1
|
Davidson WS, Vaisar T, Heinecke JW, Bornfeldt KE. Distinct roles of size-defined HDL subpopulations in cardiovascular disease. Curr Opin Lipidol 2025; 36:111-118. [PMID: 39450930 PMCID: PMC12003705 DOI: 10.1097/mol.0000000000000959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/26/2024]
Abstract
PURPOSE OF REVIEW Doubts about whether high-density lipoprotein-cholesterol (HDL-C) levels are causally related to atherosclerotic cardiovascular disease (CVD) risk have stimulated research on identifying HDL-related metrics that might better reflect its cardioprotective functions. HDL is made up of different types of particles that vary in size, protein and lipid composition, and function. This review focuses on recent findings on the specific roles of HDL subpopulations defined by size in CVD. RECENT FINDINGS Small HDL particles are more effective than larger particles at promoting cellular cholesterol efflux because apolipoprotein A-I on their surface better engages ABCA1 (ATP binding cassette subfamily A member 1). In contrast, large HDL particles bind more effectively to scavenger receptor class B type 1 on endothelial cells, which helps prevent LDL from moving into the artery wall. The specific role of medium-sized HDL particles, the most abundant subpopulation, is still unclear. SUMMARY HDL is made up of subpopulations of different sizes of particles, with selective functional roles for small and large HDLs. The function of HDL may depend more on the size and composition of its subpopulations than on HDL-C levels. Further research is required to understand how these different HDL subpopulations influence the development of CVD.
Collapse
Affiliation(s)
- W Sean Davidson
- Department of Pathology and Laboratory Medicine, University of Cincinnati, Cincinnati, Ohio
| | - Tomas Vaisar
- Deaprtment of Medicine, University of Washington School of Medicine
- University of Washington Medicine Diabetes Institute
| | - Jay W Heinecke
- Deaprtment of Medicine, University of Washington School of Medicine
- University of Washington Medicine Diabetes Institute
| | - Karin E Bornfeldt
- Deaprtment of Medicine, University of Washington School of Medicine
- University of Washington Medicine Diabetes Institute
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, USA
| |
Collapse
|
2
|
Vaisar T, Babenko I, Horvath KV, Niisuke K, Asztalos BF. Relationships between HDL subpopulation proteome and HDL function in overweight/obese people with and without coronary heart disease. Atherosclerosis 2024; 397:118565. [PMID: 39260003 PMCID: PMC11539851 DOI: 10.1016/j.atherosclerosis.2024.118565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 08/08/2024] [Accepted: 08/09/2024] [Indexed: 09/13/2024]
Abstract
BACKGROUND AND AIMS The structure-function relationships of high-density lipoprotein (HDL) subpopulations are not well understood. Our aim was to examine the interrelationships between HDL particle proteome and HDL functionality in subjects with and without coronary heart disease (CHD). METHODS We isolated 5 different HDL subpopulations based on charge, size, and apolipoprotein A1 (APOA1) content from the plasma of 33 overweight/obese CHD patients and 33 age-and body mass index (BMI)-matched CHD-free subjects. We measured the relative molar concentration of HDL-associated proteins by liquid chromatography tandem mass spectrometry (LC-MS/MS) and assessed particle functionality. RESULTS We quantified 110 proteins associated with the 5 APOA1-containing HDL subpopulations. The relative molar concentration of these proteins spanned five orders of magnitude. Only 10 proteins were present in >1% while 73 were present in <0.1% concentration. Only 6 of the 10 most abundant proteins were apolipoproteins. Interestingly, the largest (α-1) and the smallest (preβ-1) HDL particles contained the most diverse proteomes. The protein composition of each HDL subpopulation was altered in CHD cases as compared to controls with the most prominent differences in preβ-1 and α-1 particles. APOA2 concentration was positively correlated with preβ-1 particle functionality (ABCA1-CEC/mg APOA1 in preβ-1) (R2 = 0.42, p = 0.005), while APOE concentration was inversely correlated with large-HDL particle functionality (SRBI-CEC/mg APOA1 in α-1+α-2) (R2 = 0.18, p = 0.01). CONCLUSIONS The protein composition of the different HDL subpopulations was altered differentially in CHD patients. The functionality of the small and large HDL particles correlated with the protein content of APOA2 and APOE, respectively. Our data indicate that distinct particle subspecies and specific particle associated proteins provide new information about the role of HDL in CHD.
Collapse
Affiliation(s)
- Tomas Vaisar
- UW Medicine Diabetes Institute, Department of Medicine, University of Washington, Seattle, WA, USA.
| | - Ilona Babenko
- UW Medicine Diabetes Institute, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Katalin V Horvath
- Cardiovascular Nutrition Laboratory, Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, USA
| | - Katrin Niisuke
- Cardiovascular Nutrition Laboratory, Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, USA
| | - Bela F Asztalos
- Cardiovascular Nutrition Laboratory, Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, USA
| |
Collapse
|
3
|
Sacher S, Mukherjee A, Ray A. Deciphering structural aspects of reverse cholesterol transport: mapping the knowns and unknowns. Biol Rev Camb Philos Soc 2023; 98:1160-1183. [PMID: 36880422 DOI: 10.1111/brv.12948] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 02/03/2023] [Accepted: 02/24/2023] [Indexed: 03/08/2023]
Abstract
Atherosclerosis is a major contributor to the onset and progression of cardiovascular disease (CVD). Cholesterol-loaded foam cells play a pivotal role in forming atherosclerotic plaques. Induction of cholesterol efflux from these cells may be a promising approach in treating CVD. The reverse cholesterol transport (RCT) pathway delivers cholesteryl ester (CE) packaged in high-density lipoproteins (HDL) from non-hepatic cells to the liver, thereby minimising cholesterol load of peripheral cells. RCT takes place via a well-organised interplay amongst apolipoprotein A1 (ApoA1), lecithin cholesterol acyltransferase (LCAT), ATP binding cassette transporter A1 (ABCA1), scavenger receptor-B1 (SR-B1), and the amount of free cholesterol. Unfortunately, modulation of RCT for treating atherosclerosis has failed in clinical trials owing to our lack of understanding of the relationship between HDL function and RCT. The fate of non-hepatic CEs in HDL is dependent on their access to proteins involved in remodelling and can be regulated at the structural level. An inadequate understanding of this inhibits the design of rational strategies for therapeutic interventions. Herein we extensively review the structure-function relationships that are essential for RCT. We also focus on genetic mutations that disturb the structural stability of proteins involved in RCT, rendering them partially or completely non-functional. Further studies are necessary for understanding the structural aspects of RCT pathway completely, and this review highlights alternative theories and unanswered questions.
Collapse
Affiliation(s)
- Sukriti Sacher
- Department of Computational Biology, Indraprastha Institute of Information Technology, Okhla Phase III, New Delhi, 110019, India
| | - Abhishek Mukherjee
- Dhiti Life Sciences Pvt Ltd, B-107, Okhla Phase I, New Delhi, 110020, India
| | - Arjun Ray
- Department of Computational Biology, Indraprastha Institute of Information Technology, Okhla Phase III, New Delhi, 110019, India
| |
Collapse
|
4
|
Arias‐Alpizar G, Papadopoulou P, Rios X, Pulagam KR, Moradi M, Pattipeiluhu R, Bussmann J, Sommerdijk N, Llop J, Kros A, Campbell F. Phase-Separated Liposomes Hijack Endogenous Lipoprotein Transport and Metabolism Pathways to Target Subsets of Endothelial Cells In Vivo. Adv Healthc Mater 2023; 12:e2202709. [PMID: 36565694 PMCID: PMC11469146 DOI: 10.1002/adhm.202202709] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 12/14/2022] [Indexed: 12/25/2022]
Abstract
Plasma lipid transport and metabolism are essential to ensure correct cellular function throughout the body. Dynamically regulated in time and space, the well-characterized mechanisms underpinning plasma lipid transport and metabolism offers an enticing, but as yet underexplored, rationale to design synthetic lipid nanoparticles with inherent cell/tissue selectivity. Herein, a systemically administered liposome formulation, composed of just two lipids, that is capable of hijacking a triglyceride lipase-mediated lipid transport pathway resulting in liposome recognition and uptake within specific endothelial cell subsets is described. In the absence of targeting ligands, liposome-lipase interactions are mediated by a unique, phase-separated ("parachute") liposome morphology. Within the embryonic zebrafish, selective liposome accumulation is observed at the developing blood-brain barrier. In mice, extensive liposome accumulation within the liver and spleen - which is reduced, but not eliminated, following small molecule lipase inhibition - supports a role for endothelial lipase but highlights these liposomes are also subject to significant "off-target" by reticuloendothelial system organs. Overall, these compositionally simplistic liposomes offer new insights into the discovery and design of lipid-based nanoparticles that can exploit endogenous lipid transport and metabolism pathways to achieve cell selective targeting in vivo.
Collapse
Affiliation(s)
- Gabriela Arias‐Alpizar
- Supramolecular and Biomaterials ChemistryLeiden Institute of ChemistryLeiden UniversityP.O. Box 9502Leiden2300The Netherlands
- Division of BioTherapeuticsLeiden Academic Centre for Drug ResearchLeiden UniversityP.O. Box 9502Leiden2300The Netherlands
| | - Panagiota Papadopoulou
- Supramolecular and Biomaterials ChemistryLeiden Institute of ChemistryLeiden UniversityP.O. Box 9502Leiden2300The Netherlands
| | - Xabier Rios
- CIC biomaGUNEBasque Research and Technology Alliance (BRTA)San Sebastián20014Spain
| | | | - Mohammad‐Amin Moradi
- Materials and Interface ChemistryDepartment of Chemical Engineering and ChemistryEindhoven University of TechnologyP.O. Box 513Eindhoven5600The Netherlands
| | - Roy Pattipeiluhu
- Supramolecular and Biomaterials ChemistryLeiden Institute of ChemistryLeiden UniversityP.O. Box 9502Leiden2300The Netherlands
| | - Jeroen Bussmann
- Supramolecular and Biomaterials ChemistryLeiden Institute of ChemistryLeiden UniversityP.O. Box 9502Leiden2300The Netherlands
- Division of BioTherapeuticsLeiden Academic Centre for Drug ResearchLeiden UniversityP.O. Box 9502Leiden2300The Netherlands
| | - Nico Sommerdijk
- Department of BiochemistryRadboud Institute for Molecular Life SciencesRadboud University Medical CenterNijmegen6525The Netherlands
- Electron Microscopy CentreRadboudumc Technology Center MicroscopyRadboud University Medical CenterGeert Grooteplein Zuid 28Nijmegen6525The Netherlands
| | - Jordi Llop
- Materials and Interface ChemistryDepartment of Chemical Engineering and ChemistryEindhoven University of TechnologyP.O. Box 513Eindhoven5600The Netherlands
| | - Alexander Kros
- Supramolecular and Biomaterials ChemistryLeiden Institute of ChemistryLeiden UniversityP.O. Box 9502Leiden2300The Netherlands
| | - Frederick Campbell
- Supramolecular and Biomaterials ChemistryLeiden Institute of ChemistryLeiden UniversityP.O. Box 9502Leiden2300The Netherlands
- Present address:
NanoVation Therapeutics2405 Wesbrook Mall 4th floorVancouverBCV6T 1Z3Canada
| |
Collapse
|
5
|
Abe RJ, Abe JI, Nguyen MTH, Olmsted-Davis EA, Mamun A, Banerjee P, Cooke JP, Fang L, Pownall H, Le NT. Free Cholesterol Bioavailability and Atherosclerosis. Curr Atheroscler Rep 2022; 24:323-336. [PMID: 35332444 PMCID: PMC9050774 DOI: 10.1007/s11883-022-01011-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/02/2022] [Indexed: 11/30/2022]
Abstract
PURPOSE OF REVIEW As both a cholesterol acceptor and carrier in the reverse cholesterol transport (RCT) pathway, high-density lipoprotein (HDL) is putatively atheroprotective. However, current pharmacological therapies to increase plasma HDL cholesterol (HDL-c) concentration have paradoxically failed to prevent or reduce atherosclerosis and cardiovascular disease (CVD). Given that free cholesterol (FC) transfer between surfaces of lipoproteins and cells is reversible, excess plasma FC can be transferred to the cells of peripheral tissue sites resulting in atherosclerosis. Here, we summarize potential mechanisms contributing to this paradox and highlight the role of excess free cholesterol (FC) bioavailability in atherosclerosis vs. atheroprotection. RECENT FINDINGS Recent findings have established a complex relationship between HDL-c concentration and atherosclerosis. Systemic scavenger receptor class B type 1 (SR-B1) knock out (KO) mice exhibit with increased diet-induced atherosclerosis despite having an elevated plasma HDL-c concentration compared to wild type (WT) mice. The greater bioavailability of HDL-FC in SR-B1 vs. WT mice is associated with a higher FC content in multiple cell types and tissue sites. These results suggest that dysfunctional HDL with high FC bioavailability is atheroprone despite high HDL-c concentration. Past oversimplification of HDL-c involvement in cholesterol transport has led to the failures in HDL targeted therapy. Evidence suggests that FC-mediated functionality of HDL is of higher importance than its quantity; as a result, deciphering the regulatory mechanisms by which HDL-FC bioavailability can induce atherosclerosis can have far-reaching clinical implications.
Collapse
Affiliation(s)
- Rei J Abe
- Center for Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, USA
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
| | - Jun-Ichi Abe
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Minh T H Nguyen
- Center for Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, USA
- University of Science and Technology of Hanoi, Vietnam Academy of Science and Technology, Hanoi, Vietnam
| | | | - Abrar Mamun
- Center for Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, USA
| | - Priyanka Banerjee
- Center for Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, USA
| | - John P Cooke
- Center for Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, USA
- Weill Cornell Medicine, New York, NY, USA
| | - Longhou Fang
- Center for Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, USA
- Weill Cornell Medicine, New York, NY, USA
| | - Henry Pownall
- Weill Cornell Medicine, New York, NY, USA
- Center for Bioenergetics, Department of Medicine, Houston Methodist Research Institute, Houston, TX, USA
| | - Nhat-Tu Le
- Center for Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, USA.
- Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
6
|
Juhl AD, Wüstner D. Pathways and Mechanisms of Cellular Cholesterol Efflux-Insight From Imaging. Front Cell Dev Biol 2022; 10:834408. [PMID: 35300409 PMCID: PMC8920967 DOI: 10.3389/fcell.2022.834408] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 02/04/2022] [Indexed: 12/24/2022] Open
Abstract
Cholesterol is an essential molecule in cellular membranes, but too much cholesterol can be toxic. Therefore, mammalian cells have developed complex mechanisms to remove excess cholesterol. In this review article, we discuss what is known about such efflux pathways including a discussion of reverse cholesterol transport and formation of high-density lipoprotein, the function of ABC transporters and other sterol efflux proteins, and we highlight their role in human diseases. Attention is paid to the biophysical principles governing efflux of sterols from cells. We also discuss recent evidence for cholesterol efflux by the release of exosomes, microvesicles, and migrasomes. The role of the endo-lysosomal network, lipophagy, and selected lysosomal transporters, such as Niemann Pick type C proteins in cholesterol export from cells is elucidated. Since oxysterols are important regulators of cellular cholesterol efflux, their formation, trafficking, and secretion are described briefly. In addition to discussing results obtained with traditional biochemical methods, focus is on studies that use established and novel bioimaging approaches to obtain insight into cholesterol efflux pathways, including fluorescence and electron microscopy, atomic force microscopy, X-ray tomography as well as mass spectrometry imaging.
Collapse
Affiliation(s)
| | - Daniel Wüstner
- Department of Biochemistry and Molecular Biology, PhyLife, Physical Life Sciences, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
7
|
Chang X, Zhao Y, Qin S, Wang H, Wang B, Zhai L, Liu B, Gu HM, