y Nutricion (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain
| | - Magdalena P. Cardelo
- Lipids and Atherosclerosis Unit, Department of Internal Medicine, Maimonides Biomedical Research Institute of Córdoba (IMIBIC), Reina Sofía University Hospital, University of Córdoba, Córdoba, Spain
- Centro de Investigación Biomédica en Red de Fisiología de la Obesidad y Nutricion (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain
| | - Raul M. Luque
- Department of Cell Biology, Physiology, and Immunology, Maimonides Biomedical Research Institute of Córdoba (IMIBIC), Reina Sofía University Hospital, University of Córdoba, Córdoba, Spain
| | - Jose M. Ordovas
- Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA, United States
- Instituto Madrileño de Estudios Avanzados en Alimentación (IMDEA-Food), Madrid, Spain
- International Advisory Board, University Camilo José Cela, Madrid, Spain
| | - Pablo Perez-Martinez
- Lipids and Atherosclerosis Unit, Department of Internal Medicine, Maimonides Biomedical Research Institute of Córdoba (IMIBIC), Reina Sofía University Hospital, University of Córdoba, Córdoba, Spain
- Centro de Investigación Biomédica en Red de Fisiología de la Obesidad y Nutricion (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain
| | - Javier Delgado-Lista
- Lipids and Atherosclerosis Unit, Department of Internal Medicine, Maimonides Biomedical Research Institute of Córdoba (IMIBIC), Reina Sofía University Hospital, University of Córdoba, Córdoba, Spain
- Centro de Investigación Biomédica en Red de Fisiología de la Obesidad y Nutricion (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain
| | - Jose Lopez-Miranda
- Lipids and Atherosclerosis Unit, Department of Internal Medicine, Maimonides Biomedical Research Institute of Córdoba (IMIBIC), Reina Sofía University Hospital, University of Córdoba, Córdoba, Spain
- Centro de Investigación Biomédica en Red de Fisiología de la Obesidad y Nutricion (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain
- *Correspondence: Jose Lopez-Miranda,
| |
Collapse
|
15
|
van Meijeren AR, Ties D, de Koning MSL, van Dijk R, van Blokland IV, Lizana Veloz P, van Woerden G, Vliegenthart R, Pundziute G, Westenbrink DB, van der Harst P. Association of epicardial adipose tissue with different stages of coronary artery disease: A cross-sectional UK Biobank cardiovascular magnetic resonance imaging substudy. IJC HEART & VASCULATURE 2022; 40:101006. [PMID: 35372662 PMCID: PMC8971641 DOI: 10.1016/j.ijcha.2022.101006] [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: 12/06/2021] [Revised: 03/07/2022] [Accepted: 03/14/2022] [Indexed: 11/28/2022]
Abstract
Objective Increased epicardial adipose tissue (EAT) has been identified as a risk factor for the development of coronary artery disease (CAD). However, the exact role of EAT in the development of CAD is unclear. This study aims to compare EAT volumes between healthy controls and individuals with stable CAD and a history of myocardial infarction (MI). Furthermore, associations between clinical and biochemical parameters with EAT volumes are examined. Methods This retrospective cross-sectional study included 171 participants from the United Kingdom Biobank (56 healthy controls; 60 stable CAD; 55 post MI), whom were balanced for age, sex and body mass index (BMI). EAT volumes were quantified on end-diastolic cardiac magnetic resonance (CMR) imaging short-axis slices along the left and right ventricle and indexed for body surface area (iEAT) and iEAT volumes were compared between groups. Results iEAT volumes were comparable between control, CAD and MI cases (median [IQR]: 66.1[54.4-77.0] vs. 70.9[55.8-85.5] vs. 67.6[58.6-82.3] mL/m2, respectively (p > 0.005 for all). Increased HDL-cholesterol was associated with decreased iEAT volume (β = -14.8, CI = -24.6 to -4.97, p = 0.003) and suggestive associations (P-value < 0.05 and ≥ 0.005) were observed between iEAT and triglycerides (β = 3.26, CI = 0.42 to 6.09, p = 0.02), Apo-lipoprotein A (β = -16.3, CI = -30.3 to -2.24, p = 0.02) and LDL-cholesterol (β = 3.99, CI = -7.15 to -0.84, p = 0.01). Conclusions No significant differences in iEAT volumes were observed between patients with CAD, MI and healthy controls. Our results indicate the importance of correcting for confounding by CVD risk factors, including circulating lipid levels, when studying the relationship between EAT volume and CAD. Further mechanistic studies on causal pathways and the role of EAT composition are warranted.
Collapse
Affiliation(s)
- Anne Ruth van Meijeren
- University of Groningen, University Medical Center Groningen, Department of Cardiology, Groningen, the Netherlands
| | - Daan Ties
- University of Groningen, University Medical Center Groningen, Department of Cardiology, Groningen, the Netherlands
| | - Marie-Sophie L.Y. de Koning
- University of Groningen, University Medical Center Groningen, Department of Cardiology, Groningen, the Netherlands
| | - Randy van Dijk
- University of Groningen, University Medical Center Groningen, Department of Cardiology, Groningen, the Netherlands
| | - Irene V. van Blokland
- University of Groningen, University Medical Center Groningen, Department of Cardiology, Groningen, the Netherlands
| | | | - Gijs van Woerden
- University of Groningen, University Medical Center Groningen, Department of Cardiology, Groningen, the Netherlands
| | - Rozemarijn Vliegenthart
- University of Groningen, University Medical Center Groningen, Department of Radiology, Groningen, the Netherlands
| | - Gabija Pundziute
- University of Groningen, University Medical Center Groningen, Department of Cardiology, Groningen, the Netherlands
| | - Daan B. Westenbrink
- University of Groningen, University Medical Center Groningen, Department of Cardiology, Groningen, the Netherlands
| | - Pim van der Harst
- University of Groningen, University Medical Center Groningen, Department of Cardiology, Groningen, the Netherlands
- Department of Cardiology, University Medical Center Utrecht, Utrecht, the Netherlands
| |
Collapse
|
16
|
Liu X, Bao Y, Lin Z, Tang L, Mao P. Platelets inhibit development of atherosclerosis in atherosclerotic mice. Cell Cycle 2022; 21:1222-1232. [PMID: 35213268 PMCID: PMC9103360 DOI: 10.1080/15384101.2022.2044703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Platelets can protect from lipopolysaccharide-induced septic shock by inhibiting inflammation, but it is unknown whether platelets have an anti-atherosclerotic effect. The aim of this study was to investigate the effect of platelet transfusion on atherosclerosis (AS) in a mouse model of AS. Apolipoprotein E deficiency (ApoE-/-) mice were fed with a high-fat diet (HFD) for 8 weeks to establish a mouse model of AS. Mice weekly underwent bi-weekly injection with or without platelets during AS induction (HFD+platelet). Hematoxylin-eosin (H&E), Oil Red O, and Sudan IV stainings were used to assess pathological and morphological changes in the aortic tissue. Lipid levels, and liver and kidney function were examined using an automatic biochemical analyzer. Immune histochemical assays were used to detect the infiltration and distribution of inflammatory cytokines, including tumor necrosis factor-α (TNF-α), interleukin 1β (IL-1β), IL-6, and monocyte chemotactic protein-1 (MCP-1) in the aortic arch. Western blot and enzyme-linked immunosorbent assay (ELISA) were used to examine the expression levels of TNF-α, IL-1β, IL-6, and MCP-1 in the aorta or the peripheral blood, respectively. Compared with the HFD group, AS pathological lesions from the aortic arch in the HFD+platelet group were significantly smaller and alterations in the lipid metabolism were also less pronounced. Furthermore, TNF-α, IL-1β, IL-6, and MCP-1 levels were all significantly reduced in mice that received platelet injection. Platelets transfusion can effectively ameliorate lipid metabolism, suppress the inflammatory response in the vascular wall, and inhibit the development of AS in mice.
Collapse
Affiliation(s)
- Xiaowei Liu
- Department of Cardiology, Zhejiang Hospital, Hangzhou, P. R. China
| | - Yizhong Bao
- Zhejiang Provincial Key Laboratory of Geriatrics, Department of Geriatrics, Zhejiang Hospital, Hangzhou, P. R. China
| | - Zhang Lin
- Department of Basic Medicine, Zhejiang Chinese Medical University, Hangzhou, P. R. China
| | - Lijiang Tang
- Department of Cardiology, Zhejiang Hospital, Hangzhou, P. R. China,Lijiang Tang Department of Cardiology, Zhejiang Hospital, 12 Lingying Road, Hangzhou, Zhejiang 310013, P. R. China Department of Cardiology, Zhejiang Hospital, Hangzhou, P. R. China
| | - Ping Mao
- Department of Cardiology, Zhejiang Hospital, Hangzhou, P. R. China,CONTACT Ping Mao Department of Cardiology, Zhejiang Hospital, 12 Lingying Road, Hangzhou, Zhejiang310013, P. R. China
| |
Collapse
|
17
|
Al-Waili K, Al-Rasadi K, Al-Bulushi M, Habais M, Al-Mujaini A, Al-Yaarubi S, Rimbert A, Zadjali R, Khaniabadi PM, Al-Barwani H, Hasary S, Al-Dahmani ZM, Al-Badi H, Al-Maawali A, Zadjali F. The Genetic Spectrum of Familial Hypertriglyceridemia in Oman. Front Genet 2022; 13:886182. [PMID: 35669187 PMCID: PMC9163817 DOI: 10.3389/fgene.2022.886182] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 04/11/2022] [Indexed: 11/15/2022] Open
Abstract
Familial hypertriglyceridemia (F-HTG) is an autosomal disorder that causes severe elevation of serum triglyceride levels. It is caused by genetic alterations in LPL, APOC2, APOA5, LMF1, and GPIHBP1 genes. The mutation spectrum of F-HTG in Arabic populations is limited. Here, we report the genetic spectrum of six families of F-HTG of Arab ancestry in Oman. Methods: six Omani families affected with triglyceride levels >11.2 mmol/L were included in this study. Ampli-Seq sequencing of the selected gene panels was performed. Whole-exome sequencing and copy number variant analysis were also performed in cases with negative exome results. Three novel pathogenic missense variants in the LPL gene were identified, p.M328T, p.H229L, and p.S286G, along with a novel splice variant c.1322+15T > G. The LPL p.H229L variant existed in double heterozygous mutation with the APOA5 gene p.V153M variant. One family had a homozygous mutation in the LMF1 gene (c.G107A; p.G36D) and a heterozygous mutation in the LPL gene (c.G106A; p.D36N). All affected subjects did not have a serum deficiency of LPL protein. Genetic analysis in one family did not show any pathogenic variants even after whole-exome sequencing. These novel LPL and APOA5 mutations are not reported in other ethnic groups. This suggests that patients with F-HTG in Oman have a founder effect and are genetically unique. This warrants further analysis of patients of F-HTG in the Middle East for preventative and counseling purposes to limit the spread of the disease in a population of high consanguinity.
