1
|
Kosugi T, Eriguchi M, Yoshida H, Tamaki H, Uemura T, Tasaki H, Furuyama R, Fukata F, Nishimoto M, Matsui M, Samejima KI, Iseki K, Fujimoto S, Konta T, Moriyama T, Yamagata K, Narita I, Kasahara M, Shibagaki Y, Kondo M, Asahi K, Watanabe T, Tsuruya K. Serum High-Density Lipoprotein Cholesterol Levels and the Risk of Kidney Function Decline: The Japan Specific Health Checkups (J‑SHC) Study. J Atheroscler Thromb 2025; 32:407-420. [PMID: 39313383 PMCID: PMC11973525 DOI: 10.5551/jat.65107] [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] [Received: 05/08/2024] [Accepted: 08/08/2024] [Indexed: 09/25/2024] Open
Abstract
AIMS Both low and high serum levels of high-density lipoprotein cholesterol (HDL-C) were reported to be associated with adverse kidney outcomes. However, this association has not been well investigated in the general Japanese population. METHODS This nationwide longitudinal study used data from the Japan Specific Health Checkups Study conducted between 2008-2014. The association between serum HDL-C levels and 40% decline in estimated glomerular filtration rate (eGFR) was analyzed using Cox regression analysis. Trajectories of eGFR were compared using mixed-effects model. RESULTS Among 768,495 participants, 6,249 developed 40% decline in eGFR during the median follow-up period of 34.6 (interquartile range: 14.8-48.4) months. Using serum HDL-C levels of 40-59 mg/dL as a reference, the adjusted hazard ratios (95% confidence intervals) for the kidney outcome of serum HDL-C levels of <40, 60-79 and ≥ 80 mg/dL were 1.26 (1.14-1.39), 0.91 (0.86-0.96), and 0.86 (0.78-0.93), respectively. Restricted cubic spline analysis showed that HDL-C levels of less than approximately 60 mg/dL were associated with an increased risk of kidney outcomes. Subgroup analysis showed that baseline eGFR and proteinuria modified the effects of serum HDL-C levels on kidney outcomes. The mixed-effects model showed that the lower category of HDL-C level was associated with a higher eGFR decline rate (p for interaction <0.001). CONCLUSIONS Low HDL-C levels were associated with kidney function decline; however, high HDL-C levels were not associated with adverse kidney outcomes in the general Japanese population.
Collapse
Affiliation(s)
- Takaaki Kosugi
- Department of Nephrology, Nara Medical University, Kashihara, Nara, Japan
| | - Masahiro Eriguchi
- Department of Nephrology, Nara Medical University, Kashihara, Nara, Japan
| | - Hisako Yoshida
- Department of Medical Statistics, Osaka Metropolitan University Graduate School of Medicine, Osaka, Osaka, Japan
| | - Hiroyuki Tamaki
- Department of Nephrology, Nara Medical University, Kashihara, Nara, Japan
| | - Takayuki Uemura
- Department of Nephrology, Nara Medical University, Kashihara, Nara, Japan
| | - Hikari Tasaki
- Department of Nephrology, Nara Medical University, Kashihara, Nara, Japan
| | - Riri Furuyama
- Department of Nephrology, Nara Medical University, Kashihara, Nara, Japan
| | - Fumihiro Fukata
- Department of Nephrology, Nara Medical University, Kashihara, Nara, Japan
| | | | - Masaru Matsui
- Department of Nephrology, Nara Medical University, Kashihara, Nara, Japan
| | - Ken-ichi Samejima
- Department of Nephrology, Nara Medical University, Kashihara, Nara, Japan
| | - Kunitoshi Iseki
- Steering Committee of The Japan Specific Health Checkups (J-SHC) Study, Fukushima, Japan
| | - Shouichi Fujimoto
- Steering Committee of The Japan Specific Health Checkups (J-SHC) Study, Fukushima, Japan
| | - Tsuneo Konta
- Steering Committee of The Japan Specific Health Checkups (J-SHC) Study, Fukushima, Japan
| | - Toshiki Moriyama
- Steering Committee of The Japan Specific Health Checkups (J-SHC) Study, Fukushima, Japan
| | - Kunihiro Yamagata
- Steering Committee of The Japan Specific Health Checkups (J-SHC) Study, Fukushima, Japan
| | - Ichiei Narita
- Steering Committee of The Japan Specific Health Checkups (J-SHC) Study, Fukushima, Japan
| | - Masato Kasahara
- Steering Committee of The Japan Specific Health Checkups (J-SHC) Study, Fukushima, Japan
| | - Yugo Shibagaki
- Steering Committee of The Japan Specific Health Checkups (J-SHC) Study, Fukushima, Japan
| | - Masahide Kondo
- Steering Committee of The Japan Specific Health Checkups (J-SHC) Study, Fukushima, Japan
| | - Koichi Asahi
- Steering Committee of The Japan Specific Health Checkups (J-SHC) Study, Fukushima, Japan
| | - Tsuyoshi Watanabe
- Steering Committee of The Japan Specific Health Checkups (J-SHC) Study, Fukushima, Japan
| | - Kazuhiko Tsuruya
- Department of Nephrology, Nara Medical University, Kashihara, Nara, Japan
- Steering Committee of The Japan Specific Health Checkups (J-SHC) Study, Fukushima, Japan
| |
Collapse
|
2
|
Lui DTW, Li L, Liu X, Xiong X, Tang EHM, Lee CH, Woo YC, Lang BHH, Wong CKH, Tan KCB. The association of HDL-cholesterol levels with incident major adverse cardiovascular events and mortality in 0.6 million individuals with type 2 diabetes: a population-based retrospective cohort study. BMC Med 2024; 22:586. [PMID: 39696353 DOI: 10.1186/s12916-024-03810-4] [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: 06/19/2024] [Accepted: 12/03/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND High levels of high-density lipoprotein cholesterol (HDL-C) are previously considered protective against cardiovascular diseases (CVD), but recent studies suggest an increased risk of adverse events at very high HDL-C levels in the general population. It remains to be elucidated such a relationship in diabetes, a condition with high cardiovascular risks. We examined the association of HDL-C levels with the risk of major adverse cardiovascular events (MACE) and mortality in type 2 diabetes. METHODS This retrospective cohort study identified individuals with type 2 diabetes who had HDL-C records (2008-2020) from the electronic health record database of the Hong Kong Hospital Authority. They were classified into three groups based on their first-recorded HDL-C levels following diabetes diagnosis: low (≤ 40 mg/dL), medium (> 40 and ≤ 80 mg/dL) and high HDL-C (> 80 mg/dL) groups. The primary outcome was incident MACE (composite of myocardial infarction, stroke, heart failure, and cardiovascular mortality). Cox regression model and restricted cubic spline analysis were employed to assess the relationship between HDL-C and adverse outcomes. RESULTS Among 596,943 individuals with type 2 diabetes included, 168,931 (28.30%), 412,863 (69.16%), and 15,149 (2.54%) were classified as low HDL-C, medium HDL-C, and high HDL-C groups, respectively. Over a median follow-up of 79.5 months, both low and high HDL-C groups had higher risk of incident MACE compared to the medium HDL-C group (HR 1.24, 95% CI 1.23-1.26, P < 0.001; HR 1.09, 95% CI 1.04-1.13, P < 0.001). The spline curves revealed a U-shaped association between HDL-C levels and incident MACE (non-linear p < 0.001). Similar U-shaped relationship was observed for all-cause and non-cardiovascular mortality. CONCLUSIONS Our study demonstrated a U-shaped association between HDL-C levels and incident MACEs and all-cause and non-cardiovascular mortality in individuals with type 2 diabetes, highlighting the need for mechanistic studies on the adverse outcomes seen at high HDL-C levels in type 2 diabetes.
Collapse
Affiliation(s)
- David Tak Wai Lui
- Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong SAR, China
| | - Lanlan Li
- Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Xiaodong Liu
- Department of Surgery, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- Laboratory of Data Discovery for Health (D24H), Hong Kong SAR, China
| | - Xi Xiong
- Laboratory of Data Discovery for Health (D24H), Hong Kong SAR, China
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- Research Department of Practice and Policy, School of Pharmacy, University College London, London, UK
| | - Eric Ho Man Tang
- Laboratory of Data Discovery for Health (D24H), Hong Kong SAR, China
- Department of Family Medicine and Primary Care, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Chi Ho Lee
- Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong SAR, China
| | - Yu Cho Woo
- Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Brian Hung Hin Lang
- Department of Surgery, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Carlos King Ho Wong
- Laboratory of Data Discovery for Health (D24H), Hong Kong SAR, China.
- Department of Family Medicine and Primary Care, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China.
- Department of Infectious Disease Epidemiology & Dynamics, Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine, London, UK.
| | - Kathryn Choon Beng Tan
- Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
3
|
Gugliucci A. Angiopoietin-like Proteins and Lipoprotein Lipase: The Waltz Partners That Govern Triglyceride-Rich Lipoprotein Metabolism? Impact on Atherogenesis, Dietary Interventions, and Emerging Therapies. J Clin Med 2024; 13:5229. [PMID: 39274442 PMCID: PMC11396212 DOI: 10.3390/jcm13175229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 08/28/2024] [Accepted: 08/29/2024] [Indexed: 09/16/2024] Open
Abstract
Over 50% of patients who take statins are still at risk of developing atherosclerotic cardiovascular disease (ASCVD) and do not achieve their goal LDL-C levels. This residual risk is largely dependent on triglyceride-rich lipoproteins (TRL) and their remnants. In essence, remnant cholesterol-rich chylomicron (CM) and very-low-density lipoprotein (VLDL) particles play a role in atherogenesis. These remnants increase when lipoprotein lipase (LPL) activity is inhibited. ApoCIII has been thoroughly studied as a chief inhibitor and therapeutic options to curb its effect are available. On top of apoCIII regulation of LPL activity, there is a more precise control of LPL in various tissues, which makes it easier to physiologically divide the TRL burden according to the body's requirements. In general, oxidative tissues such as skeletal and cardiac muscle preferentially take up lipids during fasting. Conversely, LPL activity in adipocytes increases significantly after feeding, while its activity in oxidative tissues decreases concurrently. This perspective addresses the recent improvements in our understanding of circadian LPL regulations and their therapeutic implications. Three major tissue-specific lipolysis regulators have been identified: ANGPTL3, ANGPTL4, and ANGPTL8. Briefly, during the postprandial phase, liver ANGPTL8 acts on ANGPTL3 (which is released continuously from the liver) to inhibit LPL in the heart and muscle through an endocrine mechanism. On the other hand, when fasting, ANGPTL4, which is released by adipocytes, inhibits lipoprotein lipase in adipose tissue in a paracrine manner. ANGPTL3 inhibitors may play a therapeutic role in the treatment of hypertriglyceridemia. Several approaches are under development. We look forward to future studies to clarify (a) the nature of hormonal and nutritional factors that determine ANGPTL3, 4, and 8 activities, along with what long-term impacts may be expected if their regulation is impaired pharmacologically; (b) the understanding of the quantitative hierarchy and interaction of the regulatory actions of apoCIII, apoAV, and ANGPTL on LPL activity; (c) strategies for the safe and proper treatment of postprandial lipemia; and (d) the effect of fructose restriction on ANGPTL3, ANGPTL4, and ANGPTL8.
Collapse
Affiliation(s)
- Alejandro Gugliucci
- Glycation, Oxidation and Disease Laboratory, Touro University California, Vallejo, CA 94592, USA
| |
Collapse
|
4
|
Apsan J, Lekarev O, Thomas C, Zhu YS, Cohan K, Lin-Su K. Relationship between adipokines and androgens in children and young adults with congenital adrenal hyperplasia. Front Endocrinol (Lausanne) 2024; 15:1433378. [PMID: 39175574 PMCID: PMC11338854 DOI: 10.3389/fendo.2024.1433378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 07/15/2024] [Indexed: 08/24/2024] Open
Abstract
Introduction Children and young adults with congenital adrenal hyperplasia (CAH) are at increased risk of obesity and insulin resistance. There is evidence that children with CAH have increased visceral adiposity, which has been linked to metabolic syndrome and cardiovascular disease (CVD). The adipokine adiponectin has been shown to correlate with reduced metabolic risk, whereas the adipokines visfatin and leptin have been linked to visceral fat and adipocyte inflammation and can serve as biomarkers of increased metabolic risk. Few studies to date have characterized adipokine levels in children and young adults with congenital adrenal hyperplasia. We sought to investigate the relationship between adiponectin, leptin and visfatin levels to metabolic risk factors and androgen levels in children and young adults with CAH. Methods Fasting blood was obtained for visfatin, leptin, adiponectin, glucose, insulin, CRP, lipid panel, total cholesterol (TC), triglycerides (TG) and HbA1c, as well as standard laboratory tests to assess adrenal control, from children with CAH due to 21-hydroxylase deficiency. HOMA-IR was calculated based on fasting glucose and insulin. Anthropomorphic measurements of BMI and waist-to-hip ratio were also obtained. Results Adiponectin and androstenedione were inversely correlated (R = -0.57, p =0.016). There was a positive correlation between leptin and BMI percentile (R = 0.63, p <0.001) as well as leptin and HOMA-IR (R = 0.63, p <0.01). Glucocorticoid dose had a positive correlation with HOMA-IR (R=0.56, p = 0.021). Visfatin was inversely correlated with HDL cholesterol (R = -0.54, p = 0.026) and total cholesterol (R = -0.49, p <0.05). Overweight children and young adults had a significantly higher leptin (p = 0.02) and HOMA-IR (p=0.001) than non-overweight children and young adults. Conclusion The inverse relationship between adiponectin and androstenedione suggests that better CAH control can reduce the risk of insulin resistance and metabolic syndrome. However, a high glucocorticoid dose appears to increase the risk of insulin resistance, underscoring the delicate balance required when treating CAH.
Collapse
Affiliation(s)
- Jennifer Apsan
- Division of Pediatric Endocrinology, Department of Pediatrics, Weill Cornell Medicine, New York, NY, United States
| | - Oksana Lekarev
- Division of Pediatric Endocrinology, Department of Pediatrics, Weill Cornell Medicine, New York, NY, United States
| | - Charlene Thomas
- Division of Biostatistics, Department of Pediatrics, Weill Cornell Medicine, New York, NY, United States
| | - Yuan-Shan Zhu
- Clinical and Translational Science Center and Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Weill Cornell Medicine, New York, NY, United States
| | - Kaela Cohan
- Division of Pediatric Endocrinology, Department of Pediatrics, Weill Cornell Medicine, New York, NY, United States
| | - Karen Lin-Su
- Division of Pediatric Endocrinology, Department of Pediatrics, Weill Cornell Medicine, New York, NY, United States
| |
Collapse
|
5
|
Lui DTW, Tan KCB. High-density lipoprotein in diabetes: Structural and functional relevance. J Diabetes Investig 2024; 15:805-816. [PMID: 38416054 PMCID: PMC11215696 DOI: 10.1111/jdi.14172] [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: 01/27/2024] [Revised: 02/14/2024] [Accepted: 02/15/2024] [Indexed: 02/29/2024] Open
Abstract
Low levels of high-density lipoprotein-cholesterol (HDL-C) is considered a major cardiovascular risk factor. However, recent studies have suggested a more U-shaped association between HDL-C and cardiovascular disease. It has been shown that the cardioprotective effect of HDL is related to the functions of HDL particles rather than their cholesterol content. HDL particles are highly heterogeneous and have multiple functions relevant to cardiometabolic conditions including cholesterol efflux capacity, anti-oxidative, anti-inflammatory, and vasoactive properties. There are quantitative and qualitative changes in HDL as well as functional abnormalities in both type 1 and type 2 diabetes. Non-enzymatic glycation, carbamylation, oxidative stress, and systemic inflammation can modify the HDL composition and therefore the functions, especially in situations of poor glycemic control. Studies of HDL proteomics and lipidomics have provided further insights into the structure-function relationship of HDL in diabetes. Interestingly, HDL also has a pleiotropic anti-diabetic effect, improving glycemic control through improvement in insulin sensitivity and β-cell function. Given the important role of HDL in cardiometabolic health, HDL-based therapeutics are being developed to enhance HDL functions rather than to increase HDL-C levels. Among these, recombinant HDL and small synthetic apolipoprotein A-I mimetic peptides may hold promise for preventing and treating diabetes and cardiovascular disease.
Collapse
Affiliation(s)
- David Tak Wai Lui
- Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of MedicineThe University of Hong KongHong Kong SARChina
| | - Kathryn Choon Beng Tan
- Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of MedicineThe University of Hong KongHong Kong SARChina
| |
Collapse
|
6
|
Jiang Y, Pang S, Liu X, Wang L, Liu Y. The Gut Microbiome Affects Atherosclerosis by Regulating Reverse Cholesterol Transport. J Cardiovasc Transl Res 2024; 17:624-637. [PMID: 38231373 DOI: 10.1007/s12265-024-10480-3] [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: 08/27/2023] [Accepted: 01/07/2024] [Indexed: 01/18/2024]
Abstract
The human system's secret organ, the gut microbiome, has received considerable attention. Emerging research has yielded substantial scientific evidence indicating that changes in gut microbial composition and microbial metabolites may contribute to the development of atherosclerotic cardiovascular disease. The burden of cardiovascular disease on healthcare systems is exacerbated by atherosclerotic cardiovascular disease, which continues to be the leading cause of mortality globally. Reverse cholesterol transport is a powerful protective mechanism that effectively prevents excessive accumulation of cholesterol for atherosclerotic cardiovascular disease. It has been revealed how the gut microbiota modulates reverse cholesterol transport in patients with atherosclerotic risk. In this review, we highlight the complex interactions between microbes, their metabolites, and their potential impacts in reverse cholesterol transport. We also explore the feasibility of modulating gut microbes and metabolites to facilitate reverse cholesterol transport as a novel therapy for atherosclerosis.
Collapse
Affiliation(s)
- Yangyang Jiang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300193, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Shuchao Pang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China.
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300193, China.
| | - Xiaoyu Liu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300193, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Lixin Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300193, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yi Liu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China.
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300193, China.
| |
Collapse
|
7
|
Yang L, Wang Y, Xu Y, Li K, Yin R, Zhang L, Wang D, Wei L, Lang J, Cheng Y, Wang L, Ke J, Zhao D. ANGPTL3 is a novel HDL component that regulates HDL function. J Transl Med 2024; 22:263. [PMID: 38462608 PMCID: PMC10926621 DOI: 10.1186/s12967-024-05032-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 02/24/2024] [Indexed: 03/12/2024] Open
Abstract
BACKGROUND Angiopoietin-like protein 3 (ANGPTL3) is secreted by hepatocytes and inhibits lipoprotein lipase and endothelial lipase activity. Previous studies reported the correlation between plasma ANGPTL3 levels and high-density lipoprotein (HDL). Recently ANGPTL3 was found to preferentially bind to HDL in healthy human circulation. Here, we examined whether ANGPTL3, as a component of HDL, modulates HDL function and affects HDL other components in human and mice with non-diabetes or type 2 diabetes mellitus. METHODS HDL was isolated from the plasma of female non-diabetic subjects and type-2 diabetic mellitus (T2DM) patients. Immunoprecipitation, western blot, and ELISA assays were used to examine ANGPTL3 levels in HDL. Db/m and db/db mice, AAV virus mediated ANGPTL3 overexpression and knockdown models and ANGPTL3 knockout mice were used. The cholesterol efflux capacity induced by HDL was analyzed in macrophages preloaded with fluorescent cholesterol. The anti-inflammation capacity of HDL was assessed using flow cytometry to measure VCAM-1 and ICAM-1 expression levels in TNF-α-stimulated endothelial cells pretreated with HDL. RESULTS ANGPTL3 was found to bind to HDL and be a component of HDL in both non-diabetic subjects and T2DM patients. Flag-ANGPTL3 was found in the HDL of transgenic mice overexpressing Flag-ANGPTL3. ANGPLT3 of HDL was positively associated with cholesterol efflux in female non-diabetic controls (r = 0.4102, p = 0.0117) but not in female T2DM patients (r = - 0.1725, p = 0.3224). Lower ANGPTL3 levels of HDL were found in diabetic (db/db) mice compared to control (db/m) mice and were associated with reduced cholesterol efflux and inhibition of VCAM-1 and ICAM-1 expression in endothelial cells (p < 0.05 for all). Following AAV-mediated ANGPTL3 cDNA transfer in db/db mice, ANGPTL3 levels were found to be increased in HDL, and corresponded to increased cholesterol efflux and decreased ICAM-1 expression. In contrast, knockdown of ANGPTL3 levels in HDL by AAV-mediated shRNA transfer led to a reduction in HDL function (p < 0.05 for both). Plasma total cholesterol, total triglycerides, HDL-c, protein components of HDL and the cholesterol efflux function of HDL were lower in ANGPTL3-/- mice than ANGPTL3+/+ mice, suggesting that ANGPTL3 in HDL may regulate HDL function by disrupting the balance of protein components in HDL. CONCLUSION ANGPTL3 was identified as a component of HDL in humans and mice. ANGPTL3 of HDL regulated cholesterol efflux and the anti-inflammatory functions of HDL in T2DM mice. Both the protein components of HDL and cholesterol efflux capacity of HDL were decreased in ANGPTL3-/- mice. Our findings suggest that ANGPTL3 in HDL may regulate HDL function by disrupting the balance of protein components in HDL. Our study contributes to a more comprehensive understanding of the role of ANGPTL3 in lipid metabolism.
Collapse
Affiliation(s)
- Longyan Yang
- Center for Endocrine Metabolism and Immune Diseases, Beijing Luhe Hospital Capital Medical University, Beijing, China
- Beijing Key Laboratory of Diabetes Research and Care, Beijing, China
| | - Yan Wang
- Center for Endocrine Metabolism and Immune Diseases, Beijing Luhe Hospital Capital Medical University, Beijing, China
- Beijing Key Laboratory of Diabetes Research and Care, Beijing, China
| | - Yongsong Xu
- Center for Endocrine Metabolism and Immune Diseases, Beijing Luhe Hospital Capital Medical University, Beijing, China
- Beijing Key Laboratory of Diabetes Research and Care, Beijing, China
| | - Kun Li
- Center for Endocrine Metabolism and Immune Diseases, Beijing Luhe Hospital Capital Medical University, Beijing, China
- Beijing Key Laboratory of Diabetes Research and Care, Beijing, China
| | - Ruili Yin
- Center for Endocrine Metabolism and Immune Diseases, Beijing Luhe Hospital Capital Medical University, Beijing, China
- Beijing Key Laboratory of Diabetes Research and Care, Beijing, China
| | - Lijie Zhang
- Center for Endocrine Metabolism and Immune Diseases, Beijing Luhe Hospital Capital Medical University, Beijing, China
- Beijing Key Laboratory of Diabetes Research and Care, Beijing, China
| | - Di Wang
- Center for Endocrine Metabolism and Immune Diseases, Beijing Luhe Hospital Capital Medical University, Beijing, China
- Beijing Key Laboratory of Diabetes Research and Care, Beijing, China
| | - Lingling Wei
- Center for Endocrine Metabolism and Immune Diseases, Beijing Luhe Hospital Capital Medical University, Beijing, China
- Beijing Key Laboratory of Diabetes Research and Care, Beijing, China
| | - Jianan Lang
- Center for Endocrine Metabolism and Immune Diseases, Beijing Luhe Hospital Capital Medical University, Beijing, China
- Beijing Key Laboratory of Diabetes Research and Care, Beijing, China
| | - Yanan Cheng
- Center for Endocrine Metabolism and Immune Diseases, Beijing Luhe Hospital Capital Medical University, Beijing, China
- Beijing Key Laboratory of Diabetes Research and Care, Beijing, China
| | - Lu Wang
- Center for Endocrine Metabolism and Immune Diseases, Beijing Luhe Hospital Capital Medical University, Beijing, China
- Beijing Key Laboratory of Diabetes Research and Care, Beijing, China
| | - Jing Ke
- Center for Endocrine Metabolism and Immune Diseases, Beijing Luhe Hospital Capital Medical University, Beijing, China.
- Beijing Key Laboratory of Diabetes Research and Care, Beijing, China.
| | - Dong Zhao
- Center for Endocrine Metabolism and Immune Diseases, Beijing Luhe Hospital Capital Medical University, Beijing, China.
- Beijing Key Laboratory of Diabetes Research and Care, Beijing, China.
| |
Collapse
|
8
|
Gangwar A, Deodhar SS, Saldanha S, Melander O, Abbasi F, Pearce RW, Collier TS, McPhaul MJ, Furtado JD, Sacks FM, Merrill NJ, McDermott JE, Melchior JT, Rohatgi A. Proteomic Determinants of Variation in Cholesterol Efflux: Observations from the Dallas Heart Study. Int J Mol Sci 2023; 24:15526. [PMID: 37958510 PMCID: PMC10648649 DOI: 10.3390/ijms242115526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 10/12/2023] [Accepted: 10/20/2023] [Indexed: 11/15/2023] Open
Abstract
High-density lipoproteins (HDLs) are promising targets for predicting and treating atherosclerotic cardiovascular disease (ASCVD), as they mediate removal of excess cholesterol from lipid-laden macrophages that accumulate in the vasculature. This functional property of HDLs, termed cholesterol efflux capacity (CEC), is inversely associated with ASCVD. HDLs are compositionally diverse, associating with >250 different proteins, but their relative contribution to CEC remains poorly understood. Our goal was to identify and define key HDL-associated proteins that modulate CEC in humans. The proteomic signature of plasma HDL was quantified in 36 individuals in the multi-ethnic population-based Dallas Heart Study (DHS) cohort that exhibited persistent extremely high (>=90th%) or extremely low CEC (<=10th%) over 15 years. Levels of apolipoprotein (Apo)A-I associated ApoC-II, ApoC-III, and ApoA-IV were differentially correlated with CEC in high (r = 0.49, 0.41, and -0.21 respectively) and low (r = -0.46, -0.41, and 0.66 respectively) CEC groups (p for heterogeneity (pHet) = 0.03, 0.04, and 0.003 respectively). Further, we observed that levels of ApoA-I with ApoC-III, complement C3 (CO3), ApoE, and plasminogen (PLMG) were inversely associated with CEC in individuals within the low CEC group (r = -0.11 to -0.25 for subspecies with these proteins vs. r = 0.58 to 0.65 for subspecies lacking these proteins; p < 0.05 for heterogeneity). These findings suggest that enrichment of specific proteins on HDLs and, thus, different subspecies of HDLs, differentially modulate the removal of cholesterol from the vasculature.
Collapse
Affiliation(s)
- Anamika Gangwar
- Department of Internal Medicine, Division of Cardiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (A.G.); (S.S.D.); (S.S.)
| | - Sneha S. Deodhar
- Department of Internal Medicine, Division of Cardiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (A.G.); (S.S.D.); (S.S.)
| | - Suzanne Saldanha
- Department of Internal Medicine, Division of Cardiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (A.G.); (S.S.D.); (S.S.)
| | - Olle Melander
- Department of Clinical Sciences, Lund University, 221 00 Malmö, Sweden;
| | - Fahim Abbasi
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA;
| | - Ryan W. Pearce
- Quest Diagnostics Cardiometabolic Center of Excellence, Cleveland HeartLab, Cleveland, OH 44103, USA; (R.W.P.); (T.S.C.)
| | - Timothy S. Collier
- Quest Diagnostics Cardiometabolic Center of Excellence, Cleveland HeartLab, Cleveland, OH 44103, USA; (R.W.P.); (T.S.C.)
| | - Michael J. McPhaul
- Quest Diagnostics Nichols Institute, San Juan Capistrano, CA 92675, USA;
| | - Jeremy D. Furtado
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; (J.D.F.); (F.M.S.)
- Biogen Inc., Cambridge, MA 02115, USA
| | - Frank M. Sacks
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; (J.D.F.); (F.M.S.)
| | - Nathaniel J. Merrill
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA; (N.J.M.); (J.E.M.); (J.T.M.)
| | - Jason E. McDermott
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA; (N.J.M.); (J.E.M.); (J.T.M.)
| | - John T. Melchior
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA; (N.J.M.); (J.E.M.); (J.T.M.)
- Center for Lipid and Arteriosclerosis Science, Department of Pathology and Laboratory Medicine, University of Cincinnati, Cincinnati, OH 45237, USA
- Department of Neurology, Oregon Health and Science University, Portland, OR 97239, USA
| | - Anand Rohatgi
- Department of Internal Medicine, Division of Cardiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (A.G.); (S.S.D.); (S.S.)
| |
Collapse
|
9
|
Sato M, Neufeld EB, Playford MP, Lei Y, Sorokin AV, Aponte AM, Freeman LA, Gordon SM, Dey AK, Jeiran K, Hamasaki M, Sampson ML, Shamburek RD, Tang J, Chen MY, Kotani K, Anderson JL, Dullaart RP, Mehta NN, Tietge UJ, Remaley AT. Cell-free, high-density lipoprotein-specific phospholipid efflux assay predicts incident cardiovascular disease. J Clin Invest 2023; 133:e165370. [PMID: 37471145 PMCID: PMC10503808 DOI: 10.1172/jci165370] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 07/18/2023] [Indexed: 07/22/2023] Open
Abstract
BACKGROUNDCellular cholesterol efflux capacity (CEC) is a better predictor of cardiovascular disease (CVD) events than HDL-cholesterol (HDL-C) but is not suitable as a routine clinical assay.METHODSWe developed an HDL-specific phospholipid efflux (HDL-SPE) assay to assess HDL functionality based on whole plasma HDL apolipoprotein-mediated solubilization of fluorescent phosphatidylethanolamine from artificial lipid donor particles. We first assessed the association of HDL-SPE with prevalent coronary artery disease (CAD): study I included NIH severe-CAD (n = 50) and non-CAD (n = 50) participants, who were frequency matched for sex, BMI, type 2 diabetes mellitus, and smoking; study II included Japanese CAD (n = 70) and non-CAD (n = 154) participants. We also examined the association of HDL-SPE with incident CVD events in the Prevention of Renal and Vascular End-stage Disease (PREVEND) study comparing 340 patients with 340 controls individually matched for age, sex, smoking, and HDL-C levels.RESULTSReceiver operating characteristic curves revealed stronger associations of HDL-SPE with prevalent CAD. The AUCs in study I were as follows: HDL-SPE, 0.68; apolipoprotein A-I (apoA-I), 0.62; HDL-C, 0.63; and CEC, 0.52. The AUCs in study II were as follows: HDL-SPE, 0.83; apoA-I, 0.64; and HDL-C, 0.53. Also longitudinally, HDL-SPE was significantly associated with incident CVD events independent of traditional risk factors with ORs below 0.2 per SD increment in the PREVEND study (P < 0.001).CONCLUSIONHDL-SPE could serve as a routine clinical assay for improving CVD risk assessment and drug discovery.TRIAL REGISTRATIONClinicalTrials.gov NCT01621594.FUNDINGNHLBI Intramural Research Program, NIH (HL006095-06).
