1
|
Proteomic analysis of postprandial high-density lipoproteins in healthy subjects. Int J Biol Macromol 2023; 225:1280-1290. [PMID: 36427620 DOI: 10.1016/j.ijbiomac.2022.11.187] [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: 09/15/2022] [Revised: 11/17/2022] [Accepted: 11/18/2022] [Indexed: 11/25/2022]
Abstract
The relationship between the functionality and composition of high-density lipoproteins (HDL) is yet not fully studied, and little is known about the influence of the diet in HDL proteome. Therefore, the aim of this research was to elucidate the HDL proteome associated to postprandial hyperlipidemia. Male volunteers were recruited for an interventional study with high fatty acid-based meals. Blood samples were collected before the intake (baseline), and 2-3 (postprandial peak) and 5-6 (postprandial post peak) hours later. HDL were purified and the protein composition was quantified by LC-MS/MS. Statistical analysis was performed by lineal models (amica) and by ANOVA and multi-t-test of the different conditions (MetaboAnalyst). Additionally, a clustering of the expression profiles of each protein was done with coseq R package (RStudio). Initially, 320 proteins were identified but only 119 remained after the filtering. APOM, APOE, APOB, and APOA2, proteins previously identified in the HDL proteome, were the only proteins with a statistically significant altered expression in postprandial hyperlipidemia when compared to baseline (p values <0.05 and logFC >1). In conclusion, we have been able to describe several behaviors of the whole HDL proteome during the postprandial hyperlipidemic metabolism.
Collapse
|
2
|
Gao H, Wu J, Sun Z, Zhang F, Shi T, Lu K, Qian D, Yin Z, Zhao Y, Qin J, Xue B. Influence of lecithin cholesterol acyltransferase alteration during different pathophysiologic conditions: A 45 years bibliometrics analysis. Front Pharmacol 2022; 13:1062249. [PMID: 36588724 PMCID: PMC9795195 DOI: 10.3389/fphar.2022.1062249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 12/06/2022] [Indexed: 12/15/2022] Open
Abstract
Background: Lecithin cholesterol acyltransferase (LCAT) is an important enzyme responsible for free cholesterol (FC) esterification, which is critical for high density lipoprotein (HDL) maturation and the completion of the reverse cholesterol transport (RCT) process. Plasma LCAT activity and concentration showed various patterns under different physiological and pathological conditions. Research on LCAT has grown rapidly over the past 50 years, but there are no bibliometric studies summarizing this field as a whole. This study aimed to use the bibliometric analysis to demonstrate the trends in LCAT publications, thus offering a brief perspective with regard to future developments in this field. Methods: We used the Web of Science Core Collection to retrieve LCAT-related studies published from 1975 to 2020. The data were further analyzed in the number of studies, the journal which published the most LCAT-related studies, co-authorship network, co-country network, co-institute network, co-reference and the keywords burst by CiteSpace V 5.7. Results: 2584 publications contained 55,311 references were used to analyzed. The number of included articles fluctuated in each year. We found that Journal of lipid research published the most LCAT-related studies. Among all the authors who work on LCAT, they tend to collaborate with a relatively stable group of collaborators to generate several major authors clusters which Albers, J. published the most studies (n = 53). The United States of America contributed the greatest proportion (n = 1036) of LCAT-related studies. The LCAT-related studies have been focused on the vascular disease, lecithin-cholesterol acyltransferase reaction, phospholipid, cholesterol efflux, chronic kidney disease, milk fever, nephrotic syndrome, platelet-activating factor acetylhydrolase, reconstituted lpa-i, reverse cholesterol transport. Four main research frontiers in terms of burst strength for LCAT-related studies including "transgenic mice", "oxidative stress", "risk", and "cholesterol metabolism "need more attention. Conclusion: This is the first study that demonstrated the trends and future development in LCAT publications. Further studies should focus on the accurate metabolic process of LCAT dependent or independent of RCT using metabolic marker tracking techniques. It was also well worth to further studying the possibility that LCAT may qualify as a biomarker for risk prediction and clinical treatment.
Collapse
Affiliation(s)
- Hongliang Gao
- Core Laboratory, Sir Run Run Hospital, Nanjing Medical University, Nanjing, China,School of Clinical Medicine, Wannan Medical College, Wuhu, China,Collaborative Innovation Center of Sustainable Forestry in Southern China, College of Forestry, Nanjing Forestry University, Nanjing, China
| | - Jing Wu
- Core Laboratory, Sir Run Run Hospital, Nanjing Medical University, Nanjing, China
| | - Zhenyu Sun
- School of Health Policy and Management, Center for Global Health, Nanjing Medical University, Nanjing, China
| | - Furong Zhang
- Core Laboratory, Sir Run Run Hospital, Nanjing Medical University, Nanjing, China
| | - Tianshu Shi
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Ke Lu
- Research Center for Computer-Aided Drug Discovery, Chinese Academy of Sciences, Shenzhen, China
| | - Dongfu Qian
- School of Health Policy and Management, Center for Global Health, Nanjing Medical University, Nanjing, China
| | - Zicheng Yin
- Nanjing Foreign Language School, Nanjing, China
| | - Yinjuan Zhao
- Collaborative Innovation Center of Sustainable Forestry in Southern China, College of Forestry, Nanjing Forestry University, Nanjing, China,*Correspondence: Bin Xue, ; Jian Qin, ; Yinjuan Zhao,
| | - Jian Qin
- Core Laboratory, Sir Run Run Hospital, Nanjing Medical University, Nanjing, China,*Correspondence: Bin Xue, ; Jian Qin, ; Yinjuan Zhao,
| | - Bin Xue
- Core Laboratory, Sir Run Run Hospital, Nanjing Medical University, Nanjing, China,*Correspondence: Bin Xue, ; Jian Qin, ; Yinjuan Zhao,
| |
Collapse
|
3
|
Holzer M, Ljubojevic-Holzer S, Souza Junior DR, Stadler JT, Rani A, Scharnagl H, Ronsein GE, Marsche G. HDL Isolated by Immunoaffinity, Ultracentrifugation, or Precipitation is Compositionally and Functionally Distinct. J Lipid Res 2022; 63:100307. [PMID: 36511335 PMCID: PMC9720336 DOI: 10.1016/j.jlr.2022.100307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 10/19/2022] [Accepted: 10/20/2022] [Indexed: 11/06/2022] Open
Abstract
The HDL proteome has been widely recognized as an important mediator of HDL function. While a variety of HDL isolation methods exist, their impact on the HDL proteome and its associated function remain largely unknown. Here, we compared three of the most common methods for HDL isolation, namely immunoaffinity (IA), density gradient ultracentrifugation (UC), and dextran-sulfate precipitation (DS), in terms of their effects on the HDL proteome and associated functionalities. We used state-of-the-art mass spectrometry to identify 171 proteins across all three isolation methods. IA-HDL contained higher levels of paraoxonase 1, apoB, clusterin, vitronectin, and fibronectin, while UC-HDL had higher levels of apoA2, apoC3, and α-1-antytrypsin. DS-HDL was enriched with apoA4 and complement proteins, while the apoA2 content was very low. Importantly, size-exclusion chromatography analysis showed that IA-HDL isolates contained subspecies in the size range above 12 nm, which were entirely absent in UC-HDL and DS-HDL isolates. Analysis of these subspecies indicated that they primarily consisted of apoA1, IGκC, apoC1, and clusterin. Functional analysis revealed that paraoxonase 1 activity was almost completely lost in IA-HDL, despite high paraoxonase content. We observed that the elution conditions, using 3M thiocyanate, during IA resulted in an almost complete loss of paraoxonase 1 activity. Notably, the cholesterol efflux capacity of UC-HDL and DS-HDL was significantly higher compared to IA-HDL. Together, our data clearly demonstrate that the isolation procedure has a substantial impact on the composition, subclass distribution, and functionality of HDL. In summary, our data show that the isolation procedure has a significant impact on the composition, subclass distribution and functionality of HDL. Our data can be helpful in the comparison, replication and analysis of proteomic datasets of HDL.
Collapse
Affiliation(s)
- Michael Holzer
- Division of Pharmacology, Otto-Loewi Research Centre, Medical University of Graz, Graz, Austria,BioTechMed Graz, Graz, Austria,For correspondence: Michael Holzer
| | - Senka Ljubojevic-Holzer
- BioTechMed Graz, Graz, Austria,Department of Cardiology, Medical University of Graz, Graz, Austria,Division of Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria
| | | | - Julia T. Stadler
- Division of Pharmacology, Otto-Loewi Research Centre, Medical University of Graz, Graz, Austria
| | - Alankrita Rani
- Division of Pharmacology, Otto-Loewi Research Centre, Medical University of Graz, Graz, Austria
| | - Hubert Scharnagl
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Graziella Eliza Ronsein
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Gunther Marsche
- Division of Pharmacology, Otto-Loewi Research Centre, Medical University of Graz, Graz, Austria,BioTechMed Graz, Graz, Austria
| |
Collapse
|
4
|
Schekatolina S, Lahovska V, Bekshaev A, Kontush S, Le Goff W, Kontush A. Mathematical Modelling of Material Transfer to High-Density Lipoprotein (HDL) upon Triglyceride Lipolysis by Lipoprotein Lipase: Relevance to Cardioprotective Role of HDL. Metabolites 2022; 12:metabo12070623. [PMID: 35888747 PMCID: PMC9317498 DOI: 10.3390/metabo12070623] [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: 11/16/2021] [Revised: 06/14/2022] [Accepted: 07/01/2022] [Indexed: 02/06/2023] Open
Abstract
High-density lipoprotein (HDL) contributes to lipolysis of triglyceride-rich lipoprotein (TGRL) by lipoprotein lipase (LPL) via acquirement of surface lipids, including free cholesterol (FC), released upon lipolysis. According to the reverse remnant-cholesterol transport (RRT) hypothesis recently developed by us, acquirement of FC by HDL is reduced at both low and extremely high HDL concentrations, potentially underlying the U-shaped relationship between HDL-cholesterol and cardiovascular disease. Mechanisms underlying impaired FC transfer however remain indeterminate. We developed a mathematical model of material transfer to HDL upon TGRL lipolysis by LPL. Consistent with experimental observations, mathematical modelling showed that surface components of TGRL, including FC, were accumulated in HDL upon lipolysis. The modelling successfully reproduced major features of cholesterol accumulation in HDL observed experimentally, notably saturation of this process over time and appearance of a maximum as a function of HDL concentration. The calculations suggested that the both phenomena resulted from competitive fluxes of FC through the HDL pool, including primarily those driven by FC concentration gradient between TGRL and HDL on the one hand and mediated by lecithin-cholesterol acyltransferase (LCAT) and cholesteryl ester transfer protein (CETP) on the other hand. These findings provide novel opportunities to revisit our view of HDL in the framework of RRT.