Collapse
Affiliation(s)
- Khalid Al-Waili
- Department of Clinical Biochemistry, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat, Oman
| | - Khalid Al-Rasadi
- Medical Research Centre, College of Medicine and Health Sciences, Department of Biochemistry, Sultan Qaboos University, Muscat, Oman
| | - Muna Al-Bulushi
- Department of Clinical Biochemistry, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat, Oman
| | - Mohammed Habais
- Department of Clinical Biochemistry, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat, Oman
| | - Abdullah Al-Mujaini
- Department of Ophthalmology, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat, Oman
| | - Saif Al-Yaarubi
- Department of Child Health, Sultan Qaboos University Hospital, Sultan Qaboos University, Muscat, Oman
| | - Antoine Rimbert
- Nantes Université, CHU Nantes, CNRS, INSERM, L’institut du Thorax, Nantes, France
| | - Razan Zadjali
- Department of Clinical Biochemistry, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat, Oman
| | - Pegah Moradi Khaniabadi
- Department of Clinical Biochemistry, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat, Oman
| | | | - Sana Hasary
- Department of Clinical Biochemistry, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat, Oman
| | - Zayana M. Al-Dahmani
- Department of Clinical Biochemistry, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat, Oman
| | - Hala Al-Badi
- Department of Clinical Biochemistry, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat, Oman
| | - Almundher Al-Maawali
- Department of Genetics, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat, Oman
| | - Fahad Zadjali
- Department of Clinical Biochemistry, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat, Oman
- *Correspondence: Fahad Zadjali,
| |
Collapse
|
18
|
Boucheniata C, Tessier N, Martel C. Editorial: Highlights in Lipids in Cardiovascular Disease: 2021. Front Cardiovasc Med 2022; 9:915262. [PMID: 35600471 PMCID: PMC9121121 DOI: 10.3389/fcvm.2022.915262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 04/19/2022] [Indexed: 11/13/2022] Open
Affiliation(s)
- Chaymae Boucheniata
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
- Research Center, Montreal Heart Institute, Montreal, QC, Canada
| | - Nolwenn Tessier
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
- Research Center, Montreal Heart Institute, Montreal, QC, Canada
| | - Catherine Martel
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
- Research Center, Montreal Heart Institute, Montreal, QC, Canada
| |
Collapse
|
19
|
Childhood Hypertriglyceridemia: Is It Time for a New Approach? Curr Atheroscler Rep 2022; 24:265-275. [PMID: 35107763 DOI: 10.1007/s11883-022-01000-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/06/2022] [Indexed: 11/03/2022]
Abstract
PURPOSE OF REVIEW Hypertriglyceridemia (HTG) is widely prevalent in youth. There is an unmet need for effective medications in the management of HTG in youth. The purpose of this review is to summarize the approach to HTG in acute and chronic settings, and highlight emerging therapies targeted at specific genes, proteins, and enzymes to selectively alter triglyceride (TG) metabolism. RECENT FINDINGS Genetic and lifestyle factors play a significant role in the pathophysiology of HTG. Severe elevation of TG poses a risk of acute pancreatitis, while mild-to-moderate HTG increases the risk for premature atherosclerotic cardiovascular disease (ASCVD) and, increasingly, has been linked with non-alcoholic fatty liver disease. Although a variety of therapeutic agents are in development, strict adherence to a heart healthy lifestyle, including dietary changes, remain the cornerstone of management for youth with HTG. In addition to lifestyle changes, pharmacological interventions, including fibrates, omega 3 fatty acids, and statins may be considered for management of moderate-to-severe HTG. In view of its association with premature cardiovascular disease (CVD), non-high-density-lipoprotein-C (non-HDL-C) is an important target for therapy in children with moderate HTG. Management of HTG is dependent on its etiology, concomitant symptoms, and degree of TG elevation. The last two decades have seen remarkable changes in drug development, specifically those that act through the lipoprotein lipase complex, including new targeted treatments such as inhibitors of apolipoprotein C3 and angiopoietin-like protein 3.
Collapse
|
20
|
Zhang M, Yin T, Xia F, Xia S, Zhou W, Zhang Y, Han X, Zhao K, Feng L, Dong R, Tian D, Yu Y, Liao J. Hypertriglyceridemia may contribute to stroke and pancreatitis: A case report and review of the literature. Front Endocrinol (Lausanne) 2022; 13:960343. [PMID: 36531479 PMCID: PMC9751361 DOI: 10.3389/fendo.2022.960343] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 11/17/2022] [Indexed: 12/03/2022] Open
Abstract
Hypertriglyceridemia (HTG) is one of the most common clinical dyslipidemia. Nevertheless, stroke and acute pancreatitis co-occurrence due to hypertriglyceridemia are extremely rare. We present a case of hypertriglyceridemia-associated stroke and pancreatitis in a 39-year-old woman. The patient's laboratory tests reported high triglyceride concentrations beyond the instrument's detection range, and radiological examination showed typical signs of cerebral infarction and acute pancreatitis. The patient received combined blood purification therapy, intravenous thrombolysis with urokinase, and conservative treatment of pancreatitis. We discuss the clinical features, pathogenesis, diagnosis, and treatment of hypertriglyceridemic stroke and pancreatitis combined with the relevant literature. We reviewed the mechanisms by which triglycerides contribute to atherosclerosis and acute pancreatitis. We point out the superiority of combined blood purification therapy and caution physicians about the effects of prescribed drugs on blood lipids.
Collapse
Affiliation(s)
- Mingyu Zhang
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Taoyuan Yin
- Department of Biliary-Pancreatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Feng Xia
- Department of Hepatic Surgery Center, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Suhong Xia
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Wangdong Zhou
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yu Zhang
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xu Han
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Kai Zhao
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Lina Feng
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ruonan Dong
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Dean Tian
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yan Yu
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- *Correspondence: Yan Yu, ; Jiazhi Liao,
| | - Jiazhi Liao
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- *Correspondence: Yan Yu, ; Jiazhi Liao,
| |
Collapse
|
21
|
Reilly NA, Lutgens E, Kuiper J, Heijmans BT, Jukema JW. Effects of fatty acids on T cell function: role in atherosclerosis. Nat Rev Cardiol 2021; 18:824-837. [PMID: 34253911 DOI: 10.1038/s41569-021-00582-9] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/01/2021] [Indexed: 01/08/2023]
Abstract
T cells are among the most common cell types present in atherosclerotic plaques and are increasingly being recognized as a central mediator in atherosclerosis development and progression. At the same time, triglycerides and fatty acids have re-emerged as crucial risk factors for atherosclerosis. Triglycerides and fatty acids are important components of the milieu to which the T cell is exposed from the circulation to the plaque, and increasing evidence shows that fatty acids influence T cell function. In this Review, we discuss the effects of fatty acids on four components of the T cell response - metabolism, activation, proliferation and polarization - and the influence of these changes on the pathogenesis of atherosclerosis. We also discuss how quiescent T cells can undergo a type of metabolic reprogramming induced by exposure to fatty acids in the circulation that influences the subsequent functions of T cells after activation, such as in atherosclerotic plaques.
Collapse
Affiliation(s)
- Nathalie A Reilly
- Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Centre, Leiden, Netherlands
- Department of Cardiology, Leiden University Medical Centre, Leiden, Netherlands
| | - Esther Lutgens
- Department of Medical Biochemistry, Amsterdam University Medical Centre, Amsterdam, Netherlands
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians Universität, Munich, Germany
- German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany
| | - Johan Kuiper
- Leiden Academic Centre for Drug Research, Division of Biotherapeutics, Leiden University, Leiden, Netherlands
| | - Bastiaan T Heijmans
- Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Centre, Leiden, Netherlands
| | - J Wouter Jukema
- Department of Cardiology, Leiden University Medical Centre, Leiden, Netherlands.
- Netherlands Heart Institute, Utrecht, Netherlands.
| |
Collapse
|
22
|
Kexin W, Yaodong D, Wen G, Rui W, Jiaxin Y, Xiaoli L, Hua S, Hailong G. Association of Increased Remnant Cholesterol and the Risk of Coronary Artery Disease: A Retrospective Study. Front Cardiovasc Med 2021; 8:740596. [PMID: 34778402 PMCID: PMC8585757 DOI: 10.3389/fcvm.2021.740596] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 08/31/2021] [Indexed: 12/28/2022] Open
Abstract
Background and Aims: Low-density lipoprotein cholesterol (LDL-C) is the primary target of lipid-lowering therapy in coronary artery disease (CAD). But some patients with the normal levels of LDL-C still suffer from CAD progression and malignant outcomes (e.g., major adverse cardiovascular events [MACEs]), and the mechanism is unclear. The previous prospective studies demonstrated that the remnant cholesterol (RC) and non-high-density lipoprotein cholesterol (non-HDL-C) were capable to predict the risk of CAD. This study evaluated the association between RC and non-HDL-C with the risk of CAD. Methods: In our study, 12,563 patients were enrolled. We categorized patients into four concordance/discordance groups according to the median of RC, LDL-C, and non-HDL-C. Then, we performed a propensity score matching (PSM) strategy. The unadjusted and adjusted multivariate logistic regression models were used to evaluate the relationship between the lipid concentrations. Results: In this study, 8,658 (68.9%) patients were male with a median age of 61 (54 and 67) years. The multivariate logistic regression showed the odds ratio (OR) of RC was 1.952 (CI = 1.276–2.988, p = 0.002). The OR of the low RC/high LDL-C group was 0.626 (CI = 0.504–0.778, p < 0.001) and the OR of the low RC/high non-HDL-C group was 0.574 (CI = 0.462–0.714, p < 0.001). The p-values for interaction between the RC and hypertension, diabetes were both < 0.001. Conclusion: Our study showed a significant association between the RC and CAD. The level of RC was more capable to reflect the risk of CAD than LDL-C and non-HDL-C. There was an interaction relationship between RC and age, gender, hypertension, diabetes, in CAD. But we did not find whether there was a relationship between the non-HDL-C and CAD.