Collapse
Affiliation(s)
- Masaki Sato
- Lipoprotein Metabolism Laboratory, National Heart, Lung, and Blood Institute (NHLBI), NIH, Bethesda, Maryland, USA
- Division of Community and Family Medicine and Department of Clinical Laboratory Medicine, Jichi Medical University, Shimotsuke-City, Tochigi, Japan
- Biochemical Research Laboratory II, Eiken Chemical Co., Ltd., Shimotsuga-gun, Tochigi, Japan
| | - Edward B. Neufeld
- Lipoprotein Metabolism Laboratory, National Heart, Lung, and Blood Institute (NHLBI), NIH, Bethesda, Maryland, USA
| | - Martin P. Playford
- Section of Inflammation and Cardiometabolic Diseases, NHLBI, NIH, Bethesda, Maryland, USA
| | - Yu Lei
- Division of Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden
| | - Alexander V. Sorokin
- Lipoprotein Metabolism Laboratory, National Heart, Lung, and Blood Institute (NHLBI), NIH, Bethesda, Maryland, USA
- Section of Inflammation and Cardiometabolic Diseases, NHLBI, NIH, Bethesda, Maryland, USA
| | - Angel M. Aponte
- Proteomics Core Facility, NHLBI, NIH, Bethesda, Maryland, USA
| | - Lita A. Freeman
- Lipoprotein Metabolism Laboratory, National Heart, Lung, and Blood Institute (NHLBI), NIH, Bethesda, Maryland, USA
| | - Scott M. Gordon
- Saha Cardiovascular Research Center and Department of Physiology, University of Kentucky, Lexington, Kentucky, USA
| | - Amit K. Dey
- Section of Inflammation and Cardiometabolic Diseases, NHLBI, NIH, Bethesda, Maryland, USA
| | - Kianoush Jeiran
- Lipoprotein Metabolism Laboratory, National Heart, Lung, and Blood Institute (NHLBI), NIH, Bethesda, Maryland, USA
| | - Masato Hamasaki
- Division of Community and Family Medicine and Department of Clinical Laboratory Medicine, Jichi Medical University, Shimotsuke-City, Tochigi, Japan
- Biochemical Research Laboratory II, Eiken Chemical Co., Ltd., Shimotsuga-gun, Tochigi, Japan
| | | | - Robert D. Shamburek
- Lipoprotein Metabolism Laboratory, National Heart, Lung, and Blood Institute (NHLBI), NIH, Bethesda, Maryland, USA
| | - Jingrong Tang
- Lipoprotein Metabolism Laboratory, National Heart, Lung, and Blood Institute (NHLBI), NIH, Bethesda, Maryland, USA
| | - Marcus Y. Chen
- Laboratory of Cardiovascular CT, NHLBI, NIH, Bethesda, Maryland, USA
| | - Kazuhiko Kotani
- Division of Community and Family Medicine and Department of Clinical Laboratory Medicine, Jichi Medical University, Shimotsuke-City, Tochigi, Japan
| | - Josephine L.C. Anderson
- Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Robin P.F. Dullaart
- Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Nehal N. Mehta
- Section of Inflammation and Cardiometabolic Diseases, NHLBI, NIH, Bethesda, Maryland, USA
| | - Uwe J.F. Tietge
- Division of Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden
- Clinical Chemistry, Karolinska University Laboratory, Karolinska University Hospital, Stockholm, Sweden
| | - Alan T. Remaley
- Lipoprotein Metabolism Laboratory, National Heart, Lung, and Blood Institute (NHLBI), NIH, Bethesda, Maryland, USA
- The NIH Clinical Center and
| |
Collapse
|
10
|
Gugliucci A. Sugar and Dyslipidemia: A Double-Hit, Perfect Storm. J Clin Med 2023; 12:5660. [PMID: 37685728 PMCID: PMC10488931 DOI: 10.3390/jcm12175660] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 08/10/2023] [Accepted: 08/29/2023] [Indexed: 09/10/2023] Open
Abstract
The availability of sugar has expanded over the past 50 years, due to improved industrial processes and corn subsidies, particularly in the form of sweetened beverages. This correlates with a surge in the prevalence of cardiometabolic disorders, which has brought this issue back into the spotlight for public health. In this narrative review, we focus on the role of fructose in the genesis of cardiometabolic dyslipidemia (an increase in serum triglyceride-rich lipoproteins (TRL): VLDL, chylomicrons (CM), and their remnants) bringing together the most recent data on humans, which demonstrates the crucial interaction between glucose and fructose, increasing the synthesis while decreasing the catabolism of these particles in a synergistic downward spiral. After reviewing TRL metabolism, we discuss the fundamental principles governing the metabolism of fructose in the intestine and liver and the effects of dysregulated fructolysis, in conjunction with the activation of carbohydrate-responsive element-binding protein (ChREBP) by glucose and the resulting crosstalk. The first byproduct of fructose catabolism, fructose-1-P, is highlighted for its function as a signaling molecule that promotes fat synthesis. We emphasize the role of fructose/glucose interaction in the liver, which enhances de novo lipogenesis, triglyceride (TG) synthesis, and VLDL production. In addition, we draw attention to current research that demonstrates how fructose affects the activity of lipoprotein lipase by increasing the concentration of inhibitors such as apolipoprotein CIII (apoCIII) and angiopoietin-like protein 3 (ANGPTL3), which reduce the catabolism of VLDL and chylomicrons and cause the building up of their atherogenic remnants. The end outcome is a dual, synergistic, and harmful action that encourages atherogenesis. Thus, considering the growing concerns regarding the connection between sugar consumption and cardiometabolic disease, current research strongly supports the actions of public health organizations aimed at reducing sugar intake, including dietary guidance addressing "safe" limits for sugar consumption.
Collapse
Affiliation(s)
- Alejandro Gugliucci
- Glycation, Oxidation and Disease Laboratory, Touro University California, Vallejo, CA 94592, USA
| |
Collapse
|
11
|
García-González M, Gómez-Bernal F, Quevedo-Abeledo JC, Fernández-Cladera Y, González-Rivero AF, López-Mejías R, Díaz-González F, González-Gay MÁ, Ferraz-Amaro I. HDL Cholesterol Efflux and the Complement System Are Linked in Systemic Lupus Erythematosus. J Clin Med 2023; 12:5405. [PMID: 37629447 PMCID: PMC10455830 DOI: 10.3390/jcm12165405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 08/07/2023] [Accepted: 08/17/2023] [Indexed: 08/27/2023] Open
Abstract
Cholesterol efflux capacity (CEC), the ability of high-density lipoprotein (HDL) cholesterol to accept cholesterol from macrophages, has been linked to cardiovascular events. Systemic lupus erythematosus (SLE) is characterized by the consumption of complement (C) proteins and has been associated with an increased risk of cardiovascular disease. CEC is reduced in SLE patients compared to controls. In the present work, our objective was to analyze whether the disruption of C influences CEC in patients with SLE. New-generation functional assays of the three pathways of the C system were performed in 207 patients with SLE. Additionally, serum levels of inactive (C1q, C2, C3, C4, and factor D) and activated (C3a) molecules, and regulators (C1-inhibitor and factor H) of C system were measured. CEC, using an in vitro assay, and lipoprotein serum concentrations were assessed. Multivariable linear regression analysis was performed to assess the relationship between C system and CEC. After full multivariable analysis, the alternative C cascade functional test showed a significant and negative relationship with CEC. This was also the case for C2 and C3, in which the associations were found to be positive and statistically significant, after adjustment for covariates. In conclusion, C system and CEC are interconnected in patients with SLE.
Collapse
Affiliation(s)
- María García-González
- Division of Rheumatology, Hospital Universitario de Canarias, 38320 Tenerife, Spain; (M.G.-G.); (F.D.-G.)
| | - Fuensanta Gómez-Bernal
- Division of Central Laboratory, Hospital Universitario de Canarias, 38320 Tenerife, Spain; (F.G.-B.); (Y.F.-C.); (A.F.G.-R.)
| | | | - Yolanda Fernández-Cladera
- Division of Central Laboratory, Hospital Universitario de Canarias, 38320 Tenerife, Spain; (F.G.-B.); (Y.F.-C.); (A.F.G.-R.)
| | - Agustín F. González-Rivero
- Division of Central Laboratory, Hospital Universitario de Canarias, 38320 Tenerife, Spain; (F.G.-B.); (Y.F.-C.); (A.F.G.-R.)
| | - Raquel López-Mejías
- Epidemiology, Genetics and Atherosclerosis Research Group on Systemic Inflammatory Diseases, Instituto de Investigación sanitaria Marqués de Valdecilla (IDIVAL), 39011 Santander, Spain;
| | - Federico Díaz-González
- Division of Rheumatology, Hospital Universitario de Canarias, 38320 Tenerife, Spain; (M.G.-G.); (F.D.-G.)
- Department of Internal Medicine, University of La Laguna (ULL), 38200 Tenerife, Spain
| | - Miguel Á. González-Gay
- Division of Rheumatology, IIS-Fundación Jiménez Díaz, 28040 Madrid, Spain
- Department of Medicine and Psychiatry, University of Cantabria, 39005 Santander, Spain
| | - Iván Ferraz-Amaro
- Division of Rheumatology, Hospital Universitario de Canarias, 38320 Tenerife, Spain; (M.G.-G.); (F.D.-G.)
- Department of Internal Medicine, University of La Laguna (ULL), 38200 Tenerife, Spain
| |
Collapse
|
12
|
Zafaranieh S, Stadler JT, Pammer A, Marsche G, van Poppel MNM, Desoye G. The Association of Physical Activity and Sedentary Behavior with Maternal and Cord Blood Anti-Oxidative Capacity and HDL Functionality: Findings of DALI Study. Antioxidants (Basel) 2023; 12:antiox12040827. [PMID: 37107203 PMCID: PMC10135087 DOI: 10.3390/antiox12040827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 03/13/2023] [Accepted: 03/20/2023] [Indexed: 03/30/2023] Open
Abstract
Obesity is one of the most common health issues in pregnancy with short and long-term consequences for both mother and her offspring. Promoting moderate to vigorous physical activity (MVPA) and decreasing sedentary time (ST) could have a positive impact on weight and obesity management, and therefore adiposity-induced oxidative stress, inflammation, and atherogenesis. However, the effects of MVPA and ST on anti-oxidative and anti-atherogenic markers in pregnancy have not been studied to date. This study aimed to assess the association of longitudinally and objectively measured MVPA and ST in 122 overweight/obese women (BMI ≥ 29 kg/m2) with maternal and cord blood markers of oxidative stress measured by advanced oxidation protein products (AOPP), anti-oxidative capacity, as well as high-density lipoproteins (HDL) related paraoxonase-1 (PON-1) activity and cholesterol efflux. Linear regression models showed no associations of MVPA and ST with outcomes in maternal blood. In contrast, MVPA at <20 weeks and 24–28 weeks of gestation were positively associated with anti-oxidative capacity, as well as PON-1 activity of HDL in cord blood. MVPA at 35–37 weeks correlated with higher AOPP, as well as higher anti-oxidative capacity. ST <20 weeks was also positively associated with inhibition of oxidation in cord blood. We speculate that increasing MVPA of overweight/obese women during pregnancy attenuates the oxidative stress state in the new-born.
Collapse
|
13
|
Matsumoto E, Oniki K, Ota-Kontani A, Seguchi Y, Sakamoto Y, Kaneko T, Imafuku T, Maeda H, Watanabe H, Maruyama T, Ogata Y, Yoshida M, Harada-Shiba M, Saruwatari J, Ogura M. Additive Effects of Drinking Habits and a Susceptible Genetic Polymorphism on Cholesterol Efflux Capacity. J Atheroscler Thromb 2023; 30:23-38. [PMID: 35249931 PMCID: PMC9899708 DOI: 10.5551/jat.63277] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 01/30/2022] [Indexed: 01/11/2023] Open
Abstract
AIMS High levels of high-density lipoprotein cholesterol (HDL-C) are not necessarily effective in preventing atherosclerotic cardiovascular disease, and cholesterol efflux capacity (CEC) has attracted attention regarding HDL functionality. We aimed to elucidate whether drinking habits are associated with CEC levels, while also paying careful attention to confounding factors including serum HDL-C levels, other life style factors, and rs671 (*2), a genetic polymorphism of the aldehyde dehydrogenase 2 (ALDH2) gene determining alcohol consumption habit. METHODS A cross-sectional study was performed in 505 Japanese male subjects who were recruited from a health screening program. Associations of HDL-C and CEC levels with drinking habits and ALDH2 genotypes were examined. RESULTS The genotype frequencies of ALDH2 *1/*1 (homozygous wild-type genotype), *1/*2 and *2/*2 (homozygous mutant genotype) were 55%, 37% and 8%, respectively. Both HDL-C and CEC levels were higher in ALDH2 *1/*1 genotype carriers than in *2 allele carriers. Although HDL-C levels were higher in subjects who had a drinking habit than in non-drinkers, CEC levels tended to be lower in subjects with ≥ 46 g/day of alcohol consumption than in non-drinkers. Furthermore, CEC levels tended to be lower in ALDH2 *1/*1 genotype carriers with a drinking habit of ≥ 46 g/day than non-drinkers, while for *2 allele carriers, CEC levels tended to be lower with a drinking habit of 23-45.9 g/day compared to no drinking habit. CONCLUSIONS Our results suggest that heavy drinking habits may tend to decrease CEC levels, and in the ALDH2 *2 allele carriers, even moderate drinking habits may tend to decrease CEC levels.