Collapse
Affiliation(s)
| | - Viktoriia Lahovska
- Odessa National Technological University, 65000 Odessa, Ukraine; (S.S.); (V.L.)
| | - Aleksandr Bekshaev
- Physics Research Institute, I.I. Mechnikov Odessa National University, 65082 Odessa, Ukraine; (A.B.); (S.K.)
| | - Sergey Kontush
- Physics Research Institute, I.I. Mechnikov Odessa National University, 65082 Odessa, Ukraine; (A.B.); (S.K.)
| | - Wilfried Le Goff
- Unité de Recherche sur les Maladies Cardiovasculaires, Institut National de la Santé et de la Recherche Médicale (INSERM), le Métabolisme et la Nutrition, ICAN, Sorbonne Université, F-75013 Paris, France;
| | - Anatol Kontush
- Unité de Recherche sur les Maladies Cardiovasculaires, Institut National de la Santé et de la Recherche Médicale (INSERM), le Métabolisme et la Nutrition, ICAN, Sorbonne Université, F-75013 Paris, France;
- Correspondence: ; Tel.: +33-(1)-40-77-96-33; Fax: +33-(1)-40-77-96-45
| |
Collapse
|
5
|
Intestinal and Hepatic Uptake of Dietary Peroxidized Lipids and Their Decomposition Products, and Their Subsequent Effects on Apolipoprotein A1 and Paraoxonase1. Antioxidants (Basel) 2021; 10:antiox10081258. [PMID: 34439506 PMCID: PMC8389297 DOI: 10.3390/antiox10081258] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 07/28/2021] [Accepted: 08/03/2021] [Indexed: 11/23/2022] Open
Abstract
Both pro- and antiatherosclerotic effects have been ascribed to dietary peroxidized lipids. Confusion on the role of peroxidized lipids in atherosclerotic cardiovascular disease is punctuated by a lack of understanding regarding the metabolic fate and potential physiological effects of dietary peroxidized lipids and their decomposition products. This study sought to determine the metabolic fate and physiological ramifications of 13-hydroperoxyoctadecadienoic acid (13-HPODE) and 13-HODE (13-hydroxyoctadecadienoic acid) supplementation in intestinal and hepatic cell lines, as well as any effects resulting from 13-HPODE or 13-HODE degradation products. In the presence of Caco-2 cells, 13-HPODE was rapidly reduced to 13-HODE. Upon entering the cell, 13-HODE appears to undergo decomposition, followed by esterification. Moreover, 13-HPODE undergoes autodecomposition to produce aldehydes such as 9-oxononanoic acid (9-ONA). Results indicate that 9-ONA was oxidized to azelaic acid (AzA) rapidly in cell culture media, but AzA was poorly absorbed by intestinal cells and remained detectable in cell culture media for up to 18 h. An increased apolipoprotein A1 (ApoA1) secretion was observed in Caco-2 cells in the presence of 13-HPODE, 9-ONA, and AzA, whereas such induction was not observed in HepG2 cells. However, 13-HPODE treatments suppressed paraoxonase 1 (PON1) activity, suggesting the induction of ApoA1 secretion by 13-HPODE may not represent functional high-density lipoprotein (HDL) capable of reducing oxidative stress. Alternatively, AzA induced both ApoA1 secretion and PON1 activity while suppressing ApoB secretion in differentiated Caco-2 cells but not in HepG2. These results suggest oxidation of 9-ONA to AzA might be an important phenomenon, resulting in the accumulation of potentially beneficial dietary peroxidized lipid-derived aldehydes.
Collapse
|
6
|
Ma F, Darabi M, Lhomme M, Tubeuf E, Canicio A, Brerault J, Medadje N, Rached F, Lebreton S, Frisdal E, Brites F, Serrano C, Santos R, Gautier E, Huby T, El Khoury P, Carrié A, Abifadel M, Bruckert E, Guerin M, Couvert P, Giral P, Lesnik P, Le Goff W, Guillas I, Kontush A. Phospholipid transfer to high-density lipoprotein (HDL) upon triglyceride lipolysis is directly correlated with HDL-cholesterol levels and is not associated with cardiovascular risk. Atherosclerosis 2021; 324:1-8. [PMID: 33798922 DOI: 10.1016/j.atherosclerosis.2021.03.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 02/05/2021] [Accepted: 03/04/2021] [Indexed: 10/22/2022]
Abstract
BACKGROUND AND AIMS While low concentrations of high-density lipoprotein-cholesterol (HDL-C) represent a well-established cardiovascular risk factor, extremely high HDL-C is paradoxically associated with elevated cardiovascular risk, resulting in the U-shape relationship with cardiovascular disease. Free cholesterol transfer to HDL upon lipolysis of triglyceride-rich lipoproteins (TGRL) was recently reported to underlie this relationship, linking HDL-C to triglyceride metabolism and atherosclerosis. In addition to free cholesterol, other surface components of TGRL, primarily phospholipids, are transferred to HDL during lipolysis. It remains indeterminate as to whether such transfer is linked to HDL-C and cardiovascular disease. METHODS AND RESULTS When TGRL was labelled with fluorescent phospholipid 1,1'-dioctadecyl-3,3,3',3'-tetramethylindocarbocyanine perchlorate (DiI), time- and dose-dependent transfer of DiI to HDL was observed upon incubations with lipoprotein lipase (LPL). The capacity of HDL to acquire DiI was decreased by -36% (p<0.001) in low HDL-C patients with acute myocardial infarction (n = 22) and by -95% (p<0.001) in low HDL-C subjects with Tangier disease (n = 7), unchanged in low HDL-C patients with Type 2 diabetes (n = 17) and in subjects with high HDL-C (n = 20), and elevated in subjects with extremely high HDL-C (+11%, p<0.05) relative to healthy normolipidemic controls. Across all the populations combined, HDL capacity to acquire DiI was directly correlated with HDL-C (r = 0.58, p<0.001). No relationship of HDL capacity to acquire DiI with both overall and cardiovascular mortality obtained from epidemiological studies for the mean HDL-C levels observed in the studied populations was obtained. CONCLUSIONS These data indicate that the capacity of HDL to acquire phospholipid from TGRL upon LPL-mediated lipolysis is proportional to HDL-C and does not reflect cardiovascular risk in subjects widely differing in HDL-C levels.
Collapse
Affiliation(s)
- Feng Ma
- National Institute for Health and Medical Research (INSERM) UMR_S 1166, Faculty of Medicine Pitie-Salpetriere, 91 Bld de L'Hopital, 75013, Paris, France; Sorbonne University, Paris, France.
| | - Maryam Darabi
- National Institute for Health and Medical Research (INSERM) UMR_S 1166, Faculty of Medicine Pitie-Salpetriere, 91 Bld de L'Hopital, 75013, Paris, France; Sorbonne University, Paris, France
| | - Marie Lhomme
- Institute of Cardiometabolism and Nutrition (ICAN), Paris, F-75013, France
| | - Emilie Tubeuf
- National Institute for Health and Medical Research (INSERM) UMR_S 1166, Faculty of Medicine Pitie-Salpetriere, 91 Bld de L'Hopital, 75013, Paris, France; Sorbonne University, Paris, France
| | - Aurélie Canicio
- National Institute for Health and Medical Research (INSERM) UMR_S 1166, Faculty of Medicine Pitie-Salpetriere, 91 Bld de L'Hopital, 75013, Paris, France; Sorbonne University, Paris, France
| | - Jean Brerault
- National Institute for Health and Medical Research (INSERM) UMR_S 1166, Faculty of Medicine Pitie-Salpetriere, 91 Bld de L'Hopital, 75013, Paris, France; Sorbonne University, Paris, France
| | - Narcisse Medadje
- National Institute for Health and Medical Research (INSERM) UMR_S 1166, Faculty of Medicine Pitie-Salpetriere, 91 Bld de L'Hopital, 75013, Paris, France; Sorbonne University, Paris, France
| | - Fabiana Rached
- National Institute for Health and Medical Research (INSERM) UMR_S 1166, Faculty of Medicine Pitie-Salpetriere, 91 Bld de L'Hopital, 75013, Paris, France; Sorbonne University, Paris, France; Heart Institute-InCor, University of Sao Paulo, Sao Paulo, Brazil
| | | | - Eric Frisdal
- National Institute for Health and Medical Research (INSERM) UMR_S 1166, Faculty of Medicine Pitie-Salpetriere, 91 Bld de L'Hopital, 75013, Paris, France; Sorbonne University, Paris, France
| | - Fernando Brites
- Laboratory of Lipids and Atherosclerosis, Department of Clinical Biochemistry, INFIBIOC, University of Buenos Aires, CONICET. Buenos Aires, Argentina
| | - Carlos Serrano
- Heart Institute-InCor, University of Sao Paulo, Sao Paulo, Brazil
| | - Raul Santos
- Heart Institute-InCor, University of Sao Paulo, Sao Paulo, Brazil
| | - Emmanuel Gautier
- National Institute for Health and Medical Research (INSERM) UMR_S 1166, Faculty of Medicine Pitie-Salpetriere, 91 Bld de L'Hopital, 75013, Paris, France; Sorbonne University, Paris, France
| | - Thierry Huby
- National Institute for Health and Medical Research (INSERM) UMR_S 1166, Faculty of Medicine Pitie-Salpetriere, 91 Bld de L'Hopital, 75013, Paris, France; Sorbonne University, Paris, France
| | - Petra El Khoury
- Laboratory of Biochemistry and Molecular Therapeutics, Faculty of Pharmacy, Pôle Technologie-Santé, Saint Joseph University, Beirut, Lebanon; INSERM LVTS U1148, Hôpital Bichat-Claude Bernard, Paris, France
| | - Alain Carrié
- National Institute for Health and Medical Research (INSERM) UMR_S 1166, Faculty of Medicine Pitie-Salpetriere, 91 Bld de L'Hopital, 75013, Paris, France; Sorbonne University, Paris, France
| | - Marianne Abifadel
- Laboratory of Biochemistry and Molecular Therapeutics, Faculty of Pharmacy, Pôle Technologie-Santé, Saint Joseph University, Beirut, Lebanon; INSERM LVTS U1148, Hôpital Bichat-Claude Bernard, Paris, France
| | - Eric Bruckert
- National Institute for Health and Medical Research (INSERM) UMR_S 1166, Faculty of Medicine Pitie-Salpetriere, 91 Bld de L'Hopital, 75013, Paris, France; Sorbonne University, Paris, France; Institute of Cardiometabolism and Nutrition (ICAN), Paris, F-75013, France; AP-HP, Groupe Hospitalier Pitié-Salpétrière, Paris, F-75013, France
| | - Maryse Guerin
- National Institute for Health and Medical Research (INSERM) UMR_S 1166, Faculty of Medicine Pitie-Salpetriere, 91 Bld de L'Hopital, 75013, Paris, France; Sorbonne University, Paris, France
| | - Philippe Couvert
- National Institute for Health and Medical Research (INSERM) UMR_S 1166, Faculty of Medicine Pitie-Salpetriere, 91 Bld de L'Hopital, 75013, Paris, France; Sorbonne University, Paris, France
| | - Philippe Giral