Collapse
Affiliation(s)
- Wang Kexin
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Ding Yaodong
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Gao Wen
- Department of Cardiology, Bayannaoer City Hospital, Bayannaoer, China
| | - Wang Rui
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Yang Jiaxin
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Liu Xiaoli
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Shen Hua
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Ge Hailong
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
23
|
Favor OK, Pestka JJ, Bates MA, Lee KSS. Centrality of Myeloid-Lineage Phagocytes in Particle-Triggered Inflammation and Autoimmunity. FRONTIERS IN TOXICOLOGY 2021; 3:777768. [PMID: 35295146 PMCID: PMC8915915 DOI: 10.3389/ftox.2021.777768] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 10/19/2021] [Indexed: 12/14/2022] Open
Abstract
Exposure to exogenous particles found as airborne contaminants or endogenous particles that form by crystallization of certain nutrients can activate inflammatory pathways and potentially accelerate autoimmunity onset and progression in genetically predisposed individuals. The first line of innate immunological defense against particles are myeloid-lineage phagocytes, namely macrophages and neutrophils, which recognize/internalize the particles, release inflammatory mediators, undergo programmed/unprogrammed death, and recruit/activate other leukocytes to clear the particles and resolve inflammation. However, immunogenic cell death and release of damage-associated molecules, collectively referred to as "danger signals," coupled with failure to efficiently clear dead/dying cells, can elicit unresolved inflammation, accumulation of self-antigens, and adaptive leukocyte recruitment/activation. Collectively, these events can promote loss of immunological self-tolerance and onset/progression of autoimmunity. This review discusses critical molecular mechanisms by which exogenous particles (i.e., silica, asbestos, carbon nanotubes, titanium dioxide, aluminum-containing salts) and endogenous particles (i.e., monosodium urate, cholesterol crystals, calcium-containing salts) may promote unresolved inflammation and autoimmunity by inducing toxic responses in myeloid-lineage phagocytes with emphases on inflammasome activation and necrotic and programmed cell death pathways. A prototypical example is occupational exposure to respirable crystalline silica, which is etiologically linked to systemic lupus erythematosus (SLE) and other human autoimmune diseases. Importantly, airway instillation of SLE-prone mice with crystalline silica elicits severe pulmonary pathology involving accumulation of particle-laden alveolar macrophages, dying and dead cells, nuclear and cytoplasmic debris, and neutrophilic inflammation that drive cytokine, chemokine, and interferon-regulated gene expression. Silica-induced immunogenic cell death and danger signal release triggers accumulation of T and B cells, along with IgG-secreting plasma cells, indicative of ectopic lymphoid tissue neogenesis, and broad-spectrum autoantibody production in the lung. These events drive early autoimmunity onset and accelerate end-stage autoimmune glomerulonephritis. Intriguingly, dietary supplementation with ω-3 fatty acids have been demonstrated to be an intervention against silica-triggered murine autoimmunity. Taken together, further insight into how particles drive immunogenic cell death and danger signaling in myeloid-lineage phagocytes and how these responses are influenced by the genome will be essential for identification of novel interventions for preventing and treating inflammatory and autoimmune diseases associated with these agents.
Collapse
Affiliation(s)
- Olivia K. Favor
- Department of Pharmacology and Toxicology, College of Osteopathic Medicine, Michigan State University, East Lansing, MI, United States
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, United States
| | - James J. Pestka
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, United States
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI, United States
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI, United States
| | - Melissa A. Bates
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, United States
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI, United States
| | - Kin Sing Stephen Lee
- Department of Pharmacology and Toxicology, College of Osteopathic Medicine, Michigan State University, East Lansing, MI, United States
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, United States
- Department of Chemistry, Michigan State University, East Lansing, MI, United States
| |
Collapse
|
24
|
Wang A, Tian X, Zuo Y, Wu J, Tang H, Wang Y, Zhao X. Association of remnant cholesterol with intra- and extra-cranial atherosclerosis in Chinese community population. ATHEROSCLEROSIS PLUS 2021; 46:20-26. [PMID: 36643726 PMCID: PMC9833241 DOI: 10.1016/j.athplu.2021.10.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 08/27/2021] [Accepted: 10/22/2021] [Indexed: 01/18/2023]
Abstract
Background Multifocal atherosclerosis has dramatically increased annual risk of adverse cardiovascular events than single artery affected. Triglyceride-rich lipoprotein (TRL) has been implicated in the early development of atherosclerosis.However, evidence on the effect of remnant cholesterol, a major atherogenic component of TRL, on multifocal atherosclerosis in Chinese health asymptomatic subjects is insufficient. This study aimed to investigate the association of remnant cholesterol with intra- and extra-cranial atherosclerosis in Chinese population. Methods This study enrolled 3665 participants (median age 52.31 years) from the Asymptomatic Polyvascular Abnormalities Community study. Parameters of intra- and extra-cranial atherosclerosis includes intracranial artery stenosis (ICAS), carotid plaque, carotid artery stenosis (CAS), and carotid hypertrophy (intima-media thickness >0.9 mm). Logistic regression was used to assess these associations. Results The prevalence of vascular atherosclerosis significantly increased with increasing remnant cholesterol quartiles (P for trend <0.0001). In the multivariable-adjusted model, the odds ratio with 95% confidence interval comparing participants in Q4 versus Q1 of remnant cholesterol was 1.73 (1.29-2.31) for ICAS, 1.54 (1.22-1.94) for carotid plaque, 1.47 (1.17-1.84) for CAS, and 1.93 (1.48-2.52) for carotid hypertrophy, respectively. Furthermore, multivariable-adjusted spline regression showed S-shaped associations between remnant cholesterol and these outcomes. Conclusion Individuals with a high level of remnant cholesterol had a higher risk of intra- and extra-cranial atherosclerosis in Chinese population. Interventions aimed at reducing remnant cholesterol to prevent atherosclerotic diseases warrant further investigations.
Collapse
Affiliation(s)
- Anxin Wang
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Xue Tian
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, Beijing, China
- Beijing Municipal Key Laboratory of Clinical Epidemiology, Beijing, China
| | - Yingting Zuo
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, Beijing, China
- Beijing Municipal Key Laboratory of Clinical Epidemiology, Beijing, China
| | - Jianwei Wu
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Hefei Tang
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yongjun Wang
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Corresponding author. China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, No.119 South 4th Ring West Road, Fengtai District, Beijing, 100070, China.
| | - Xingquan Zhao
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Research Unit of Artificial Intelligence in Cerebrovascular Disease, Chinese Academy of Medical Sciences, Beijing, China
- Corresponding author. China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, No. 119 South 4th Ring West Road, Fengtai District, Beijing 100070, China.
| |
Collapse
|
25
|
Jiao H, Zhao Y, Zhou Z, Li W, Li B, Gu G, Luo Y, Shuai X, Fan C, Wu L, Chen J, Huang Q, Wang F, Liu J. Identifying Circular RNAs in HepG2 Expressing Genotype IV Swine Hepatitis E Virus ORF3 Via Whole Genome Sequencing. Cell Transplant 2021; 30:9636897211055042. [PMID: 34699255 PMCID: PMC8552397 DOI: 10.1177/09636897211055042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Swine hepatitis E (SHE) is a new type of zoonotic infectious disease caused by swine hepatitis E virus (SHEV). Open reading frame 3 (ORF3) is a key regulatory and virulent protein of SHEV. Circular RNAs (circRNAs) are a special kind of non-coding RNA molecule, which has a closed ring structure. In this study, to identify the circRNA profile in host cells affected by SHEV ORF3, adenovirus ADV4-ORF3 mediated the overexpression of ORF3 in HepG2 cells, whole genome sequencing was used to investigate the differentially expressed circRNAs, GO and KEGG were performed to enrichment analyze of differentially expressed circRNA-hosting gene, and Targetscan and miRanda softwares were used to analyze the interaction between circRNA and miRNA. The results showed adenovirus successfully mediated the overexpression of ORF3 in HepG2 cells, 1,105 up-regulation circRNAs and 1,556 down-regulation circRNAs were identified in ADV4-ORF3 infection group compared with the control. GO function enrichment analysis of differentially expressed circRNAs-hosting genes classified three main categories (cellular component, biological process and molecular function). KEGG pathway enrichment analysis scatter plot showed the pathway term of top20. The circRNAs with top10 number of BS sites for qRT-PCR validation were selected to confirmed, the results indicated that the up-regulated hsa_circ_0001423 and hsa_circ_0006404, and down-regulated of hsa_circ_0004833 and hsa_circ_0007444 were consistent with the sequencing data. Our findings first preliminarily found that ORF3 protein may affect triglyceride activation (GO:0006642) and riboflavin metabolism (ko00740) in HepG2 cells, which provides a scientific basis for further elucidating the effect of ORF3 on host lipid metabolism and the mechanism of SHEV infection.
Collapse
Affiliation(s)
- Hanwei Jiao
- College of Veterinary Medicine, Southwest University, Chongqing, China.,Immunology Research Center, Medical Research Institute, Southwest University, Chongqing, China.,Chongqing Veterinary Scientific Engineering Research Center, Southwest University, Chongqing, China
| | - Yu Zhao
- College of Veterinary Medicine, Southwest University, Chongqing, China.,Institute of Animal Husbandry and Veterinary Medicine of Guizhou Academy of Agricultural Science, Guiyang, China
| | - Zhixiong Zhou
- College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Wenjie Li
- College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Bowen Li
- College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Guojing Gu
- College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Yichen Luo
- College of Veterinary Medicine, Southwest University, Chongqing, China.,Immunology Research Center, Medical Research Institute, Southwest University, Chongqing, China.,Chongqing Veterinary Scientific Engineering Research Center, Southwest University, Chongqing, China
| | - Xuehong Shuai
- College of Veterinary Medicine, Southwest University, Chongqing, China.,Immunology Research Center, Medical Research Institute, Southwest University, Chongqing, China.,Chongqing Veterinary Scientific Engineering Research Center, Southwest University, Chongqing, China
| | - Cailiang Fan
- Rongchang Animal Epidemic Prevention and Control Center, Chongqing, Rongchang, China
| | - Li Wu
- College of Veterinary Medicine, Southwest University, Chongqing, China.,Chongqing Veterinary Scientific Engineering Research Center, Southwest University, Chongqing, China
| | - Jixuan Chen
- College of Veterinary Medicine, Southwest University, Chongqing, China.,Chongqing Veterinary Scientific Engineering Research Center, Southwest University, Chongqing, China
| | - Qingzhou Huang
- College of Veterinary Medicine, Southwest University, Chongqing, China.,Chongqing Veterinary Scientific Engineering Research Center, Southwest University, Chongqing, China
| | - Fengyang Wang
- Hainan Key Lab of Tropical Animal Reproduction and Breeding and Epidemic Disease Research, College of Animal Science and Technology, Hainan University, Haikou, China
| | - Juan Liu
- College of Veterinary Medicine, Southwest University, Chongqing, China.,Immunology Research Center, Medical Research Institute, Southwest University, Chongqing, China.,Chongqing Veterinary Scientific Engineering Research Center, Southwest University, Chongqing, China
| |
Collapse
|
26
|
Khetarpal SA, Vitali C, Levin MG, Klarin D, Park J, Pampana A, Millar JS, Kuwano T, Sugasini D, Subbaiah PV, Billheimer JT, Natarajan P, Rader DJ. Endothelial lipase mediates efficient lipolysis of triglyceride-rich lipoproteins. PLoS Genet 2021; 17:e1009802. [PMID: 34543263 PMCID: PMC8483387 DOI: 10.1371/journal.pgen.1009802] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 09/30/2021] [Accepted: 09/02/2021] [Indexed: 11/18/2022] Open
Abstract
Triglyceride-rich lipoproteins (TRLs) are circulating reservoirs of fatty acids used as vital energy sources for peripheral tissues. Lipoprotein lipase (LPL) is a predominant enzyme mediating triglyceride (TG) lipolysis and TRL clearance to provide fatty acids to tissues in animals. Physiological and human genetic evidence support a primary role for LPL in hydrolyzing TRL TGs. We hypothesized that endothelial lipase (EL), another extracellular lipase that primarily hydrolyzes lipoprotein phospholipids may also contribute to TRL metabolism. To explore this, we studied the impact of genetic EL loss-of-function on TRL metabolism in humans and mice. Humans carrying a loss-of-function missense variant in LIPG, p.Asn396Ser (rs77960347), demonstrated elevated plasma TGs and elevated phospholipids in TRLs, among other lipoprotein classes. Mice with germline EL deficiency challenged with excess dietary TG through refeeding or a high-fat diet exhibited elevated TGs, delayed dietary TRL clearance, and impaired TRL TG lipolysis in vivo that was rescued by EL reconstitution in the liver. Lipidomic analyses of postprandial plasma from high-fat fed Lipg-/- mice demonstrated accumulation of phospholipids and TGs harboring long-chain polyunsaturated fatty acids (PUFAs), known substrates for EL lipolysis. In vitro and in vivo, EL and LPL together promoted greater TG lipolysis than either extracellular lipase alone. Our data positions EL as a key collaborator of LPL to mediate efficient lipolysis of TRLs in humans and mice.
Collapse
Affiliation(s)
- Sumeet A. Khetarpal
- Departments of Medicine and Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America,Cardiovascular Research Center, Massachusetts General Hospital, Boston, Massachusetts, United States of America
| | - Cecilia Vitali
- Departments of Medicine and Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Michael G. Levin
- Division of Cardiovascular Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America,Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America,Corporal Michael J. Crescenz VA Medical Center, Philadelphia, Pennsylvania, United States of America
| | - Derek Klarin
- Boston VA Healthcare System, Boston, Massachusetts, United States of America,Center for Genomic Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America,Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America
| | - Joseph Park
- Departments of Medicine and Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Akhil Pampana
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, Massachusetts, United States of America,Center for Genomic Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America,Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America,Department of Medicine, Harvard Medical School, Boston, Massachusetts, United States of America
| | - John S. Millar
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Takashi Kuwano
- Departments of Medicine and Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Dhavamani Sugasini
- Section of Endocrinology, Department of Medicine, University of Illinois at Chicago; Jesse Brown VA Medical Center, Chicago, Illinois, United States of America
| | - Papasani V. Subbaiah
- Section of Endocrinology, Department of Medicine, University of Illinois at Chicago; Jesse Brown VA Medical Center, Chicago, Illinois, United States of America
| | - Jeffrey T. Billheimer
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Pradeep Natarajan
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, Massachusetts, United States of America,Center for Genomic Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America,Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America,Department of Medicine, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Daniel J. Rader
- Departments of Medicine and Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America,* E-mail:
| |
Collapse
|
27
|
Duran EK, Pradhan AD. Triglyceride-Rich Lipoprotein Remnants and Cardiovascular Disease. Clin Chem 2021; 67:183-196. [PMID: 33409533 DOI: 10.1093/clinchem/hvaa296] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 10/29/2020] [Indexed: 12/15/2022]
Abstract
BACKGROUND Triglycerides, cholesterol, and their metabolism are linked due to shared packaging and transport within circulating lipoprotein particles. While a case for a causal role of cholesterol-carrying low-density lipoproteins (LDLs) in atherosclerosis is well made, the body of scientific evidence for a causal role of triglyceride-rich lipoproteins (TRLs) is rapidly growing, with multiple lines of evidence (old and new) providing robust support. CONTENT This review will discuss current perspectives and accumulated evidence that an overabundance of remnant lipoproteins stemming from intravascular remodeling of nascent TRLs-chylomicrons and very low-density lipoproteins (VLDL)-results in a proatherogenic milieu that augments cardiovascular risk. Basic mechanisms of TRL metabolism and clearance will be summarized, assay methods reviewed, and pivotal clinical studies highlighted. SUMMARY Remnant lipoproteins are rendered highly atherogenic by their high cholesterol content, altered apolipoprotein composition, and physicochemical properties. The aggregate findings from multiple lines of evidence suggest that TRL remnants play a central role in residual cardiovascular risk.
Collapse
Affiliation(s)
- Edward K Duran
- Cardiovascular Division, University of Minnesota Medical Center, Minneapolis, MN
| | - Aruna D Pradhan
- Division of Preventive Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA.,Division of Cardiovascular Medicine, VA Boston Medical Center, Boston, MA
| |
Collapse
|
28
|
Abstract
PURPOSE OF REVIEW Triglycerides (TGs) are measured as part of routine lipid profiles but their relationship to cardiovascular disease (CVD) risk has been controversial and overshadowed by high-density lipoprotein cholesterol (HDL-C). RECENT FINDINGS Epidemiological studies show a clear relationship of TG-containing lipoproteins including remnant particles with CVD risk with the effect being most clearly demonstrated through the excess risk captured by non-HDL-C compared with low-density lipoprotein-cholesterol (LDL-C). Mendelian randomisation studies show a consistent relationship of gene variants linked to TG metabolism with rates of CVD. Furthermore, meta-analyses of intervention trials with statins and other nonstatin drugs also suggest that reducing TGs is associated with benefits on rates of CVD events. Historical subgroup data from fibrate trials suggest benefits in patients with high TG:HDL ratios but seem to add little to optimized statin therapy. Recent trials with omega-3 fatty acids (specifically eicosapentaenoic acid) have suggested that high-dose formulations in contrast to low dose formulations have benefits on CVD outcomes. SUMMARY Further studies with newer agents are required to determine the place of TG-lowering drugs in therapeutic pathways. Trials with agents such as pemafibrate and vupanorsen may finally answer these questions.
Collapse
Affiliation(s)
| | - Anthony S Wierzbicki
- Metabolic Medicine/Chemical Pathology, Guy's & St Thomas Hospitals, London SE1 7EH, UK
| |
Collapse
|
29
|
Määttä J, Serpi R, Hörkkö S, Izzi V, Myllyharju J, Dimova EY, Koivunen P. Genetic Ablation of Transmembrane Prolyl 4-Hydroxylase Reduces Atherosclerotic Plaques in Mice. Arterioscler Thromb Vasc Biol 2021; 41:2128-2140. [PMID: 34039020 DOI: 10.1161/atvbaha.121.316034] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Jenni Määttä
- Biocenter Oulu, Faculty of Biochemistry and Molecular Medicine, Oulu Center for Cell-Matrix Research (J. Määttä, R.S., V.I., J. Myllyharju, E.Y.D., P.K.), University of Oulu, Finland
| | - Raisa Serpi
- Biocenter Oulu, Faculty of Biochemistry and Molecular Medicine, Oulu Center for Cell-Matrix Research (J. Määttä, R.S., V.I., J. Myllyharju, E.Y.D., P.K.), University of Oulu, Finland
| | - Sohvi Hörkkö
- Institute of Biomedicine (S.H.), University of Oulu, Finland
| | - Valerio Izzi
- Faculty of Medicine (V.I.), University of Oulu, Finland
- Finnish Cancer Institute, Helsinki, Finland (V.I.)
| | - Johanna Myllyharju
- Biocenter Oulu, Faculty of Biochemistry and Molecular Medicine, Oulu Center for Cell-Matrix Research (J. Määttä, R.S., V.I., J. Myllyharju, E.Y.D., P.K.), University of Oulu, Finland
| | - Elitsa Y Dimova
- Biocenter Oulu, Faculty of Biochemistry and Molecular Medicine, Oulu Center for Cell-Matrix Research (J. Määttä, R.S., V.I., J. Myllyharju, E.Y.D., P.K.), University of Oulu, Finland
| | - Peppi Koivunen
- Biocenter Oulu, Faculty of Biochemistry and Molecular Medicine, Oulu Center for Cell-Matrix Research (J. Määttä, R.S., V.I., J. Myllyharju, E.Y.D., P.K.), University of Oulu, Finland
| |
Collapse
|
30
|
Abstract
PURPOSE OF REVIEW Hypertriglyceridemia is a common dyslipidemia associated with an increased risk of cardiovascular disease and pancreatitis. Severe hypertriglyceridemia may sometimes be a monogenic condition. However, in the vast majority of patients, hypertriglyceridemia is due to the cumulative effect of multiple genetic risk variants along with lifestyle factors, medications, and disease conditions that elevate triglyceride levels. In this review, we will summarize recent progress in the understanding of the genetic basis of hypertriglyceridemia. RECENT FINDINGS More than 300 genetic loci have been identified for association with triglyceride levels in large genome-wide association studies. Studies combining the loci into polygenic scores have demonstrated that some hypertriglyceridemia phenotypes previously attributed to monogenic inheritance have a polygenic basis. The new genetic discoveries have opened avenues for the development of more effective triglyceride-lowering treatments and raised interest towards genetic screening and tailored treatments against hypertriglyceridemia. The discovery of multiple genetic loci associated with elevated triglyceride levels has led to improved understanding of the genetic basis of hypertriglyceridemia and opened new translational opportunities.
Collapse
Affiliation(s)
- Germán D. Carrasquilla
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Mærsk Building, Blegdamsvej 3B, 2200 Copenhagen, Denmark
| | - Malene Revsbech Christiansen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Mærsk Building, Blegdamsvej 3B, 2200 Copenhagen, Denmark
| | - Tuomas O. Kilpeläinen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Mærsk Building, Blegdamsvej 3B, 2200 Copenhagen, Denmark
| |
Collapse
|
31
|
Goldberg IJ, Cabodevilla AG, Samovski D, Cifarelli V, Basu D, Abumrad NA. Lipolytic enzymes and free fatty acids at the endothelial interface. Atherosclerosis 2021; 329:1-8. [PMID: 34130222 DOI: 10.1016/j.atherosclerosis.2021.05.018] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 05/12/2021] [Accepted: 05/21/2021] [Indexed: 01/17/2023]
Abstract
Lipids released from circulating lipoproteins by intravascular action of lipoprotein lipase (LpL) reach parenchymal cells in tissues with a non-fenestrated endothelium by transfer through or around endothelial cells. The actions of LpL are controlled at multiple sites, its synthesis and release by myocytes and adipocytes, its transit and association with the endothelial cell luminal surface, and finally its activation and inhibition by a number of proteins and by its product non-esterified fatty acids. Multiple pathways mediate endothelial transit of lipids into muscle and adipose tissues. These include movement of fatty acids via the endothelial cell fatty acid transporter CD36 and movement of whole or partially LpL-hydrolyzed lipoproteins via other apical endothelial cell receptors such as SR-B1and Alk1. Lipids also likely change the barrier function of the endothelium and operation of the paracellular pathway around endothelial cells. This review summarizes in vitro and in vivo support for the key role of endothelial cells in delivery of lipids and highlights incompletely understood processes that are the focus of active investigation.