Collapse
Affiliation(s)
- Erika Matsumoto
- Division of Pharmacology and Therapeutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Kentaro Oniki
- Division of Pharmacology and Therapeutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Ami Ota-Kontani
- Department of Molecular Innovation in Lipidology, National Cerebral and Cardiovascular Center Research Institute, Osaka, Japan
| | - Yuri Seguchi
- Division of Pharmacology and Therapeutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yuki Sakamoto
- Division of Pharmacology and Therapeutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Tetsuya Kaneko
- Division of Pharmacology and Therapeutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Tadashi Imafuku
- Department of Molecular Pathophysiology, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, Japan
| | - Hitoshi Maeda
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Hiroshi Watanabe
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Toru Maruyama
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yasuhiro Ogata
- Japanese Red Cross Kumamoto Health Care Center, Kumamoto, Japan
| | - Minoru Yoshida
- Japanese Red Cross Kumamoto Health Care Center, Kumamoto, Japan
| | - Mariko Harada-Shiba
- Department of Molecular Innovation in Lipidology, National Cerebral and Cardiovascular Center Research Institute, Osaka, Japan
- Department of Molecular Pathogenesis, National Cerebral and Cardiovascular Center Research Institute, Osaka, Japan
| | - Junji Saruwatari
- Division of Pharmacology and Therapeutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Masatsune Ogura
- Department of Molecular Innovation in Lipidology, National Cerebral and Cardiovascular Center Research Institute, Osaka, Japan
- Department of General Medical Science, Chiba University Graduate School of Medicine, Chiba, Japan
- Department of Metabolism and Endocrinology, Eastern Chiba Medical Center, Togane, Japan
| |
Collapse
|
14
|
Fadaei R, Mohassel Azadi S, Rhéaume E, Khazaie H. High-density lipoprotein cholesterol efflux capacity in patients with obstructive sleep apnea and its relation with disease severity. Lipids Health Dis 2022; 21:116. [PMID: 36344946 PMCID: PMC9639319 DOI: 10.1186/s12944-022-01723-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 10/17/2022] [Indexed: 11/09/2022] Open
Abstract
Background Obstructive sleep apnea (OSA) is linked to an accelerated risk of cardiovascular disease (CVD). Some key CVD risk factors are present in patients suffering from OSA such as hypertension, inflammation, oxidative stress, and dyslipidemia. High-density lipoprotein (HDL) cholesterol efflux capacity (CEC) is proposed as a reliable biomarker of HDL function and the present study aimed to quantify this biomarker in patients with OSA. Methods ATP binding cassette subfamily A member 1 (ABCA1), non-ABCA1, and total CEC were determined in 69 polysomnographic-confirmed OSA patients and 23 controls. Moreover, paraoxonase (PON) activities, high-sensitivity C-reactive protein (hsCRP), apolipoprotein B (apo B), and apolipoprotein A-I (apo A-I) circulating levels were quantified in the studied population. Results: All CEC measures were reduced in the OSA group compared to the control group. Strikingly, ABCA1 CEC was diminished in severe OSA in comparison with mild OSA. Furthermore, PON activities and apo A-I showed lower levels, while hsCRP and apo B were elevated in OSA patients compared to controls. Moreover, ABCA1 CEC showed an inverse association with hsCRP and a positive association with apo A-I, while non-ABCA1 CEC presented an association with HDL-C. Conclusion These results suggest the presence of an impaired HDL function in OSA. In particular, ABCA1 CEC was associated with disease severity and inflammation which could be a factor increasing the risk of CVD. Supplementary Information The online version contains supplementary material available at 10.1186/s12944-022-01723-w.
Collapse
Affiliation(s)
- Reza Fadaei
- grid.412112.50000 0001 2012 5829Sleep Disorders Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Samaneh Mohassel Azadi
- grid.411705.60000 0001 0166 0922Department of Clinical Biochemistry, Faculty of Medicine Tehran University of Medical Sciences, Tehran, Iran
| | - Eric Rhéaume
- grid.482476.b0000 0000 8995 9090Montreal Heart Institute, 5000 Belanger Street, Montreal, H1T 1C8 Canada ,grid.14848.310000 0001 2292 3357Department of medicine, Université de Montréal, 2900 Edouard-Montpetit boulevard, Montreal, H3T 1J4 Canada
| | - Habibolah Khazaie
- grid.412112.50000 0001 2012 5829Sleep Disorders Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
15
|
Abstract
Lipid disorders involving derangements in serum cholesterol, triglycerides, or both are commonly encountered in clinical practice and often have implications for cardiovascular risk and overall health. Recent advances in knowledge, recommendations, and treatment options have necessitated an updated approach to these disorders. Older classification schemes have outlived their usefulness, yielding to an approach based on the primary lipid disturbance identified on a routine lipid panel as a practical starting point. Although monogenic dyslipidemias exist and are important to identify, most individuals with lipid disorders have polygenic predisposition, often in the context of secondary factors such as obesity and type 2 diabetes. With regard to cardiovascular disease, elevated low-density lipoprotein cholesterol is essentially causal, and clinical practice guidelines worldwide have recommended treatment thresholds and targets for this variable. Furthermore, recent studies have established elevated triglycerides as a cardiovascular risk factor, whereas depressed high-density lipoprotein cholesterol now appears less contributory than was previously believed. An updated approach to diagnosis and risk assessment may include measurement of secondary lipid variables such as apolipoprotein B and lipoprotein(a), together with selective use of genetic testing to diagnose rare monogenic dyslipidemias such as familial hypercholesterolemia or familial chylomicronemia syndrome. The ongoing development of new agents-especially antisense RNA and monoclonal antibodies-targeting dyslipidemias will provide additional management options, which in turn motivates discussion on how best to incorporate them into current treatment algorithms.
Collapse
Affiliation(s)
- Amanda J Berberich
- Department of Medicine; Schulich School of Medicine and Dentistry, Western University, London, ON, Canada, N6A 5C1.,Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada, N6A 5B7
| | - Robert A Hegele
- Department of Medicine; Schulich School of Medicine and Dentistry, Western University, London, ON, Canada, N6A 5C1.,Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada, N6A 5B7
| |
Collapse
|
16
|
Role of ABCA1 in Cardiovascular Disease. J Pers Med 2022; 12:jpm12061010. [PMID: 35743794 PMCID: PMC9225161 DOI: 10.3390/jpm12061010] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Revised: 06/17/2022] [Accepted: 06/17/2022] [Indexed: 11/17/2022] Open
Abstract
Cholesterol homeostasis plays a significant role in cardiovascular disease. Previous studies have indicated that ATP-binding cassette transporter A1 (ABCA1) is one of the most important proteins that maintains cholesterol homeostasis. ABCA1 mediates nascent high-density lipoprotein biogenesis. Upon binding with apolipoprotein A-I, ABCA1 facilitates the efflux of excess intracellular cholesterol and phospholipids and controls the rate-limiting step of reverse cholesterol transport. In addition, ABCA1 interacts with the apolipoprotein receptor and suppresses inflammation through a series of signaling pathways. Thus, ABCA1 may prevent cardiovascular disease by inhibiting inflammation and maintaining lipid homeostasis. Several studies have indicated that post-transcriptional modifications play a critical role in the regulation of ABCA1 transportation and plasma membrane localization, which affects its biological function. Meanwhile, carriers of the loss-of-function ABCA1 gene are often accompanied by decreased expression of ABCA1 and an increased risk of cardiovascular diseases. We summarized the ABCA1 transcription regulation mechanism, mutations, post-translational modifications, and their roles in the development of dyslipidemia, atherosclerosis, ischemia/reperfusion, myocardial infarction, and coronary heart disease.
Collapse
|
17
|
HDL as Bidirectional Lipid Vectors: Time for New Paradigms. Biomedicines 2022; 10:biomedicines10051180. [PMID: 35625916 PMCID: PMC9138557 DOI: 10.3390/biomedicines10051180] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 05/10/2022] [Accepted: 05/13/2022] [Indexed: 02/06/2023] Open
Abstract
The anti-atherogenic properties of high-density lipoproteins (HDL) have been explained mainly by reverse cholesterol transport (RCT) from peripheral tissues to the liver. The RCT seems to agree with most of the negative epidemiological correlations between HDL cholesterol levels and coronary artery disease. However, therapies designed to increase HDL cholesterol failed to reduce cardiovascular risk, despite their capacity to improve cholesterol efflux, the first stage of RCT. Therefore, the cardioprotective role of HDL may not be explained by RCT, and it is time for new paradigms about the physiological function of these lipoproteins. It should be considered that the main HDL apolipoprotein, apo AI, has been highly conserved throughout evolution. Consequently, these lipoproteins play an essential physiological role beyond their capacity to protect against atherosclerosis. We propose HDL as bidirectional lipid vectors carrying lipids from and to tissues according to their local context. Lipid influx mediated by HDL appears to be particularly important for tissue repair right on site where the damage occurs, including arteries during the first stages of atherosclerosis. In contrast, the HDL-lipid efflux is relevant for secretory cells where the fusion of intracellular vesicles drastically enlarges the cytoplasmic membrane with the potential consequence of impairment of cell function. In such circumstances, HDL could deliver some functional lipids and pick up not only cholesterol but an integral part of the membrane in excess, restoring the viability of the secretory cells. This hypothesis is congruent with the beneficial effects of HDL against atherosclerosis as well as with their capacity to induce insulin secretion and merits experimental exploration.
Collapse
|
18
|
Liu H, Peng D. Update on dyslipidemia in hypothyroidism: the mechanism of dyslipidemia in hypothyroidism. Endocr Connect 2022; 11:e210002. [PMID: 35015703 PMCID: PMC8859969 DOI: 10.1530/ec-21-0002] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Accepted: 01/11/2022] [Indexed: 11/18/2022]
Abstract
Hypothyroidism is often associated with elevated serum levels of total cholesterol, LDL-C and triglycerides. Thyroid hormone (TH) affects the production, clearance and transformation of cholesterol, but current research shows that thyroid-stimulating hormone (TSH) also participates in lipid metabolism independently of TH. Therefore, the mechanism of hypothyroidism-related dyslipidemia is associated with the decrease of TH and the increase of TSH levels. Some newly identified regulatory factors, such as proprotein convertase subtilisin/kexin type 9, angiogenin-like proteins and fibroblast growth factors are the underlying causes of dyslipidemia in hypothyroidism. HDL serum concentration changes were not consistent, and its function was reportedly impaired. The current review focuses on the updated understanding of the mechanism of hypothyroidism-related dyslipidemia.
