1
|
Georgiev D, Fernández-Galiana Á, Vilms Pedersen S, Papadopoulos G, Xie R, Stevens MM, Barahona M. Hyperspectral unmixing for Raman spectroscopy via physics-constrained autoencoders. Proc Natl Acad Sci U S A 2024; 121:e2407439121. [PMID: 39471214 DOI: 10.1073/pnas.2407439121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 09/10/2024] [Indexed: 11/01/2024] Open
Abstract
Raman spectroscopy is widely used across scientific domains to characterize the chemical composition of samples in a nondestructive, label-free manner. Many applications entail the unmixing of signals from mixtures of molecular species to identify the individual components present and their proportions, yet conventional methods for chemometrics often struggle with complex mixture scenarios encountered in practice. Here, we develop hyperspectral unmixing algorithms based on autoencoder neural networks, and we systematically validate them using both synthetic and experimental benchmark datasets created in-house. Our results demonstrate that unmixing autoencoders provide improved accuracy, robustness, and efficiency compared to standard unmixing methods. We also showcase the applicability of autoencoders to complex biological settings by showing improved biochemical characterization of volumetric Raman imaging data from a monocytic cell.
Collapse
Affiliation(s)
- Dimitar Georgiev
- Department of Computing, Faculty of Engineering, Imperial College London, London SW7 2AZ, United Kingdom
- UKRI Centre for Doctoral Training in AI for Healthcare, Imperial College London, London SW7 2AZ, United Kingdom
- Department of Materials, Faculty of Engineering, Imperial College London, London SW7 2AZ, United Kingdom
- Department of Bioengineering, Faculty of Engineering, Imperial College London, London SW7 2AZ, United Kingdom
- Institute of Biomedical Engineering, Faculty of Engineering, Imperial College London, London SW7 2AZ, United Kingdom
| | - Álvaro Fernández-Galiana
- Department of Materials, Faculty of Engineering, Imperial College London, London SW7 2AZ, United Kingdom
- Department of Bioengineering, Faculty of Engineering, Imperial College London, London SW7 2AZ, United Kingdom
- Institute of Biomedical Engineering, Faculty of Engineering, Imperial College London, London SW7 2AZ, United Kingdom
| | - Simon Vilms Pedersen
- Department of Materials, Faculty of Engineering, Imperial College London, London SW7 2AZ, United Kingdom
- Department of Bioengineering, Faculty of Engineering, Imperial College London, London SW7 2AZ, United Kingdom
- Institute of Biomedical Engineering, Faculty of Engineering, Imperial College London, London SW7 2AZ, United Kingdom
| | - Georgios Papadopoulos
- Department of Computing, Faculty of Engineering, Imperial College London, London SW7 2AZ, United Kingdom
- UKRI Centre for Doctoral Training in AI for Healthcare, Imperial College London, London SW7 2AZ, United Kingdom
- Department of Materials, Faculty of Engineering, Imperial College London, London SW7 2AZ, United Kingdom
- Department of Bioengineering, Faculty of Engineering, Imperial College London, London SW7 2AZ, United Kingdom
- Institute of Biomedical Engineering, Faculty of Engineering, Imperial College London, London SW7 2AZ, United Kingdom
| | - Ruoxiao Xie
- Department of Materials, Faculty of Engineering, Imperial College London, London SW7 2AZ, United Kingdom
- Department of Bioengineering, Faculty of Engineering, Imperial College London, London SW7 2AZ, United Kingdom
- Institute of Biomedical Engineering, Faculty of Engineering, Imperial College London, London SW7 2AZ, United Kingdom
| | - Molly M Stevens
- Department of Materials, Faculty of Engineering, Imperial College London, London SW7 2AZ, United Kingdom
- Department of Bioengineering, Faculty of Engineering, Imperial College London, London SW7 2AZ, United Kingdom
- Institute of Biomedical Engineering, Faculty of Engineering, Imperial College London, London SW7 2AZ, United Kingdom
- Medical Sciences Division, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3QU, United Kingdom
- Mathematical, Physical & Life Sciences Division, Department of Engineering Science, University of Oxford, Oxford OX1 3QU, United Kingdom
- Medical Sciences Division and Mathematical, Physical & Life Sciences Division, Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford OX1 3QU, United Kingdom
| | - Mauricio Barahona
- Department of Mathematics, Faculty of Natural Sciences, Imperial College London, London SW7 2AZ, United Kingdom
| |
Collapse
|
2
|
Chambers KL, Myerscough MR, Watson MG, Byrne HM. Blood Lipoproteins Shape the Phenotype and Lipid Content of Early Atherosclerotic Lesion Macrophages: A Dual-Structured Mathematical Model. Bull Math Biol 2024; 86:112. [PMID: 39093509 PMCID: PMC11297092 DOI: 10.1007/s11538-024-01342-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 07/16/2024] [Indexed: 08/04/2024]
Abstract
Macrophages in atherosclerotic lesions exhibit a spectrum of behaviours or phenotypes. The phenotypic distribution of monocyte-derived macrophages (MDMs), its correlation with MDM lipid content, and relation to blood lipoprotein densities are not well understood. Of particular interest is the balance between low density lipoproteins (LDL) and high density lipoproteins (HDL), which carry bad and good cholesterol respectively. To address these issues, we have developed a mathematical model for early atherosclerosis in which the MDM population is structured by phenotype and lipid content. The model admits a simpler, closed subsystem whose analysis shows how lesion composition becomes more pathological as the blood density of LDL increases relative to the HDL capacity. We use asymptotic analysis to derive a power-law relationship between MDM phenotype and lipid content at steady-state. This relationship enables us to understand why, for example, lipid-laden MDMs have a more inflammatory phenotype than lipid-poor MDMs when blood LDL lipid density greatly exceeds HDL capacity. We show further that the MDM phenotype distribution always attains a local maximum, while the lipid content distribution may be unimodal, adopt a quasi-uniform profile or decrease monotonically. Pathological lesions exhibit a local maximum in both the phenotype and lipid content MDM distributions, with the maximum at an inflammatory phenotype and near the lipid content capacity respectively. These results illustrate how macrophage heterogeneity arises in early atherosclerosis and provide a framework for future model validation through comparison with single-cell RNA sequencing data.
Collapse
Affiliation(s)
- Keith L Chambers
- Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford, Andrew Wiles Building, Radcliffe Observatory Quarter, Woodstock Road, Oxford, Oxfordshire, OX2 6GG, UK.
| | - Mary R Myerscough
- School of Mathematics and Statistics, University of Sydney, Carslaw Building, Eastern Avenue, Camperdown, Sydney, NSW, 2006, Australia
| | - Michael G Watson
- School of Mathematics and Statistics, University of New South Wales, Anita B. Lawrence Centre, University Mall, UNSW, Kensington, Sydney, NSW, 2052, Australia
| | - Helen M Byrne
- Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford, Andrew Wiles Building, Radcliffe Observatory Quarter, Woodstock Road, Oxford, Oxfordshire, OX2 6GG, UK
- Ludwig Institute for Cancer Research, University of Oxford, Old Road Campus Research Build, Roosevelt Dr, Headington, Oxford, Oxfordshire, OX3 7DQ, UK
| |
Collapse
|
3
|
Zhang Y, Xiao W, He S, Xia X, Yang W, Yang Z, Hu H, Wang Y, Wang X, Li H, Huang Y, Gao H. Lipid-mediated protein corona regulation with increased apolipoprotein A-I recruitment for glioma targeting. J Control Release 2024; 368:42-51. [PMID: 38365180 DOI: 10.1016/j.jconrel.2024.02.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 02/13/2024] [Indexed: 02/18/2024]
Abstract
Protein corona has long been a source of concern, as it might impair the targeting efficacy of targeted drug delivery systems. However, engineered up-regulating the adsorption of certain functional serum proteins could provide nanoparticles with specific targeting drug delivery capacity. Herein, apolipoprotein A-I absorption increased nanoparticles (SPC-PLGA NPs), composed with the Food and Drug Administration approved intravenously injectable soybean phosphatidylcholine (SPC) and poly (DL-lactide-co-glycolide) (PLGA), were fabricated for enhanced glioma targeting. Due to the high affinity of SPC and apolipoprotein A-I, the percentage of apolipoprotein A-I in the protein corona of SPC-PLGA NPs was 2.19-fold higher than that of nanoparticles without SPC, which made SPC-PLGA NPs have superior glioma targeting ability through binding to scavenger receptor class BI on blood-brain barrier and glioma cells both in vitro and in vivo. SPC-PLGA NPs loaded with paclitaxel could effectively reduce glioma invasion and prolong the survival time of glioma-bearing mice. In conclusion, we provided a good example of the direction of achieving targeting drug delivery based on protein corona regulation.
Collapse
Affiliation(s)
- Yiwei Zhang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Wei Xiao
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Siqin He
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Xue Xia
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Wenqin Yang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Zhihang Yang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Haili Hu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Yushan Wang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Xiaorong Wang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Hanmei Li
- School of Food and Biological Engineering, Chengdu University, Chengdu 610106, China
| | - Yuan Huang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Huile Gao
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
4
|
Sharma P, Venkatachalam K, Binesh A. Decades Long Involvement of THP-1 Cells as a Model for Macrophage Research: A Comprehensive Review. Antiinflamm Antiallergy Agents Med Chem 2024; 23:85-104. [PMID: 38676532 DOI: 10.2174/0118715230294413240415054610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 03/22/2024] [Accepted: 03/26/2024] [Indexed: 04/29/2024]
Abstract
Over the years, researchers have endeavored to identify dependable and reproducible in vitro models for examining macrophage behavior under controlled conditions. The THP-1 cell line has become a significant and widely employed tool in macrophage research within these models. Originating from the peripheral blood of individuals with acute monocytic leukemia, this human monocytic cell line can undergo transformation into macrophage-like cells, closely mirroring primary human macrophages when exposed to stimulants. Macrophages play a vital role in the innate immune system, actively regulating inflammation, responding to infections, and maintaining tissue homeostasis. A comprehensive understanding of macrophage biology and function is crucial for gaining insights into immunological responses, tissue healing, and the pathogenesis of diseases such as viral infections, autoimmune disorders, and neoplastic conditions. This review aims to thoroughly evaluate and emphasize the extensive history of THP-1 cells as a model for macrophage research. Additionally, it will delve into the significance of THP-1 cells in advancing our comprehension of macrophage biology and their invaluable contributions to diverse scientific domains.
Collapse
Affiliation(s)
- Prakhar Sharma
- Institute of Fisheries Post Graduate Studies, Tamil Nadu Dr. J. Jayalalithaa Fisheries University (TNJFU), OMR Campus, Vaniyanchavadi, Chennai, 603103, Tamil Nadu, India
| | - Kaliyamurthi Venkatachalam
- Institute of Fisheries Post Graduate Studies, Tamil Nadu Dr. J. Jayalalithaa Fisheries University (TNJFU), OMR Campus, Vaniyanchavadi, Chennai, 603103, Tamil Nadu, India
| | - Ambika Binesh
- Institute of Fisheries Post Graduate Studies, Tamil Nadu Dr. J. Jayalalithaa Fisheries University (TNJFU), OMR Campus, Vaniyanchavadi, Chennai, 603103, Tamil Nadu, India
| |
Collapse
|
5
|
Lethcoe K, Fox CA, Hafiane A, Kiss RS, Ryan RO. Isolation of recombinant apolipoprotein E4 N-terminal domain by foam fractionation. Protein Expr Purif 2023; 210:106319. [PMID: 37290717 PMCID: PMC10330888 DOI: 10.1016/j.pep.2023.106319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 06/01/2023] [Accepted: 06/05/2023] [Indexed: 06/10/2023]
Abstract
Apolipoprotein (apo) E functions in lipoprotein metabolism as a low density lipoprotein receptor ligand. ApoE is comprised of two structural domains, a 22 kDa N-terminal (NT) domain that adopts a helix bundle conformation and a 10 kDa C-terminal domain with strong lipid binding affinity. The NT domain is capable of transforming aqueous phospholipid dispersions into discoidal reconstituted high density lipoprotein (rHDL) particles. Given the utility of apoE-NT as a structural component of rHDL, expression studies were conducted. A plasmid construct encoding a pelB leader sequence fused to the N-terminus of human apoE4 (residues 1-183) was transformed into Escherichia coli. Upon expression, the fusion protein is directed to the periplasmic space where leader peptidase cleaves the pelB sequence, generating mature apoE4-NT. In shaker flask expression cultures, apoE4-NT escapes the bacteria and accumulates in the medium. In a bioreactor setting, however, apoE4-NT was found to combine with gas and liquid components in the culture medium to generate large quantities of foam. When this foam was collected in an external vessel and collapsed into a liquid foamate, analysis revealed that apoE4-NT was the sole major protein present. The product protein was further isolated by heparin affinity chromatography (60-80 mg/liter bacterial culture), shown to be active in rHDL formulation, and documented to serve as an acceptor of effluxed cellular cholesterol. Thus, foam fractionation provides a streamlined process to produce recombinant apoE4-NT for biotechnology applications.
Collapse
Affiliation(s)
- Kyle Lethcoe
- Department of Biochemistry and Molecular Biology, University of Nevada, Reno, NV, 89557, USA
| | - Colin A Fox
- Department of Biochemistry and Molecular Biology, University of Nevada, Reno, NV, 89557, USA
| | - Anouar Hafiane
- Department of Medicine, Division of Cardiology, McGill University, Montreal, QC, Canada
| | - Robert S Kiss
- Department of Medicine, Division of Cardiology, McGill University, Montreal, QC, Canada
| | - Robert O Ryan
- Department of Biochemistry and Molecular Biology, University of Nevada, Reno, NV, 89557, USA.
| |
Collapse
|
6
|
Paez I, Prado Y, Loren P, Ubilla CG, Rodríguez N, Salazar LA. Cholesterol-Related lncRNAs as Response Predictors of Atorvastatin Treatment in Chilean Hypercholesterolemic Patients: A Pilot Study. Biomedicines 2023; 11:biomedicines11030742. [PMID: 36979720 PMCID: PMC10045917 DOI: 10.3390/biomedicines11030742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 02/23/2023] [Accepted: 02/24/2023] [Indexed: 03/05/2023] Open
Abstract
Statins are currently the treatment of choice for hypercholesterolemia. However, wide interindividual variability has been observed in the response to treatment. Recent studies have reported the role of lncRNAs in the metabolism of lipids; nevertheless, there are few studies to date that show their role in the response to treatment with statins. Thus, the aim of this study was to assess the levels of expression of three lncRNAs (RP1-13D10.2; MANTIS; lncHR1) associated with genes involved in cholesterol homeostasis in leukocyte cells of hypercholesterolemic patients after treatment with atorvastatin and compare them with levels in subjects with normal cholesterol levels. A secondary aim was to assess the levels of expression in monocytic THP-1 cells differentiated to macrophages. The study included 20 subjects with normal cholesterol (NC) levels and 20 individuals with hypercholesterolemia (HC). The HC patients were treated with atorvastatin (20 mg/day/4 weeks). THP-1 cells were differentiated to macrophages with PMA and treated with different doses of atorvastatin for 24 h. Expression of lncRNAs was determined by RT-qPCR. The lncRNAs RP1-13D10.2 (p < 0.0001), MANTIS (p = 0.0013) and lncHR1 (p < 0.0001) presented increased expression in HC subjects compared with NC subjects. Furthermore, atorvastatin had a negative regulatory effect on the expression of lncHR1 (p < 0.0001) in HC subjects after treatment. In vitro, all the lncRNAs showed significant differences in expression after atorvastatin treatment. Our findings show that the lncRNAs tested present differential expression in HC patients and play a role in the variability reported in the response to atorvastatin treatment. Further research is needed to clarify the biological impact of these lncRNAs on cholesterol homeostasis and treatment with statins.