Collapse
Affiliation(s)
- Ira J Goldberg
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA.
| | - Ainara G Cabodevilla
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA
| | - Dmitri Samovski
- Department of Medicine, Center for Human Nutrition, Washington University School of Medicine, Saint Louis, MO, USA
| | - Vincenza Cifarelli
- Department of Medicine, Center for Human Nutrition, Washington University School of Medicine, Saint Louis, MO, USA
| | - Debapriya Basu
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA
| | - Nada A Abumrad
- Department of Medicine, Center for Human Nutrition, Washington University School of Medicine, Saint Louis, MO, USA.
| |
Collapse
|
32
|
Lahoz C, Mostaza JM. Familial hypertriglyceridemia/polygenic hypertrigliceridemia. CLINICA E INVESTIGACION EN ARTERIOSCLEROSIS : PUBLICACION OFICIAL DE LA SOCIEDAD ESPANOLA DE ARTERIOSCLEROSIS 2021; 33 Suppl 2:37-42. [PMID: 34006352 DOI: 10.1016/j.arteri.2020.12.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 12/29/2020] [Indexed: 12/26/2022]
Abstract
For decades, familial hypertriglyceridemia (HTG) has been considered a specific entity characterized by an increase in VLDL particles and an autosomal dominant inheritance pattern. In the genomics era, it has been proven that familial HTG, although it could be grouped in families, had a polygenic inheritance in which the phenotype would be determined by concomitant environmental factors. Hence its inclusion in the group of polygenic HTGs. Clinically, they are characterized by moderate HTG, with the consequent increase in cardiovascular risk, and in rare cases, by severe HTG with risk of acute pancreatitis. Treatment will be based on controlling environmental factors, implementing hygienic-dietetic measures and sometimes drugs, to reduce cardiovascular risk in moderate HTGs and acute pancreatitis risk in severe HTGs.
Collapse
Affiliation(s)
- Carlos Lahoz
- Unidad de Lípidos y Riesgo Vascular, Hospital Carlos III, Madrid, España.
| | - José María Mostaza
- Unidad de Lípidos y Riesgo Vascular, Hospital Carlos III, Madrid, España
| |
Collapse
|
33
|
Abstract
PURPOSE OF REVIEW The relationship between elevated triglyceride levels (i.e. hypertriglyceridemia) and risk of atherosclerotic cardiovascular disease (ASCVD) has been investigated for decades. Recent genetic studies have sought to resolve the decades-old question of a causal relationship. RECENT FINDINGS Genetic studies seem to demonstrate associations between elevated triglyceride levels and ASCVD risk. Mendelian randomization studies suggest this association may be causal. However, simultaneous pleiotropic effects of metabolically linked lipid variables - such as non-HDL cholesterol, apolipoprotein B and HDL cholesterol -- often go unaccounted for in these studies. Complex underlying pleiotropic interactions of triglycerides with these lipid fractions together with unmeasured intercalated nonlipid-related mechanisms, such as inflammation and coagulation, impair the ability of genetic studies to implicate a direct role for triglycerides on ASCVD risk. One potential mechanism seems largely driven by the cholesterol carried within triglyceride-rich lipoproteins and their remnants, rather than their triglyceride content. SUMMARY Although the exact mechanisms linking elevated triglyceride levels to ASCVD remain to be determined, new therapeutics that reduce triglyceride levels might be advantageous in certain patients. Newer investigational triglyceride-lowering therapies derived from human genetics target key proteins, such as apo C-III and ANGPTL3. Although these treatments clearly lower triglyceride levels, their efficacy in atherosclerotic risk reduction remains unproven.
Collapse
|
34
|
Kowara M, Cudnoch-Jedrzejewska A. Pathophysiology of Atherosclerotic Plaque Development-Contemporary Experience and New Directions in Research. Int J Mol Sci 2021; 22:ijms22073513. [PMID: 33805303 PMCID: PMC8037897 DOI: 10.3390/ijms22073513] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 03/22/2021] [Accepted: 03/22/2021] [Indexed: 01/12/2023] Open
Abstract
Atherosclerotic plaque is the pathophysiological basis of important and life-threatening diseases such as myocardial infarction. Although key aspects of the process of atherosclerotic plaque development and progression such as local inflammation, LDL oxidation, macrophage activation, and necrotic core formation have already been discovered, many molecular mechanisms affecting this process are still to be revealed. This minireview aims to describe the current directions in research on atherogenesis and to summarize selected studies published in recent years-in particular, studies on novel cellular pathways, epigenetic regulations, the influence of hemodynamic parameters, as well as tissue and microorganism (microbiome) influence on atherosclerotic plaque development. Finally, some new and interesting ideas are proposed (immune cellular heterogeneity, non-coding RNAs, and immunometabolism) which will hopefully bring new discoveries in this area of investigation.
Collapse
|
35
|
Read RW, Schlauch KA, Lombardi VC, Cirulli ET, Washington NL, Lu JT, Grzymski JJ. Genome-Wide Identification of Rare and Common Variants Driving Triglyceride Levels in a Nevada Population. Front Genet 2021; 12:639418. [PMID: 33763119 PMCID: PMC7982958 DOI: 10.3389/fgene.2021.639418] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 02/12/2021] [Indexed: 01/08/2023] Open
Abstract
Clinical conditions correlated with elevated triglyceride levels are well-known: coronary heart disease, hypertension, and diabetes. Underlying genetic and phenotypic mechanisms are not fully understood, partially due to lack of coordinated genotypic-phenotypic data. Here we use a subset of the Healthy Nevada Project, a population of 9,183 sequenced participants with longitudinal electronic health records to examine consequences of altered triglyceride levels. Specifically, Healthy Nevada Project participants sequenced by the Helix Exome+ platform were cross-referenced to their electronic medical records to identify: (1) rare and common single-variant genome-wide associations; (2) gene-based associations using a Sequence Kernel Association Test; (3) phenome-wide associations with triglyceride levels; and (4) pleiotropic variants linked to triglyceride levels. The study identified 549 significant single-variant associations (p < 8.75 × 10-9), many in chromosome 11's triglyceride hotspot: ZPR1, BUD13, APOC3, APOA5. A well-known protective loss-of-function variant in APOC3 (R19X) was associated with a 51% decrease in triglyceride levels in the cohort. Sixteen gene-based triglyceride associations were identified; six of these genes surprisingly did not include a single variant with significant associations. Results at the variant and gene level were validated with the UK Biobank. The combination of a single-variant genome-wide association, a gene-based association method, and phenome wide-association studies identified rare and common variants, genes, and phenotypes associated with elevated triglyceride levels, some of which may have been overlooked with standard approaches.
Collapse
Affiliation(s)
- Robert W. Read
- Center for Genomic Medicine, Desert Research Institute, Reno, NV, United States
| | - Karen A. Schlauch
- Center for Genomic Medicine, Desert Research Institute, Reno, NV, United States
| | - Vincent C. Lombardi
- Department of Microbiology and Immunology, School of Medicine, University of Nevada, Reno, Reno, NV, United States
| | | | | | - James T. Lu
- Helix Opco, LLC., San Mateo, CA, United States
| | - Joseph J. Grzymski
- Center for Genomic Medicine, Desert Research Institute, Reno, NV, United States
- Renown Health, Reno, NV, United States
| |
Collapse
|
36
|
Efficacy of Polyphenols in the Management of Dyslipidemia: A Focus on Clinical Studies. Nutrients 2021; 13:nu13020672. [PMID: 33669729 PMCID: PMC7922034 DOI: 10.3390/nu13020672] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/15/2021] [Accepted: 02/16/2021] [Indexed: 02/06/2023] Open
Abstract
Polyphenols (PLPs), phytochemicals found in a wide range of plant-based foods, have gained extensive attention in view of their antioxidant, anti-inflammatory, immunomodulatory and several additional beneficial activities. The health-promoting effects noted in animal models of various non-communicable diseases explain the growing interest in these molecules. In particular, in vitro and animal studies reported an attenuation of lipid disorders in response to PLPs. However, despite promising preclinical investigations, the effectiveness of PLPs in human dyslipidemia (DLP) is less clear and necessitates revision of available literature. Therefore, the present review analyzes the role of PLPs in managing clinical DLP, notably by dissecting their potential in ameliorating lipid/lipoprotein metabolism and alleviating hyperlipidemia, both postprandially and in long-term interventions. To this end, PubMed was used for article search. The search terms included polyphenols, lipids, triglycerides, cholesterol, LDL-cholesterol and /or HDL-cholesterol. The critical examination of the trials published to date illustrates certain benefits on blood lipids along with co-morbidities in participant’s health status. However, inconsistent results document significant research gaps, potentially owing to study heterogeneity and lack of rigor in establishing PLP bioavailability during supplementation. This underlines the need for further efforts in order to elucidate and support a potential role of PLPs in fighting DLP.
Collapse
|
37
|
Josefs T, Basu D, Vaisar T, Arets B, Kanter JE, Huggins LA, Hu Y, Liu J, Clouet-Foraison N, Heinecke JW, Bornfeldt KE, Goldberg IJ, Fisher EA. Atherosclerosis Regression and Cholesterol Efflux in Hypertriglyceridemic Mice. Circ Res 2021; 128:690-705. [PMID: 33530703 DOI: 10.1161/circresaha.120.317458] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Tatjana Josefs
- Division of Cardiology (T.J., J.L., E.A.F.), Department of Medicine, New York University School of Medicine.,Department of Internal Medicine, MUMC, Maastricht, the Netherlands (T.J., B.A.).,CARIM, MUMC, Maastricht, the Netherlands (T.J., B.A.)
| | - Debapriya Basu
- Division of Endocrinology, Diabetes and Metabolism (D.B., L.-A.H., Y.H., I.J.G.), Department of Medicine, New York University School of Medicine.,Department of Internal Medicine, MUMC, Maastricht, the Netherlands (T.J., B.A.).,CARIM, MUMC, Maastricht, the Netherlands (T.J., B.A.)
| | - Tomas Vaisar
- Department of Medicine, UW Medicine Diabetes Institute, University of Washington, Seattle (T.V., J.E.K., N.C.-F., J.W.H., K.E.B.)
| | | | - Jenny E Kanter
- Department of Medicine, UW Medicine Diabetes Institute, University of Washington, Seattle (T.V., J.E.K., N.C.-F., J.W.H., K.E.B.)