Collapse
Affiliation(s)
- Huixing Liu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Daoquan Peng
- Department of Cardiovascular Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Correspondence should be addressed to D Peng:
| |
Collapse
|
19
|
Triglycerides and low HDL cholesterol predict coronary heart disease risk in patients with stable angina. Sci Rep 2021; 11:20714. [PMID: 34671067 PMCID: PMC8528835 DOI: 10.1038/s41598-021-00020-3] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 10/05/2021] [Indexed: 12/12/2022] Open
Abstract
We assessed whether high triglycerides (TG) and low high-density lipoprotein cholesterol (HDL-C) levels, expressed by an increased TG/HDL-C ratio, predict coronary atherosclerotic disease (CAD) outcomes in patients with stable angina. We studied 355 patients (60 ± 9 years, 211 males) with stable angina who underwent coronary computed tomography angiography (CTA), were managed clinically and followed for 4.5 ± 0.9 years. The primary composite outcome was all-cause mortality and non-fatal myocardial infarction. At baseline, the proportion of males, patients with metabolic syndrome, diabetes and obstructive CAD increased across TG/HDL-C ratio quartiles, together with markers of insulin resistance, hepatic and adipose tissue dysfunction and myocardial damage, with no difference in total cholesterol or LDL-C. At follow-up, the global CTA risk score (HR 1.06, 95% confidence interval (CI) 1.03–1.09, P = 0.001) and the IV quartile of the TG/HDL-C ratio (HR 2.85, 95% CI 1.30–6.26, P < 0.01) were the only independent predictors of the primary outcome. The TG/HDL-C ratio and the CTA risk score progressed over time despite increased use of lipid-lowering drugs and reduction in LDL-C. In patients with stable angina, high TG and low HDL-C levels are associated with CAD related outcomes independently of LDL-C and treatments. Trial registration. EVINCI study: ClinicalTrials.gov NCT00979199, registered September 17, 2009; SMARTool study: ClinicalTrials.gov NCT04448691, registered June 26, 2020.
Collapse
|
20
|
Schachtl-Riess JF, Coassin S, Lamina C, Demetz E, Streiter G, Hilbe R, Kronenberg F. Lysis reagents, cell numbers, and calculation method influence high-throughput measurement of HDL-mediated cholesterol efflux capacity. J Lipid Res 2021; 62:100125. [PMID: 34571016 PMCID: PMC8521207 DOI: 10.1016/j.jlr.2021.100125] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 09/09/2021] [Accepted: 09/15/2021] [Indexed: 11/23/2022] Open
Abstract
HDL-mediated cholesterol efflux capacity (CEC) may protect against cardiovascular disease. However, CEC assays are not standardized, hampering their application in large cohorts and comparison between studies. To improve standardization, we systematically investigated technical differences between existing protocols that influence assay performance that have not been previously addressed. CEC was measured in 96-well plates using J774A.1 macrophages labeled with BODIPY-cholesterol and incubated for 4 h with 2% apolipoprotein B-depleted human serum. The time zero method, which calculates CEC using control wells, and the per-well method, which calculates CEC based on the actual content of BODIPY-cholesterol in each well, were compared in 506 samples. We showed that the per-well method had a considerably lower sample rejection rate (4.74% vs. 13.44%) and intra-assay (4.48% vs. 5.28%) and interassay coefficients of variation (two controls: 7.85%, 9.86% vs. 13.58%, 15.29%) compared with the time zero method. Correction for plate-to-plate differences using four controls on each plate also improved assay performance of both methods. In addition, we observed that the lysis reagent used had a significant effect. Compared with cholic acid, lysis with sodium hydroxide results in higher (P = 0.0082) and Triton X-100 in lower (P = 0.0028) CEC values. Furthermore, large cell seeding errors (30% variation) greatly biased CEC for both referencing methods (P < 0.0001) as measured by a resazurin assay. In conclusion, lysis reagents, cell numbers, and assay setup greatly impact the quality and reliability of CEC quantification and should be considered when this method is newly established in a laboratory.
Collapse
Affiliation(s)
- Johanna F Schachtl-Riess
- Department of Genetics and Pharmacology, Institute of Genetic Epidemiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Stefan Coassin
- Department of Genetics and Pharmacology, Institute of Genetic Epidemiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Claudia Lamina
- Department of Genetics and Pharmacology, Institute of Genetic Epidemiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Egon Demetz
- Department of Internal Medicine II, Medical University of Innsbruck, Innsbruck, Austria
| | - Gertraud Streiter
- Department of Genetics and Pharmacology, Institute of Genetic Epidemiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Richard Hilbe
- Department of Internal Medicine II, Medical University of Innsbruck, Innsbruck, Austria
| | - Florian Kronenberg
- Department of Genetics and Pharmacology, Institute of Genetic Epidemiology, Medical University of Innsbruck, Innsbruck, Austria.
| |
Collapse
|
21
|
El-Ghazali A, Deodhar S, Saldanha S, Smyth B, Izbrand M, Gangwar A, Pahlavani M, Rohatgi A. Molecular Patterns of Extreme and Persistent Cholesterol Efflux Capacity. Arterioscler Thromb Vasc Biol 2021; 41:2588-2597. [PMID: 34433296 DOI: 10.1161/atvbaha.120.315648] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Objective: Cholesterol efflux capacity (CEC), the ability of extracellular acceptors to pick-up cholesterol from macrophages, is a clinically relevant cardiovascular biomarker. CEC is inversely associated with incident atherosclerotic cardiovascular disease events. However, CEC is only modestly associated with HDL-C (high-density lipoprotein cholesterol) levels, which may explain the failure of HDL-C raising therapies to improve atherosclerotic cardiovascular disease outcomes. Determinants of variation in CEC are not well understood. Thus, we sought to establish whether extreme high and low CEC is a robust persistent phenotype and to characterize associations with cholesterol, protein, and phospholipids across the particle size distribution.
Approach and Results: CEC was previously measured in 2924 participants enrolled in the Dallas Heart Study, a multi-ethnic population-based study from 2000 to 2002. We prospectively recruited those who were below the 10th and above 90th percentile of CEC. Our study revealed that extreme low and high CEC are persistent, robust phenotypes after 15 years of follow-up. Using size exclusion chromatography, CEC to fractionated plasma depleted of apolipoprotein B (fraction-specific CEC) demonstrated significant differences in CEC patterns between persistent high and low efflux groups. Fraction-specific CEC was correlated with fraction-specific total phospholipid but not apolipoprotein A-I, cholesterol, or total protein. These correlations varied across the size distribution and differed among persistent high versus low efflux groups.
Conclusions: Extreme high and low CEC are persistent and robust phenotypes. CEC patterns in fractionated plasma reveal marked variation across the size distribution. Future studies are warranted to determine specific molecular species linked to CEC in a size-specific manner.
Collapse
Affiliation(s)
- Ayea El-Ghazali
- Department of Internal Medicine, Division of Cardiology. University of Texas Southwestern Medical Center, Dallas (A.E.-G., S.D., S.S., B.S., A.G., M.P., A.R.)
| | - Sneha Deodhar
- Department of Internal Medicine, Division of Cardiology. University of Texas Southwestern Medical Center, Dallas (A.E.-G., S.D., S.S., B.S., A.G., M.P., A.R.)
| | - Suzanne Saldanha
- Department of Internal Medicine, Division of Cardiology. University of Texas Southwestern Medical Center, Dallas (A.E.-G., S.D., S.S., B.S., A.G., M.P., A.R.)
| | - Brooke Smyth
- Department of Internal Medicine, Division of Cardiology. University of Texas Southwestern Medical Center, Dallas (A.E.-G., S.D., S.S., B.S., A.G., M.P., A.R.)
| | - Mark Izbrand
- Department of Pediatrics, Division of Infectious Disease, University of Colorado Anschutz Medical Campus, Aurora (M.I.)
| | - Anamika Gangwar
- Department of Internal Medicine, Division of Cardiology. University of Texas Southwestern Medical Center, Dallas (A.E.-G., S.D., S.S., B.S., A.G., M.P., A.R.)
| | - Mandana Pahlavani
- Department of Internal Medicine, Division of Cardiology. University of Texas Southwestern Medical Center, Dallas (A.E.-G., S.D., S.S., B.S., A.G., M.P., A.R.)
| | - Anand Rohatgi
- Department of Internal Medicine, Division of Cardiology. University of Texas Southwestern Medical Center, Dallas (A.E.-G., S.D., S.S., B.S., A.G., M.P., A.R.)
| |
Collapse
|
22
|
Robert J, Osto E, von Eckardstein A. The Endothelium Is Both a Target and a Barrier of HDL's Protective Functions. Cells 2021; 10:1041. [PMID: 33924941 PMCID: PMC8146309 DOI: 10.3390/cells10051041] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 04/19/2021] [Accepted: 04/26/2021] [Indexed: 12/11/2022] Open
Abstract
The vascular endothelium serves as a barrier between the intravascular and extravascular compartments. High-density lipoproteins (HDL) have two kinds of interactions with this barrier. First, bloodborne HDL must pass the endothelium to access extravascular tissues, for example the arterial wall or the brain, to mediate cholesterol efflux from macrophages and other cells or exert other functions. To complete reverse cholesterol transport, HDL must even pass the endothelium a second time to re-enter circulation via the lymphatics. Transendothelial HDL transport is a regulated process involving scavenger receptor SR-BI, endothelial lipase, and ATP binding cassette transporters A1 and G1. Second, HDL helps to maintain the integrity of the endothelial barrier by (i) promoting junction closure as well as (ii) repair by stimulating the proliferation and migration of endothelial cells and their progenitor cells, and by preventing (iii) loss of glycocalix, (iv) apoptosis, as well as (v) transmigration of inflammatory cells. Additional vasoprotective functions of HDL include (vi) the induction of nitric oxide (NO) production and (vii) the inhibition of reactive oxygen species (ROS) production. These vasoprotective functions are exerted by the interactions of HDL particles with SR-BI as well as specific agonists carried by HDL, notably sphingosine-1-phophate (S1P), with their specific cellular counterparts, e.g., S1P receptors. Various diseases modify the protein and lipid composition and thereby the endothelial functionality of HDL. Thorough understanding of the structure-function relationships underlying the multiple interactions of HDL with endothelial cells is expected to elucidate new targets and strategies for the treatment or prevention of various diseases.
Collapse
Affiliation(s)
| | | | - Arnold von Eckardstein
- Institute of Clinical Chemistry, University of Zurich and University Hospital of Zurich, 8091 Zurich, Switzerland; (J.R.); (E.O.)
| |
Collapse
|
23
|
Tereshkina YA, Kostryukova LV, Torkhovskaya TI, Khudoklinova YY, Tikhonova EG. [Plasma high density lipoproteins phospholipds as an indirect indicator of their cholesterol efflux capacity - new suspected atherosclerosis risk factor]. BIOMEDIT︠S︡INSKAI︠A︡ KHIMII︠A︡ 2021; 67:119-129. [PMID: 33860768 DOI: 10.18097/pbmc20216702119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
High density lipoproteins (HDL) are a unique natural structure, protecting the body from the development of atherosclerotic vascular lesions and cardiovascular diseases due to this ability to remove cholesterol from cells. Plasma HDL level estimated by their cholesterol content, is a common lipid parameter, and its decrease is considered as an established atherosclerosis risk factor. However, a number of studies have shown the absence of positive clinical effects after drug-induced increase in HDL cholesterol. There is increasing evidence that not only HDL concentration, but also HDL properties, considered in this review are important. Many studies showed the decrease of HDL cholesterol efflux capacity in patients with coronary heart diseases and its association with disease severity. Some authors consider a decrease of this HDL capacity as a new additional risk factor of atherosclerosis. The review summarizes existing information on various protein and lipid components of HDL with a primary emphasis on the HDL. Special attention is paid to correlation between the HDL cholesterol efflux capacity and HDL phospholipids and the ratio "phospholipids/free cholesterol". The accumulated information indicates importance of evaluation in the HDL fraction not only in terms of their cholesterol, but also phospholipids. In addition to the traditionally used lipid criteria, this would provide more comprehensive information about the activity of the reverse cholesterol transport process in the body and could contribute to the targeted correction of the detected disorders.