- National Institute for Health and Medical Research (INSERM) UMR_S 1166, Faculty of Medicine Pitie-Salpetriere, 91 Bld de L'Hopital, 75013, Paris, France; Sorbonne University, Paris, France; Institute of Cardiometabolism and Nutrition (ICAN), Paris, F-75013, France; AP-HP, Groupe Hospitalier Pitié-Salpétrière, Paris, F-75013, France
| | - Philippe Lesnik
- National Institute for Health and Medical Research (INSERM) UMR_S 1166, Faculty of Medicine Pitie-Salpetriere, 91 Bld de L'Hopital, 75013, Paris, France; Sorbonne University, Paris, France
| | - Wilfried Le Goff
- National Institute for Health and Medical Research (INSERM) UMR_S 1166, Faculty of Medicine Pitie-Salpetriere, 91 Bld de L'Hopital, 75013, Paris, France; Sorbonne University, Paris, France
| | - Isabelle Guillas
- National Institute for Health and Medical Research (INSERM) UMR_S 1166, Faculty of Medicine Pitie-Salpetriere, 91 Bld de L'Hopital, 75013, Paris, France; Sorbonne University, Paris, France
| | - Anatol Kontush
- National Institute for Health and Medical Research (INSERM) UMR_S 1166, Faculty of Medicine Pitie-Salpetriere, 91 Bld de L'Hopital, 75013, Paris, France; Sorbonne University, Paris, France
| |
Collapse
|
7
|
HDL and Reverse Remnant-Cholesterol Transport (RRT): Relevance to Cardiovascular Disease. Trends Mol Med 2020; 26:1086-1100. [DOI: 10.1016/j.molmed.2020.07.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 07/17/2020] [Accepted: 07/20/2020] [Indexed: 12/17/2022]
|
8
|
Feng M, Darabi M, Tubeuf E, Canicio A, Lhomme M, Frisdal E, Lanfranchi-Lebreton S, Matheron L, Rached F, Ponnaiah M, Serrano CV, Santos RD, Brites F, Bolbach G, Gautier E, Huby T, Carrie A, Bruckert E, Guerin M, Couvert P, Giral P, Lesnik P, Le Goff W, Guillas I, Kontush A. Free cholesterol transfer to high-density lipoprotein (HDL) upon triglyceride lipolysis underlies the U-shape relationship between HDL-cholesterol and cardiovascular disease. Eur J Prev Cardiol 2019; 27:1606-1616. [PMID: 31840535 DOI: 10.1177/2047487319894114] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Low concentrations of high-density lipoprotein cholesterol (HDL-C) represent a well-established cardiovascular risk factor. Paradoxically, extremely high HDL-C levels are equally associated with elevated cardiovascular risk, resulting in the U-shape relationship of HDL-C with cardiovascular disease. Mechanisms underlying this association are presently unknown. We hypothesised that the capacity of high-density lipoprotein (HDL) to acquire free cholesterol upon triglyceride-rich lipoprotein (TGRL) lipolysis by lipoprotein lipase underlies the non-linear relationship between HDL-C and cardiovascular risk. METHODS To assess our hypothesis, we developed a novel assay to evaluate the capacity of HDL to acquire free cholesterol (as fluorescent TopFluor® cholesterol) from TGRL upon in vitro lipolysis by lipoprotein lipase. RESULTS When the assay was applied to several populations markedly differing in plasma HDL-C levels, transfer of free cholesterol was significantly decreased in low HDL-C patients with acute myocardial infarction (-45%) and type 2 diabetes (-25%), and in subjects with extremely high HDL-C of >2.59 mmol/L (>100 mg/dL) (-20%) versus healthy normolipidaemic controls. When these data were combined and plotted against HDL-C concentrations, an inverse U-shape relationship was observed. Consistent with these findings, animal studies revealed that the capacity of HDL to acquire cholesterol upon lipolysis was reduced in low HDL-C apolipoprotein A-I knock-out mice and was negatively correlated with aortic accumulation of [3H]-cholesterol after oral gavage, attesting this functional characteristic as a negative metric of postprandial atherosclerosis. CONCLUSIONS Free cholesterol transfer to HDL upon TGRL lipolysis may underlie the U-shape relationship between HDL-C and cardiovascular disease, linking HDL-C to triglyceride metabolism and atherosclerosis.
Collapse
Affiliation(s)
- Ma Feng
- National Institute for Health and Medical Research (INSERM) UMR_S 1166, Faculty of Medicine Pitié-Salpétrière, Paris, France.,Sorbonne University, Paris, France
| | - Maryam Darabi
- National Institute for Health and Medical Research (INSERM) UMR_S 1166, Faculty of Medicine Pitié-Salpétrière, Paris, France.,Sorbonne University, Paris, France.,Institute of Cardiometabolism and Nutrition (ICAN), Paris, France
| | - Emilie Tubeuf
- National Institute for Health and Medical Research (INSERM) UMR_S 1166, Faculty of Medicine Pitié-Salpétrière, Paris, France.,Sorbonne University, Paris, France
| | - Aurélie Canicio
- National Institute for Health and Medical Research (INSERM) UMR_S 1166, Faculty of Medicine Pitié-Salpétrière, Paris, France.,Sorbonne University, Paris, France.,Institute of Cardiometabolism and Nutrition (ICAN), Paris, France
| | - Marie Lhomme
- Institute of Cardiometabolism and Nutrition (ICAN), Paris, France
| | - Eric Frisdal
- National Institute for Health and Medical Research (INSERM) UMR_S 1166, Faculty of Medicine Pitié-Salpétrière, Paris, France.,Sorbonne University, Paris, France
| | | | | | - Fabiana Rached
- National Institute for Health and Medical Research (INSERM) UMR_S 1166, Faculty of Medicine Pitié-Salpétrière, Paris, France.,Sorbonne University, Paris, France.,Heart Institute-InCor, University of Sao Paulo, Brazil
| | | | | | - Raul D Santos
- Heart Institute-InCor, University of Sao Paulo, Brazil
| | - Fernando Brites
- Heart Institute-InCor, University of Sao Paulo, Brazil.,Laboratory of Lipids and Atherosclerosis, Department of Clinical Biochemistry, INFIBIOC, University of Buenos Aires, CONICET, Argentina
| | | | - Emmanuel Gautier
- National Institute for Health and Medical Research (INSERM) UMR_S 1166, Faculty of Medicine Pitié-Salpétrière, Paris, France.,Sorbonne University, Paris, France
| | - Thierry Huby
- National Institute for Health and Medical Research (INSERM) UMR_S 1166, Faculty of Medicine Pitié-Salpétrière, Paris, France.,Sorbonne University, Paris, France
| | - Alain Carrie
- National Institute for Health and Medical Research (INSERM) UMR_S 1166, Faculty of Medicine Pitié-Salpétrière, Paris, France.,Sorbonne University, Paris, France
| | - Eric Bruckert
- National Institute for Health and Medical Research (INSERM) UMR_S 1166, Faculty of Medicine Pitié-Salpétrière, Paris, France.,Sorbonne University, Paris, France.,Institute of Cardiometabolism and Nutrition (ICAN), Paris, France.,AP-HP, Groupe hospitalier Pitié-Salpétrière, Paris, France
| | - Maryse Guerin
- National Institute for Health and Medical Research (INSERM) UMR_S 1166, Faculty of Medicine Pitié-Salpétrière, Paris, France.,Sorbonne University, Paris, France
| | - Philippe Couvert
- National Institute for Health and Medical Research (INSERM) UMR_S 1166, Faculty of Medicine Pitié-Salpétrière, Paris, France.,Sorbonne University, Paris, France
| | - Philippe Giral
- National Institute for Health and Medical Research (INSERM) UMR_S 1166, Faculty of Medicine Pitié-Salpétrière, Paris, France.,Sorbonne University, Paris, France.,Institute of Cardiometabolism and Nutrition (ICAN), Paris, France.,AP-HP, Groupe hospitalier Pitié-Salpétrière, Paris, France
| | - Philippe Lesnik
- National Institute for Health and Medical Research (INSERM) UMR_S 1166, Faculty of Medicine Pitié-Salpétrière, Paris, France.,Sorbonne University, Paris, France
| | - Wilfried Le Goff
- National Institute for Health and Medical Research (INSERM) UMR_S 1166, Faculty of Medicine Pitié-Salpétrière, Paris, France.,Sorbonne University, Paris, France
| | - Isabelle Guillas
- National Institute for Health and Medical Research (INSERM) UMR_S 1166, Faculty of Medicine Pitié-Salpétrière, Paris, France.,Sorbonne University, Paris, France
| | - Anatol Kontush
- National Institute for Health and Medical Research (INSERM) UMR_S 1166, Faculty of Medicine Pitié-Salpétrière, Paris, France.,Sorbonne University, Paris, France
| |
Collapse
|
9
|
High-density lipoprotein metabolism and reverse cholesterol transport: strategies for raising HDL cholesterol. Anatol J Cardiol 2019; 18:149-154. [PMID: 28766509 PMCID: PMC5731265 DOI: 10.14744/anatoljcardiol.2017.7608] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
A key to effective treatment of cardiovascular disease is to understand the body’s complex lipoprotein transport system. Reverse cholesterol transport (RCT) is the process of cholesterol movement from the extrahepatic tissues back to the liver. Lipoproteins containing apoA-I [high-density lipoprotein (HDL)] are key mediators in RCT, whereas non-high-density lipoproteins (non-HDL, lipoproteins containing apoB) are involved in the lipid delivery pathway. HDL particles are heterogeneous; they differ in proportion of proteins and lipids, size, shape, and charge. HDL heterogeneity is the result of the activity of several factors that assemble and remodel HDL particles in plasma: ATP-binding cassette transporter A1 (ABCA1), lecithin cholesterol acyltransferase (LCAT), cholesteryl ester transfer protein (CETP), hepatic lipase (HL), phospholipid transfer protein (PLTP), endothelial lipase (EL), and scavenger receptor class B type I (SR-BI). The RCT pathway consists of the following steps: 1. Cholesterol efflux from peripheral tissues to plasma, 2. LCAT-mediated esterification of cholesterol and remodeling of HDL particles, 3. direct pathway of HDL cholesterol delivery to the liver, and 4. indirect pathway of HDL cholesterol delivery to the liver via CETP-mediated transfer There are several established strategies for raising HDL cholesterol in humans, such as lifestyle changes; use of drugs including fibrates, statins, and niacin; and new therapeutic approaches. The therapeutic approaches include CETP inhibition, peroxisome proliferator-activated receptor (PPAR) agonists, synthetic farnesoid X receptor agonists, and gene therapy. Results of clinical trials should be awaited before further clinical management of atherosclerotic cardiovascular disease.