| | - Lesley-Ann Huggins
- Division of Endocrinology, Diabetes and Metabolism (D.B., L.-A.H., Y.H., I.J.G.), Department of Medicine, New York University School of Medicine
| | - Yunying Hu
- Division of Endocrinology, Diabetes and Metabolism (D.B., L.-A.H., Y.H., I.J.G.), Department of Medicine, New York University School of Medicine
| | - Jianhua Liu
- Division of Cardiology (T.J., J.L., E.A.F.), Department of Medicine, New York University School of Medicine
| | - Noemie Clouet-Foraison
- Department of Medicine, UW Medicine Diabetes Institute, University of Washington, Seattle (T.V., J.E.K., N.C.-F., J.W.H., K.E.B.)
| | - Jay W Heinecke
- Department of Medicine, UW Medicine Diabetes Institute, University of Washington, Seattle (T.V., J.E.K., N.C.-F., J.W.H., K.E.B.)
| | - Karin E Bornfeldt
- Department of Medicine, UW Medicine Diabetes Institute, University of Washington, Seattle (T.V., J.E.K., N.C.-F., J.W.H., K.E.B.)
| | - Ira J Goldberg
- Division of Endocrinology, Diabetes and Metabolism (D.B., L.-A.H., Y.H., I.J.G.), Department of Medicine, New York University School of Medicine
| | - Edward A Fisher
- Division of Cardiology (T.J., J.L., E.A.F.), Department of Medicine, New York University School of Medicine
| |
Collapse
|
38
|
Farnier M, Zeller M, Masson D, Cottin Y. Triglycerides and risk of atherosclerotic cardiovascular disease: An update. Arch Cardiovasc Dis 2021; 114:132-139. [PMID: 33546998 DOI: 10.1016/j.acvd.2020.11.006] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 11/05/2020] [Accepted: 11/06/2020] [Indexed: 12/22/2022]
Abstract
Low-density lipoprotein cholesterol is a well-known causal factor for atherosclerotic cardiovascular disease, and is the primary target of lipid-lowering therapy. There is, however, still a substantial risk of atherosclerotic cardiovascular disease events despite intensive statin therapy, and data from clinical trials suggest that an elevated concentration of triglycerides is a marker of residual cardiovascular risk on low-density lipoprotein-lowering therapy. Serum triglycerides are a biomarker for triglyceride-rich lipoproteins, and several lines of evidence indicate that triglyceride-rich lipoproteins and their cholesterol-enriched remnant particles are associated with atherogenesis. Moreover, genetic data in humans strongly suggest that the remnants of triglyceride-rich lipoproteins are a causal cardiovascular risk factor. Although lifestyle changes remain the cornerstone of management of hypertriglyceridaemia, a recent trial with high doses of the omega-3 fatty acid icosapent ethyl showed a significant reduction in cardiovascular events that was not explained by the reduction in triglycerides alone. In patients with elevated triglycerides, several novel drugs are in development to reduce the residual risk on statin therapy linked to an excess of atherogenic triglyceride-rich lipoproteins. In this review, we provide an update on the biology, epidemiology and genetics of triglycerides, and the risk of atherosclerotic cardiovascular disease.
Collapse
Affiliation(s)
- Michel Farnier
- PEC2, EA 7460, University of Bourgogne Franche-Comté, 21000 Dijon, France; Cardiology Department, University Hospital Centre of Dijon Bourgogne, 21000 Dijon, France.
| | - Marianne Zeller
- PEC2, EA 7460, University of Bourgogne Franche-Comté, 21000 Dijon, France; Cardiology Department, University Hospital Centre of Dijon Bourgogne, 21000 Dijon, France
| | - David Masson
- Inserm, LNC UMR 1231, FCS Bourgogne Franche-Comté, LipSTIC LabEx, 21078 Dijon, France
| | - Yves Cottin
- PEC2, EA 7460, University of Bourgogne Franche-Comté, 21000 Dijon, France; Cardiology Department, University Hospital Centre of Dijon Bourgogne, 21000 Dijon, France
| |
Collapse
|
39
|
Functional Haplotype of LIPC Induces Triglyceride-Mediated Suppression of HDL-C Levels According to Genome-Wide Association Studies. Genes (Basel) 2021; 12:genes12020148. [PMID: 33499410 PMCID: PMC7910859 DOI: 10.3390/genes12020148] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 01/11/2021] [Accepted: 01/19/2021] [Indexed: 01/08/2023] Open
Abstract
Hepatic lipase (encoded by LIPC) is a glycoprotein in the triacylglycerol lipase family and mainly synthesized in and secreted from the liver. Previous studies demonstrated that hepatic lipase is crucial for reverse cholesterol transport and modulating metabolism and the plasma levels of several lipoproteins. This study was conducted to investigate the suppression effect of high-density lipoprotein cholesterol (HDL-C) levels in a genome-wide association study and explore the possible mechanisms linking triglyceride (TG) to LIPC variants and HDL-C. Genome-wide association data for TG and HDL-C were available for 4657 Taiwan-biobank participants. The prevalence of haplotypes in the LIPC promoter region and their effects were calculated. The cloned constructs of the haplotypes were expressed transiently in HepG2 cells and evaluated in a luciferase reporter assay. Genome-wide association analysis revealed that HDL-C was significantly associated with variations in LIPC after adjusting for TG. Three haplotypes (H1: TCG, H2: CTA and H3: CCA) in LIPC were identified. H2: CTA was significantly associated with HDL-C levels and H1: TCG suppressed HDL-C levels when a third factor, TG, was included in mediation analysis. The luciferase reporter assay further showed that the H2: CTA haplotype significantly inhibited luciferase activity compared with the H1: TCG haplotype. In conclusion, we identified a suppressive role for TG in the genome-wide association between LIPC and HDL-C. A functional haplotype of hepatic lipase may reduce HDL-C levels and is suppressed by TG.
Collapse
|
40
|
Gill PK, Dron JS, Dilliott AA, McIntyre AD, Cao H, Wang J, Movsesyan IG, Malloy MJ, Pullinger CR, Kane JP, Hegele RA. Ancestry-specific profiles of genetic determinants of severe hypertriglyceridemia. J Clin Lipidol 2021; 15:88-96. [PMID: 33303403 DOI: 10.1016/j.jacl.2020.11.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Revised: 10/31/2020] [Accepted: 11/17/2020] [Indexed: 12/01/2022]
Abstract
BACKGROUND Susceptibility to severe hypertriglyceridemia (HTG), defined as plasma triglyceride (TG) levels ≥10 mmol/L (880 mg/dL), is conferred by both heterozygous rare variants in five genes involved in TG metabolism and numerous common single-nucleotide polymorphisms (SNPs) associated with TG levels. OBJECTIVE To date, these genetic susceptibility factors have been comprehensively assessed primarily in severe HTG patients of European ancestry. Here, we expand our analysis to HTG patients of East Asian and Hispanic ancestry. METHODS The genomic DNA of 336, 63 and 199 severe HTG patients of European, East Asian and Hispanic ancestry, respectively, was evaluated using a targeted next-generation sequencing panel to screen for: 1) rare variants in LPL, APOA5, APOC2, GPIHBP1 and LMF1; 2) common, small-to-moderate effect SNPs, quantified using a polygenic score; and 3) common, large-effect polymorphisms, APOA5 p.G185C and p.S19W. RESULTS While the proportion of individuals with high polygenic scores was similar, frequency of rare variant carriers varied across ancestries. Compared with ancestry-matched controls, Hispanic patients were the most likely to have a rare variant (OR = 5.02; 95% CI 3.07-8.21; p < 0.001), while European patients were the least likely (OR = 2.56; 95% CI 1.58-4.13; p < 0.001). The APOA5 p.G185C polymorphism, exclusive to East Asians, was significantly enriched in patients compared with controls (OR = 10.1; 95% CI 5.6-18.3; p < 0.001), showing the highest enrichment among the measured genetic factors. CONCLUSION While TG-associated rare variants and common SNPs are both found in statistical excess in severe HTG patients of different ancestral backgrounds, the overall genetic profiles of each ancestry group were distinct.
Collapse
Affiliation(s)
- Praneet K Gill
- Department of Biochemistry, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada; Blackburn Cardiovascular Genetics Laboratory, Robarts Research Institute, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
| | - Jacqueline S Dron
- Department of Biochemistry, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada; Blackburn Cardiovascular Genetics Laboratory, Robarts Research Institute, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
| | - Allison A Dilliott
- Department of Biochemistry, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada; Blackburn Cardiovascular Genetics Laboratory, Robarts Research Institute, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
| | - Adam D McIntyre
- Blackburn Cardiovascular Genetics Laboratory, Robarts Research Institute, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
| | - Henian Cao
- Blackburn Cardiovascular Genetics Laboratory, Robarts Research Institute, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
| | - Jian Wang
- Blackburn Cardiovascular Genetics Laboratory, Robarts Research Institute, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
| | - Irina G Movsesyan
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA, USA
| | - Mary J Malloy
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA, USA
| | - Clive R Pullinger
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA, USA
| | - John P Kane
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA, USA
| | - Robert A Hegele
- Department of Biochemistry, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada; Blackburn Cardiovascular Genetics Laboratory, Robarts Research Institute, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada.
| |
Collapse
|
41
|
Burnett JR, Hooper AJ, Hegele RA. Remnant Cholesterol and Atherosclerotic Cardiovascular Disease Risk. J Am Coll Cardiol 2020; 76:2736-2739. [DOI: 10.1016/j.jacc.2020.10.029] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 10/19/2020] [Indexed: 12/22/2022]
|
42
|
Joshi R, Wannamethee SG, Engmann J, Gaunt T, Lawlor DA, Price J, Papacosta O, Shah T, Tillin T, Chaturvedi N, Kivimaki M, Kuh D, Kumari M, Hughes AD, Casas JP, Humphries S, Hingorani AD, Schmidt AF. Triglyceride-containing lipoprotein sub-fractions and risk of coronary heart disease and stroke: A prospective analysis in 11,560 adults. Eur J Prev Cardiol 2020; 27:1617-1626. [PMID: 31996015 PMCID: PMC7707881 DOI: 10.1177/2047487319899621] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 12/17/2019] [Indexed: 11/16/2022]
Abstract
AIMS Elevated low-density lipoprotein cholesterol (LDL-C) is a risk factor for cardiovascular disease; however, there is uncertainty about the role of total triglycerides and the individual triglyceride-containing lipoprotein sub-fractions. We measured 14 triglyceride-containing lipoprotein sub-fractions using nuclear magnetic resonance and examined associations with coronary heart disease and stroke. METHODS Triglyceride-containing sub-fraction measures were available in 11,560 participants from the three UK cohorts free of coronary heart disease and stroke at baseline. Multivariable logistic regression was used to estimate the association of each sub-fraction with coronary heart disease and stroke expressed as the odds ratio per standard deviation increment in the corresponding measure. RESULTS The 14 triglyceride-containing sub-fractions were positively correlated with one another and with total triglycerides, and inversely correlated with high-density lipoprotein cholesterol (HDL-C). Thirteen sub-fractions were positively associated with coronary heart disease (odds ratio in the range 1.12 to 1.22), with the effect estimates for coronary heart disease being comparable in subgroup analysis of participants with and without type 2 diabetes, and were attenuated after adjustment for HDL-C and LDL-C. There was no evidence for a clear association of any triglyceride lipoprotein sub-fraction with stroke. CONCLUSIONS Triglyceride sub-fractions are associated with increased risk of coronary heart disease but not stroke, with attenuation of effects on adjustment for HDL-C and LDL-C.