Collapse
|
24
|
Rhainds D, Packard CJ, Brodeur MR, Niesor EJ, Sacks FM, Jukema JW, Wright RS, Waters DD, Heinonen T, Black DM, Laghrissi-Thode F, Dubé MP, Pfeffer MA, Tardif JC. Role of Adenylate Cyclase 9 in the Pharmacogenomic Response to Dalcetrapib: Clinical Paradigm and Molecular Mechanisms in Precision Cardiovascular Medicine. CIRCULATION-GENOMIC AND PRECISION MEDICINE 2021; 14:e003219. [PMID: 33794646 DOI: 10.1161/circgen.121.003219] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Following the neutral results of the dal-OUTCOMES trial, a genome-wide study identified the rs1967309 variant in the adenylate cyclase type 9 (ADCY9) gene on chromosome 16 as being associated with the risk of future cardiovascular events only in subjects taking dalcetrapib, a CETP (cholesterol ester transfer protein) modulator. Homozygotes for the minor A allele (AA) were protected from recurrent cardiovascular events when treated with dalcetrapib, while homozygotes for the major G allele (GG) had increased risk. Here, we present the current state of knowledge regarding the impact of rs1967309 in ADCY9 on clinical observations and biomarkers in dalcetrapib trials and the effects of mouse ADCY9 gene inactivation on cardiovascular physiology. Finally, we present our current model of the interaction between dalcetrapib and ADCY9 gene variants in the arterial wall macrophage, based on the intracellular role of CETP in the transfer of complex lipids from endoplasmic reticulum membranes to lipid droplets. Briefly, the concept is that dalcetrapib would inhibit CETP-mediated transfer of cholesteryl esters, resulting in a progressive inhibition of cholesteryl ester synthesis and free cholesterol accumulation in the endoplasmic reticulum. Reduced ADCY9 activity, by paradoxically leading to higher cyclic AMP levels and in turn increased cellular cholesterol efflux, could impart cardiovascular protection in rs1967309 AA patients. The ongoing dal-GenE trial recruited 6145 patients with the protective AA genotype and will provide a definitive answer to whether dalcetrapib will be protective in this population.
Collapse
Affiliation(s)
- David Rhainds
- Montreal Heart Institute (D.R., M.R.B., M.-P.D., J.-C.T.)
| | | | | | | | - Frank M Sacks
- Harvard School of Public Health, Boston, MA (F.M.S.)
| | | | | | - David D Waters
- School of Medicine, University of California, San Francisco (D.D.W.)
| | - Therese Heinonen
- DalCor Pharmaceuticals, Leatherhead, United Kingdom & Zug, Switzerland (T.H., D.M.B., F.L.-T.)
| | - Donald M Black
- DalCor Pharmaceuticals, Leatherhead, United Kingdom & Zug, Switzerland (T.H., D.M.B., F.L.-T.)
| | - Fouzia Laghrissi-Thode
- DalCor Pharmaceuticals, Leatherhead, United Kingdom & Zug, Switzerland (T.H., D.M.B., F.L.-T.)
| | - Marie-Pierre Dubé
- Montreal Heart Institute (D.R., M.R.B., M.-P.D., J.-C.T.).,Université de Montréal, Montreal, Canada (M.-P.D., J.-C.T.)
| | - Marc A Pfeffer
- Brigham and Women's Hospital & Harvard Medical School, Boston, MA (M.A.P.)
| | - Jean-Claude Tardif
- Montreal Heart Institute (D.R., M.R.B., M.-P.D., J.-C.T.).,Université de Montréal, Montreal, Canada (M.-P.D., J.-C.T.)
| |
Collapse
|
25
|
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
|
26
|
Ke J, Wang Y, Liu S, Li K, Xu Y, Yang L, Zhao D. Relationship of Para and Perirenal Fat and High-Density Lipoprotein and Its Function in Patients with Type 2 Diabetes Mellitus. Int J Endocrinol 2021; 2021:9286492. [PMID: 34976052 PMCID: PMC8716211 DOI: 10.1155/2021/9286492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 11/15/2021] [Accepted: 12/08/2021] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Para and perirenal fat is a fat pad surrounding the kidneys. Recent studies showed the association between para and perirenal fat and cardiovascular diseases including atherosclerosis and hypertension. We aimed to assess the relationship between para-perirenal ultrasonographic fat thickness and serum high-density lipoprotein (HDL) level and cholesterol efflux capacity of HDL in patients with type 2 diabetes mellitus (T2DM). METHODS We recruited 58 subjects with T2DM and collected anthropometric indices including height, weight, waist circumference, and other clinical data. Para-perirenal ultrasonographic fat thickness (PUFT) was measured via ultrasound. Serum lipid profile and other metabolic indices were determined as well. Correlation analysis and regression analysis were performed to analyze the relationship between PUFT and HDL level and cholesterol efflux capacity in all patients and subgroups. RESULTS Patients with higher PUFT have lower serum HDL level but increased cholesterol efflux capacity. Further analysis showed that PUFT negatively correlated with the serum HDL level in all patients, with no difference in groups divided by body mass index (BMI). In addition, PUFT was positively correlated with cholesterol efflux capacity in all patients. Multiple stepwise regression analysis showed an independent association of PUFT and serum HDL level and cholesterol efflux capacity. CONCLUSIONS PUFT is closely correlated with the serum HDL level and cholesterol efflux capacity in patients with T2DM.
Collapse
Affiliation(s)
- Jing Ke
- Center for Endocrine Metabolism and Immune Diseases, Beijing Luhe Hospital, Capital Medical University, Beijing 101149, China
- Beijing Key Laboratory of Diabetes Research and Care, Beijing 101149, China
| | - Yan Wang
- Center for Endocrine Metabolism and Immune Diseases, Beijing Luhe Hospital, Capital Medical University, Beijing 101149, China
- Beijing Key Laboratory of Diabetes Research and Care, Beijing 101149, China
| | - Simo Liu
- Center for Endocrine Metabolism and Immune Diseases, Beijing Luhe Hospital, Capital Medical University, Beijing 101149, China
- Beijing Key Laboratory of Diabetes Research and Care, Beijing 101149, China
| | - Kun Li
- Center for Endocrine Metabolism and Immune Diseases, Beijing Luhe Hospital, Capital Medical University, Beijing 101149, China
- Beijing Key Laboratory of Diabetes Research and Care, Beijing 101149, China
| | - YueChao Xu
- Center for Endocrine Metabolism and Immune Diseases, Beijing Luhe Hospital, Capital Medical University, Beijing 101149, China
| | - Longyan Yang
- Center for Endocrine Metabolism and Immune Diseases, Beijing Luhe Hospital, Capital Medical University, Beijing 101149, China
- Beijing Key Laboratory of Diabetes Research and Care, Beijing 101149, China
| | - Dong Zhao
- Center for Endocrine Metabolism and Immune Diseases, Beijing Luhe Hospital, Capital Medical University, Beijing 101149, China
- Beijing Key Laboratory of Diabetes Research and Care, Beijing 101149, China
| |
Collapse
|
27
|
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]
|
28
|
Peterson SJ, Choudhary A, Kalsi AK, Zhao S, Alex R, Abraham NG. OX-HDL: A Starring Role in Cardiorenal Syndrome and the Effects of Heme Oxygenase-1 Intervention. Diagnostics (Basel) 2020; 10:E976. [PMID: 33233550 PMCID: PMC7699797 DOI: 10.3390/diagnostics10110976] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 11/17/2020] [Accepted: 11/19/2020] [Indexed: 12/14/2022] Open
Abstract
In this review, we will evaluate how high-density lipoprotein (HDL) and the reverse cholesterol transport (RCT) pathway are critical for proper cardiovascular-renal physiology. We will begin by reviewing the basic concepts of HDL cholesterol synthesis and pathway regulation, followed by cardiorenal syndrome (CRS) pathophysiology. After explaining how the HDL and RCT pathways become dysfunctional through oxidative processes, we will elaborate on the potential role of HDL dysfunction in CRS. We will then present findings on how HDL function and the inducible antioxidant gene heme oxygenase-1 (HO-1) are interconnected and how induction of HO-1 is protective against HDL dysfunction and important for the proper functioning of the cardiovascular-renal system. This will substantiate the proposal of HO-1 as a novel therapeutic target to prevent HDL dysfunction and, consequently, cardiovascular disease, renal dysfunction, and the onset of CRS.
Collapse
Affiliation(s)
- Stephen J. Peterson
- Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA;
- Department of Medicine, New York Presbyterian Brooklyn Methodist Hospital, Brooklyn, NY 11215, USA; (A.C.); (A.K.K.); (S.Z.)
| | - Abu Choudhary
- Department of Medicine, New York Presbyterian Brooklyn Methodist Hospital, Brooklyn, NY 11215, USA; (A.C.); (A.K.K.); (S.Z.)
| | - Amardeep K. Kalsi
- Department of Medicine, New York Presbyterian Brooklyn Methodist Hospital, Brooklyn, NY 11215, USA; (A.C.); (A.K.K.); (S.Z.)
| | - Shuyang Zhao
- Department of Medicine, New York Presbyterian Brooklyn Methodist Hospital, Brooklyn, NY 11215, USA; (A.C.); (A.K.K.); (S.Z.)
| | - Ragin Alex
- Department of Medicine, New York Medical College, Valhalla, NY 10595, USA;
| | - Nader G. Abraham
- Department of Medicine, New York Medical College, Valhalla, NY 10595, USA;
- Department of Pharmacology, New York Medical College, Valhalla, NY 10595, USA
- Department of Medicine, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25701, USA
| |
Collapse
|
29
|
Breder I, Cunha Breder J, Bonilha I, Munhoz DB, Medorima STK, Oliveira DC, do Carmo HR, Moreira C, Kontush A, Zimetti F, Zanotti I, Carvalho LSF, Nadruz W, Muscelli E, Quinaglia T, Sposito AC. Rationale and design of the expanded combination of evolocumab plus empagliflozin in diabetes: EXCEED-BHS3 trial. Ther Adv Chronic Dis 2020; 11:2040622320959248. [PMID: 33062236 PMCID: PMC7534094 DOI: 10.1177/2040622320959248] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 08/21/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Patients with type 2 diabetes mellitus (T2DM) remain at increased cardiovascular residual risk and endothelial dysfunction, even after optimizing metabolic control and treatment by sodium-glucose-2 transporter inhibitors (SGLT2-is). The present study was based on the hypothesis that proprotein convertase subtilisin/kexin 9 inhibitor (PCSK9i) therapy may mitigate endothelial dysfunction in T2DM patients who are on regular treatment by SGLT2-i. METHODS The EXCEED-BHS3 is a prospective, single-center, investigator-blinded, open-label, randomized clinical trial. Participants (n = 110) will be randomized (1:1) to either empagliflozin 25 mg/day alone or empagliflozin 25 mg/day plus evolocumab 140 mg every 2 weeks in addition to optimal medical care. The primary endpoint was defined as the change in the 1-min flow-mediated dilation (FMD) after 16 weeks of treatment. The secondary endpoint is the FMD change after ischemia/reperfusion injury protocol (reserve FMD) after 16 weeks of treatment. Exploratory outcomes comprise the change in FMD and reserve FMD after 8 weeks of treatment and the change after 16 weeks of treatment in the following parameters: plasma levels of nitric oxide, vascular cell adhesion molecule-1 and isoprostane, high-density lipoprotein (HDL) and low-density lipoprotein subfractions profile, HDL function, blood pressure, body mass index, waist circumference and adipokines. CONCLUSION This will be the first study to evaluate the add-on effect of PCSK9i on endothelial function of T2DM patients under regular use of empagliflozin. TRIAL REGISTRATION ClinicalTrials.gov identifier: NCT03932721.