Collapse
|
10
|
Gaida JE, Alfredson H, Scott A, Mousavizadeh R, Forsgren S. Apolipoprotein A1 distribution pattern in the human Achilles tendon. Scand J Med Sci Sports 2018; 28:1506-1513. [PMID: 29315811 DOI: 10.1111/sms.13051] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/02/2018] [Indexed: 11/27/2022]
Abstract
Metabolic factors such as cholesterol appear to play an important role in the development of Achilles tendinopathy. There is, however, no morphologic proof explaining the link between high cholesterol and tendinopathy. As apolipoprotein A1 (Apo-A1) is essential for reverse cholesterol transport, it may be related to cholesterol overload in tendon. Nothing is known about Apo-A1 expression in tendon tissue. We examined the distribution of Apo-A1 protein in biopsies from normal and tendinopathy-affected human Achilles tendons, and APOA1 mRNA production from cultured human hamstring tenocytes. Specific immunoreactions for Apo-A1 were detected. The tenocytes showed specific Apo-A1 immunoreactions. These reactions were usually distinct in the tendinopathy specimens. While the tendinopathy specimens often showed granular/small deposit reactions, the slender tenocytes of control specimens did not show this pattern. The magnitude of Apo-A1 immunoreactivity was especially marked in the tendinopathy specimens, as there is a high number of tenocytes. Reactions were also seen in the walls of blood vessels located within the tendon tissue proper of both the normal and tendinopathy tendons and within the peritendinous/fatty tissue of the tendinopathy tendons. The reactions were predominantly in the form of deposit reactions within the smooth muscle layer of the vessel walls. Cultured hamstring tenocytes produced APOA1 mRNA. We demonstrated the presence of Apo-A1 in human tendon tissue. This suggests there may be a link between Achilles tendinopathy and cholesterol metabolism. We hypothesize that Apo-A1 may be important for tenocyte and blood vessel function within tendons.
Collapse
Affiliation(s)
- J E Gaida
- University of Canberra Research Institute for Sport and Exercise (UCRISE), Canberra, ACT, Australia.,Discipline of Physiotherapy, University of Canberra, Canberra, ACT, Australia.,Department of Integrative Medical Biology, Anatomy Section, Umeå University, Umeå, Sweden.,Department of Surgical and Perioperative Sciences, Sports Medicine, Umeå University, Umeå, Sweden
| | - H Alfredson
- Department of Community Medicine and Rehabilitation, Umeå University, Umeå, Sweden.,Institute of Sport Exercise and Health, University College Hospital London, London, UK
| | - A Scott
- Department of Physical Therapy, University of British Columbia, Vancouver, BC, Canada
| | - R Mousavizadeh
- Department of Physical Therapy, University of British Columbia, Vancouver, BC, Canada
| | - S Forsgren
- Department of Integrative Medical Biology, Anatomy Section, Umeå University, Umeå, Sweden
| |
Collapse
|
11
|
Darandale SS, Shevalkar GB, Vavia PR. Effect of Lipid Composition in Propofol Formulations: Decisive Component in Reducing the Free Propofol Content and Improving Pharmacodynamic Profiles. AAPS PharmSciTech 2017; 18:441-450. [PMID: 27055535 DOI: 10.1208/s12249-016-0524-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2015] [Accepted: 03/28/2016] [Indexed: 11/30/2022] Open
Abstract
Current endeavor was aimed towards studying significance of lipid composition on free propofol concentration in aqueous phase and associated pain on injection. Three different nanoformulations, namely long-chain triglyceride (LCT)/medium-chain glyceride (MCG)-based nanoemulsion (ProNano), MCG-based self-nanoemulsifying formulation (PSNE), and lipid-free nanoformulation (PNS) were accessed for the same. In vitro and in vivo performances of developed formulations were compared with Diprivan®. ProNano showed minimum free propofol concentration (0.13%) and hence lower pain on injection (rat paw-lick test, 6 ± 2 s) compared to Diprivan®, PSNE, and PNS (0.21%, 0.23%, and 0.51% free propofol, respectively, and rat paw-lick test; 12 ± 3, 14 ± 2, and 22 ± 3 s, respectively). These results conjecture the role of MCG in effective encapsulation of propofol. Anesthetic action assessed by measuring duration of loss of righting reflex (LORR), which was found similar in case of ProNano and PSNE (14 ± 3 and 15 ± 3 min, respectively) compared to Diprivan® (13 ± 3 min). In case of lipid-free formulation, PNS, extended anesthetic action (21 ± 2 min) was observed which may be due to sustained release of propofol from nanosponges. Studies on effect of lipoproteins on propofol release highlighted significance of HDL (100% release with maximum concentration of about 1.2 μg/ml of HDL) from all three formulations.
Collapse
|
12
|
Chistiakov DA, Orekhov AN, Bobryshev YV. ApoA1 and ApoA1-specific self-antibodies in cardiovascular disease. J Transl Med 2016; 96:708-18. [PMID: 27183204 DOI: 10.1038/labinvest.2016.56] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2016] [Revised: 03/21/2016] [Accepted: 04/03/2016] [Indexed: 12/15/2022] Open
Abstract
Apolipoprotein A1 (ApoA1) is a main protein moiety in high-density lipoprotein (HDL) particles. Generally, ApoA1 and HDL are considered as atheroprotective. In prooxidant and inflammatory microenvironment in the vicinity to the atherosclerotic lesion, ApoA1/HDL are subjected to modification. The chemical modifications such as oxidation, nitration, etc result in altering native architecture of ApoA1 toward dysfunctionality and abnormality. Neutrophil myeloperoxidase has a prominent role in this mechanism. Neo-epitopes could be formed and then exposed that makes them immunogenic. Indeed, these epitopes may be recognized by immune cells and induce production of proatherogenic ApoA1-specific IgG antibodies. These antibodies are biologically relevant because they are able to react with Toll-like receptor (TLR)-2 and TLR4 in target cells and induce a variety of pro-inflammatory responses. Epidemiological and functional studies underline a prognostic value of ApoA1 self-antibodies for several cardiovascular diseases, including myocardial infarction, acute coronary syndrome, and severe carotid stenosis.
Collapse
Affiliation(s)
- Dimitry A Chistiakov
- Department of Molecular Genetic Diagnostics and Cell Biology, Division of Laboratory Medicine, Institute of Pediatrics, Research Center for Children's Health, Moscow, Russia
| | - Alexander N Orekhov
- Institute of General Pathology and Pathophysiology, Russian Academy of Sciences, Moscow, Russia.,Faculty of Biology, Department of Biophysics, Lomonosov Moscow State University, Moscow, Russia
| | - Yuri V Bobryshev
- Institute of General Pathology and Pathophysiology, Russian Academy of Sciences, Moscow, Russia.,Faculty of Medicine, School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia.,School of Medicine, University of Western Sydney, Campbelltown, NSW, Australia
| |
Collapse
|
13
|
Rosales C, Patel N, Gillard BK, Yelamanchili D, Yang Y, Courtney HS, Santos RD, Gotto AM, Pownall HJ. Apolipoprotein AI deficiency inhibits serum opacity factor activity against plasma high density lipoprotein via a stabilization mechanism. Biochemistry 2015; 54:2295-302. [PMID: 25790332 DOI: 10.1021/bi501486z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The reaction of Streptococcal serum opacity factor (SOF) against plasma high-density lipoproteins (HDL) produces a large cholesteryl ester-rich microemulsion (CERM), a smaller neo HDL that is apolipoprotein (apo) AI-poor, and lipid-free apo AI. SOF is active versus both human and mouse plasma HDL. In vivo injection of SOF into mice reduces plasma cholesterol ∼40% in 3 h while forming the same products observed in vitro, but at different ratios. Previous studies supported the hypothesis that labile apo AI is required for the SOF reaction vs HDL. Here we further tested that hypothesis by studies of SOF against HDL from apo AI-null mice. When injected into apo AI-null mice, SOF reduced plasma cholesterol ∼35% in 3 h. The reaction of SOF vs apo AI-null HDL in vitro produced a CERM and neo HDL, but no lipid-free apo. Moreover, according to the rate of CERM formation, the extent and rate of the SOF reaction versus apo AI-null mouse HDL were less than that against wild-type (WT) mouse HDL. Chaotropic perturbation studies using guanidine hydrochloride showed that apo AI-null HDL was more stable than WT HDL. Human apo AI added to apo AI-null HDL was quantitatively incorporated, giving reconstituted HDL. Both SOF and guanidine hydrochloride displaced apo AI from the reconstituted HDL. These results support the conclusion that apo AI-null HDL is more stable than WT HDL because it lacks apo AI, a labile protein that is readily displaced by physicochemical and biochemical perturbations. Thus, apo AI-null HDL is less SOF-reactive than WT HDL. The properties of apo AI-null HDL can be partially restored to those of WT HDL by the spontaneous incorporation of human apo AI. It remains to be determined what other HDL functions are affected by apo AI deletion.
Collapse
Affiliation(s)
- Corina Rosales
- †Laboratory of Atherosclerosis and Lipoprotein Research, Department of Cardiology, Houston Methodist Research Institute, 6670 Bertner Avenue, Houston, Texas 77030, United States
| | - Niket Patel
- †Laboratory of Atherosclerosis and Lipoprotein Research, Department of Cardiology, Houston Methodist Research Institute, 6670 Bertner Avenue, Houston, Texas 77030, United States
| | - Baiba K Gillard
- †Laboratory of Atherosclerosis and Lipoprotein Research, Department of Cardiology, Houston Methodist Research Institute, 6670 Bertner Avenue, Houston, Texas 77030, United States
| | - Dedipya Yelamanchili
- †Laboratory of Atherosclerosis and Lipoprotein Research, Department of Cardiology, Houston Methodist Research Institute, 6670 Bertner Avenue, Houston, Texas 77030, United States
| | - Yaliu Yang
- †Laboratory of Atherosclerosis and Lipoprotein Research, Department of Cardiology, Houston Methodist Research Institute, 6670 Bertner Avenue, Houston, Texas 77030, United States
| | - Harry S Courtney
- ‡Veterans Affairs Medical Center and Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee 38104, United States
| | - Raul D Santos
- §Heart Institute-INCOR, University of Sao Paulo, 05409-003 Sao Paulo, Brazil
| | - Antonio M Gotto
- †Laboratory of Atherosclerosis and Lipoprotein Research, Department of Cardiology, Houston Methodist Research Institute, 6670 Bertner Avenue, Houston, Texas 77030, United States.,⊥Weill Cornell Medical College, 1305 York Avenue, New York, New York 10021, United States
| | - Henry J Pownall
- †Laboratory of Atherosclerosis and Lipoprotein Research, Department of Cardiology, Houston Methodist Research Institute, 6670 Bertner Avenue, Houston, Texas 77030, United States.,⊥Weill Cornell Medical College, 1305 York Avenue, New York, New York 10021, United States
| |
Collapse
|
14
|
Handa D, Kimura H, Oka T, Takechi Y, Okuhira K, Phillips MC, Saito H. Kinetic and thermodynamic analyses of spontaneous exchange between high-density lipoprotein-bound and lipid-free apolipoprotein A-I. Biochemistry 2015; 54:1123-31. [PMID: 25564321 DOI: 10.1021/bi501345j] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
It is thought that apolipoprotein A-I (apoA-I) spontaneously exchanges between high-density lipoprotein (HDL)-bound and lipid-free states, which is relevant to the occurrence of preβ-HDL particles in plasma. To improve our understanding of the mechanistic basis for this phenomenon, we performed kinetic and thermodynamic analyses for apoA-I exchange between discoidal HDL-bound and lipid-free forms using fluorescence-labeled apoA-I variants. Gel filtration experiments demonstrated that addition of excess lipid-free apoA-I to discoidal HDL particles promotes exchange of apoA-I between HDL-associated and lipid-free pools without alteration of the steady-state HDL particle size. Kinetic analysis of time-dependent changes in NBD fluorescence upon the transition of NBD-labeled apoA-I from HDL-bound to lipid-free state indicates that the exchange kinetics are independent of the collision frequency between HDL-bound and lipid-free apoA-I, in which the lipid binding ability of apoA-I affects the rate of association of lipid-free apoA-I with the HDL particles and not the rate of dissociation of HDL-bound apoA-I. Thus, C-terminal truncations or mutations that reduce the lipid binding affinity of apoA-I strongly impair the transition of lipid-free apoA-I to the HDL-bound state. Thermodynamic analysis of the exchange kinetics demonstrated that the apoA-I exchange process is enthalpically unfavorable but entropically favorable. These results explain the thermodynamic basis of the spontaneous exchange reaction of apoA-I associated with HDL particles. The altered exchangeability of dysfunctional apoA-I would affect HDL particle rearrangement, leading to perturbed HDL metabolism.
Collapse
Affiliation(s)
- Daisuke Handa
- Institute of Health Biosciences, Graduate School of Pharmaceutical Sciences, Tokushima University , 1-78-1 Shomachi, Tokushima 770-8505, Japan
| | | | | | | | | | | | | |
Collapse
|
15
|
Borja MS, Zhao L, Hammerson B, Tang C, Yang R, Carson N, Fernando G, Liu X, Budamagunta MS, Genest J, Shearer GC, Duclos F, Oda MN. HDL-apoA-I exchange: rapid detection and association with atherosclerosis. PLoS One 2013; 8:e71541. [PMID: 24015188 PMCID: PMC3756009 DOI: 10.1371/journal.pone.0071541] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2013] [Accepted: 06/28/2013] [Indexed: 01/23/2023] Open
Abstract
High density lipoprotein (HDL) cholesterol levels are associated with decreased risk of cardiovascular disease, but not all HDL are functionally equivalent. A primary determinant of HDL functional status is the conformational adaptability of its main protein component, apoA-I, an exchangeable apolipoprotein. Chemical modification of apoA-I, as may occur under conditions of inflammation or diabetes, can severely impair HDL function and is associated with the presence of cardiovascular disease. Chemical modification of apoA-I also impairs its ability to exchange on and off HDL, a critical process in reverse cholesterol transport. In this study, we developed a method using electron paramagnetic resonance spectroscopy (EPR) to quantify HDL-apoA-I exchange. Using this approach, we measured the degree of HDL-apoA-I exchange for HDL isolated from rabbits fed a high fat, high cholesterol diet, as well as human subjects with acute coronary syndrome and metabolic syndrome. We observed that HDL-apoA-I exchange was markedly reduced when atherosclerosis was present, or when the subject carries at least one risk factor of cardiovascular disease. These results show that HDL-apoA-I exchange is a clinically relevant measure of HDL function pertinent to cardiovascular disease.
Collapse
Affiliation(s)
- Mark S. Borja
- Children's Hospital Oakland Research Institute, Oakland, California, United States of America
| | - Lei Zhao
- Cardiovascular Drug Discovery, Bristol-Myers Squibb Company, Pennington, New Jersey, United States of America
| | - Bradley Hammerson
- Accent Assays, Inc., Sacramento, California, United States of America
| | - Chongren Tang
- Department of Medicine, University of Washington, Seattle, Washington, United States of America
| | - Richard Yang
- Cardiovascular Drug Discovery, Bristol-Myers Squibb Company, Pennington, New Jersey, United States of America
| | - Nancy Carson
- Cardiovascular Drug Discovery, Bristol-Myers Squibb Company, Pennington, New Jersey, United States of America
| | - Gayani Fernando
- Cardiovascular Drug Discovery, Bristol-Myers Squibb Company, Pennington, New Jersey, United States of America
| | - Xiaoqin Liu
- Cardiovascular Drug Discovery, Bristol-Myers Squibb Company, Pennington, New Jersey, United States of America
| | - Madhu S. Budamagunta
- Department of Biochemistry and Molecular Medicine, University of California Davis, Davis, California, United States of America
| | - Jacques Genest
- Royal Victoria Hospital, McGill University Health Centre, Montreal, Quebec, Canada
| | - Gregory C. Shearer
- Cardiovascular Health Research Center, Sanford Research/USD, Sioux Falls, South Dakota, United States of America
- Department of Internal Medicine and Department of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Sioux Falls, South Dakota, United States of America
| | - Franck Duclos
- Cardiovascular Drug Discovery, Bristol-Myers Squibb Company, Pennington, New Jersey, United States of America
| | - Michael N. Oda
- Children's Hospital Oakland Research Institute, Oakland, California, United States of America
- * E-mail:
| |
Collapse
|
16
|
Charles MA, Kane JP. New molecular insights into CETP structure and function: a review. J Lipid Res 2012; 53:1451-8. [PMID: 22679067 DOI: 10.1194/jlr.r027011] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Cholesteryl ester transfer protein (CETP) is important clinically and is the current target for new drug development. Its structure and mechanism of action has not been well understood. We have combined current new structural and functional methods to compare with relevant prior data. These analyses have led us to propose several steps in CETP's function at the molecular level, in the context of its interactions with lipoproteins, e.g., sensing, penetration, docking, selectivity, ternary complex formation, lipid transfer, and HDL dissociation. These new molecular insights improve our understanding of CETP's mechanisms of action.
Collapse
Affiliation(s)
- M Arthur Charles
- Department of Medicine University of California, San Francisco, CA 94158, USA.
| | | |
Collapse
|
17
|
Gorshkova IN, Atkinson D. Enhanced binding of apolipoprotein A-I variants associated with hypertriglyceridemia to triglyceride-rich particles. Biochemistry 2011; 50:2040-7. [PMID: 21288012 PMCID: PMC6128146 DOI: 10.1021/bi200158b] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Hypertriglyceridemia (HTG) is a common lipid abnormality in humans. However, its etiology remains largely unknown. It was shown that severe HTG can be induced in mice by overexpression of wild-type (WT) apolipoprotein E (apoE) or specific apoA-I mutants. Certain mutations in apoE4 were found to affect plasma triglyceride (TG) levels in mice overexpressing the protein. HTG appeared to positively correlate with the ability of the apoE4 variants to bind to TG-rich particles, protein destabilization, and the exposure of protein hydrophobic surface in solution. Here, we propose that the apoA-I mutations that cause HTG may also lead to changes in the conformation and stability that promote binding of apoA-I to TG-rich lipoproteins. To test this hypothesis, we studied binding to TG-rich emulsion and biophysical properties of the apoA-I mutants that induce HTG, apoA-I[E110A/E111A] and apoA-I[Δ(61-78)], and compared them to those of WT apoA-I and another apoA-I mutant, apoA-I[Δ(89-99)], that does not induce HTG but causes hypercholesterolemia in mice. We found that the apoA-I[E110A/E111A] and apoA-I[Δ(61-78)] mutations lead to enhanced binding of apoA-I to TG-rich particles, destabilization, and greater exposure of the hydrophobic surface of the protein. The apoA-I[Δ(89-99)] mutant did not show enhanced binding to the emulsion or a more exposed hydrophobic surface. Thus, like apoE4, the apoA-I variants that cause HTG in mice have the altered conformation and stability that facilitate their binding to TG-rich lipoproteins and thereby may lead to the reduced level of lipolysis of these lipoproteins. While many factors may be involved in induction of HTG, we suggest that an increased level of association of destabilized loosely folded apolipoproteins with TG-rich lipoproteins may contribute to some cases of HTG in humans.
Collapse
Affiliation(s)
- Irina N Gorshkova
- Department of Physiology and Biophysics, Boston University School of Medicine, 700 Albany Street, Boston, Massachusetts 02118, United States.
| | | |
Collapse
|
18
|
Courtney HS, Pownall HJ. The structure and function of serum opacity factor: a unique streptococcal virulence determinant that targets high-density lipoproteins. J Biomed Biotechnol 2010; 2010:956071. [PMID: 20671930 PMCID: PMC2910554 DOI: 10.1155/2010/956071] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2010] [Accepted: 05/26/2010] [Indexed: 12/04/2022] Open
Abstract
Serum opacity factor (SOF) is a virulence determinant expressed by a variety of streptococcal and staphylococcal species including both human and animal pathogens. SOF derives its name from its ability to opacify serum where it targets and disrupts the structure of high-density lipoproteins resulting in formation of large lipid vesicles that cause the serum to become cloudy. SOF is a multifunctional protein and in addition to its opacification activity, it binds to a number of host proteins that mediate adhesion of streptococci to host cells, and it plays a role in resistance to phagocytosis in human blood. This article will provide an overview of the structure and function of SOF, its role in the pathogenesis of streptococcal infections, its vaccine potential, its prevalence and distribution in bacteria, and the molecular mechanism whereby SOF opacifies serum and how an understanding of this mechanism may lead to therapies for reducing high-cholesterol concentrations in blood, a major risk factor for cardiovascular disease.
Collapse
Affiliation(s)
- Harry S Courtney
- Veterans Affairs Medical Center and Department of Medicine, University of Tennessee Health Science Center, 1030 Jefferson Avenue, Memphis, TN 38104, USA.
| | | |
Collapse
|
19
|
Abstract
PURPOSE OF REVIEW HDL is a cardioprotective lipoprotein, at least in part, because of its ability to mediate reverse cholesterol transport (RCT). It is becoming increasingly clear that the antiatherogenic effects of HDL are not only dependent on its concentration in circulating blood but also on its biological 'quality'. This review summarizes our current understanding of how the biological activities of individual subclasses of HDL particles contribute to overall HDL performance in RCT. RECENT FINDINGS Recent work indicates that apolipoprotein A-I-containing nascent HDL particles are heterogeneous and that such particles exert different effects on the RCT pathway. RCT from macrophages has been examined in detail in mice and the roles of plasma factors (lecithin-cholesterol acyltransferase, cholesterol ester transfer protein, phospholipid transfer protein) and cell factors (ATP-binding cassette transporter A1, ATP-binding cassette transporter G1, scavenger receptor class B type 1) have been evaluated. Manipulation of such factors has consistent effects on RCT and atherosclerosis, but the level of plasma HDL does not reliably predict the degree of RCT. Furthermore, HDL cholesterol or apolipoprotein A-I levels do not necessarily correlate with the magnitude of cholesterol efflux from macrophages; more understanding of the contributions of specific HDL subspecies is required. SUMMARY The antiatherogenic quality of HDL is defined by the functionality of HDL subspecies. In the case of RCT, the rate of cholesterol movement through the pathway is critical and the contributions of particular types of HDL particles to this process are becoming better defined.
Collapse
Affiliation(s)
- George H Rothblat
- Gastroenterology, Hepatology and Nutrition Division, Children's Hospital of Philadelphia, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104-4318, USA
| | | |
Collapse
|
20
|
Abstract
High density lipoprotein (HDL) possesses important anti-atherogenic properties and this review addresses the molecular mechanisms underlying these functions. The structures and cholesterol transport abilities of HDL particles are determined by the properties of their exchangeable apolipoprotein (apo) components. ApoA-I and apoE, which are the best characterized in structural terms, contain a series of amphipathic alpha-helical repeats. The helices located in the amino-terminal two-thirds of the molecule adopt a helix bundle structure while the carboxy-terminal segment forms a separately folded, relatively disorganized, domain. The latter domain initiates lipid binding and this interaction induces changes in conformation; the alpha-helix content increases and the amino-terminal helix bundle can open subsequently. These conformational changes alter the abilities of apoA-I and apoE to function as ligands for their receptors. The apoA-I and apoE molecules possess detergent-like properties and they can solubilize vesicular phospholipid to create discoidal HDL particles with hydrodynamic diameters of ~10 nm. In the case of apoA-I, such a particle is stabilized by two protein molecules arranged in an anti-parallel, double-belt, conformation around the edge of the disc. The abilities of apoA-I and apoE to solubilize phospholipid and stabilize HDL particles enable these proteins to be partners with ABCA1 in mediating efflux of cellular phospholipid and cholesterol, and the biogenesis of HDL particles. ApoA-I-containing nascent HDL particles play a critical role in cholesterol transport in the circulation whereas apoE-containing HDL particles mediate cholesterol transport in the brain. The mechanisms by which HDL particles are remodeled by lipases and lipid transfer proteins, and interact with SR-BI to deliver cholesterol to cells, are reviewed.
Collapse
|
21
|
Molecular characterization of EmABP, an apolipoprotein A-I binding protein secreted by the Echinococcus multilocularis metacestode. Infect Immun 2009; 77:5564-71. [PMID: 19805524 DOI: 10.1128/iai.00653-09] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Cestodes are unable to synthesize de novo most of their own membrane lipids, including cholesterol, and have to take them up from the host during an infection. The underlying molecular mechanisms are so far unknown. Here we report the identification and characterization of a novel gene, Emabp, which is expressed by larval stages and adults of the fox tapeworm Echinococcus multilocularis. The encoded protein, EmABP, displays significant homologies to apolipoprotein A-I binding protein (AI-BP) of mammalian origin and to metazoan YjeF_N domain proteins. Like mammalian AI-BP, EmABP carries an export-directing signal sequence which is absent in predicted AI-BP orthologs from the related flatworms Schistosoma japonicum and Schmidtea mediterranea. Using a specific antibody and immunoprecipitation techniques, we demonstrate that EmABP is secreted into the extraparasitic environment and into the hydatid fluid of in vitro-cultivated metacestode vesicles. Furthermore, we show that apolipoprotein A-I (apoA-I), a major constituent of cholesterol-transporting high-density lipoproteins, is present in hydatid fluid. By pulldown experiments, we demonstrate that recombinantly expressed, purified EmABP interacts with purified human apoA-I and is able to precipitate apoA-I from human serum. On the basis of these features and the suggested function of AI-BP in cholesterol transport in higher eukaryotes, we propose a role for EmABP in cholesterol and lipid uptake mechanisms of larval E. multilocularis.
Collapse
|
22
|
Lund-Katz S, Nguyen D, Dhanasekaran P, Kono M, Nickel M, Saito H, Phillips MC. Surface plasmon resonance analysis of the mechanism of binding of apoA-I to high density lipoprotein particles. J Lipid Res 2009; 51:606-17. [PMID: 19786567 DOI: 10.1194/jlr.m002055] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The partitioning of apolipoprotein A-I (apoA-I) molecules in plasma between HDL-bound and -unbound states is an integral part of HDL metabolism. We used the surface plasmon resonance (SPR) technique to monitor in real time the reversible binding of apoA-I to HDL. Biotinylated human HDL(2) and HDL(3) were immobilized on a streptavidin-coated SPR sensor chip, and apoA-I solutions at different concentrations were flowed across the surface. The wild-type (WT) human and mouse apoA-I/HDL interaction involves a two-step process; apoA-I initially binds to HDL with fast association and dissociation rates, followed by a step exhibiting slower kinetics. The isolated N-terminal helix bundle domains of human and mouse apoA-I also exhibit a two-step binding process, consistent with the second slower step involving opening of the helix bundle domain. The results of fluorescence experiments with pyrene-labeled apoA-I are consistent with the N-terminal helix bundle domain interacting with proteins resident on the HDL particle surface. Dissociation constants (K(d)) measured for WT human apoA-I interactions with HDL(2) and HDL(3) are about 10 microM, indicating that the binding is low affinity. This K(d) value does not apply to all of the apoA-I molecules on the HDL particle but only to a relatively small, labile pool.
Collapse
Affiliation(s)
- Sissel Lund-Katz
- Lipid Research Group, Division of Gastroenterology, Hepatology, and Nutrition, The Children's Hospital of Philadelphia, University of Pennsylvania School of Medicine, Philadelphia, PA 19104-4318, USA
| | | | | | | | | | | | | |
Collapse
|
23
|
Block R, Corsetti J, Goldenberg I, Vorobiof G, McNitt S, Ryan D, Zareba W, Moss AJ. The common apolipoprotein A-1 polymorphism -75A>G is associated with ethnic differences in recurrent coronary events after recovery from an acute myocardial infarction. Heart Int 2009; 4:e8. [PMID: 21152377 PMCID: PMC2997744 DOI: 10.4081/hi.2009.e8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2009] [Accepted: 10/05/2009] [Indexed: 11/22/2022] Open
Abstract
Since data regarding the relationship between a common polymorphism (SNP) of the apoA1 gene with apoA1 levels and risk of coronary artery disease are inconsistent, we hypothesized that its association with recurrent coronary events differs for White and Black individuals with diagnosed coronary heart disease. The apoA1 -75G>A SNP was genotyped in a cohort of 834 Black (n=129) and White (n=705) post-myocardial infarction patients. Recurrent coronary events (coronary-related death, non-fatal myocardial infarction, or unstable angina) were documented during an average follow-up of 28 months. Thirty percent of White and 21% of Black patients carried the SNP. Cox proportional-hazards regression analysis, adjusting for clinical and laboratory covariates, demonstrated that the SNP was not associated with recurrent events in the total cohort (HR=1.37, 95% CI 0.95-1.97; p= 0.09) but was the only variable associated with an increased risk of recurrent cardiac events in Blacks (HR=2.40, 95% CI 1.07-5.40; p= 0.034). Conversely in Whites, the SNP was not associated with recurrent events (HR=1.12, 95% CI 0.75-1.67; p= 0.59) whereas apoB (HR=1.78, 95% CI 1.20 -2.65; p= 0.0042) and calcium channel blocker use (HR=2.53, 95% CI 1.72-3.72; p<0.001) were associated; p= 0.0024 for interaction between ethnicity and the SNP. A common apoA1 SNP is associated with a significantly increased risk of recurrent cardiac events among Black, but not White, postmyocardial infarction patients. Relationships with lipoproteins may help explain this finding.
Collapse
Affiliation(s)
- Robert Block
- Department of Community and Preventive Medicine, University of Rochester, Rochester, NY
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Han M, Gillard BK, Courtney HS, Ward K, Rosales C, Khant H, Ludtke SJ, Pownall HJ. Disruption of human plasma high-density lipoproteins by streptococcal serum opacity factor requires labile apolipoprotein A-I. Biochemistry 2009; 48:1481-7. [PMID: 19191587 DOI: 10.1021/bi802287q] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Human plasma high-density lipoproteins (HDL), the primary vehicle for reverse cholesterol transport, are the target of serum opacity factor (SOF), a virulence determinant of Streptococcus pyogenes that turns serum opaque. HDL comprise a core of neutral lipidscholesteryl esters and some triglyceridesurrounded by a surface monolayer of cholesterol, phospholipids, and specialized proteins [apolipoproteins (apos) A-I and A-II]. A HDL is an unstable particle residing in a kinetic trap from which it can escape via chaotropic, detergent, or thermal perturbation. Recombinant (r) SOF catalyzes the transfer of nearly all neutral lipids of approximately 100,000 HDL particles (D approximately 8.5 nm) into a single, large cholesteryl ester-rich microemulsion (CERM; D > 100 nm), leaving a new HDL-like particle [neo HDL (D approximately 5.8 nm)] while releasing lipid-free (LF) apo A-I. CERM formation and apo A-I release have similar kinetics, suggesting parallel or rapid consecutive steps. By using complementary physicochemical methods, we have refined the mechanistic model for HDL opacification. According to size exclusion chromatography, a HDL containing nonlabile apo A-I resists rSOF-mediated opacification. On the basis of kinetic cryo-electron microscopy, rSOF (10 nM) catalyzes the conversion of HDL (4 microM) to neo HDL via a stepwise mechanism in which intermediate-sized particles are seen. Kinetic turbidimetry revealed opacification as a rising exponential reaction with a rate constant k of (4.400 +/- 0.004) x 10(-2) min(-1). Analysis of the kinetic data using transition state theory gave an enthalpy (DeltaH()), entropy (DeltaS(++)), and free energy (DeltaG()) of activation of 73.9 kJ/mol, -66.87 J/K, and 94.6 kJ/mol, respectively. The free energy of activation for opacification is nearly identical to that for the displacement of apo A-I from HDL by guanidine hydrochloride. We conclude that apo A-I lability is required for HDL opacification, LF apo A-I desorption is the rate-limiting step, and nearly all HDL particles contain at least one labile copy of apo A-I.
Collapse
Affiliation(s)
- Mikyung Han
- Section of Atherosclerosis and Vascular Medicine, Department of Medicine, Baylor College of Medicine, Houston, Texas 77030, USA
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Differential stability of high-density lipoprotein subclasses: effects of particle size and protein composition. J Mol Biol 2009; 387:628-38. [PMID: 19236880 DOI: 10.1016/j.jmb.2009.02.036] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2009] [Revised: 02/09/2009] [Accepted: 02/13/2009] [Indexed: 01/17/2023]
Abstract
High-density lipoproteins (HDLs) are complexes of proteins (mainly apoA-I and apoA-II) and lipids that remove cholesterol and prevent atherosclerosis. Understanding the distinct properties of the heterogeneous HDL population may aid the development of new diagnostic tools and therapies for atherosclerosis. Mature human HDLs form two major subclasses differing in particle diameter and metabolic properties, HDL(2) (large) and HDL(3) (small). These subclasses are comprised of HDL(A-I) containing only apoA-I, and HDL(A-I/A-II) containing apoA-I and apoA-II. ApoA-I is strongly cardioprotective, but the function of the smaller, more hydrophobic apoA-II is unclear. ApoA-II is thought to counteract the cardioprotective action of apoA-I by stabilizing HDL particles and inhibiting their remodeling. To test this notion, we performed the first kinetic stability study of human HDL subclasses. The results revealed that the stability of plasma spherical HDL decreases with increasing particle diameter; which may facilitate preferential cholesterol ester uptake from large lipid-loaded HDL(2). Surprisingly, size-matched plasma HDL(A-I/A-II) showed comparable or slightly lower stability than HDL(A-I); this is consistent with the destabilization of model discoidal HDL observed upon increasing the A-II to A-I ratio. These results clarify the roles of the particle size and protein composition in HDL remodeling, and help reconcile conflicting reports regarding the role of apoA-II in this remodeling.
Collapse
|
26
|
Graves TL, Scott JE. A high throughput serum paraoxonase assay for discovery of small molecule modulators of PON1 activity. CURRENT CHEMICAL GENOMICS 2008; 2:51-61. [PMID: 20161844 PMCID: PMC2803440 DOI: 10.2174/1875397300802010051] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/01/2008] [Revised: 10/17/2008] [Accepted: 10/20/2008] [Indexed: 01/08/2023]
Abstract
PON1 has been demonstrated to be the serum enzyme responsible for detoxifying organophosphate chemical weapons and plays a protective role against atherosclerosis. In order to identify small molecules that modulate PON1 activity in serum, we developed a high throughput kinetic absorbance assay using mouse serum and the organophosphate paraoxon. The IC(50) value obtained for the known PON1 inhibitor, 2-hydroxyquinoline, matched the value reported for purified PON1. A compound library was screened resulting in no confirmed activators, but 12 confirmed inhibitors. Seven of these hits also inhibited purified human PON1. One compound was only two-fold less potent than 2-hydroxyquinoline in the serum assay, but 10-fold more potent against purified PON1. This compound (IC(50) = 420 nM) may be useful towards a chemical probe for PON1. Therefore, this assay has utility as a high throughput assay for discovery of small molecule modulators of PON1 activity that maintain activity in serum.
Collapse
Affiliation(s)
| | - John E Scott
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute and Technology Enterprise, North Carolina Central University, 1801 Fayetteville Street, Durham, NC 27707, USA
| |
Collapse
|
27
|
Kono M, Okumura Y, Tanaka M, Nguyen D, Dhanasekaran P, Lund-Katz S, Phillips MC, Saito H. Conformational flexibility of the N-terminal domain of apolipoprotein a-I bound to spherical lipid particles. Biochemistry 2008; 47:11340-7. [PMID: 18831538 DOI: 10.1021/bi801503r] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Lipid binding of human apolipoprotein A-I (apoA-I) occurs initially through the C-terminal alpha-helices followed by conformational reorganization of the N-terminal helix bundle. This led us to hypothesize that apoA-I has multiple lipid-bound conformations, in which the N-terminal helix bundle adopts either open or closed conformations anchored by the C-terminal domain. To investigate such possible conformations of apoA-I at the surface of a spherical lipid particle, site-specific labeling of the N- and C-terminal helices in apoA-I by N-(1-pyrene)maleimide was employed after substitution of a Cys residue for Val-53 or Phe-229. Neither mutagenesis nor the pyrene labeling caused discernible changes in the lipid-free structure and lipid interaction of apoA-I. Taking advantage of a significant increase in fluorescence when a pyrene-labeled helix is in contact with the lipid surface, we monitored the behaviors of the N- and C-terminal helices upon binding of apoA-I to egg PC small unilamellar vesicles. Comparison of the binding isotherms for pyrene-labeled apoA-I as well as a C-terminal helical peptide suggests that an increase in surface concentration of apoA-I causes dissociation of the N-terminal helix from the surface leaving the C-terminal helix attached. Consistent with this, isothermal titration calorimetry measurements showed that the enthalpy of apoA-I binding to the lipid surface under near saturated conditions is much less exothermic than that for binding at a low surface concentration, indicating the N-terminal helix bundle is out of contact with lipid at high apoA-I surface concentrations. Interestingly, the presence of cholesterol significantly induces the open conformation of the helix bundle. These results provide insight into the multiple lipid-bound conformations that the N-terminal helix bundle of apoA-I can adopt on a lipid or lipoprotein particle, depending upon the availability of space on the surface and the surface composition.
Collapse
Affiliation(s)
- Momoe Kono
- Department of Biophysical Chemistry, Kobe Pharmaceutical University, Kobe 658-8558, Japan
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Sethi AA, Stonik JA, Thomas F, Demosky SJ, Amar M, Neufeld E, Brewer HB, Davidson WS, D'Souza W, Sviridov D, Remaley AT. Asymmetry in the lipid affinity of bihelical amphipathic peptides. A structural determinant for the specificity of ABCA1-dependent cholesterol efflux by peptides. J Biol Chem 2008; 283:32273-82. [PMID: 18805791 DOI: 10.1074/jbc.m804461200] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
ApoA-I contains a tandem array of amphipathic helices with varying lipid affinity, which are critical in its ability to bind and remove lipids from cells by the ABCA1 transporter. In this study, the effect of asymmetry in the lipid affinity of amphipathic helices in a bihelical apoA-I mimetic peptide, 37pA, on lipid efflux by the ABCA1 transporter was examined. Seven peptide variants of 37pA were produced by substituting a varying number of hydrophobic amino acids for alanine on either one or both helices. The 5A peptide with five alanine substitutions in the second helix had decreased helical content compared with 37pA (5A, 12+/-1% helicity; 37pA, 28+/-2% helicity) and showed less self-association but, similar to the parent peptide, was able to readily solubilize phospholipid vesicles. Furthermore, 5A, unlike the parent peptide 37pA, was not hemolytic (37pA, 27+/-2% RBC lysis, 2 h, 18 microm). Finally, the 5A peptide stimulated cholesterol and phospholipid efflux by the ABCA1 transporter with higher specificity (ABCA1-transfected versus untransfected cells) than 37pA (5A, 9.7+/-0.77%, 18 h, 18 microm versus 1.5+/-0.27%, 18 h, 18 microm (p<0.0001); 37pA, 7.4+/-0.85%, 18 h, 18 microm versus 5.8+/-0.20%, 18 h, 18 microm (p=0.03)). In summary, we describe a novel bihelical peptide with asymmetry in the lipid affinity of its helices and properties similar to apoA-I in terms of specificity for cholesterol efflux by the ABCA1 transporter and low cytotoxicity.
Collapse
Affiliation(s)
- Amar A Sethi
- Lipoprotein Metabolism Section, Pulmonary and Vascular Medicine Branch, NHLBI, National Institutes of Health, Bethesda, Maryland 20892-1508, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Schicher M, Polsinger M, Hermetter A, Prassl R, Zimmer A. In vitro release of propofol and binding capacity with regard to plasma constituents. Eur J Pharm Biopharm 2008; 70:882-8. [PMID: 18640268 DOI: 10.1016/j.ejpb.2008.06.018] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2007] [Revised: 06/18/2008] [Accepted: 06/23/2008] [Indexed: 11/28/2022]
Abstract
PURPOSE New evidence suggests that the anesthetic effect of parenteral propofol emulsions varies between commercial preparations. We examined and compared different propofol preparations to determine propofol release and binding capacity with regard to plasma lipoproteins and albumin. METHODS We created a novel assay consisting of microtiter plates coated with either low-density lipoprotein (LDL) or albumin to analyze propofol binding kinetics. Using high performance liquid chromatography (HPLC), we measured propofol release from the oily phase and the corresponding amount of propofol bound to the plates in a time-dependent manner and at equilibrium conditions attained after 30 min of incubation at 37 degrees C. The concentrations of free propofol in the aqueous phase of different propofol preparations - Diprivan, and the generic formulations Propofol "Fresenius" (1% and 2% propofol) and Propofol-Lipuro - were analyzed using ultracentrifugation or dialysis for phase separation. Finally, we investigated the effect of isolated lipoprotein fractions on propofol release. RESULTS Propofol bound to LDL-coated plates with approximately twofold higher affinity than to albumin-coated plates. No significant differences in total propofol release were observed between preparations. Moreover, similar amounts of free propofol were observed in the aqueous phase of all products tested (1% propofol preparations: 18 microg/ml; 2% propofol preparations: 35 microg/ml), except for the medium-chain and long-chain triglyceride (MCT/LCT) preparation studied, in which the concentration of free propofol was lower. Lipoproteins had no effect on propofol release, except for high-density lipoprotein (HDL), which triggered almost 100% release from the oily phase at HDL concentrations above 1000 microg/ml. CONCLUSIONS No differences were observed between the binding/release capacity and lipoprotein interactions of any of the propofol preparations tested. We propose that clinical observations of inconsistent propofol activity are related to variations in the lipoprotein profile, enzyme activity or genetic disorders of individual patients, rather than to the propofol preparation itself.
Collapse
|
30
|
Santo AS, Cunningham AM, Alhassan S, Browne RW, Burton H, Leddy JJ, Grandjean PW, Horvath SM, Horvath PJ. NMR analysis of lipoprotein particle size does not increase sensitivity to the effect of soy protein on CVD risk when compared with the traditional lipid profile. Appl Physiol Nutr Metab 2008; 33:489-500. [DOI: 10.1139/h08-023] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The traditional lipid profile compared with nuclear magnetic resonance (NMR) may underestimate the risk for cardiovascular disease and may explain some of the discrepancies in results between studies analyzing the salubrious effects of soy. Our purpose was to compare the traditional lipid profile with NMR quantification of the number of lipoprotein particles, subclasses, and diameters or sizes in 30 sedentary males, between 18 and 30 years of age, consuming 1 of the following 3 supplements daily for 28 days: milk protein (Milk), isoflavone-poor soy protein (Soy–), or isoflavone-rich soy protein (Soy+). The study used a double-blind, parallel-arm design with random assignment to 1 of the 3 protein supplement groups. Fasting EDTA blood samples were collected at baseline and after 28 days of supplementation and analyzed for the number and size of very low-density lipoprotein (VLDL), low-density lipoprotein (LDL), and high-density lipoprotein (HDL) particles, respectively. Fasting serum samples were analyzed for concentrations of total cholesterol (TC), LDL cholesterol (LDL-C), total HDL cholesterol (HDL-C), HDL2-C, HDL3-C, triglycerides (TGs), free fatty acids (FFAs), and glucose. Fasting heparin blood samples were collected at baseline and after supplementation and analyzed for apolipoproteins A-I, A-II, B, C-II, C-III, and E, as well as hepatic and lipoprotein lipase concentrations. HDL3-C increased by 47.2% after Soy+ supplementation and hepatic lipase decreased 19.2% after Soy– supplementation (p < 0.05). HDL-C and apolipoproteins A-I and A-II were found to increase in all 3 groups (p < 0.05). Results support that NMR analysis of lipoprotein particle number and size are not more sensitive to the effect of soy protein on CVD risk compared with the traditional lipid profile. Furthermore, the lack of isoflavones in soy protein seems to have a deleterious effect on hepatic lipase.
Collapse
Affiliation(s)
- Antonio S. Santo
- Division of Healthful Living and Sports Studies, School of Health Sciences, Lenoir-Rhyne College, Moretz 104 – Exercise Physiology Laboratory, Hickory, NC 28603, USA
- The Center for Diabetes Self-Management Care, Frye Regional Medical Center, 420 North Center St., Hickory, NC 28601, USA
- Stanford Prevention Research Center, Stanford University School of Medicine, Hoover Pavilion, N229, 211 Quarry Rd., Stanford, CA 94305, USA
- Biotechnical and Clinical Laboratory Sciences, University at Buffalo, State University of New York, 26 Cary Hall, Buffalo, NY 14214, USA
- Department of Exercise and Nutrition Sciences, University at Buffalo, State University of New York, Kimball Tower 108, Buffalo, NY 14214, USA
| | - Ariana M. Cunningham
- Division of Healthful Living and Sports Studies, School of Health Sciences, Lenoir-Rhyne College, Moretz 104 – Exercise Physiology Laboratory, Hickory, NC 28603, USA
- The Center for Diabetes Self-Management Care, Frye Regional Medical Center, 420 North Center St., Hickory, NC 28601, USA
- Stanford Prevention Research Center, Stanford University School of Medicine, Hoover Pavilion, N229, 211 Quarry Rd., Stanford, CA 94305, USA
- Biotechnical and Clinical Laboratory Sciences, University at Buffalo, State University of New York, 26 Cary Hall, Buffalo, NY 14214, USA
- Department of Exercise and Nutrition Sciences, University at Buffalo, State University of New York, Kimball Tower 108, Buffalo, NY 14214, USA
| | - Sofiya Alhassan
- Division of Healthful Living and Sports Studies, School of Health Sciences, Lenoir-Rhyne College, Moretz 104 – Exercise Physiology Laboratory, Hickory, NC 28603, USA
- The Center for Diabetes Self-Management Care, Frye Regional Medical Center, 420 North Center St., Hickory, NC 28601, USA
- Stanford Prevention Research Center, Stanford University School of Medicine, Hoover Pavilion, N229, 211 Quarry Rd., Stanford, CA 94305, USA
- Biotechnical and Clinical Laboratory Sciences, University at Buffalo, State University of New York, 26 Cary Hall, Buffalo, NY 14214, USA
- Department of Exercise and Nutrition Sciences, University at Buffalo, State University of New York, Kimball Tower 108, Buffalo, NY 14214, USA
| | - Richard W. Browne
- Division of Healthful Living and Sports Studies, School of Health Sciences, Lenoir-Rhyne College, Moretz 104 – Exercise Physiology Laboratory, Hickory, NC 28603, USA
- The Center for Diabetes Self-Management Care, Frye Regional Medical Center, 420 North Center St., Hickory, NC 28601, USA
- Stanford Prevention Research Center, Stanford University School of Medicine, Hoover Pavilion, N229, 211 Quarry Rd., Stanford, CA 94305, USA
- Biotechnical and Clinical Laboratory Sciences, University at Buffalo, State University of New York, 26 Cary Hall, Buffalo, NY 14214, USA
- Department of Exercise and Nutrition Sciences, University at Buffalo, State University of New York, Kimball Tower 108, Buffalo, NY 14214, USA
| | - Harold Burton
- Division of Healthful Living and Sports Studies, School of Health Sciences, Lenoir-Rhyne College, Moretz 104 – Exercise Physiology Laboratory, Hickory, NC 28603, USA
- The Center for Diabetes Self-Management Care, Frye Regional Medical Center, 420 North Center St., Hickory, NC 28601, USA
- Stanford Prevention Research Center, Stanford University School of Medicine, Hoover Pavilion, N229, 211 Quarry Rd., Stanford, CA 94305, USA
- Biotechnical and Clinical Laboratory Sciences, University at Buffalo, State University of New York, 26 Cary Hall, Buffalo, NY 14214, USA
- Department of Exercise and Nutrition Sciences, University at Buffalo, State University of New York, Kimball Tower 108, Buffalo, NY 14214, USA
| | - John J. Leddy
- Division of Healthful Living and Sports Studies, School of Health Sciences, Lenoir-Rhyne College, Moretz 104 – Exercise Physiology Laboratory, Hickory, NC 28603, USA
- The Center for Diabetes Self-Management Care, Frye Regional Medical Center, 420 North Center St., Hickory, NC 28601, USA
- Stanford Prevention Research Center, Stanford University School of Medicine, Hoover Pavilion, N229, 211 Quarry Rd., Stanford, CA 94305, USA
- Biotechnical and Clinical Laboratory Sciences, University at Buffalo, State University of New York, 26 Cary Hall, Buffalo, NY 14214, USA
- Department of Exercise and Nutrition Sciences, University at Buffalo, State University of New York, Kimball Tower 108, Buffalo, NY 14214, USA
| | - Peter W. Grandjean
- Division of Healthful Living and Sports Studies, School of Health Sciences, Lenoir-Rhyne College, Moretz 104 – Exercise Physiology Laboratory, Hickory, NC 28603, USA
- The Center for Diabetes Self-Management Care, Frye Regional Medical Center, 420 North Center St., Hickory, NC 28601, USA
- Stanford Prevention Research Center, Stanford University School of Medicine, Hoover Pavilion, N229, 211 Quarry Rd., Stanford, CA 94305, USA
- Biotechnical and Clinical Laboratory Sciences, University at Buffalo, State University of New York, 26 Cary Hall, Buffalo, NY 14214, USA
- Department of Exercise and Nutrition Sciences, University at Buffalo, State University of New York, Kimball Tower 108, Buffalo, NY 14214, USA
| | - Steven M. Horvath
- Division of Healthful Living and Sports Studies, School of Health Sciences, Lenoir-Rhyne College, Moretz 104 – Exercise Physiology Laboratory, Hickory, NC 28603, USA
- The Center for Diabetes Self-Management Care, Frye Regional Medical Center, 420 North Center St., Hickory, NC 28601, USA
- Stanford Prevention Research Center, Stanford University School of Medicine, Hoover Pavilion, N229, 211 Quarry Rd., Stanford, CA 94305, USA
- Biotechnical and Clinical Laboratory Sciences, University at Buffalo, State University of New York, 26 Cary Hall, Buffalo, NY 14214, USA
- Department of Exercise and Nutrition Sciences, University at Buffalo, State University of New York, Kimball Tower 108, Buffalo, NY 14214, USA
| | - Peter J. Horvath
- Division of Healthful Living and Sports Studies, School of Health Sciences, Lenoir-Rhyne College, Moretz 104 – Exercise Physiology Laboratory, Hickory, NC 28603, USA
- The Center for Diabetes Self-Management Care, Frye Regional Medical Center, 420 North Center St., Hickory, NC 28601, USA
- Stanford Prevention Research Center, Stanford University School of Medicine, Hoover Pavilion, N229, 211 Quarry Rd., Stanford, CA 94305, USA
- Biotechnical and Clinical Laboratory Sciences, University at Buffalo, State University of New York, 26 Cary Hall, Buffalo, NY 14214, USA
- Department of Exercise and Nutrition Sciences, University at Buffalo, State University of New York, Kimball Tower 108, Buffalo, NY 14214, USA
| |
Collapse
|
31
|
Bhat S, Sorci-Thomas MG, Tuladhar R, Samuel MP, Thomas MJ. Conformational adaptation of apolipoprotein A-I to discretely sized phospholipid complexes. Biochemistry 2007; 46:7811-21. [PMID: 17563120 PMCID: PMC2553278 DOI: 10.1021/bi700384t] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The conformational constraints for apoA-I bound to recombinant phospholipid complexes (rHDL) were attained from a combination of chemical cross-linking and mass spectrometry. Molecular distances were then used to refine models of lipid-bound apoA-I on both 80 and 96 A diameter rHDL particles. To obtain molecular constraints on the protein bound to phospholipid complexes, three different lysine-selective homo-bifunctional cross-linkers with increasing spacer arm lengths (i.e., 7.7, 12.0, and 16.1 A) were reacted with purified, homogeneous recombinant 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine (POPC) apoA-I rHDL complexes of each diameter. Cross-linked dimeric apoA-I products were separated from monomeric apoprotein using 12% SDS-PAGE, then subjected to in-gel trypsin digest, and identified by MS/MS sequencing. These studies aid in the refinement of our previously published molecular model of two apoA-I molecules bound to approximately 150 molecules of POPC and suggest that the protein hydrophobic interactions at the N- and C-terminal domains decrease as the number of phospholipid molecules or "lipidation state" of apoA-I increases. Thus, it appears that these incremental changes in the interaction between the N- and C-terminal ends of apoA-I stabilize its tertiary conformation in the lipid-free state as well as allowing it to unfold and sequester discrete amounts of phospholipid molecules.
Collapse
Affiliation(s)
- Shaila Bhat
- Department of Pathology, Center for Lipid Science, Wake Forest University Medical Center, 391 Technology Way, Building A1, Winston-Salem, NC 27101
| | - Mary G Sorci-Thomas
- Department of Biochemistry, Wake Forest University Medical Center, 391 Technology Way, Building A1, Winston-Salem, NC 27101
- Department of Pathology, Center for Lipid Science, Wake Forest University Medical Center, 391 Technology Way, Building A1, Winston-Salem, NC 27101
| | - Rubina Tuladhar
- Department of Biochemistry, Wake Forest University Medical Center, 391 Technology Way, Building A1, Winston-Salem, NC 27101
| | - Michael P. Samuel
- Department of Biochemistry, Wake Forest University Medical Center, 391 Technology Way, Building A1, Winston-Salem, NC 27101
| | - Michael J. Thomas
- Department of Biochemistry, Wake Forest University Medical Center, 391 Technology Way, Building A1, Winston-Salem, NC 27101
| |
Collapse
|
32
|
Robinson JG, Davidson MH. Investigational drugs targeting HDL-C metabolism and reverse cholesterol transport. ACTA ACUST UNITED AC 2007. [DOI: 10.2217/17460875.2.3.285] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|