Collapse
Affiliation(s)
- Roshni Joshi
- Institute of Cardiovascular Science, Faculty of Population Health, University College London, UK
| | - S Goya Wannamethee
- Department of Primary Care & Population Health, Faculty of Population Health, University College London, UK
| | - Jorgen Engmann
- Institute of Cardiovascular Science, Faculty of Population Health, University College London, UK
| | - Tom Gaunt
- MRC Integrative Epidemiology Unit at the University of Bristol, UK
| | - Deborah A Lawlor
- MRC Integrative Epidemiology Unit at the University of Bristol, UK
- Bristol NIHR Biomedical Research Centre, UK
- Population Health Science, Bristol Medical School, UK
| | - Jackie Price
- The Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, UK
| | - Olia Papacosta
- Department of Primary Care & Population Health, Faculty of Population Health, University College London, UK
| | - Tina Shah
- Institute of Cardiovascular Science, Faculty of Population Health, University College London, UK
| | - Therese Tillin
- Department of Epidemiology and Public Health, University College London, UK
| | - Nishi Chaturvedi
- Department of Epidemiology and Public Health, University College London, UK
| | - Mika Kivimaki
- Department of Epidemiology and Public Health, University College London, UK
| | - Diana Kuh
- MRC Unit for Lifelong Health and Ageing, UK
| | - Meena Kumari
- Institute for Social and Economic Research, University of Essex, UK
| | - Alun D Hughes
- Department of Epidemiology and Public Health, University College London, UK
| | - Juan P Casas
- Massachusetts Veterans Epidemiology Research and Information Center (MAVERIC), VA Boston Healthcare, USA
| | - Steve Humphries
- Institute of Cardiovascular Science, Faculty of Population Health, University College London, UK
| | - Aroon D Hingorani
- Institute of Cardiovascular Science, Faculty of Population Health, University College London, UK
| | - A Floriaan Schmidt
- Institute of Cardiovascular Science, Faculty of Population Health, University College London, UK
- Department of Cardiology, Division Heart and Lungs, University Medical Centre Utrecht, The Netherlands
| |
Collapse
|
43
|
Lu X. Structure and Function of Angiopoietin-like Protein 3 (ANGPTL3) in Atherosclerosis. Curr Med Chem 2020; 27:5159-5174. [PMID: 31223079 DOI: 10.2174/0929867326666190621120523] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 04/24/2019] [Accepted: 04/30/2019] [Indexed: 02/08/2023]
Abstract
BACKGROUND Angiopoietin-Like Proteins (ANGPTLs) are structurally related to the angiopoietins. A total of eight ANGPTLs (from ANGPTL1 to ANGPTL8) have been identified so far. Most ANGPTLs possess multibiological functions on lipid metabolism, atherosclerosis, and cancer. Among them, ANGPTL3 has been shown to regulate the levels of Very Low-Density Lipoprotein (VLDL) made by the liver and play a crucial role in human lipoprotein metabolism. METHOD A systematic appraisal of ANGPTLs was conducted, focusing on the main features of ANGPTL3 that has a significant role in atherosclerosis. RESULTS Angiopoietins including ANGPTL3 are vascular growth factors that are highly specific for endothelial cells, perform a variety of other regulatory activities to influence inflammation, and have been shown to possess both pro-atherosclerotic and atheroprotective effects. CONCLUSION ANGPTL3 has been demonstrated as a promising target in the pharmacological management of atherosclerosis. However, many questions remain about its biological functions.
Collapse
Affiliation(s)
- Xinjie Lu
- The Mary and Garry Weston Molecular Immunology Laboratory, Thrombosis Research Institute, London SW3 6LR, England, United Kingdom
| |
Collapse
|
44
|
Boden WE, Baum S, Toth PP, Fazio S, Bhatt DL. Impact of expanded FDA indication for icosapent ethyl on enhanced cardiovascular residual risk reduction. Future Cardiol 2020; 17:155-174. [PMID: 32959713 DOI: 10.2217/fca-2020-0106] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Hypertriglyceridemia is associated with increased cardiovascular disease (CVD) risk. The Reduction of Cardiovascular Events with Icosapent Ethyl-Intervention Trial (REDUCE-IT) demonstrated that the purified, stable ethyl ester of eicosapentaenoic acid, icosapent ethyl (IPE), added to statins reduced CVD events by 25% (p < 0.001), leading to an expanded indication in the USA. IPE is now approved as an adjunct to maximally tolerated statins to reduce CVD event risk in adults with triglyceride (TG) levels ≥150 mg/dl and either established CVD or diabetes mellitus plus ≥2 additional CVD risk factors. The new indication allows co-administration of IPE for elevated TG levels with statin treatment, enabling effective residual risk reduction in a broader at-risk population beyond what can be achieved with intensive low-density lipoprotein cholesterol control alone.
Collapse
Affiliation(s)
- William E Boden
- VA New England Healthcare System, Boston, MA, & Boston University School of Medicine, Boston, MA 02130, USA
| | - Seth Baum
- Boca Raton Regional Hospital, Boca Raton, FL 33486, USA
| | - Peter P Toth
- CGH Medical Center, Sterling, IL, Johns Hopkins University School of Medicine, Baltimore, MD 61081, USA
| | - Sergio Fazio
- Oregon Health & Science University, Portland, OR 97239, USA
| | - Deepak L Bhatt
- Brigham & Women's Hospital Heart & Vascular Center & Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
45
|
Hegele RA, Dron JS. 2019 George Lyman Duff Memorial Lecture: Three Decades of Examining DNA in Patients With Dyslipidemia. Arterioscler Thromb Vasc Biol 2020; 40:1970-1981. [PMID: 32762461 DOI: 10.1161/atvbaha.120.313065] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Dyslipidemias include both rare single gene disorders and common conditions that have a complex underlying basis. In London, ON, there is fortuitous close physical proximity between the Lipid Genetics Clinic and the London Regional Genomics Centre. For >30 years, we have applied DNA sequencing of clinical samples to help answer scientific questions. More than 2000 patients referred with dyslipidemias have participated in an ongoing translational research program. In 2013, we transitioned to next-generation sequencing; our targeted panel is designed to concurrently assess both monogenic and polygenic contributions to dyslipidemias. Patient DNA is screened for rare variants underlying 25 mendelian dyslipidemias, including familial hypercholesterolemia, hepatic lipase deficiency, abetalipoproteinemia, and familial chylomicronemia syndrome. Furthermore, polygenic scores for LDL (low-density lipoprotein) and HDL (high-density lipoprotein) cholesterol, and triglycerides are calculated for each patient. We thus simultaneously document both rare and common genetic variants, allowing for a broad view of genetic predisposition for both individual patients and cohorts. For instance, among patients referred with severe hypertriglyceridemia, defined as ≥10 mmol/L (≥885 mg/dL), <1% have a mendelian disorder (ie, autosomal recessive familial chylomicronemia syndrome), ≈15% have heterozygous rare variants (a >3-fold increase over normolipidemic individuals), and ≈35% have an extreme polygenic score (a >3-fold increase over normolipidemic individuals). Other dyslipidemias show a different mix of genetic determinants. Genetic results are discussed with patients and can support clinical decision-making. Integrating DNA testing into clinical care allows for a bidirectional flow of information, which facilitates scientific discoveries and clinical translation.
Collapse
Affiliation(s)
- Robert A Hegele
- From the Department of Medicine (R.A.H.), Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
- Department of Biochemistry (R.A.H., J.S.D.), Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
- Robarts Research Institute (R.A.H., J.S.D.), Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Jacqueline S Dron
- Department of Biochemistry (R.A.H., J.S.D.), Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
- Robarts Research Institute (R.A.H., J.S.D.), Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| |
Collapse
|
46
|
Toth PP, Shah PK, Lepor NE. Targeting hypertriglyceridemia to mitigate cardiovascular risk: A review. Am J Prev Cardiol 2020; 3:100086. [PMID: 32929418 PMCID: PMC7481317 DOI: 10.1016/j.ajpc.2020.100086] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 08/21/2020] [Accepted: 08/25/2020] [Indexed: 12/12/2022] Open
Abstract
A causal relationship between elevated triglycerides and cardiovascular disease is controversial, as trials of triglyceride-lowering treatments have not shown significant impact on cardiovascular outcomes. However, hypertriglyceridemia is associated with atherogenesis and risk for acute cardiovascular events that persist despite optimal statin treatment. Although most trials of triglyceride-lowering treatments have been negative, in trials of niacin and fibrates, subgroup analyses in patients with higher baseline triglycerides and lower HDL-C levels suggest reduced incidence of cardiovascular endpoints. The REDUCE-IT trial demonstrated that addition of purified prescription eicosapentaenoic acid (icosapent ethyl) 4 g/day in high-risk patients with triglyceride levels 135-499 mg/dL and optimized statin treatment significantly reduced cardiovascular events versus placebo (hazard ratio 0.75; 95% confidence interval 0.68-0.83; P < 0.001). Benefit was seen regardless of baseline and on-treatment triglyceride levels, suggesting that other effects of eicosapentaenoic acid besides triglyceride reduction may have played a role.
Collapse
Affiliation(s)
- Peter P. Toth
- CGH Medical Center, Sterling, IL, USA
- Cicarrone Center for the Prevention of Cardiovascular Disease, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Prediman K. Shah
- Smidt Heart Institute at Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Norman E. Lepor
- Smidt Heart Institute at Cedars-Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|
47
|
Dron JS, Dilliott AA, Lawson A, McIntyre AD, Davis BD, Wang J, Cao H, Movsesyan I, Malloy MJ, Pullinger CR, Kane JP, Hegele RA. Loss-of-Function
CREB3L3
Variants in Patients With Severe Hypertriglyceridemia. Arterioscler Thromb Vasc Biol 2020; 40:1935-1941. [DOI: 10.1161/atvbaha.120.314168] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Objective:
Genetic determinants of severe hypertriglyceridemia include both common variants with small effects (assessed using polygenic risk scores) plus heterozygous and homozygous rare variants in canonical genes directly affecting triglyceride metabolism. Here, we broadened our scope to detect associations with rare loss-of-function variants in genes affecting noncanonical pathways, including those known to affect triglyceride metabolism indirectly.
Approach and Results:
From targeted next-generation sequencing of 69 metabolism-related genes in 265 patients of European descent with severe hypertriglyceridemia (≥10 mmol/L or ≥885 mg/dL) and 477 normolipidemic controls, we focused on the association of rare heterozygous loss-of-function variants in individual genes. We observed that compared with controls, severe hypertriglyceridemia patients were 20.2× (95% CI, 1.11–366.1;
P
=0.03) more likely than controls to carry a rare loss-of-function variant in
CREB3L3
, which encodes a transcription factor that regulates several target genes with roles in triglyceride metabolism.
Conclusions:
Our findings indicate that rare variants in a noncanonical gene for triglyceride metabolism, namely
CREB3L3
, contribute significantly to severe hypertriglyceridemia. Secondary genes and pathways should be considered when evaluating the genetic architecture of this complex trait.
Collapse
Affiliation(s)
- Jacqueline S. Dron
- From the Robarts Research Institute (J.S.D., A.A.D., A.L., A.D.M., J.W., H.C., R.A.H.), Western University, London, ON, Canada
- Department of Biochemistry (J.S.D., A.A.D., A.L., R.A.H.), Western University, London, ON, Canada
| | - Allison A. Dilliott
- From the Robarts Research Institute (J.S.D., A.A.D., A.L., A.D.M., J.W., H.C., R.A.H.), Western University, London, ON, Canada
- Department of Biochemistry (J.S.D., A.A.D., A.L., R.A.H.), Western University, London, ON, Canada
| | - Arden Lawson
- From the Robarts Research Institute (J.S.D., A.A.D., A.L., A.D.M., J.W., H.C., R.A.H.), Western University, London, ON, Canada
- Department of Biochemistry (J.S.D., A.A.D., A.L., R.A.H.), Western University, London, ON, Canada
| | - Adam D. McIntyre
- From the Robarts Research Institute (J.S.D., A.A.D., A.L., A.D.M., J.W., H.C., R.A.H.), Western University, London, ON, Canada
| | - Brent D. Davis
- Schulich School of Medicine and Dentistry, and Department of Computer Science (B.D.D.), Western University, London, ON, Canada
| | - Jian Wang
- From the Robarts Research Institute (J.S.D., A.A.D., A.L., A.D.M., J.W., H.C., R.A.H.), Western University, London, ON, Canada
| | - Henian Cao
- From the Robarts Research Institute (J.S.D., A.A.D., A.L., A.D.M., J.W., H.C., R.A.H.), Western University, London, ON, Canada
| | - Irina Movsesyan
- Cardiovascular Research Institute, University of California, San Francisco (I.M., M.J.M., C.R.P., J.P.K.)
| | - Mary J. Malloy
- Cardiovascular Research Institute, University of California, San Francisco (I.M., M.J.M., C.R.P., J.P.K.)
| | - Clive R. Pullinger
- Cardiovascular Research Institute, University of California, San Francisco (I.M., M.J.M., C.R.P., J.P.K.)
| | - John P. Kane
- Cardiovascular Research Institute, University of California, San Francisco (I.M., M.J.M., C.R.P., J.P.K.)
| | - Robert A. Hegele
- From the Robarts Research Institute (J.S.D., A.A.D., A.L., A.D.M., J.W., H.C., R.A.H.), Western University, London, ON, Canada
- Department of Biochemistry (J.S.D., A.A.D., A.L., R.A.H.), Western University, London, ON, Canada
- Department of Medicine (R.A.H.), Western University, London, ON, Canada
| |
Collapse
|
48
|
Park S, Kang S. A minor allele of the haplotype located in the 19q13 loci is associated with a decreased risk of hyper-LDL-cholesterolemia, and a balanced diet and high protein intake can reduce the risk. Lipids Health Dis 2020; 19:178. [PMID: 32727492 PMCID: PMC7391697 DOI: 10.1186/s12944-020-01352-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 07/20/2020] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Although the human chromosome 19q13 loci are reported to be associated with hyper-LDL-cholesterolemia, the haplotype of single nucleotide polymorphism (SNP) has not been studied. Therefore, the association of the haplotype in 19q13 loci with hyper-LDL-cholesterolemia was determined and their interactions with lifestyles and nutrient intakes were evaluated in 28,445 Koreans aged > 40 years. METHODS SNPs were selected from 19q13 loci that had an association with hyper-LDL-cholesterolemia with the adjustment of confounders (age, gender, area of residence, and body mass index). Haplotype was constructed from the selected SNPs. An adjusted odds ratio of the haplotype for hyper-LDL-cholesterolemia and the interaction between haplotype and lifestyles was analyzed after adjusting for covariates. RESULTS Hyper-LDL-cholesterolemia had an association with apolipoprotein E (APOE)_ rs7259620, translocase of outer mitochondrial membrane 40(TOMM40)_rs157581, poliovirus receptor-related 2(PVRL2)_rs403155, exocyst complex component 3-like 2(EXOC3L2)_ rs10406604 and CD3e molecule-associated protein (CD3EAP)_rs3212986 in 19q13. The haplotype of these SNPs had a negative association with hyper-total-cholesterolemia and hyper-LDL-cholesterolemia by 0.669 and 0.684 times, respectively, after adjusting for covariates. The incidence of cardiovascular diseases, especially myocardial infarction, had a negative association with the minor alleles. The balanced diet pattern (BD) and protein intake had a significant interaction with the haplotype: the major-allele of the haplotype exhibited a positive association with hyper-LDL-cholesterolemia, compared to the minor allele, only when combined with a high intake of BD. The participants with the minor allele exhibited a lower hyper-LDL-cholesterolemia risk compared to those with the major allele only with high protein intake. CONCLUSION The minor allele of haplotype located in 19q13 loci protected against hyper-LDL-cholesterolemia, especially with BD and high protein intake. The minor allele also had a negative association with myocardial infarction events.
Collapse
Affiliation(s)
- Sunmin Park
- Department of Food and Nutrition, Obesity/Diabetes Research Center, Hoseo University, 165 Sechul-Ri, BaeBang-Yup, Asan-Si, ChungNam-Do, 31499, South Korea.
| | - Suna Kang
- Department of Food and Nutrition, Obesity/Diabetes Research Center, Hoseo University, 165 Sechul-Ri, BaeBang-Yup, Asan-Si, ChungNam-Do, 31499, South Korea
| |
Collapse
|
49
|
Lazarte J, Hegele RA. Pediatric Dyslipidemia-Beyond Familial Hypercholesterolemia. Can J Cardiol 2020; 36:1362-1371. [PMID: 32640212 DOI: 10.1016/j.cjca.2020.03.020] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 03/04/2020] [Accepted: 03/16/2020] [Indexed: 12/17/2022] Open
Abstract
Dyslipidemia is seen with increasing prevalence in young Canadians, mainly mild to moderate hypertriglyceridemia secondary to obesity. This review focuses on pediatric dyslipidemias excluding familial hypercholesterolemia (FH), but including both severe and mild to moderate hypertriglyceridemia, combined hyperlipidemia, and elevated lipoprotein(a) [Lp(a)]. We suggest that for Canadian children and adolescents with dyslipidemia, atherosclerotic cardiovascular disease (ASCVD) risk assessment should include both low-density lipoprotein cholesterol and triglyceride measurement. To further stratify risk, determination of non-high-density lipoprotein cholesterol is recommended, for both its ability to predict ASCVD and convenience for the patient because fasting is not required. Similarly, apolipoprotein B measurement (fasting or nonfasting), where available, can be helpful. Lp(a) measurement should not be routine in childhood, but it can be considered in special circumstances. After ruling out secondary causes, the foundation for management of pediatric dyslipidemia includes weight regulation, optimizing diet, and increasing activity level. At present, randomized clinical trial data to guide pharmaceutical management of pediatric hypertriglyceridemia or other non-FH pediatric dyslipidemias are scarce. Pharmaceutical management should be reserved for special situations in which risk of complications such as acute pancreatitis or ASCVD over the intermediate term is high and conservative lifestyle-based interventions have been ineffective.
Collapse
Affiliation(s)
- Julieta Lazarte
- Departments of Medicine and Biochemistry, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada; Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Robert A Hegele
- Departments of Medicine and Biochemistry, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada; Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada.
| |
Collapse
|
50
|
Tremblay K, Gaudet D, Khoury E, Brisson D. Dissection of Clinical and Gene Expression Signatures of Familial versus Multifactorial Chylomicronemia. J Endocr Soc 2020; 4:bvaa056. [PMID: 32537545 PMCID: PMC7278277 DOI: 10.1210/jendso/bvaa056] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 05/08/2020] [Indexed: 12/18/2022] Open
Abstract
Familial chylomicronemia syndrome (FCS) is a rare disorder associated with chylomicronemia (CM) and an increased risk of pancreatitis. Most individuals with CM do not have FCS but exhibit multifactorial CM (MCM), which differs from FCS in terms of risk and disease management. This study aimed to investigate clinical and gene expression profiles of FCS and MCM patients. Anthropometrics, clinical, and biochemical variables were analyzed in 57 FCS and 353 MCM patients. Gene expression analyses were performed in a subsample of 19 FCS, 28 MCM, and 15 normolipidemic controls. Receiver operating characteristic (ROC) curve analyses were performed to analyze the capacity of variables to discriminate FCS from MCM. Sustained fasting triglycerides ≥20 mmol/L (>15 mmol/L with eruptive xanthomas), history of pancreatitis, poor response to fibrates, diagnosis of CM at childhood, body mass index <22 kg/m2, and delipidated apolipoprotein B or glycerol levels <0.9 g/L and <0.05 mmol/L, respectively, had an area under the ROC curve ≥0.7. Gene expression analyses identified 142 probes differentially expressed in FCS and 32 in MCM compared with controls. Among them, 13 probes are shared between FCS and MCM; 63 are specific to FCS and 2 to MCM. Most FCS-specific or shared biomarkers are involved in inflammatory, immune, circadian, postprandial metabolism, signaling, docking systems, or receptor-mediated clearance mechanisms. This study reveals differential signatures of FCS and MCM. It opens the door to the identification of key mechanisms of CM expression and potential targets for the development of new treatments.
Collapse
Affiliation(s)
- Karine Tremblay
- Lipidology Unit, Community Genomic Medicine Center, Department of Medicine, Université de Montréal and ECOGENE-21 Clinical and Translational Research Center, Chicoutimi, Quebec, Canada.,Centre intégré universitaire de santé et de services sociaux du Saguenay-Lac-Saint-Jean, Saguenay, Quebec, Canada
| | - Daniel Gaudet
- Lipidology Unit, Community Genomic Medicine Center, Department of Medicine, Université de Montréal and ECOGENE-21 Clinical and Translational Research Center, Chicoutimi, Quebec, Canada
| | - Etienne Khoury
- Lipidology Unit, Community Genomic Medicine Center, Department of Medicine, Université de Montréal and ECOGENE-21 Clinical and Translational Research Center, Chicoutimi, Quebec, Canada
| | - Diane Brisson
- Lipidology Unit, Community Genomic Medicine Center, Department of Medicine, Université de Montréal and ECOGENE-21 Clinical and Translational Research Center, Chicoutimi, Quebec, Canada
| |
Collapse
|