Collapse
Affiliation(s)
- Ikaro Breder
- Atherosclerosis and Vascular Biology Laboratory, Cardiology Department, State University of Campinas, SP, Brazil
| | - Jessica Cunha Breder
- Atherosclerosis and Vascular Biology Laboratory, Cardiology Department, State University of Campinas, SP, Brazil
| | - Isabella Bonilha
- Atherosclerosis and Vascular Biology Laboratory, Cardiology Department, State University of Campinas, SP, Brazil
| | - Daniel B. Munhoz
- Atherosclerosis and Vascular Biology Laboratory, Cardiology Department, State University of Campinas, SP, Brazil
| | - Sheila T. Kimura Medorima
- Atherosclerosis and Vascular Biology Laboratory, Cardiology Department, State University of Campinas, SP, Brazil
| | - Daniela C. Oliveira
- Atherosclerosis and Vascular Biology Laboratory, Cardiology Department, State University of Campinas, SP, Brazil
| | - Helison R. do Carmo
- Atherosclerosis and Vascular Biology Laboratory, Cardiology Department, State University of Campinas, SP, Brazil
| | - Camila Moreira
- Atherosclerosis and Vascular Biology Laboratory, Cardiology Department, State University of Campinas, SP, Brazil
| | - Anatol Kontush
- UMR-ICAN 1166, National Institute for Health and Medical Research (INSERM), Sorbonne University, Paris, France
| | - Francesca Zimetti
- Department of Food and Drugs, University of Parma, Parma, Emilia-Romagna, Italy
- BHS – Brazilian Heart Study Group, State University of Campinas, São Paulo, Brazil
| | - Ilaria Zanotti
- Department of Food and Drugs, University of Parma, Parma, Emilia-Romagna, Italy
- BHS – Brazilian Heart Study Group, State University of Campinas, São Paulo, Brazil
| | - Luiz Sergio F. Carvalho
- Atherosclerosis and Vascular Biology Laboratory, Cardiology Department, State University of Campinas, SP, Brazil
| | - Wilson Nadruz
- Atherosclerosis and Vascular Biology Laboratory, Cardiology Department, State University of Campinas, SP, Brazil
| | - Elza Muscelli
- Atherosclerosis and Vascular Biology Laboratory, Cardiology Department, State University of Campinas, SP, Brazil
| | - Thiago Quinaglia
- Atherosclerosis and Vascular Biology Laboratory, Cardiology Department, State University of Campinas, SP, Brazil
| | - Andrei C. Sposito
- Brazilian Heart Study Group, Cardiology Division, State University of Campinas, Rua Tessália Vieira de Camargo, 126., Cidade Universitária Zeferino Vaz, Campinas, São Paulo, 13084-971, Brazil
| | | |
Collapse
|
30
|
Cardner M, Yalcinkaya M, Goetze S, Luca E, Balaz M, Hunjadi M, Hartung J, Shemet A, Kränkel N, Radosavljevic S, Keel M, Othman A, Karsai G, Hornemann T, Claassen M, Liebisch G, Carreira E, Ritsch A, Landmesser U, Krützfeldt J, Wolfrum C, Wollscheid B, Beerenwinkel N, Rohrer L, von Eckardstein A. Structure-function relationships of HDL in diabetes and coronary heart disease. JCI Insight 2020; 5:131491. [PMID: 31830004 PMCID: PMC7030825 DOI: 10.1172/jci.insight.131491] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 12/04/2019] [Indexed: 12/24/2022] Open
Abstract
High-density lipoproteins (HDL) contain hundreds of lipid species and proteins and exert many potentially vasoprotective and antidiabetogenic activities on cells. To resolve structure-function-disease relationships of HDL, we characterized HDL of 51 healthy subjects and 98 patients with diabetes (T2DM), coronary heart disease (CHD), or both for protein and lipid composition, as well as functionality in 5 cell types. The integration of 40 clinical characteristics, 34 nuclear magnetic resonance (NMR) features, 182 proteins, 227 lipid species, and 12 functional read-outs by high-dimensional statistical modeling revealed, first, that CHD and T2DM are associated with different changes of HDL in size distribution, protein and lipid composition, and function. Second, different cellular functions of HDL are weakly correlated with each other and determined by different structural components. Cholesterol efflux capacity (CEC) was no proxy of other functions. Third, 3 potentially novel determinants of HDL function were identified and validated by the use of artificially reconstituted HDL, namely the sphingadienine-based sphingomyelin SM 42:3 and glycosylphosphatidylinositol-phospholipase D1 for the ability of HDL to inhibit starvation-induced apoptosis of human aortic endothelial cells and apolipoprotein F for the ability of HDL to promote maximal respiration of brown adipocytes.
Collapse
Affiliation(s)
- Mathias Cardner
- Department of Biosystems Science and Engineering, Swiss Federal Institute of Technology in Zurich (ETH Zurich), Basel, Switzerland
- Swiss Institute of Bioinformatics (SIB), Basel, Switzerland
| | - Mustafa Yalcinkaya
- Institute of Clinical Chemistry, University of Zurich and University Hospital of Zurich, Zurich, Switzerland
| | - Sandra Goetze
- Department of Health Sciences and Technology and
- Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland
| | - Edlira Luca
- Department of Diabetology and Endocrinology, University of Zurich and University Hospital of Zurich, Zurich, Switzerland
| | | | - Monika Hunjadi
- Department of Internal Medicine, Medical University of Innsbruck, Innsbruck, Austria
| | - Johannes Hartung
- Department of Cardiology, University Medicine Charité Berlin, Berlin, Germany
| | | | - Nicolle Kränkel
- Department of Cardiology, University Medicine Charité Berlin, Berlin, Germany
| | - Silvija Radosavljevic
- Institute of Clinical Chemistry, University of Zurich and University Hospital of Zurich, Zurich, Switzerland
| | - Michaela Keel
- Institute of Clinical Chemistry, University of Zurich and University Hospital of Zurich, Zurich, Switzerland
| | - Alaa Othman
- Institute of Clinical Chemistry, University of Zurich and University Hospital of Zurich, Zurich, Switzerland
| | - Gergely Karsai
- Institute of Clinical Chemistry, University of Zurich and University Hospital of Zurich, Zurich, Switzerland
| | - Thorsten Hornemann
- Institute of Clinical Chemistry, University of Zurich and University Hospital of Zurich, Zurich, Switzerland
| | - Manfred Claassen
- Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland
- Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Gerhard Liebisch
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, Regensburg, Germany
| | | | - Andreas Ritsch
- Department of Internal Medicine, Medical University of Innsbruck, Innsbruck, Austria
| | - Ulf Landmesser
- Department of Cardiology, University Medicine Charité Berlin, Berlin, Germany
| | - Jan Krützfeldt
- Department of Diabetology and Endocrinology, University of Zurich and University Hospital of Zurich, Zurich, Switzerland
| | | | - Bernd Wollscheid
- Department of Health Sciences and Technology and
- Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland
| | - Niko Beerenwinkel
- Department of Biosystems Science and Engineering, Swiss Federal Institute of Technology in Zurich (ETH Zurich), Basel, Switzerland
- Swiss Institute of Bioinformatics (SIB), Basel, Switzerland
| | - Lucia Rohrer
- Institute of Clinical Chemistry, University of Zurich and University Hospital of Zurich, Zurich, Switzerland
| | - Arnold von Eckardstein
- Institute of Clinical Chemistry, University of Zurich and University Hospital of Zurich, Zurich, Switzerland
| |
Collapse
|
31
|
Choi S, Park YE, Cheon EJ, Kim KY, Kim M, Ann SJ, Noh HM, Lee J, Lee CJ, Lee ST, Lee C, Lee JE, Lee SH. Novel Associations between Related Proteins and Cellular Effects of High-Density Lipoprotein. Korean Circ J 2019; 50:236-247. [PMID: 31845554 PMCID: PMC7043958 DOI: 10.4070/kcj.2019.0195] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 09/05/2019] [Accepted: 10/02/2019] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND AND OBJECTIVES Recent studies have examined the structure-function relationship of high-density lipoprotein (HDL). This study aimed to identify and rank HDL-associated proteins involved in several biological function of HDL. METHODS HDLs isolated from 48 participants were analyzed. Cholesterol efflux capacity, effect of HDL on nitric oxide production, and vascular cell adhesion molecule-1 expression were assessed. The relative abundance of identified proteins in the highest vs. lowest quartile was expressed using the normalized spectral abundance factor ratio. RESULTS After adjustment by multiple testing, six proteins, thyroxine-binding globulin, alpha-1B-glycoprotein, plasma serine protease inhibitor, vitronectin, angiotensinogen, and serum amyloid A-4, were more abundant (relative abundance ratio ≥2) in HDLs with the highest cholesterol efflux capacity. In contrast, three proteins, complement C4-A, alpha-2-macroglobulin, and immunoglobulin mu chain C region, were less abundant (relative abundance ratio <0.5). In terms of nitric oxide production and vascular cell adhesion molecule-1 expression, no proteins showed abundance ratios ≥2 or <0.5 after adjustment. Proteins correlated with the functional parameters of HDL belonged to diverse biological categories. CONCLUSIONS In summary, this study ranked proteins showing higher or lower abundance in HDLs with high functional capacities and newly identified multiple proteins linked to cholesterol efflux capacity.
Collapse
Affiliation(s)
- Seungbum Choi
- Cardiovascular Research Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Yae Eun Park
- Center for Theragnosis, Biomedical Research Institute, Korean Institute of Science and Technology, Seoul, Korea.,Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
| | - Eun Jeong Cheon
- Cardiovascular Research Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Kyeong Yeon Kim
- Center for Theragnosis, Biomedical Research Institute, Korean Institute of Science and Technology, Seoul, Korea.,Department of Chemistry, Sookmyung Women's University, Seoul, Korea.,Proteometech Inc., Seoul, Korea
| | - Miso Kim
- Cardiovascular Research Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Soo Jin Ann
- Cardiovascular Research Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Hye Min Noh
- Cardiovascular Research Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Jaeho Lee
- Cardiovascular Research Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Chan Joo Lee
- Cardiovascular Research Institute, Yonsei University College of Medicine, Seoul, Korea.,Division of Cardiology, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Seung Taek Lee
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
| | - Cheolju Lee
- Center for Theragnosis, Biomedical Research Institute, Korean Institute of Science and Technology, Seoul, Korea.,KHU-KIST Department of Converging Science and Technology, Kyung Hee University, Seoul, Korea.,Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology, Seoul, Korea
| | - Ji Eun Lee
- Center for Theragnosis, Biomedical Research Institute, Korean Institute of Science and Technology, Seoul, Korea.
| | - Sang Hak Lee
- Cardiovascular Research Institute, Yonsei University College of Medicine, Seoul, Korea.,Division of Cardiology, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea.
| |
Collapse
|