Collapse
|
7
|
Hafiane A, Favari E, Bortnick AE. Measures of high-density lipoprotein function in men and women with severe aortic stenosis. Lipids Health Dis 2022; 21:48. [PMID: 35643498 PMCID: PMC9148512 DOI: 10.1186/s12944-022-01653-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 04/26/2022] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND Calcification of the aortic valve is a common heart valve disorder, in some cases leading to clinically impactful severe aortic stenosis (AS). Sex-specific differences in aortic valve calcification (ACV) exist, with women having a lower burden of calcification than men as measured by computed tomography; however, the pathophysiological mechanism that leads to these differences remains unclear. METHODS Using cultured human Tamm-Horsfall protein 1 (THP-1) macrophages and human aortic valve interstitial cells, the effects of high-density lipoprotein (HDL) particles isolated from the plasma of men and women with severe AS were studied for cholesterol efflux capacity (CEC). RESULTS HDL-CEC was assessed in 46 patients with severe AS, n = 30 men, n = 16 women. ATP-Binding Cassette A1 (ABCA1)-mediated HDL-CEC was measured from human cultured THP-1 macrophages to plasma HDL samples. Women with severe AS had more ABCA1-mediated HDL-CEC, as compared to men (8.50 ± 3.90% cpm vs. 6.80 ± 1.50% cpm, P = 0.04). HDL pre-β1 and α-particles were higher in woman than in men by spectral density, (pre-β1 HDL, 20298.29 ± 1076.15 vs. 15,661.74 ± 789.00, P = 0.002, and α-HDL, 63006.35 ± 756.81 vs. 50,447.00 ± 546.52, P = 0.03). Lecithin-cholesterol acyltransferase conversion of free cholesterol into cholesteryl esters was higher in women than men (16.44 ± 9.11%/h vs. 12.00 ± 8.07%/h, P = 0.03). CONCLUSIONS Sex-specific changes in various parameters of HDL-CEC were found in patients with severe AS. Sex-based modifications in HDL functionality by HDL-CEC might account for the reduced burden of calcification in women vs. men with severe AS. Therefore, future studies should target sex-related pathways in AS to help to improve understanding and treatment of AS. Sex specifc differences in AVC and differences associated with HDL function in men and women with severe AS. When compared to men, women had higher preβ-HDL and α-HDL migrating particles, higher cholesterol efflux to HDL, and higher lecithin cholesterol acyl transferase (LCAT) activity, possibly indicating that improved reverse cholesterol transport may be protective against worsened calcification.
Collapse
Affiliation(s)
- Anouar Hafiane
- Department of Medicine, Faculty of Medicine, Research Institute of the McGill University Health Centre, 1001 Boulevard Decarie, Montreal, Québec, H3A 1A1, Canada.
| | - Elda Favari
- Department of Food and Drug, University of Parma, Parma, Italy
| | - Anna E Bortnick
- Department of Medicine, Division of Cardiology, Bronx, New York, USA
- Division of Geriatrics, Montefiore Medical Center and Albert Einstein College of Medicine, Bronx, New York, USA
| |
Collapse
|
8
|
Becker PH, Le Guillou E, Duque M, Blondel A, Gons C, Ben Souna H, Imbard A, Fournier N, Gaignard P, Thérond P. Cholesterol accumulation induced by acetylated LDL exposure modifies the enzymatic activities of the TCA cycle without impairing the respiratory chain functionality in macrophages. Biochimie 2022; 200:87-98. [PMID: 35618159 DOI: 10.1016/j.biochi.2022.05.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Revised: 03/31/2022] [Accepted: 05/19/2022] [Indexed: 11/27/2022]
Abstract
The unregulated uptake of modified low-density lipoproteins (LDL) by macrophages leads to foam cell formation, promoting atherosclerotic plaque progression. The cholesterol efflux capacity of macrophages by the ATP-Binding Cassette transporters depends on the ATP mitochondrial production. Therefore, the mitochondrial function maintenance is crucial in limiting foam cell formation. Thus, we aimed to investigate the mechanisms involved in the mitochondrial dysfunction that may occur in cholesterol-laden macrophages. We incubated THP-1 macrophages with acetylated LDL (acLDL) to obtain cholesterol-laden cells or with mildly oxidized LDL (oxLDL) to generate cholesterol- and oxidized lipids-laden cells. Cellular cholesterol content was measured in each condition. Mitochondrial function was evaluated by measurement of several markers of energetic metabolism, oxidative phosphorylation, oxidative stress, mitochondrial biogenesis and dynamics. OxLDL-exposed macrophages exhibited a significantly reduced mitochondrial respiration and complexes I and III activities, associated to an oxidative stress state and a reduced mitochondrial DNA copy number. Meanwhile, acLDL-exposed macrophages featured an efficient oxidative phosphorylation despite the decreased activities of aconitase, isocitrate dehydrogenase and α-ketoglutarate dehydrogenase. Our study revealed that mitochondrial function was differently impacted according to the nature of modified LDL. Exposure to cholesterol and oxidized lipids carried by oxLDL leads to a mitochondrial dysfunction in macrophages, affecting the mitochondrial respiratory chain functional capacity, whereas the cellular cholesterol enrichment induced by acLDL exposure results in a tricarboxylic acid cycle shunt while maintaining mitochondrial energetic production, reflecting a metabolic adaptation to cholesterol intake. These new mechanistic insights are of direct relevance to the understanding of the mitochondrial dysfunction in foam cells.
Collapse
Affiliation(s)
- Pierre-Hadrien Becker
- Université Paris-Saclay, EA 7357, Lipides: systèmes analytiques et biologiques, Châtenay-Malabry, 92296, France; Hôpital Bicêtre, AP-HP, Laboratoire de Biochimie, Le Kremlin Bicêtre, 94270, France.
| | - Edouard Le Guillou
- Hôpital Bicêtre, AP-HP, Laboratoire de Biochimie, Le Kremlin Bicêtre, 94270, France
| | - Mathilde Duque
- Hôpital Bicêtre, AP-HP, Laboratoire de Biochimie, Le Kremlin Bicêtre, 94270, France
| | - Amélie Blondel
- Hôpital Bicêtre, AP-HP, Laboratoire de Biochimie, Le Kremlin Bicêtre, 94270, France
| | - Camille Gons
- Hôpital Bicêtre, AP-HP, Laboratoire de Biochimie, Le Kremlin Bicêtre, 94270, France
| | - Hajar Ben Souna
- Hôpital Bicêtre, AP-HP, Laboratoire de Biochimie, Le Kremlin Bicêtre, 94270, France
| | - Apolline Imbard
- Université Paris-Saclay, EA 7357, Lipides: systèmes analytiques et biologiques, Châtenay-Malabry, 92296, France; Hôpital Necker-Enfants Malades, AP-HP, Laboratoire de Biochimie Métabolique, Paris, 75015, France
| | - Natalie Fournier
- Université Paris-Saclay, EA 7357, Lipides: systèmes analytiques et biologiques, Châtenay-Malabry, 92296, France; Hôpital Européen Georges Pompidou, AP-HP, Laboratoire de Biochimie, Paris, 75015, France
| | - Pauline Gaignard
- Université Paris-Saclay, EA 7357, Lipides: systèmes analytiques et biologiques, Châtenay-Malabry, 92296, France; Hôpital Bicêtre, AP-HP, Laboratoire de Biochimie, Le Kremlin Bicêtre, 94270, France
| | - Patrice Thérond
- Université Paris-Saclay, EA 7357, Lipides: systèmes analytiques et biologiques, Châtenay-Malabry, 92296, France; Hôpital Bicêtre, AP-HP, Laboratoire de Biochimie, Le Kremlin Bicêtre, 94270, France
| |
Collapse
|
9
|
Choi HY, Ruel I, Choi S, Genest J. New Strategies to Promote Macrophage Cholesterol Efflux. Front Cardiovasc Med 2022; 8:795868. [PMID: 35004908 PMCID: PMC8733154 DOI: 10.3389/fcvm.2021.795868] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 12/03/2021] [Indexed: 12/11/2022] Open
Abstract
The capacity of macrophages to dispose of cholesterol deposited in the atherosclerotic plaque depends on their ability to activate cholesterol efflux pathways. To develop athero-protective therapies aimed at promoting macrophage cholesterol efflux, cholesterol metabolism in THP-1 monocyte-derived macrophages has been extensively studied, but the intrinsic sensitivity of monocytes and the lack of a standardized procedure to differentiate THP-1 monocytes into macrophages have made it difficult to utilize THP-1 macrophages in the same or similar degree of differentiation across studies. The variability has resulted in lack of understanding of how the differentiation affects cholesterol metabolism, and here we review and investigate the effects of THP-1 differentiation on cholesterol efflux. The degree of THP-1 differentiation was inversely associated with ATP binding cassette A1 (ABCA1) transporter-mediated cholesterol efflux. The differentiation-associated decrease in ABCA1-mediated cholesterol efflux occurred despite an increase in ABCA1 expression. In contrast, DSC1 expression decreased during the differentiation. DSC1 is a negative regulator of the ABCA1-mediated efflux pathway and a DSC1-targeting agent, docetaxel showed high potency and efficacy in promoting ABCA1-mediated cholesterol efflux in THP-1 macrophages. These data suggest that pharmacological targeting of DSC1 may be more effective than increasing ABCA1 expression in promoting macrophage cholesterol efflux. In summary, the comparison of THP-1 macrophage subtypes in varying degrees of differentiation provided new insights into cholesterol metabolism in macrophages and allowed us to identify a viable target DSC1 for the promotion of cholesterol efflux in differentiated macrophages. Docetaxel and other pharmacological strategies targeting DSC1 may hold significant potential for reducing atherogenic cholesterol deposition.
Collapse
Affiliation(s)
- Hong Y Choi
- Cardiovascular Research Laboratories, Research Institute of the McGill University Health Center, Montreal, QC, Canada
| | - Isabelle Ruel
- Cardiovascular Research Laboratories, Research Institute of the McGill University Health Center, Montreal, QC, Canada
| | - Shiwon Choi
- Cardiovascular Research Laboratories, Research Institute of the McGill University Health Center, Montreal, QC, Canada
| | - Jacques Genest
- Cardiovascular Research Laboratories, Research Institute of the McGill University Health Center, Montreal, QC, Canada
| |
Collapse
|
10
|
Pan X. Cholesterol Metabolism in Chronic Kidney Disease: Physiology, Pathologic Mechanisms, and Treatment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1372:119-143. [PMID: 35503178 PMCID: PMC11106795 DOI: 10.1007/978-981-19-0394-6_9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
High plasma levels of lipids and/or lipoproteins are risk factors for atherosclerosis, nonalcoholic fatty liver disease (NAFLD), obesity, and diabetes. These four conditions have also been identified as risk factors leading to the development of chronic kidney disease (CKD). Although many pathways that generate high plasma levels of these factors have been identified, most clinical and physiologic dysfunction results from aberrant assembly and secretion of lipoproteins. The results of several published studies suggest that elevated levels of low-density lipoprotein (LDL)-cholesterol are a risk factor for atherosclerosis, myocardial infarction, coronary artery calcification associated with type 2 diabetes, and NAFLD. Cholesterol metabolism has also been identified as an important pathway contributing to the development of CKD; clinical treatments designed to alter various steps of the cholesterol synthesis and metabolism pathway are currently under study. Cholesterol synthesis and catabolism contribute to a multistep process with pathways that are regulated at the cellular level in renal tissue. Cholesterol metabolism may also be regulated by the balance between the influx and efflux of cholesterol molecules that are capable of crossing the membrane of renal proximal tubular epithelial cells and podocytes. Cellular accumulation of cholesterol can result in lipotoxicity and ultimately kidney dysfunction and failure. Thus, further research focused on cholesterol metabolism pathways will be necessary to improve our understanding of the impact of cholesterol restriction, which is currently a primary intervention recommended for patients with dyslipidemia.
Collapse
Affiliation(s)
- Xiaoyue Pan
- Department of Foundations of Medicine, New York University Long Island School of Medicine, Mineola, NY, USA.
| |
Collapse
|
11
|
Dakroub H, Nowak M, Benoist JF, Noël B, Vedie B, Paul JL, Fournier N. Eicosapentaenoic acid membrane incorporation stimulates ABCA1-mediated cholesterol efflux from human THP-1 macrophages. Biochim Biophys Acta Mol Cell Biol Lipids 2021; 1866:159016. [PMID: 34332075 DOI: 10.1016/j.bbalip.2021.159016] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 06/28/2021] [Accepted: 07/25/2021] [Indexed: 12/16/2022]
Abstract
A high intake in polyunsaturated fatty acids (PUFAs), especially eicosapentaenoic acid (EPA) (C20:5 n-3), is cardioprotective. Dietary PUFAs incorporate into membrane phospholipids, which may modify the function of membrane proteins. We investigated the consequences of the membrane incorporation of several PUFAs on the key antiatherogenic ABCA1-mediated cholesterol efflux pathway. Human THP-1 macrophages were incubated with EPA, arachidonic acid (AA) (C20:4 n-6) or docosahexaenoic acid (DHA) (C22:6 n-3) for a long time to mimic a chronic exposure. EPA 70 μM, but not AA 50 μM or DHA 15 μM, increased ABCA1-mediated cholesterol efflux to apolipoprotein (apo) AI by 28% without altering aqueous diffusion. No variation in ABCA1 expression or localization was observed after EPA treatment. EPA incorporation did not affect the phenotype of THP-1 macrophages. The membrane phospholipids composition of EPA cells displayed higher levels of both EPA and its elongation product docosapentaenoic acid, which was associated with drastic lower levels of AA. Treatment by EPA increased the ATPase activity of the transporter, likely through a PKA-dependent mechanism. Eicosanoids were not involved in the stimulated ABCA1-mediated cholesterol efflux from EPA-enriched macrophages. In addition, EPA supplementation increased the apo AI binding capacity from macrophages by 38%. Moreover, the increased apo AI binding in EPA-enriched macrophages can be competed. In conclusion, EPA membrane incorporation increased ABCA1 functionality in cholesterol-normal human THP-1 macrophages, likely through a combination of different mechanisms. This beneficial in vitro effect may partly contribute to the cardioprotective effect of a diet enriched with EPA highlighted by several recent clinical trials.
Collapse
Affiliation(s)
- Hani Dakroub
- Lip(Sys) (2) - EA 7357, Athérosclérose et macrophages: impact des phospholipides et des fonctions mitochondriales sur le trafic et l'efflux du cholestérol, Université Paris-Saclay, UFR de Pharmacie, 92296 Châtenay-Malabry, France
| | - Maxime Nowak
- Lip(Sys) (2) - EA 7357, Athérosclérose et macrophages: impact des phospholipides et des fonctions mitochondriales sur le trafic et l'efflux du cholestérol, Université Paris-Saclay, UFR de Pharmacie, 92296 Châtenay-Malabry, France
| | - Jean-François Benoist
- Lip(Sys) (2) - EA 7357, Athérosclérose et macrophages: impact des phospholipides et des fonctions mitochondriales sur le trafic et l'efflux du cholestérol, Université Paris-Saclay, UFR de Pharmacie, 92296 Châtenay-Malabry, France; Laboratoire de Biochimie métabolique, AP-HP (Assistance Publique-Hôpitaux de Paris), Hôpital Necker, 75015 Paris, France
| | - Benoît Noël
- Allergie, Immunotoxicologie et Immunopathologie, INSERM UMR 996, Université Paris-Saclay, UFR de Pharmacie, 92296 Châtenay-Malabry, France
| | - Benoît Vedie
- Laboratoire de Biochimie, AP-HP (Assistance Publique-Hôpitaux de Paris), Hôpital Européen Georges Pompidou, 75015 Paris, France
| | - Jean-Louis Paul
- Lip(Sys) (2) - EA 7357, Athérosclérose et macrophages: impact des phospholipides et des fonctions mitochondriales sur le trafic et l'efflux du cholestérol, Université Paris-Saclay, UFR de Pharmacie, 92296 Châtenay-Malabry, France; Laboratoire de Biochimie, AP-HP (Assistance Publique-Hôpitaux de Paris), Hôpital Européen Georges Pompidou, 75015 Paris, France
| | - Natalie Fournier
- Lip(Sys) (2) - EA 7357, Athérosclérose et macrophages: impact des phospholipides et des fonctions mitochondriales sur le trafic et l'efflux du cholestérol, Université Paris-Saclay, UFR de Pharmacie, 92296 Châtenay-Malabry, France; Laboratoire de Biochimie, AP-HP (Assistance Publique-Hôpitaux de Paris), Hôpital Européen Georges Pompidou, 75015 Paris, France.
| |
Collapse
|
12
|
Induction of microRNA hsa-let-7d-5p, and repression of HMGA2, contribute protection against lipid accumulation in macrophage 'foam' cells. Biochim Biophys Acta Mol Cell Biol Lipids 2021; 1866:159005. [PMID: 34274506 DOI: 10.1016/j.bbalip.2021.159005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 07/08/2021] [Accepted: 07/13/2021] [Indexed: 12/14/2022]
Abstract
Accumulation of excess cholesterol and cholesteryl ester in macrophage 'foam' cells within the arterial intima characterises early 'fatty streak' atherosclerotic lesions, and is accompanied by epigenetic changes, including altered expression of microRNA sequences which determine of gene and protein expression. This study established that exposure to lipoproteins, including acetylated LDL, induced macrophage expression of microRNA hsa-let-7d-5p, a sequence previously linked with tumour suppression, and repressed expression of one of its target genes, high mobility group AT hook 2 (HMGA2). A let-7d-5p mimic repressed expression of HMGA2 (18%; p < 0.05) while a marked increase (2.9-fold; p < 0.05) in expression of HMGA2 was noted in the presence of let-7d-5p inhibitor. Under these conditions, let-7d-5p mimic significantly (p < 0.05) decreased total (10%), free (8%) and cholesteryl ester (21%) mass, while the inhibitor significantly (p < 0.05) increased total (29%) and free cholesterol (29%) mass, compared with the relevant controls. Let-7d-5p inhibition significantly (p < 0.05) increased endogenous biosynthesis of cholesterol (38%) and cholesteryl ester (39%) pools in macrophage 'foam' cells, without altering the cholesterol efflux pathway, or esterification of exogenous radiolabelled oleate. Let-7d-5p inhibition in sterol-loaded cells increased the level of HMGA2 protein (32%; p < 0.05), while SiRNA knockdown of this protein (29%; p < 0.05) resulted in a (21%, p < 0.05) reduction in free cholesterol mass. Thus, induction of let-7d-5p, and repression of its target HMGA2, in macrophages is a protective response to the challenge of increased cholesterol influx into these cells; dysregulation of this response may contribute to atherosclerosis and other disorders such as cancer.
Collapse
|
13
|
Pinto SM, Kim H, Subbannayya Y, Giambelluca MS, Bösl K, Ryan L, Sharma A, Kandasamy RK. Comparative Proteomic Analysis Reveals Varying Impact on Immune Responses in Phorbol 12-Myristate-13-Acetate-Mediated THP-1 Monocyte-to-Macrophage Differentiation. Front Immunol 2021; 12:679458. [PMID: 34234780 PMCID: PMC8255674 DOI: 10.3389/fimmu.2021.679458] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 05/25/2021] [Indexed: 01/06/2023] Open
Abstract
Macrophages are sentinels of the innate immune system, and the human monocytic cell line THP-1 is one of the widely used in vitro models to study inflammatory processes and immune responses. Several monocyte-to-macrophage differentiation protocols exist, with phorbol 12-myristate-13-acetate (PMA) being the most commonly used and accepted method. However, the concentrations and duration of PMA treatment vary widely in the published literature and could affect the probed phenotype, however their effect on protein expression is not fully deciphered. In this study, we employed a dimethyl labeling-based quantitative proteomics approach to determine the changes in the protein repertoire of macrophage-like cells differentiated from THP-1 monocytes by three commonly used PMA-based differentiation protocols. Employing an integrated network analysis, we show that variations in PMA concentration and duration of rest post-stimulation result in downstream differences in the protein expression and cellular signaling processes. We demonstrate that these differences result in altered inflammatory responses, including variation in the expression of cytokines upon stimulation with various Toll-like receptor (TLR) agonists. Together, these findings provide a valuable resource that significantly expands the knowledge of protein expression dynamics with one of the most common in vitro models for macrophages, which in turn has a profound impact on the immune as well as inflammatory responses being studied.
Collapse
Affiliation(s)
- Sneha M. Pinto
- Centre of Molecular Inflammation Research (CEMIR), and Department of Clinical and Molecular Medicine (IKOM), Norwegian University of Science and Technology, Trondheim, Norway
- Center for Systems Biology and Molecular Medicine, Yenepoya (Deemed to be University), Mangalore, India
| | - Hera Kim
- Centre of Molecular Inflammation Research (CEMIR), and Department of Clinical and Molecular Medicine (IKOM), Norwegian University of Science and Technology, Trondheim, Norway
| | - Yashwanth Subbannayya
- Centre of Molecular Inflammation Research (CEMIR), and Department of Clinical and Molecular Medicine (IKOM), Norwegian University of Science and Technology, Trondheim, Norway
| | - Miriam S. Giambelluca
- Centre of Molecular Inflammation Research (CEMIR), and Department of Clinical and Molecular Medicine (IKOM), Norwegian University of Science and Technology, Trondheim, Norway
| | - Korbinian Bösl
- Centre of Molecular Inflammation Research (CEMIR), and Department of Clinical and Molecular Medicine (IKOM), Norwegian University of Science and Technology, Trondheim, Norway
- Department of Infectious Diseases, Medical Clinic, St. Olavs Hospital, Trondheim, Norway
| | - Liv Ryan
- Centre of Molecular Inflammation Research (CEMIR), and Department of Clinical and Molecular Medicine (IKOM), Norwegian University of Science and Technology, Trondheim, Norway
| | - Animesh Sharma
- Proteomics and Modomics Experimental Core, PROMEC, Norwegian University of Science and Technology and the Central Norway Regional Health Authority, Stjørdal, Norway
| | - Richard K. Kandasamy
- Centre of Molecular Inflammation Research (CEMIR), and Department of Clinical and Molecular Medicine (IKOM), Norwegian University of Science and Technology, Trondheim, Norway
| |
Collapse
|
14
|
Effects of lipoproteins on endothelial cells and macrophages function and its possible implications on fetal adverse outcomes associated to maternal hypercholesterolemia during pregnancy. Placenta 2021; 106:79-87. [PMID: 33706211 DOI: 10.1016/j.placenta.2021.02.019] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 02/03/2021] [Accepted: 02/25/2021] [Indexed: 11/23/2022]
Abstract
Hypercholesterolemia is one of the main risk factors associated with atherosclerosis and cardiovascular disease, the leading cause of death worldwide. During pregnancy, maternal hypercholesterolemia develops, and it can occur in a physiological (MPH) or supraphysiological (MSPH) manner, where MSPH is associated with endothelial dysfunction and early atherosclerotic lesions in the fetoplacental vasculature. In the pathogenesis of atherosclerosis, endothelial activation and endothelial dysfunction, characterized by an imbalance in the bioavailability of nitric oxide, contribute to the early stages of this disease. Macrophages conversion to foam cells, cholesterol efflux from these cells and its differentiation into a pro- or anti-inflammatory phenotype are also important processes that contribute to atherosclerosis. In adults it has been reported that native and modified HDL and LDL play an important role in endothelial and macrophage function. In this review it is proposed that fetal lipoproteins could be also relevant factors involved in the detrimental vascular effects described in MSPH. Changes in the composition and function of neonatal lipoproteins compared to adults has been reported and, although in MSPH pregnancies the fetal lipid profile does not differ from MPH, differences in the lipidomic profiles of umbilical venous blood have been reported, which could have implications in the vascular function. In this review we summarize the available information regarding the effects of lipoproteins on endothelial and macrophage function, emphasizing its possible implications on fetal adverse outcomes associated to maternal hypercholesterolemia during pregnancy.
Collapse
|
15
|
Hafiane A, Daskalopoulou SS. Adiponectin's mechanisms in high-density lipoprotein biogenesis and cholesterol efflux. Metabolism 2020; 113:154393. [PMID: 33058851 DOI: 10.1016/j.metabol.2020.154393] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 09/18/2020] [Accepted: 10/07/2020] [Indexed: 12/13/2022]
Abstract
AIM Among adiponectin's beneficial properties is its ability to promote cellular cholesterol efflux, thereby generating high-density lipoprotein (HDL) particles. However, adiponectin's role in the regulation of macrophage lipid metabolism, a crucial process in atherogenesis, remains poorly investigated. The aim of this study was to characterize the adiponectin's role in HDL biogenesis. METHODS AND RESULTS We perform kinetics studies in baby hamster kidney (BHK) and Tamm-Horsfall protein 1 (THP-1) cell lines to elucidate adiponectin's role in HDL biogenesis. In cholesterol-enriched cells, specific molar doses of adiponectin stimulated cholesterol efflux with high efficiency to apoA-I. In the presence of adiponectin, BHK cells expressing ATP binding cassette transporter A1 (ABCA1) or ABCG1 generated lipidated particles having α electrophoretic mobility (α-HDL) and a molecular size of 7.5-20 nm. Interestingly, in THP-1 macrophages, cholesterol efflux was associated with more lipidated preβ1-HDL particles. Direct molecular interaction of adiponectin with apoA-I enhanced the affinity of apoA-I to free cholesterol and resulted in an increase in preβ1-HDL particles from plasma ex vivo. Adiponectin increased ABCA1 and ABCG1 protein expression and activated the formation of ABCA1-linked cholesterol oxidase sensitive plasma membrane domains. CONCLUSION Adiponectin upregulated ABCA1 and ABCG1 protein expression, reduced lipid accumulation, and efficiently promoted nascent HDL formation. These results highlight that these cellular processes are interconnected through adiponectin and ABCA1- and ABCG1-dependent. In this pathway, adiponectin increased the affinity of apoA-I to cholesterol and effectively accelerated cholesterol removal from the plasma membrane to HDL particles. Thus, by accelerating HDL biogenesis, adiponectin may have therapeutic potential for atherosclerotic cardiovascular disease prevention and management.
Collapse
Affiliation(s)
- Anouar Hafiane
- Department of Medicine, Faculty of Medicine, Research Institute of the McGill University Health Centre, McGill University, 1001 Decarie Blvd, Bloc E01.3370H, Montréal, Qc H4A 3J1, Canada.
| | - Stella S Daskalopoulou
- Department of Medicine, Faculty of Medicine, Research Institute of the McGill University Health Centre, McGill University, 1001 Decarie Blvd, Bloc E01.3370H, Montréal, Qc H4A 3J1, Canada; Department of Medicine, Division of Internal Medicine, McGill University, Research Institute of the McGill University Health Centre, 1001 Decarie Blvd, EM1.2230, Montreal, Quebec H4A 3J1, Canada.
| |
Collapse
|
16
|
He P, Gelissen IC, Ammit AJ. Regulation of ATP binding cassette transporter A1 (ABCA1) expression: cholesterol-dependent and - independent signaling pathways with relevance to inflammatory lung disease. Respir Res 2020; 21:250. [PMID: 32977800 PMCID: PMC7519545 DOI: 10.1186/s12931-020-01515-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 09/21/2020] [Indexed: 12/11/2022] Open
Abstract
The role of the ATP binding cassette transporter A1 (ABCA1) in maintaining cellular lipid homeostasis in cardiovascular disease is well established. More recently, the important beneficial role played by ABCA1 in modulating pathogenic disease mechanisms, such as inflammation, in a broad range of chronic conditions has been realised. These studies position ABCA1 as a potential therapeutic target in a diverse range of diseases where inflammation is an underlying cause. Chronic respiratory conditions such as asthma and chronic obstructive pulmonary disease (COPD) are driven by inflammation, and as such, there is now a growing recognition that we need a greater understanding of the signaling pathways responsible for regulation of ABCA1 expression in this clinical context. While the signaling pathways responsible for cholesterol-mediated ABCA1 expression have been clearly delineated through decades of studies in the atherosclerosis field, and thus far appear to be translatable to the respiratory field, less is known about the cholesterol-independent signaling pathways that can modulate ABCA1 expression in inflammatory lung disease. This review will identify the various signaling pathways and ligands that are associated with the regulation of ABCA1 expression and may be exploited in future as therapeutic targets in the setting of chronic inflammatory lung diseases.
Collapse
Affiliation(s)
- Patrick He
- Woolcock Emphysema Centre, Woolcock Institute of Medical Research, University of Sydney, Sydney, NSW, Australia
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
| | - Ingrid C Gelissen
- Sydney Pharmacy School, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, 2006, Australia
| | - Alaina J Ammit
- Woolcock Emphysema Centre, Woolcock Institute of Medical Research, University of Sydney, Sydney, NSW, Australia.
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia.
| |
Collapse
|
17
|
Jayaraj P, Narasimhulu CA, Maiseyeu A, Durairaj R, Rao S, Rajagopalan S, Parthasarathy S, Desikan R. Methoxyphenol derivatives as reversible inhibitors of myeloperoxidase as potential antiatherosclerotic agents. Future Med Chem 2020; 12:95-110. [PMID: 31769316 PMCID: PMC7333589 DOI: 10.4155/fmc-2019-0080] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 10/23/2019] [Indexed: 01/26/2023] Open
Abstract
Aim: To evaluate new chemical entities, based on ferulic acid scaffolds, as reversible myeloperoxidase inhibitors (MPOI). Methodology & results:In silico docking studies are performed with MPO protein as a target for several ferulic acid analogs followed by multiple in vitro assays to validate this approach. Two lead compounds 2a and 3 are identified with optimum docking and IC50 values: -7.95 kcal/mol, 0.9 μM and -8.35 kcal/mol, 8.5 μM, respectively. These MPOIs are able to inhibit oxidation of high-density lipoprotein and further promoted functionality of high-density lipoprotein. Conclusion: Lead analogs are potent MPOIs that exert specific effects on MPO-mediated oxidation as well as inflammatory pathways. It also acts as promoters of cholesterol efflux that sheds light on pharmacological approach in atherosclerosis treatment.
Collapse
Affiliation(s)
- Premkumar Jayaraj
- Department of Chemistry, School of Advanced Science, Vellore Institute of Technology, Vellore 632014, India
| | - Chandrakala A Narasimhulu
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Andrei Maiseyeu
- Cardiovascular Research Institute, Case Western Reserve University, School of Medicine, 10900 Euclid Ave, Cleveland, OH 44106, USA
| | - Rekha Durairaj
- Cardiovascular Research Institute, Case Western Reserve University, School of Medicine, 10900 Euclid Ave, Cleveland, OH 44106, USA
| | - Shashidhar Rao
- Department of Chemistry & Chemical Biology, Rutgers University, Piscataway, NJ 08554, USA
| | - Sanjay Rajagopalan
- Cardiovascular Research Institute, Case Western Reserve University, School of Medicine, 10900 Euclid Ave, Cleveland, OH 44106, USA
| | - Sampath Parthasarathy
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Rajagopal Desikan
- Carmel Biosciences, 3562 Habersham at Northlake, Building J, Suite A, Tucker, GA 30084, USA
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
- Department of Chemistry, School of Advanced Science, Vellore Institute of Technology, Vellore 632014, India
| |
Collapse
|
18
|
Fournier N, Benoist JF, Allaoui F, Nowak M, Dakroub H, Vedie B, Paul JL. Contrasting effects of membrane enrichment with polyunsaturated fatty acids on phospholipid composition and cholesterol efflux from cholesterol-loaded J774 mouse or primary human macrophages. Biochim Biophys Acta Mol Cell Biol Lipids 2019; 1865:158536. [PMID: 31672574 DOI: 10.1016/j.bbalip.2019.158536] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 08/30/2019] [Accepted: 09/24/2019] [Indexed: 12/15/2022]
Abstract
A high consumption of polyunsaturated fatty acids (PUFAs), particularly n-3 PUFAs, is atheroprotective. PUFAs incorporation into membrane phospholipids alters the functionality of membrane proteins. We studied the consequences of the in vitro supplementation of several PUFAs on the FA profiles and on ABCA1-dependent cholesterol efflux capacities from cholesterol-loaded macrophages. Arachidonic acid (AA, C20:4 n-6) and, to a lesser extent, eicosapentaenoic acid (EPA, C20:5 n-3), dose-dependently impaired cholesterol efflux from cholesterol-loaded J774 mouse macrophages without alterations in ABCA1 expression, whereas docosahexaenoic acid (DHA, C22:6 n-3) had no impact. AA cells exhibited higher proportions of arachidonic acid and adrenic acid (C22:4 n-6), its elongation product. EPA cells exhibited slightly higher proportions of EPA associated with much higher proportions of docosapentaenoic acid (C22:5 n-3), its elongation product and with lower proportions of AA. Conversely, both EPA and DHA and, to a lesser extent, AA decreased cholesterol efflux from cholesterol-loaded primary human macrophages (HMDM). The differences observed in FA profiles after PUFA supplementations were different from those observed for the J774 cells. In conclusion, we are the first to report that AA and EPA, but not DHA, have deleterious effects on the cardioprotective ABCA1 cholesterol efflux pathway from J774 foam cells. Moreover, the membrane incorporation of PUFAs does not have the same impact on cholesterol efflux from murine (J774) or human (HMDM) cholesterol-loaded macrophages. This finding emphasizes the key role of the cellular model in cholesterol efflux studies and may partly explain the heterogeneous literature data on the impact of PUFAs on cholesterol efflux.
Collapse
Affiliation(s)
- Natalie Fournier
- Lip(Sys)(2) - EA 7357, Athérosclérose: homéostasie et trafic du cholestérol des macrophages, Univ. Paris-Sud, Université Paris-Saclay, UFR de Pharmacie, 92290 Châtenay-Malabry, France; Laboratoire de Biochimie, AP-HP (Assistance Publique-Hôpitaux de Paris), Hôpital Européen Georges Pompidou, 75015 Paris, France.
| | - Jean-François Benoist
- Lip(Sys)(2) - EA 7357, Athérosclérose: homéostasie et trafic du cholestérol des macrophages, Univ. Paris-Sud, Université Paris-Saclay, UFR de Pharmacie, 92290 Châtenay-Malabry, France; Laboratoire de Biochimie hormonale, AP-HP (Assistance Publique-Hôpitaux de Paris), Hôpital Robert Debré, 75019 Paris, France
| | - Fatima Allaoui
- Lip(Sys)(2) - EA 7357, Athérosclérose: homéostasie et trafic du cholestérol des macrophages, Univ. Paris-Sud, Université Paris-Saclay, UFR de Pharmacie, 92290 Châtenay-Malabry, France
| | - Maxime Nowak
- Lip(Sys)(2) - EA 7357, Athérosclérose: homéostasie et trafic du cholestérol des macrophages, Univ. Paris-Sud, Université Paris-Saclay, UFR de Pharmacie, 92290 Châtenay-Malabry, France
| | - Hani Dakroub
- Lip(Sys)(2) - EA 7357, Athérosclérose: homéostasie et trafic du cholestérol des macrophages, Univ. Paris-Sud, Université Paris-Saclay, UFR de Pharmacie, 92290 Châtenay-Malabry, France
| | - Benoît Vedie
- Laboratoire de Biochimie, AP-HP (Assistance Publique-Hôpitaux de Paris), Hôpital Européen Georges Pompidou, 75015 Paris, France
| | - Jean-Louis Paul
- Lip(Sys)(2) - EA 7357, Athérosclérose: homéostasie et trafic du cholestérol des macrophages, Univ. Paris-Sud, Université Paris-Saclay, UFR de Pharmacie, 92290 Châtenay-Malabry, France; Laboratoire de Biochimie, AP-HP (Assistance Publique-Hôpitaux de Paris), Hôpital Européen Georges Pompidou, 75015 Paris, France
| |
Collapse
|
19
|
ABCA1 Agonist Mimetic Peptide CS-6253 Induces Microparticles Release From Different Cell Types by ABCA1-Efflux-Dependent Mechanism. Can J Cardiol 2019; 35:770-781. [PMID: 31151713 DOI: 10.1016/j.cjca.2019.02.018] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 02/18/2019] [Accepted: 02/18/2019] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND Small peptides based on the C-terminal domain of apo E have recently been proposed as ATP-binding cassette transporter A1 (ABCA1) agonist with therapeutic potential. Previous work has shown that a novel synthetic peptide, CS-6253, acts synergistically with apolipoprotein A-I or alone to generate high-density lipoprotein (HDL) particles; we have also shown that cells can release microparticles (50-350 nm in apparent diameter) in an ABCA1- and apolipoprotein A-I-dependent manner. The purpose of this study was to explore the ability of a novel synthetic peptide CS-6253 to induce microparticle release from various cell lines in the process of HDL biogenesis. METHODS The effects of CS-6253 on microparticle formation through the ABCA1 transporter were examined in vitro using cell-based systems and pharmacologic manipulations. RESULTS In cell-based systems combined with fast performance liquid chromatography and nano-sight-tracking analysis, we show that ABCA1 and CS-6253 mediate and increase the production of microparticles containing cholesterol. CS-6253 in baby hamster kidney cells not expressing ABCA1 (baby hamster kidney mock cells) did not alter cholesterol removal across the plasma membrane in the absence of ABCA1 expression even at high concentrations. We report that CS-6253 is not cytotoxic. CONCLUSIONS The present study shows that CS-6253 generates cholesterol containing microparticles with size heterogeneity (100-350 nm) in an ABCA1-dependent manner. We show that microparticles contribute to cell cholesterol efflux from monocyte-macrophage cells. At high doses, CS-6253 is not able to extract cholesterol from cells not expressing ABCA1, indicating that CS-6253 requires ABCA1 cooperation for cholesterol mobilization. We conclude that CS-6253 is an ABCA1 agonist peptide that promotes cellular cholesterol efflux through HDL biogenesis and microparticle formation.
Collapse
|
20
|
White CR, Datta G, Wilson L, Palgunachari MN, Anantharamaiah GM. The apoA-I mimetic peptide 4F protects apolipoprotein A-I from oxidative damage. Chem Phys Lipids 2019; 219:28-35. [PMID: 30707910 DOI: 10.1016/j.chemphyslip.2019.01.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 01/10/2019] [Accepted: 01/28/2019] [Indexed: 02/07/2023]
Abstract
High density lipoprotein (HDL) is prone to modification by the oxidizing and chlorinating agent hypochlorite anion (OCl-). Oxidation of apolipoprotein (apo) A-I, the major protein in HDL, reduces ABCA-1 mediated cholesterol efflux and other protective responses to HDL. The apoA-I mimetic peptide 4F has been shown to undergo oxidation; however, the ability of the peptide to mediate cholesterol efflux remains intact. Here, we show that 4F protects apoA-I from hypochlorite-mediated oxidation. Mass spectral analysis of apoA-I shows that tyrosine residues that are prone to hypochlorite-mediated chlorination are protected in the presence of 4F. Furthermore, 4F enhances the cholesterol efflux ability of apoA-I to a greater extent than either 4F or apoA-I alone, even after hypochlorite oxidation. These observations suggest that apoA-I in lipid complexes may be protected by the presence of 4F, resulting in the preservation of its anti-inflammatory and anti-atherogenic properties. These studies also form the basis for the future studies of nanoparticles possessing both apoA-I and 4F.
Collapse
Affiliation(s)
- C Roger White
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, United States
| | - Geeta Datta
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, United States
| | - Landon Wilson
- Targeted Metabolomics and Proteomics Laboratory, University of Alabama at Birmingham, Birmingham, AL, 35294, United States
| | - Mayakonda N Palgunachari
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, United States
| | - G M Anantharamaiah
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, United States; Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL, 35294, United States.
| |
Collapse
|
21
|
Tort O, Escribà T, Egaña-Gorroño L, de Lazzari E, Cofan M, Fernandez E, Gatell JM, Martinez E, Garcia F, Arnedo M. Cholesterol efflux responds to viral load and CD4 counts in HIV+ patients and is dampened in HIV exposed. J Lipid Res 2018; 59:2108-2115. [PMID: 30213800 PMCID: PMC6210904 DOI: 10.1194/jlr.m088153] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 08/29/2018] [Indexed: 01/30/2023] Open
Abstract
Cholesterol efflux (CE) capacity has been inversely associated with atherosclerosis and may provide an insight on inflammation occurring in human immunodeficiency virus (HIV) individuals. We address this by studying CE in HIV patients at different stages of HIV disease progression. In this cross-sectional study, CE from ApoB-depleted plasma, lipids levels, viral load (VL), CD4+/CD8+ T-cells, high-sensitive C-reactive protein (hsCRP), and lipoprotein (a) were evaluated in untreated HIV-infected patients (UHIVs; n = 43), elite controllers (ECs; n = 8), HIV-exposed seronegative individuals (HESNs; n = 32), and healthy controls (HCs; n = 14). Among UHIVs, those with CD4+ <500 cells/mm3 presented the lowest significant CE, HDL cholesterol (HDL-C), and ApoAI levels. ECs showed similar HDL-C, ApoAI, and CE compared with HCs. Among UHIVs, CE positively correlated with CD4+ T-cell counts (Beta: 1.05; 95% CI: 1.02; 1.07), and for VL higher than 3.8 log, CE was inversely associated with VL (Beta: 0.70; 95% CI: 0.51; 0.95). Remarkably, HESNs presented higher CE (0.78 ± 0.14) than UHIVs (0.65 ± 0.17; P = 0.0005), but lower than HCs (0.90 ± 0.13; P = 0.009). hsCRP levels were highest in the UHIV group (0.45 ± 0.49). CE was sensitive to HIV disease progression. Low CE in HIV patients was associated with lower CD4+ T-cells and higher VL and hsCRP. CE was also lower in HESNs compared with HCs. Our results suggest that immune status secondary to HIV progression and exposure influence plasma HDL-CE capacity.
Collapse
Affiliation(s)
- Olivia Tort
- Group of Genomics and Pharmacogenomics, Acquired Immune Deficiency Syndrome Research Group, Catalan Project for the Development of a Human Immunodeficiency Virus Vaccine (HIVACAT), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clinic de Barcelona, Barcelona, Spain
| | - Tuixent Escribà
- Group of Genomics and Pharmacogenomics, Acquired Immune Deficiency Syndrome Research Group, Catalan Project for the Development of a Human Immunodeficiency Virus Vaccine (HIVACAT), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clinic de Barcelona, Barcelona, Spain
| | - Lander Egaña-Gorroño
- Group of Genomics and Pharmacogenomics, Acquired Immune Deficiency Syndrome Research Group, Catalan Project for the Development of a Human Immunodeficiency Virus Vaccine (HIVACAT), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clinic de Barcelona, Barcelona, Spain
| | - Elisa de Lazzari
- Infectious Diseases Service, Hospital Clínic-IDIBAPS, Laboratory of Retrovirology and Viral Immunopathogenesis, University of Barcelona, Barcelona, Spain
| | - Montserrat Cofan
- Lipid Clinic, Department of Endocrinology and Nutrition, Hospital Clínic de Barcelona, IDIBAPS, Barcelona, Spain and CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Emma Fernandez
- Infectious Diseases Service, Hospital Clínic-IDIBAPS, Laboratory of Retrovirology and Viral Immunopathogenesis, University of Barcelona, Barcelona, Spain
| | - José Maria Gatell
- Hospital Clinic/IDIBAPS, University of Barcelona, ViiV Healthcare, Barcelona, Spain
| | - Esteban Martinez
- Infectious Diseases Service, Hospital Clínic-IDIBAPS, Laboratory of Retrovirology and Viral Immunopathogenesis, University of Barcelona, Barcelona, Spain
| | - Felipe Garcia
- Infectious Diseases Service, Hospital Clínic-IDIBAPS, Laboratory of Retrovirology and Viral Immunopathogenesis, University of Barcelona, Barcelona, Spain
| | - Mireia Arnedo
- Group of Genomics and Pharmacogenomics, Acquired Immune Deficiency Syndrome Research Group, Catalan Project for the Development of a Human Immunodeficiency Virus Vaccine (HIVACAT), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clinic de Barcelona, Barcelona, Spain
| |
Collapse
|
22
|
Zhang H, Reilly MP. Human Induced Pluripotent Stem Cell-Derived Macrophages for Unraveling Human Macrophage Biology. Arterioscler Thromb Vasc Biol 2017; 37:2000-2006. [PMID: 28982665 DOI: 10.1161/atvbaha.117.309195] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Accepted: 09/12/2017] [Indexed: 12/21/2022]
Abstract
Despite a substantial appreciation for the critical role of macrophages in cardiometabolic diseases, understanding of human macrophage biology has been hampered by the lack of reliable and scalable models for cellular and genetic studies. Human induced pluripotent stem cell (iPSC)-derived macrophages (IPSDM), as an unlimited source of subject genotype-specific cells, will undoubtedly play an important role in advancing our understanding of the role of macrophages in human diseases. In this review, we summarize current literature in the differentiation and characterization of IPSDM at phenotypic, functional, and transcriptomic levels. We emphasize the progress in differentiating iPSC to tissue resident macrophages, and in understanding the ontogeny of in vitro differentiated IPSDM that resembles primitive hematopoiesis, rather than adult definitive hematopoiesis. We review the application of IPSDM in modeling both Mendelian genetic disorders and host-pathogen interactions. Finally, we highlighted the potential areas of research using IPSDM in functional validation of coronary artery disease loci in genome-wide association studies, functional genomic analyses, drug testing, and cell therapeutics in cardiovascular diseases.
Collapse
Affiliation(s)
- Hanrui Zhang
- From the Division of Cardiology, Department of Medicine (H.Z., M.P.R.) and Irving Institute for Clinical and Translational Research (M.P.R.), Columbia University Medical Center, New York, NY.
| | - Muredach P Reilly
- From the Division of Cardiology, Department of Medicine (H.Z., M.P.R.) and Irving Institute for Clinical and Translational Research (M.P.R.), Columbia University Medical Center, New York, NY.
| |
Collapse
|
23
|
Abstract
Macrophages are critical to organ structure and function in health and disease. To determine mechanisms by which granulocyte/macrophage-colony stimulating factor (GM-CSF) signaling normally maintains surfactant homeostasis and how its disruption causes pulmonary alveolar proteinosis (PAP), we evaluated lipid composition in alveolar macrophages and lung surfactant, macrophage-mediated surfactant clearance kinetics/dynamics, and cholesterol-targeted pharmacotherapy of PAP in vitro and in vivo. Without GM-CSF signaling, surfactant-exposed macrophages massively accumulated cholesterol ester-rich lipid-droplets and surfactant had an increased proportion of cholesterol. GM-CSF regulated cholesterol clearance in macrophages in constitutive, dose-dependent, and reversible fashion but did not affect phospholipid clearance. PPARγ-agonist therapy increased cholesterol clearance in macrophages and reduced disease severity in PAP mice. Results demonstrate that GM-CSF is required for cholesterol clearance in macrophages, identify reduced cholesterol clearance as the primary macrophage defect driving PAP pathogenesis, and support the feasibility of translating pioglitazone as a novel pharmacotherapy of PAP.
Collapse
|
24
|
Fournier N, Sayet G, Vedie B, Nowak M, Allaoui F, Solgadi A, Caudron E, Chaminade P, Benoist JF, Paul JL. Eicosapentaenoic acid membrane incorporation impairs cholesterol efflux from cholesterol-loaded human macrophages by reducing the cholesteryl ester mobilization from lipid droplets. Biochim Biophys Acta Mol Cell Biol Lipids 2017; 1862:1079-1091. [PMID: 28739279 DOI: 10.1016/j.bbalip.2017.07.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Revised: 07/18/2017] [Accepted: 07/20/2017] [Indexed: 12/26/2022]
Abstract
A diet containing a high n-3/n-6 polyunsaturated fatty acids (PUFA) ratio has cardioprotective properties. PUFAs incorporation into membranes influences the function of membrane proteins. We investigated the impact of the membrane incorporation of PUFAs, especially eicosapentaenoic acid (EPA) (C20:5 n-3), on the anti-atherogenic cholesterol efflux pathways. We used cholesteryl esters (CE)-loaded human monocyte-derived macrophages (HMDM) to mimic foam cells exposed to the FAs for a long period of time to ensure their incorporation into cellular membranes. Phospholipid fraction of EPA cells exhibited high levels of EPA and its elongation product docosapentaenoic acid (DPA) (C22:5 n-3), which was associated with a decreased level of arachidonic acid (AA) (C20:4 n-6). EPA 70μM reduced ABCA1-mediated cholesterol efflux to apolipoprotein (apo) AI by 30% without any alteration in ABCA1 expression. The other tested PUFAs, DPA, docosahexaenoic acid (DHA) (C22:6 n-3), and AA, were also able to reduce ABCA1 functionality while the monounsaturated oleic FA slightly decreased efflux and the saturated palmitic FA had no impact. Moreover, EPA also reduced cholesterol efflux to HDL mediated by the Cla-1 and ABCG1 pathways. EPA incorporation did not hinder efflux in free cholesterol-loaded HMDM and did not promote esterification of cholesterol. Conversely, EPA reduced the neutral hydrolysis of cytoplasmic CE by 24%. The reduced CE hydrolysis was likely attributed to the increase in cellular TG contents and/or the decrease in apo E secretion after EPA treatment. In conclusion, EPA membrane incorporation reduces cholesterol efflux in human foam cells by reducing the cholesteryl ester mobilization from lipid droplets.
Collapse
Affiliation(s)
- Natalie Fournier
- Univ Paris Sud-Paris Saclay, EA 7357, Lip(Sys)(2), Athérosclérose: homéostasie et trafic du cholestérol des macrophages (FKA EA 4529), UFR de Pharmacie, 92296 Châtenay-Malabry, France; AP-HP (Assistance Publique-Hôpitaux de Paris), Hôpital Européen Georges Pompidou, Laboratoire de Biochimie, 75015 Paris, France.
| | - Guillaume Sayet
- Univ Paris Sud-Paris Saclay, EA 7357, Lip(Sys)(2), Chimie Analytique Pharmaceutique (FKA EA 4041), UFR de Pharmacie, 92296 Châtenay-Malabry, France
| | - Benoît Vedie
- AP-HP (Assistance Publique-Hôpitaux de Paris), Hôpital Européen Georges Pompidou, Laboratoire de Biochimie, 75015 Paris, France
| | - Maxime Nowak
- Univ Paris Sud-Paris Saclay, EA 7357, Lip(Sys)(2), Athérosclérose: homéostasie et trafic du cholestérol des macrophages (FKA EA 4529), UFR de Pharmacie, 92296 Châtenay-Malabry, France
| | - Fatima Allaoui
- Univ Paris Sud-Paris Saclay, EA 7357, Lip(Sys)(2), Athérosclérose: homéostasie et trafic du cholestérol des macrophages (FKA EA 4529), UFR de Pharmacie, 92296 Châtenay-Malabry, France
| | - Audrey Solgadi
- Univ Paris Sud-Paris Saclay, SFR IPSIT (Institut Paris-Saclay d'Innovation Thérapeutique), UMS IPSIT Service d'Analyse des Médicaments et Métabolites, 92296 Châtenay-Malabry, France
| | - Eric Caudron
- Univ Paris Sud-Paris Saclay, EA 7357, Lip(Sys)(2), Chimie Analytique Pharmaceutique (FKA EA 4041), UFR de Pharmacie, 92296 Châtenay-Malabry, France
| | - Pierre Chaminade
- Univ Paris Sud-Paris Saclay, EA 7357, Lip(Sys)(2), Chimie Analytique Pharmaceutique (FKA EA 4041), UFR de Pharmacie, 92296 Châtenay-Malabry, France
| | - Jean-François Benoist
- AP-HP (Assistance Publique-Hôpitaux de Paris), Hôpital Robert Debré, Laboratoire de Biochimie hormonale, 75019 Paris, France
| | - Jean-Louis Paul
- Univ Paris Sud-Paris Saclay, EA 7357, Lip(Sys)(2), Athérosclérose: homéostasie et trafic du cholestérol des macrophages (FKA EA 4529), UFR de Pharmacie, 92296 Châtenay-Malabry, France; AP-HP (Assistance Publique-Hôpitaux de Paris), Hôpital Européen Georges Pompidou, Laboratoire de Biochimie, 75015 Paris, France
| |
Collapse
|
25
|
Batt KV, Avella M, Moore EH, Jackson B, Suckling KE, Botham KM. Differential Effects of Low-Density Lipoprotein and Chylomicron Remnants on Lipid Accumulation in Human Macrophages. Exp Biol Med (Maywood) 2016; 229:528-37. [PMID: 15169972 DOI: 10.1177/153537020422900611] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
The effects of low-density lipoprotein (LDL) and chylomicron remnants on lipid accumulation in human monocyte–derived macrophages (HMDMs) and in macrophages derived from the human monocyte cell line THP-1 were compared. The HMDMs or THP-1 macrophages were incubated with LDL, oxidized LDL (oxLDL), chylomicron remnant–like particles (CMR-LPs), or oxidized CMR-LPs (oxCMR-LPs), and the amount and type of lipid accumulated were determined. As expected, the lipid content of both cell types was increased markedly by oxLDL but not LDL, and this was due to a rise in cholesterol, cholesteryl ester (CE), and triacylglycerol (TG) levels. In contrast, both CMR-LPs and oxCMR-LPs caused a considerable increase in cellular lipid in HMDMs and THP-1 macrophages, but in this case there was a greater rise in the TG than in the cholesterol or CE content. Lipid accumulation in response to oxLDL, CMR-LPs, and oxCMR-LPs was prevented by the ACAT inhibitor CI976 in HMDMs but not in THP-1 macrophages, where TG levels remained markedly elevated. The rate of incorporation of [3H]oleate into CE and TG in THP-1 macrophages was increased by oxLDL, CMR-LPs, and oxCMR-LPs, but incorporation into TG was increased to a greater extent with CMR-LPs and oxCMR-LPs compared with oxLDL. These results demonstrate that both CMR-LPs and oxCMR-LPs cause lipid accumulation in human macrophages comparable to that seen with oxLDL and that oxidation of the remnant particles does not enhance this effect. They also demonstrate that a greater proportion of the lipid accumulated in response to CMR-LPs compared with oxLDL is TG rather than cholesterol or CE and that this is associated with a higher rate of TG synthesis. This study, therefore, provides further evidence to suggest that chylomicron remnants have a role in foam cell formation that is distinct from that of oxLDL.
Collapse
Affiliation(s)
- Kelly V Batt
- Department of Veterinary Basic Sciences, The Royal Veterinary College, London NW1 0TU, England
| | | | | | | | | | | |
Collapse
|
26
|
Dinnes DLM, White MY, Kockx M, Traini M, Hsieh V, Kim M, Hou L, Jessup W, Rye K, Thaysen‐Andersen M, Cordwell SJ, Kritharides L. Human macrophage cathepsin β‐mediated C‐terminal cleavage of apolipoprotein α‐I at Ser
228
severely impairs antiatherogenic capacity. FASEB J 2016; 30:4239-4255. [DOI: 10.1096/fj.201600508r] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2016] [Accepted: 09/01/2016] [Indexed: 11/11/2022]
Affiliation(s)
| | - Melanie Y. White
- School of Molecular BioscienceDiscipline of Pathology School of Medical Sciences and Charles Perkins Centre University of Sydney Sydney New South Wales Australia
| | - Maaike Kockx
- Atherosclerosis LaboratoryANZAC Research Institute
| | | | - Victar Hsieh
- Department of CardiologySt. George Hospital Sydney New South Wales Australia
| | - Mi‐Jurng Kim
- School of Medical Sciences Sydney New South Wales Australia
| | - Liming Hou
- Lipid Research GroupSchool of Medical Sciences University of New South Wales Sydney New South Wales Australia
| | - Wendy Jessup
- Atherosclerosis LaboratoryANZAC Research Institute
| | - Kerry‐Anne Rye
- Lipid Research GroupSchool of Medical Sciences University of New South Wales Sydney New South Wales Australia
| | - Morten Thaysen‐Andersen
- Department of Chemistry and Biomolecular SciencesFaculty of Science and Engineering Macquarie University Sydney New South Wales Australia
| | - Stuart J. Cordwell
- School of Molecular BioscienceDiscipline of Pathology School of Medical Sciences and Charles Perkins Centre University of Sydney Sydney New South Wales Australia
| | - Leonard Kritharides
- Atherosclerosis LaboratoryANZAC Research Institute
- Department of CardiologyConcord Repatriation General Hospital Sydney New South Wales Australia
| |
Collapse
|
27
|
Versmissen J, Vongpromek R, Yahya R, van der Net JB, van Vark-van der Zee L, Blommesteijn-Touw J, Wattimena D, Rietveld T, Pullinger CR, Christoffersen C, Dahlbäck B, Kane JP, Mulder M, Sijbrands EJG. Familial hypercholesterolaemia: cholesterol efflux and coronary disease. Eur J Clin Invest 2016; 46:643-50. [PMID: 27208892 PMCID: PMC5113689 DOI: 10.1111/eci.12643] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 05/18/2016] [Indexed: 12/13/2022]
Abstract
BACKGROUND Coronary heart disease (CHD) risk inversely associates with levels of high-density lipoprotein cholesterol (HDL-C). The protective effect of HDL is thought to depend on its functionality, such as its ability to induce cholesterol efflux. MATERIALS AND METHODS We compared plasma cholesterol efflux capacity between male familial hypercholesterolaemia (FH) patients with and without CHD relative to their non-FH brothers, and examined HDL constituents including sphingosine-1-phosphate (S1P) and its carrier apolipoprotein M (apoM). RESULTS Seven FH patients were asymptomatic and six had experienced a cardiac event at a mean age of 39 years. Compared to their non-FH brothers, cholesterol efflux from macrophages to plasma from the FH patients without CHD was 16 ± 22% (mean ± SD) higher and to plasma from the FH patients with CHD was 7 ± 8% lower (P = 0·03, CHD vs. non-CHD). Compared to their non-FH brothers, FH patients without CHD displayed significantly higher levels of HDL-cholesterol, HDL-S1P and apoM, while FH patients with CHD displayed lower levels than their non-FH brothers. CONCLUSIONS A higher plasma cholesterol efflux capacity and higher S1P and apoM content of HDL in asymptomatic FH patients may play a role in their apparent protection from premature CHD.
Collapse
Affiliation(s)
- Jorie Versmissen
- Department of Internal Medicine, Section of Pharmacology, Vascular and Metabolic Diseases, Cardiovascular Research School COEUR, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Ranitha Vongpromek
- Department of Internal Medicine, Section of Pharmacology, Vascular and Metabolic Diseases, Cardiovascular Research School COEUR, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Reyhana Yahya
- Department of Internal Medicine, Section of Pharmacology, Vascular and Metabolic Diseases, Cardiovascular Research School COEUR, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Jeroen B van der Net
- Department of Internal Medicine, Section of Pharmacology, Vascular and Metabolic Diseases, Cardiovascular Research School COEUR, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Leonie van Vark-van der Zee
- Department of Internal Medicine, Section of Pharmacology, Vascular and Metabolic Diseases, Cardiovascular Research School COEUR, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Jeannette Blommesteijn-Touw
- Department of Internal Medicine, Section of Pharmacology, Vascular and Metabolic Diseases, Cardiovascular Research School COEUR, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Darcos Wattimena
- Department of Internal Medicine, Section of Pharmacology, Vascular and Metabolic Diseases, Cardiovascular Research School COEUR, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Trinet Rietveld
- Department of Internal Medicine, Section of Pharmacology, Vascular and Metabolic Diseases, Cardiovascular Research School COEUR, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Clive R Pullinger
- Cardiovascular Research Institute, University of California, San Francisco, CA, USA.,Department of Physiological Nursing, University of California, San Francisco, CA, USA
| | | | - Björn Dahlbäck
- Wallenberg Laboratory, Department of Laboratory Medicine, Skån University Hospital, Malmö, Sweden
| | - John P Kane
- Cardiovascular Research Institute, University of California, San Francisco, CA, USA.,Department of Medicine, University of California, San Francisco, CA, USA.,Department of Biochemistry and Biophysics, University of California, San Francisco, CA, USA
| | - Monique Mulder
- Department of Internal Medicine, Section of Pharmacology, Vascular and Metabolic Diseases, Cardiovascular Research School COEUR, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Eric J G Sijbrands
- Department of Internal Medicine, Section of Pharmacology, Vascular and Metabolic Diseases, Cardiovascular Research School COEUR, Erasmus University Medical Center, Rotterdam, the Netherlands
| |
Collapse
|
28
|
Lipid droplet-associated proteins in atherosclerosis (Review). Mol Med Rep 2016; 13:4527-34. [PMID: 27082419 PMCID: PMC4878557 DOI: 10.3892/mmr.2016.5099] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Accepted: 01/29/2016] [Indexed: 01/01/2023] Open
Abstract
Accumulation of atherosclerotic plaques in arterial walls leads to major cardiovascular diseases and stroke. Macrophages/foam cells are central components of atherosclerotic plaques, which populate the arterial wall in order to remove harmful modified low‑density lipoprotein (LDL) particles, resulting in the accumulation of lipids, mostly LDL‑derived cholesterol ester, in cytosolic lipid droplets (LDs). At present, LDs are recognized as dynamic organelles that govern cellular metabolic processes. LDs consist of an inner core of neutral lipids surrounded by a monolayer of phospholipids and free cholesterol, and contain LD‑associated proteins (LDAPs) that regulate LD functions. Foam cells are characterized by an aberrant accumulation of cytosolic LDs, and are considered a hallmark of atherosclerotic lesions through all stages of development. Previous studies have investigated the mechanisms underlying foam cell formation, aiming to discover therapeutic strategies that target foam cells and intervene against atherosclerosis. It is well established that LDAPs have a major role in the pathogenesis of metabolic diseases caused by dysfunction of lipid metabolism, and several studies have linked LDAPs to the development of atherosclerosis. In this review, several foam cell‑targeting pathways have been described, with an emphasis on the role of LDAPs in cholesterol mobilization from macrophages. In addition, the potential of LDAPs as therapeutic targets to prevent the progression and/or facilitate the regression of the disease has been discussed.
Collapse
|
29
|
Eicosapentaenoic acid membrane incorporation impairs ABCA1-dependent cholesterol efflux via a protein kinase A signaling pathway in primary human macrophages. Biochim Biophys Acta Mol Cell Biol Lipids 2016; 1861:331-41. [DOI: 10.1016/j.bbalip.2016.01.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Revised: 01/04/2016] [Accepted: 01/07/2016] [Indexed: 11/22/2022]
|
30
|
Fernandez-Ruiz I, Puchalska P, Narasimhulu CA, Sengupta B, Parthasarathy S. Differential lipid metabolism in monocytes and macrophages: influence of cholesterol loading. J Lipid Res 2016; 57:574-86. [PMID: 26839333 DOI: 10.1194/jlr.m062752] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Indexed: 11/20/2022] Open
Abstract
The influence of the hypercholesterolemia associated with atherosclerosis on monocytes is poorly understood. Monocytes are exposed to high concentrations of lipids, particularly cholesterol and lysophosphatidylcholine (lyso-PC). Indeed, in line with recent reports, we found that monocytes accumulate cholesteryl esters (CEs) in hypercholesterolemic mice, demonstrating the need for studies that analyze the effects of lipid accumulation on monocytes. Here we analyze the effects of cholesterol and lyso-PC loading in human monocytes and macrophages. We found that cholesterol acyltransferase and CE hydrolase activities are lower in monocytes. Monocytes also showed a different expression profile of cholesterol influx and efflux genes in response to lipid loading and a different pattern of lyso-PC metabolism. In monocytes, increased levels of CE slowed the conversion of lyso-PC into PC. Interestingly, although macrophages accumulated glycerophosphocholine, phosphocholine was the main water-soluble choline metabolite being generated in monocytes, suggesting a role for mono- and diacylglycerol in the chemoattractability of these cells. In summary, monocytes and macrophages show significant differences in lipid metabolism and gene expression profiles in response to lipid loading. These findings provide new insights into the mechanisms of atherosclerosis and suggest potentials for targeting monocyte chemotactic properties not only in atherosclerosis but also in other diseases.
Collapse
Affiliation(s)
- Irene Fernandez-Ruiz
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827
| | - Patrycja Puchalska
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827
| | | | - Bhaswati Sengupta
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827
| | - Sampath Parthasarathy
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827
| |
Collapse
|
31
|
Affiliation(s)
- Hanrui Zhang
- From the Department of Medicine, Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia.
| | - Muredach P Reilly
- From the Department of Medicine, Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia.
| |
Collapse
|
32
|
Hafiane A, Bielicki JK, Johansson JO, Genest J. Novel Apo E-Derived ABCA1 Agonist Peptide (CS-6253) Promotes Reverse Cholesterol Transport and Induces Formation of preβ-1 HDL In Vitro. PLoS One 2015. [PMID: 26207756 PMCID: PMC4514675 DOI: 10.1371/journal.pone.0131997] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Apolipoprotein (apo) mimetic peptides replicate some aspects of HDL function. We have previously reported the effects of compound ATI-5261 on its ability to replicate many functions of native apo A-I in the process of HDL biogenesis. ATI-5261 induced muscle toxicity in wild type C57Bl/6 mice, increased CPK, ALT and AST and increase in triglyceride (Tg) levels. Aromatic phenylalanine residues on the non-polar face of ATI-5261, together with positively charged arginine residues at the lipid-water interface were responsible for these effects. This information was used to create a novel analog (CS-6253) that was non-toxic. We evaluated this peptide designed from the carboxyl terminus of apo E, in its ability to mimic apo A-I functionality. Our data shows that the lipidated particles generated by incubating cells overexpressing ABCA1 with lipid free CS-6253 enhances the rate of ABCA1 lipid efflux with high affinity interactions with native ABCA1 oligomeric forms and plasma membrane micro-domains. Interaction between ABCA1 and lipid free CS-6253 resulted in formation of nascent HDL-CS-6253 particles that are actively remodeled in plasma. Mature HDL-CS-6253 particles deliver cholesterol to liver cells via SR-BI in-vitro. CS-6253 significantly increases cholesterol efflux in murine macrophages and in human THP-1 macrophage-derived foam cells expressing ABCA1. Addition of CS-6253 to plasma dose-dependently displaced apo A-I from α-HDL particles and led to de novo formation of preβ-1 HDL that stimulates ABCA1 dependent cholesterol efflux efficiently. When incubated with human plasma CS-6253 was also found to bind with HDL and LDL and promoted the transfer of cholesterol from HDL to LDL predominantly. Our data shows that CS-6253 mimics apo A-I in its ability to promote ABCA1-mediated formation of nascent HDL particles, and enhances formation of preβ-1 HDL with increase in the cycling of apo A-I between the preβ and α-HDL particles in-vitro. These mechanisms are potentially anti-atherogenic.
Collapse
Affiliation(s)
- Anouar Hafiane
- Cardiovascular Research Laboratories Laboratory, Research Institute of the McGill University Health Centre, Montréal, Québec H4A 3J1, Canada
| | - John K. Bielicki
- Lawrence Berkeley National Laboratory, Donner Laboratory, MS1-267, Berkeley, CA, United States of America
| | | | - Jacques Genest
- Cardiovascular Research Laboratories Laboratory, Research Institute of the McGill University Health Centre, Montréal, Québec H4A 3J1, Canada
- * E-mail:
| |
Collapse
|
33
|
Retinoic acid induces macrophage cholesterol efflux and inhibits atherosclerotic plaque formation in apoE-deficient mice. Br J Nutr 2015. [PMID: 26201974 DOI: 10.1017/s0007114515002159] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
It has been suggested that retinoic acid (RA) has a potential role in the prevention of atherosclerotic CVD. In the present study, we used J774A.1 cell lines and primary peritoneal macrophages to investigate the protective effects of RA on foam cell formation and atherogenesis in apoE-deficient (apoE- / -) mice. A total of twenty male apoE- / - mice (n 10 animals per group), aged 8 weeks, were fed on a high-fat diet (HFD) and treated with vehicle or 9-cis-RA for 8 weeks. The atherosclerotic plaque area in the aortic sinus of mice in the 9-cis-RA group was 40·7 % less than that of mice in the control group (P< 0·01). Mouse peritoneal macrophages from the 9-cis-RA group had higher protein expression levels of ATP-binding cassette transporter A1 (ABCA1) and G1 (ABCG1) than those from the control group. Serum total and LDL-cholesterol concentrations were lower in the 9-cis-RA group than in the control group (P< 0·05). In vitro studies showed that incubation of cholesterol-loaded J774A.1 macrophages with 9-cis-RA (0·1, 1 and 10 μmol/l) induced cholesterol efflux in a dose-dependent manner. The 9-cis-RA treatment markedly attenuated lipid accumulation in macrophages exposed to oxidised LDL. Moreover, treatment with 9-cis-RA significantly increased the protein expression levels of ABCA1 and ABCG1 in J774A.1 macrophages in a dose-dependent manner. Furthermore, 9-cis-RA dose-dependently enhanced the protein expression level of liver X receptor-α (LXRα), the upstream regulator of ABCA1 and ABCG1. Taken together, the present results show that 9-cis-RA suppresses foam cell formation and prevents HFD-induced atherogenesis via the LXRα-dependent up-regulation of ABCA1 and ABCG1.
Collapse
|
34
|
Ross MK, Borazjani A, Mangum LC, Wang R, Crow JA. Effects of toxicologically relevant xenobiotics and the lipid-derived electrophile 4-hydroxynonenal on macrophage cholesterol efflux: silencing carboxylesterase 1 has paradoxical effects on cholesterol uptake and efflux. Chem Res Toxicol 2014; 27:1743-56. [PMID: 25250848 PMCID: PMC4203386 DOI: 10.1021/tx500221a] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
![]()
Cholesterol
cycles between free cholesterol (unesterified) found
predominantly in membranes and cholesteryl esters (CEs) stored in
cytoplasmic lipid droplets. Only free cholesterol is effluxed from
macrophages via ATP-binding cassette (ABC) transporters to extracellular
acceptors. Carboxylesterase 1 (CES1), proposed to hydrolyze CEs, is
inactivated by oxon metabolites of organophosphorus pesticides and
by the lipid electrophile 4-hydroxynonenal (HNE). We assessed the
ability of these compounds to reduce cholesterol efflux from foam
cells. Human THP-1 macrophages were loaded with [3H]-cholesterol/acetylated
LDL and then allowed to equilibrate to enable [3H]-cholesterol
to distribute into its various cellular pools. The cholesterol-engorged
cells were then treated with toxicants in the absence of cholesterol
acceptors for 24 h, followed by a 24 h efflux period in the presence
of toxicant. A concentration-dependent reduction in [3H]-cholesterol
efflux via ABCA1 (up to 50%) was found for paraoxon (0.1–10
μM), whereas treatment with HNE had no effect. A modest reduction
in [3H]-cholesterol efflux via ABCG1 (25%) was found after
treatment with either paraoxon or chlorpyrifos oxon (10 μM each)
but not HNE. No difference in efflux rates was found after treatments
with either paraoxon or HNE when the universal cholesterol acceptor
10% (v/v) fetal bovine serum was used. When the re-esterification
arm of the CE cycle was disabled in foam cells, paraoxon treatment
increased CE levels, suggesting the neutral CE hydrolysis arm of the
cycle had been inhibited by the toxicant. However, paraoxon also partially
inhibited lysosomal acid lipase, which generates cholesterol for efflux,
and reduced the expression of ABCA1 protein. Paradoxically, silencing CES1 expression in macrophages did not affect the percent
of [3H]-cholesterol efflux. However, CES1 mRNA knockdown markedly reduced cholesterol uptake by macrophages,
with SR-A and CD36 mRNA reduced
3- and 4-fold, respectively. Immunoblots confirmed SR-A and CD36 protein
downregulation. Together, these results suggest that toxicants, e.g.,
oxons, may interfere with macrophage cholesterol homeostasis/metabolism.
Collapse
Affiliation(s)
- Matthew K Ross
- Department of Basic Sciences, Center for Environmental Health Sciences, College of Veterinary Medicine, Mississippi State University , P.O. Box 6100, Mississippi State, Mississippi 39762, United States
| | | | | | | | | |
Collapse
|
35
|
Sakai K, Igarashi M, Yamamuro D, Ohshiro T, Nagashima S, Takahashi M, Enkhtuvshin B, Sekiya M, Okazaki H, Osuga JI, Ishibashi S. Critical role of neutral cholesteryl ester hydrolase 1 in cholesteryl ester hydrolysis in murine macrophages. J Lipid Res 2014; 55:2033-40. [PMID: 24868095 DOI: 10.1194/jlr.m047787] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Hydrolysis of intracellular cholesteryl ester (CE) is the rate-limiting step in the efflux of cholesterol from macrophage foam cells. In mouse peritoneal macrophages (MPMs), this process is thought to involve several enzymes: hormone-sensitive lipase (Lipe), carboxylesterase 3 (Ces3), neutral CE hydrolase 1 (Nceh1). However, there is some disagreement over the relative contributions of these enzymes. To solve this problem, we first compared the abilities of several compounds to inhibit the hydrolysis of CE in cells overexpressing Lipe, Ces3, or Nceh1. Cells overexpressing Ces3 had negligible neutral CE hydrolase activity. We next examined the effects of these inhibitors on the hydrolysis of CE and subsequent cholesterol trafficking in MPMs. CE accumulation was increased by a selective inhibitor of Nceh1, paraoxon, and two nonselective inhibitors of Nceh1, (+)-AS115 and (-)-AS115, but not by two Lipe-selective inhibitors, orlistat and 76-0079. Paraoxon inhibited cholesterol efflux to apoA-I or HDL, while 76-0079 did not. These results suggest that Nceh1 plays a dominant role over Lipe in the hydrolysis of CE and subsequent cholesterol efflux in MPMs.
Collapse
Affiliation(s)
- Kent Sakai
- Division of Endocrinology and Metabolism, Department of Medicine, Jichi Medical University, Tochigi 329-0498, Japan
| | - Masaki Igarashi
- Department of Metabolic Diseases, Graduate School of Medicine, University of Tokyo, Tokyo 113-8655 Japan
| | - Daisuke Yamamuro
- Division of Endocrinology and Metabolism, Department of Medicine, Jichi Medical University, Tochigi 329-0498, Japan
| | - Taichi Ohshiro
- Division of Endocrinology and Metabolism, Department of Medicine, Jichi Medical University, Tochigi 329-0498, Japan
| | - Shuichi Nagashima
- Division of Endocrinology and Metabolism, Department of Medicine, Jichi Medical University, Tochigi 329-0498, Japan
| | - Manabu Takahashi
- Division of Endocrinology and Metabolism, Department of Medicine, Jichi Medical University, Tochigi 329-0498, Japan
| | - Bolormaa Enkhtuvshin
- Division of Endocrinology and Metabolism, Department of Medicine, Jichi Medical University, Tochigi 329-0498, Japan
| | - Motohiro Sekiya
- Department of Metabolic Diseases, Graduate School of Medicine, University of Tokyo, Tokyo 113-8655 Japan
| | - Hiroaki Okazaki
- Department of Metabolic Diseases, Graduate School of Medicine, University of Tokyo, Tokyo 113-8655 Japan
| | - Jun-ichi Osuga
- Division of Endocrinology and Metabolism, Department of Medicine, Jichi Medical University, Tochigi 329-0498, Japan
| | - Shun Ishibashi
- Division of Endocrinology and Metabolism, Department of Medicine, Jichi Medical University, Tochigi 329-0498, Japan
| |
Collapse
|
36
|
Amézaga N, Sanjurjo L, Julve J, Aran G, Pérez-Cabezas B, Bastos-Amador P, Armengol C, Vilella R, Escolà-Gil JC, Blanco-Vaca F, Borràs FE, Valledor AF, Sarrias MR. Human scavenger protein AIM increases foam cell formation and CD36-mediated oxLDL uptake. J Leukoc Biol 2013; 95:509-20. [PMID: 24295828 DOI: 10.1189/jlb.1212660] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
AIM is expressed by macrophages in response to agonists of the nuclear receptors LXR/RXR. In mice, it acts as an atherogenic factor by protecting macrophages from the apoptotic effects of oxidized lipids. In humans, it is detected in atherosclerotic lesions, but no role related to atherosclerosis has been reported. This study aimed to investigate whether the role of hAIM extends beyond inhibiting oxidized lipid-induced apoptosis. To accomplish this goal, functional analysis with human monocytic THP1 cells and macrophages differentiated from peripheral blood monocytes were performed. It was found that hAIM reduced oxLDL-induced macrophage apoptosis and increased macrophage adhesion to endothelial ICAM-1 by enhancing LFA-1 expression. Furthermore, hAIM increased foam cell formation, as shown by Oil Red O and Nile Red staining, as well as quantification of cholesterol content. This was not a result of decreased reverse cholesterol transport, as hAIM did not affect the efflux significantly from [(3)H] Cholesterol-laden macrophages driven by plasma, apoA-I, or HDL2 acceptors. Rather, flow cytometry studies indicated that hAIM increased macrophage endocytosis of fluorescent oxLDL, which correlated with an increase in the expression of the oxLDLR CD36. Moreover, hAIM bound to oxLDL in ELISA and enhanced the capacity of HEK-293 cells expressing CD36 to endocytose oxLDL, as studied using immunofluorescence microscopy, suggesting that hAIM serves to facilitate CD36-mediated uptake of oxLDL. Our data represent the first evidence that hAIM is involved in macrophage survival, adhesion, and foam cell formation and suggest a significant contribution to atherosclerosis-related mechanisms in the macrophage.
Collapse
Affiliation(s)
- Núria Amézaga
- 1.Ctra Can Ruti, camí de les escoles s/n, Edifici de Recerca, Planta 1, 08916 Badalona, Spain. ; Twitter: http://www.twitter.com/mrsarrias
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Sengupta B, Narasimhulu CA, Parthasarathy S. Novel technique for generating macrophage foam cells for in vitro reverse cholesterol transport studies. J Lipid Res 2013; 54:3358-72. [PMID: 24115226 PMCID: PMC3826683 DOI: 10.1194/jlr.m041327] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Generation of foam cells, an essential step for reverse cholesterol transport studies, uses the technique of receptor-dependent macrophage loading with radiolabeled acetylated LDL. In this study, we used the ability of a biologically relevant detergent molecule, lysophosphatidylcholine (lyso-PtdCho), to form mixed micelles with cholesterol or cholesteryl ester (CE) to generate macrophage foam cells. Fluorescent or radiolabeled cholesterol/lyso-PtdCho mixed micelles were prepared and incubated with RAW 264.7 or mouse peritoneal macrophages. Results showed that such micelles were quite stable at 4°C and retained the solubilized cholesterol during one month of storage. Macrophages incubated with cholesterol or CE (unlabeled, fluorescently labeled, or radiolabeled)/lyso-PtdCho mixed micelles accumulated CE as documented by microscopy, lipid staining, labeled oleate incorporation, and by TLC. Such foam cells unloaded cholesterol when incubated with HDL but not with oxidized HDL. We propose that stable cholesterol or CE/lyso-PtdCho micelles would offer advantages over existing methods.
Collapse
Affiliation(s)
- Bhaswati Sengupta
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827
| | | | | |
Collapse
|
38
|
Chai JT, Digby JE, Ruparelia N, Jefferson A, Handa A, Choudhury RP. Nicotinic acid receptor GPR109A is down-regulated in human macrophage-derived foam cells. PLoS One 2013; 8:e62934. [PMID: 23658787 PMCID: PMC3642175 DOI: 10.1371/journal.pone.0062934] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2013] [Accepted: 03/27/2013] [Indexed: 02/07/2023] Open
Abstract
Nicotinic acid (NA) regresses atherosclerosis in human imaging studies and reduces atherosclerosis in mice, mediated by myeloid cells, independent of lipoproteins. Since GPR109A is expressed by human monocytes, we hypothesized that NA may drive cholesterol efflux from foam cells. In THP-1 cells NA suppressed LPS-induced mRNA transcription of MCP-1 by 76.6±12.2% (P<0.01) and TNFα by 56.1±11.5% (P<0.01), yet restored LPS-induced suppression of PPARγ transcription by 536.5±46.4% (P<0.001) and its downstream effector CD36 by 116.8±19.8% (P<0.01). Whilst direct PPARγ-agonism promoted cholesterol efflux from THP-1 derived foam cells by 37.7±3.1% (P<0.01) and stimulated transcription of LXRα by 87.9±9.5% (P<0.001) and ABCG1 by 101.2±15.5% (P<0.01), NA showed no effect in foam cells on either cholesterol efflux or key RCT genes transcription. Upon foam cell induction, NA lost its effect on PPARγ and cAMP pathways, since its receptor, GPR109A, was down-regulated by foam cell transformation. This observation was confirmed in explanted human carotid plaques. In conclusion, despite NA's anti-inflammatory effect on human macrophages, it has no effect on foam cells in reverse cholesterol transport; due to GPR109A down-regulation.
Collapse
Affiliation(s)
- Joshua T. Chai
- Division of Cardiovascular Medicine, University of Oxford, Oxford, United Kingdom
| | - Janet E. Digby
- Division of Cardiovascular Medicine, University of Oxford, Oxford, United Kingdom
| | - Neil Ruparelia
- Division of Cardiovascular Medicine, University of Oxford, Oxford, United Kingdom
| | - Andrew Jefferson
- Division of Cardiovascular Medicine, University of Oxford, Oxford, United Kingdom
| | - Ashok Handa
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, United Kingdom
| | - Robin P. Choudhury
- Division of Cardiovascular Medicine, University of Oxford, Oxford, United Kingdom
- * E-mail:
| |
Collapse
|
39
|
White CR, Datta G, Buck AKW, Chaddha M, Reddy G, Wilson L, Palgunachari MN, Abbasi M, Anantharamaiah GM. Preservation of biological function despite oxidative modification of the apolipoprotein A-I mimetic peptide 4F. J Lipid Res 2012; 53:1576-87. [PMID: 22589558 DOI: 10.1194/jlr.m026278] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Myeloperoxidase (MPO)-derived hypochlorous acid induces changes in HDL function via redox modifications at the level of apolipoprotein A-I (apoA-I). As 4F and apoA-I share structural and functional properties, we tested the hypothesis that 4F acts as a reactive substrate for hypochlorous acid (HOCl). 4F reduced the HOCl-mediated oxidation of the fluorescent substrate APF in a concentration-dependent manner (ED(50) ∼ 56 ± 3 μM). This reaction induced changes in the physical properties of 4F. Addition of HOCl to 4F at molar ratios ranging from 1:1 to 3:1 reduced 4F band intensity on SDS-PAGE gels and was accompanied by the formation of a higher molecular weight species. Chromatographic studies showed a reduction in 4F peak area with increasing HOCl and the formation of new products. Mass spectral analyses of collected fractions revealed oxidation of the sole tryptophan (Trp) residue in 4F. 4F was equally susceptible to oxidation in the lipid-free and lipid-bound states. To determine whether Trp oxidation influenced its apoA-I mimetic properties, we monitored effects of HOCl on 4F-mediated lipid binding and ABCA1-dependent cholesterol efflux. Neither property was altered by HOCl. These results suggest that 4F serves as a reactive substrate for HOCl, an antioxidant response that does not influence the lipid binding and cholesterol effluxing capacities of the peptide.
Collapse
Affiliation(s)
- C Roger White
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Fournier N, Attia N, Rousseau-Ralliard D, Vedie B, Destaillats F, Grynberg A, Paul JL. Deleterious impact of elaidic fatty acid on ABCA1-mediated cholesterol efflux from mouse and human macrophages. Biochim Biophys Acta Mol Cell Biol Lipids 2011; 1821:303-12. [PMID: 22074701 DOI: 10.1016/j.bbalip.2011.10.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2011] [Revised: 09/07/2011] [Accepted: 10/05/2011] [Indexed: 10/16/2022]
Abstract
Consumption of trans fatty acids (TFA) increase cardiovascular risk more than do saturated FA, but the mechanisms explaining their atherogenicity are still unclear. We investigated the impact of membrane incorporation of TFA on cholesterol efflux by exposing J774 mouse macrophages or human monocyte-derived macrophages (HMDM) to media enriched or not (standard medium) with industrially produced elaidic (trans-9 18:1) acid, naturally produced vaccenic (trans-11 18:1) acid (34 h, 70 μM) or palmitic acid. In J774 macrophages, elaidic and palmitic acid, but not vaccenic acid, reduced ABCA1-mediated efflux by ~23% without affecting aqueous diffusion, SR-BI or ABCG1-mediated pathways, and this effect was maintained in cholesterol-loaded cells. The impact of elaidic acid on the ABCA1 pathway was weaker in cholesterol-normal HMDM, but elaidic acid induced a strong reduction of ABCA1-mediated efflux in cholesterol-loaded cells (-36%). In J774 cells, the FA supplies had no impact on cellular free cholesterol or cholesteryl ester masses, the abundance of ABCA1 mRNA or the total and plasma membrane ABCA1 protein content. Conversely, TFA or palmitic acid incorporation induced strong modifications of the membrane FA composition with a decrease in the ratio of (cis-monounsaturated FA+polyunsaturated FA):(saturated FA+TFA), with elaidic and vaccenic acids representing each 20% and 13% of the total FA composition, respectively. Moreover, we demonstrated that cellular ATP was required for the effect of elaidic acid, suggesting that it contributes to atherogenesis by impairing ABCA1-mediated cholesterol efflux in macrophages, likely by decreasing the membrane fluidity, which could thereby reduce ATPase activity and the function of the transporter.
Collapse
Affiliation(s)
- Natalie Fournier
- Univ Paris-Sud, EA 4529, UFR de Pharmacie, 92296 Châtenay-Malabry, France.
| | | | | | | | | | | | | |
Collapse
|
41
|
Qin Z. The use of THP-1 cells as a model for mimicking the function and regulation of monocytes and macrophages in the vasculature. Atherosclerosis 2011; 221:2-11. [PMID: 21978918 DOI: 10.1016/j.atherosclerosis.2011.09.003] [Citation(s) in RCA: 276] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2011] [Revised: 08/16/2011] [Accepted: 09/01/2011] [Indexed: 10/17/2022]
Abstract
Since their establishment thirty years ago, THP-1 cells have become one of most widely used cell lines to investigate the function and regulation of monocytes and macrophages in the cardiovascular system. However, because this cell line was derived from the blood of a patient with acute monocytic leukemia, the extent to which THP-1 cells mimic monocytes and macrophages in the vasculature is not entirely known. This article serves as a meaningful attempt to address this question by reviewing the recent publications. The interactions between THP-1 cells and various vascular cells (such as endothelial cells, smooth muscle cells, adipocytes, and T cells) provide insight into the roles of the interconnection of monocytes-macrophages with other vascular cells during vascular inflammation, particularly atherogenesis and obesity. Transcriptome, microRNA profile, and histone modifications of THP-1 cells shed new light on the regulatory mechanism of the monocytes-macrophages in response to various inflammatory mediators, such as oxidized low density lipoprotein, lipopolysaccharide, and glucose. These studies hint that under certain defined conditions, THP-1 cells not only resemble primary monocytes-macrophages isolated from healthy donors or donors with disease, such as diabetes mellitus, but also mimic the in situ alteration of macrophages in the adipose tissue of obese subjects and in atherosclerotic lesions. A potential trajectory is to use this cell line to study the novel molecular mechanisms in monocytes and macrophages in relation to the physiology and pathophysiology of the cardiovascular system, however, the conclusion of studies employing THP-1 cells requires further verification using primary cells and/or in vivo models to be generalized to monocytes and macrophages.
Collapse
Affiliation(s)
- Zhenyu Qin
- Division of Vascular Surgery, Department of Surgery, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, United States.
| |
Collapse
|
42
|
Type II interleukin-1 receptor expression is reduced in monocytes/macrophages and atherosclerotic lesions. Biochim Biophys Acta Mol Cell Biol Lipids 2011; 1811:556-63. [DOI: 10.1016/j.bbalip.2011.05.014] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2010] [Revised: 05/02/2011] [Accepted: 05/31/2011] [Indexed: 11/19/2022]
|
43
|
Maranghi M, Hiukka A, Badeau R, Sundvall J, Jauhiainen M, Taskinen MR. Macrophage cholesterol efflux to plasma and HDL in subjects with low and high homocysteine levels: a FIELD substudy. Atherosclerosis 2011; 219:259-65. [PMID: 21696738 DOI: 10.1016/j.atherosclerosis.2011.05.018] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2010] [Revised: 04/29/2011] [Accepted: 05/16/2011] [Indexed: 11/30/2022]
Abstract
OBJECTIVES Increases of homocysteine (Hcy) by fenofibrate correlated inversely to changes in HDL-C and apoA-I in the FIELD study. This finding raised the question whether high Hcy may influence HDL function and counteract benefits of fenofibrate on cardiovascular outcomes. In a subset of the FIELD study we investigated whether fenofibrate therapy or high Hcy, separately or in concert, modulate: (1) ability of plasma or HDL to facilitate cholesterol efflux from THP-1 foam cells; (2) plasma potential to generate preβ-HDL; (3) plasma phospholipid transfer protein (PLTP) activity, serum PON-1 mass and activity, HDL particle size and distribution. METHODS We selected 33 subjects in the FIELD fenofibrate arm according to quartiles of Hcy at 5th year: 17 subjects were in the lowest (Low Hcy group) and 16 subjects were in the highest quartile (High Hcy group). In addition, 14 subjects allocated to placebo were matched by close-out Hcy levels to Low Hcy group. This design allowed us to examine the effects of both fenofibrate (comparison between placebo vs Low Hcy groups) and Hcy (comparison between close-out Low and High Hcy groups) on plasma and HDL ability to facilitate cellular cholesterol removal in the efflux assay in vitro using THP-1 foam cells. RESULTS Hcy levels were 13.3±0.7 μmol/L (placebo), 13.2±2 μmol/L (Low Hcy) and 27.4±6.5 μmol/L (High Hcy). Cholesterol efflux values to HDL and plasma, percentage of plasma preβ-HDL, PLTP activity, serum PON-1 mass and HDL particle size and distribution were similar in both fenofibrate groups and comparable to those of the placebo group. CONCLUSIONS In the present study cohort fenofibrate and high Hcy levels did not modulate HDL and plasma functions in the first step of reverse cholesterol transport, cholesterol efflux from foam cells.
Collapse
Affiliation(s)
- Marianna Maranghi
- Helsinki University Central Hospital, Biomedicum, Haartmaninkatu 8 PO Box 700, FIN-00029 Helsinki, Finland.
| | | | | | | | | | | |
Collapse
|
44
|
Abstract
Cholesterol-engorged macrophage foam cells are a critical component of the atherosclerotic lesion. Reducing the sterol deposits in lesions reduces clinical events. Sterol accumulations within lysosomes have proven to be particularly hard to mobilize out of foam cells. Moreover, excess sterol accumulation in lysosomes has untoward effects, including a complete disruption of lysosome function. Recently, we demonstrated that treatment of sterol-engorged macrophages in culture with triglyceride-containing particles can reverse many of the effects of cholesterol on lysosomes and dramatically reduce the sterol burden in these cells. This article describes what is known about lysosomal sterol engorgement, discusses the possible mechanisms by which triglyceride could produce its effects, and evaluates the possible positive and negative effects of reducing the lysosomal cholesterol levels in foam cells.
Collapse
Affiliation(s)
- W Gray Jerome
- Department of Pathology, U-2206 Medical Center North Vanderbilt University School of Medicine 1161 21st Avenue, South Nashville, TN 37232-32561, USA, Tel.: +1 615 322 5530
| |
Collapse
|
45
|
Vidyashankar S, Godavarthi A, Varma RS, Nandakumar KS. Water-soluble compounds in the herbal preparation Abana inhibit lipid biosynthesis and enhance cholesterol efflux in HepG2 cells. Can J Physiol Pharmacol 2010; 88:456-64. [PMID: 20555414 DOI: 10.1139/y10-021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Higher concentrations of circulating lipids (cholesterol and triglycerides) and their decreased catabolism pose a major risk in the development of atherosclerosis and coronary heart disease (CHD). Although statins are widely used for treatment of hyperlipidemia, side effects associated with their use have prompted the search for a safer alternative for treating hyperlipidemia. The present study investigated the effect of water-soluble compounds in Abana (WSCA), a polyherbal drug formulation traditionally used in India for the treatment of hyperlipidemia, on lipid metabolism in HepG2 cells. WSCA reduced cholesterol and triglyceride content in the cells and their supernatant. WSCA inhibited the incorporation of [2-14C]acetate into cellular cholesterol and fatty acids, suggesting the inhibition of lipid synthesis. In addition, WSCA inhibited HMG-CoA reductase, a key metabolic enzyme involved in the biosynthesis of cholesterol. WSCA also increased cholesterol and fatty acid secretion into the cell supernatant, suggesting the enhanced removal of cholesterol and fatty acids. Furthermore, WSCA showed decreased linoleic acid (18:2) and arachidonic acid (20:4) content in HepG2 cells. The present study is the first to show that WSCA simultaneously inhibited cellular cholesterol biosynthesis and increased cholesterol secretion into the cell supernatant in HepG2 cells.
Collapse
Affiliation(s)
- Satyakumar Vidyashankar
- Department of Biochemistry and Immunology, Research and Development, The Himalaya Drug Company, Makali, Bangalore 562 123, India.
| | | | | | | |
Collapse
|
46
|
Bencharif K, Hoareau L, Murumalla RK, Tarnus E, Tallet F, Clerc RG, Gardes C, Cesari M, Roche R. Effect of apoA-I on cholesterol release and apoE secretion in human mature adipocytes. Lipids Health Dis 2010; 9:75. [PMID: 20642861 PMCID: PMC2917427 DOI: 10.1186/1476-511x-9-75] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2010] [Accepted: 07/20/2010] [Indexed: 11/10/2022] Open
Abstract
Background The risk of cardiovascular disease is inversely correlated to level of plasma HDL-c. Moreover, reverse cholesterol transport (RCT) from peripheral tissues to the liver is the most widely accepted mechanism linked to the anti-atherosclerotic activity of HDL. The apolipoprotein A-I (apoA-I) and the ABC transporters play a key role in this process. Adipose tissue constitutes the body's largest pool of free cholesterol. The adipose cell could therefore be regarded as a key factor in cholesterol homeostasis. The present study investigates the capacity of primary cultures of mature human adipocytes to release cholesterol and explores the relationships between apoA-I, ABCA1, and apoE as well as the signaling pathways that could be potentially involved. Results We demonstrate that apoA-I induces a strong increase in cholesterol release and apoE secretion from adipocytes, whereas it has no transcriptional effect on ABCA1 or apoE genes. Furthermore, brefeldin A (BFA), an intracellular trafficking inhibitor, reduces basal cholesterol and apoE secretion, but does not modify induction by apoA-I. The use of statins also demonstrates that apoA-I stimulated cholesterol release is independent of HMG-CoA reductase activation. Conclusion Our work highlights the fact that adipose tissue, and particularly adipocytes, may largely contribute to RCT via a mechanism specifically regulated within these cells. This further supports the argument that adipose tissue must be regarded as a major factor in the development of cardiovascular diseases, in particular atherosclerosis.
Collapse
Affiliation(s)
- Karima Bencharif
- LBGM-GEICO, Laboratoire de Biochimie et de Génétique Moléculaire - Groupe d'Etude sur l'Inflammation Chronique et l'Obésité, Plateforme CYROI, Université de La Réunion 15 avenue René Cassin 97715 Saint Denis Messag Cedex 9, France
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Guillaumot P, Luquain C, Malek M, Huber AL, Brugière S, Garin J, Grunwald D, Régnier D, Pétrilli V, Lefai E, Manié SN. Pdro, a protein associated with late endosomes and lysosomes and implicated in cellular cholesterol homeostasis. PLoS One 2010; 5:e10977. [PMID: 20544018 PMCID: PMC2882324 DOI: 10.1371/journal.pone.0010977] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2009] [Accepted: 05/13/2010] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Cellular cholesterol is a vital component of the cell membrane. Its concentration is tightly controlled by mechanisms that remain only partially characterized. In this study, we describe a late endosome/lysosomes-associated protein whose expression level affects cellular free cholesterol content. METHODOLOGY/PRINCIPAL FINDINGS Using a restricted proteomic analysis of detergent-resistant membranes (DRMs), we have identified a protein encoded by gene C11orf59. It is mainly localized to late endosome/lysosome (LE/LY) compartment through N-terminal myristoylation and palmitoylation. We named it Pdro for protein associated with DRMs and endosomes. Very recently, three studies have reported on the same protein under two other names: the human p27RF-Rho that regulates RhoA activation and actin dynamics, and its rodent orthologue p18 that controls both LE/LY dynamics through the MERK-ERK pathway and the lysosomal activation of mammalian target of rapamycin complex 1 by amino acids. We found that, consistent with the presence of sterol-responsive element consensus sequences in the promoter region of C11orf59, Pdro mRNA and protein expression levels are regulated positively by cellular cholesterol depletion and negatively by cellular cholesterol loading. Conversely, Pdro is involved in the regulation of cholesterol homeostasis, since its depletion by siRNA increases cellular free cholesterol content that is accompanied by an increased cholesterol efflux from cells. On the other hand, cells stably overexpressing Pdro display reduced cellular free cholesterol content. Pdro depletion-mediated excess cholesterol results, at least in part, from a stimulated low-density lipoprotein (LDL) uptake and an increased cholesterol egress from LE/LY. CONCLUSIONS/SIGNIFICANCE LDL-derived cholesterol release involves LE/LY motility that is linked to actin dynamics. Because Pdro regulates these two processes, we propose that modulation of Pdro expression in response to sterol levels regulates LDL-derived cholesterol through both LDL uptake and LE/LY dynamics, to ultimately control free cholesterol homeostasis.
Collapse
Affiliation(s)
- Patricia Guillaumot
- Génétique Moléculaire, Signalisation et Cancer, UMR 5201 CNRS, Centre Leon Berard, Lyon, France
| | - Céline Luquain
- Regulation Métabolique, Nutrition et Diabète, UMR 870 INSERM/Insa-Lyon, Villeurbanne, France
| | - Mouhannad Malek
- Génétique Moléculaire, Signalisation et Cancer, UMR 5201 CNRS, Centre Leon Berard, Lyon, France
| | - Anne-Laure Huber
- Génétique Moléculaire, Signalisation et Cancer, UMR 5201 CNRS, Centre Leon Berard, Lyon, France
| | - Sabine Brugière
- Laboratoire de Chimie des Protéines, ERM 201 INSERM/CEA/UJF, CEA/Grenoble, Grenoble, France
| | - Jérome Garin
- Laboratoire de Chimie des Protéines, ERM 201 INSERM/CEA/UJF, CEA/Grenoble, Grenoble, France
| | - Didier Grunwald
- Laboratoire Transduction de Signal, Unité 873, INSERM/CEA/DSV, Institut de Recherches en Technologies et Sciences pour le Vivant, Grenoble, France
| | - Daniel Régnier
- Génétique Moléculaire, Signalisation et Cancer, UMR 5201 CNRS, Centre Leon Berard, Lyon, France
| | - Virginie Pétrilli
- Génétique Moléculaire, Signalisation et Cancer, UMR 5201 CNRS, Centre Leon Berard, Lyon, France
| | | | - Serge N. Manié
- Génétique Moléculaire, Signalisation et Cancer, UMR 5201 CNRS, Centre Leon Berard, Lyon, France
- * E-mail:
| |
Collapse
|
48
|
Impact of android overweight or obesity and insulin resistance on basal and postprandial SR-BI and ABCA1-mediated serum cholesterol efflux capacities. Atherosclerosis 2010; 209:422-9. [DOI: 10.1016/j.atherosclerosis.2009.09.025] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2009] [Revised: 09/17/2009] [Accepted: 09/18/2009] [Indexed: 10/20/2022]
|
49
|
Smythies LE, White CR, Maheshwari A, Palgunachari MN, Anantharamaiah GM, Chaddha M, Kurundkar AR, Datta G. Apolipoprotein A-I mimetic 4F alters the function of human monocyte-derived macrophages. Am J Physiol Cell Physiol 2010; 298:C1538-48. [PMID: 20219948 DOI: 10.1152/ajpcell.00467.2009] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
HDL and its major protein component apolipoprotein A-I (apoA-I) exert anti-inflammatory effects, inhibit monocyte chemotaxis/adhesion, and reduce vascular macrophage content in inflammatory conditions. In this study, we tested the hypothesis that the apoA-I mimetic 4F modulates the function of monocyte-derived macrophages (MDMs) by regulating the expression of key cell surface receptors on MDMs. Primary human monocytes and THP-1 cells were treated with 4F, apoA-I, or vehicle for 7 days and analyzed for expression of cell surface markers, adhesion to human endothelial cells, phagocytic function, cholesterol efflux capacity, and lipid raft organization. 4F and apoA-I treatment decreased the expression of HLA-DR, CD86, CD11b, CD11c, CD14, and Toll-like receptor-4 (TLR-4) compared with control cells, suggesting the induction of monocyte differentiation. Both treatments abolished LPS-induced mRNA for monocyte chemotactic protein-1 (MCP-1), macrophage inflammatory protein-1 (MIP-1), regulated on activation, normal T-expressed and presumably secreted (RANTES), IL-6, and TNF-alpha but significantly upregulated LPS-induced IL-10 expression. Moreover, 4F and apoA-I induced a 90% reduction in the expression of CD49d, a ligand for the VCAM-1 receptor, with a concurrent decrease in monocyte adhesion (55% reduction) to human endothelial cells and transendothelial migration (34 and 27% for 4F and apoA-I treatments) compared with vehicle treatment. In addition, phagocytosis of dextran-FITC beads was inhibited by 4F and apoA-I, a response associated with reduced expression of CD32. Finally, 4F and apoA-I stimulated cholesterol efflux from MDMs, leading to cholesterol depletion and disruption of lipid rafts. These data provide evidence that 4F, similar to apoA-I, induces profound functional changes in MDMs, possibly due to differentiation to an anti-inflammatory phenotype.
Collapse
|
50
|
Datta G, White CR, Dashti N, Chaddha M, Palgunachari MN, Gupta H, Handattu SP, Garber DW, Anantharamaiah GM. Anti-inflammatory and recycling properties of an apolipoprotein mimetic peptide, Ac-hE18A-NH(2). Atherosclerosis 2010; 208:134-41. [PMID: 19656510 PMCID: PMC2813354 DOI: 10.1016/j.atherosclerosis.2009.07.019] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2008] [Revised: 06/11/2009] [Accepted: 07/05/2009] [Indexed: 02/02/2023]
Abstract
Apolipoprotein E (apoE) exerts prominent anti-inflammatory effects and undergoes recycling by target cells. We previously reported that the peptide Ac-hE18A-NH(2), composed of the receptor binding domain (LRKLRKRLLR) of apoE covalently linked to the Class A amphipathic peptide 18A, dramatically lowers plasma cholesterol and lipid hydroperoxides and enhances paraoxonase activity in dyslipidemic animal models. The objective of this study was to determine whether this peptide, analogous to apoE, exerts anti-inflammatory effects and undergoes recycling under in vitro conditions. Pulse chase studies using [(125)I]-Ac-hE18A-NH(2) in THP-1 derived macrophages and HepG2 cells showed greater amounts of intact peptide in the cells at later time points indicating recycling of the peptide. Ac-hE18A-NH(2) induced a 2.5-fold increase in prebeta-HDL in the conditioned media of HepG2 cells. This effect persisted for 3 days after removal of the peptide from culture medium. Ac-hE18A-NH(2) also induced the secretion of cell surface apoE from THP-1 macrophages. In addition, the peptide increased cholesterol efflux from THP-1 cells by an ABCA1 independent mechanism. Moreover, Ac-hE18A-NH(2) inhibited LPS-induced vascular cell adhesion molecule-1 (VCAM-1) expression, and reduced monocyte adhesion in human umbilical vein endothelial cells (HUVECs). It also reduced the secretion of interleukin-6 (IL-6) and monocyte chemoattractant protein-1 (MCP-1) from THP-1 macrophages even when administered post-LPS and abolished the 18-fold increase in LPS-induced mRNA levels for MCP-1 in THP-1 cells. Taken together, these results suggest that addition of the putative apoE receptor-domain to the Class A amphipathic peptide 18A results in a peptide that, similar to apoE, recycles, thus enabling the potentiation and prolongation of its anti-atherogenic and anti-inflammatory effects. Such a peptide has great potential as a therapeutic agent in the management of atherosclerosis and other inflammatory diseases.
Collapse
Affiliation(s)
- Geeta Datta
- Department of Medicine, Atherosclerosis Research Unit, Division of Gerontology, Geriatrics and Palliative Medicine, University of Alabama at Birmingham, 1808 Seventh Avenue South, Birmingham, AL 35294, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|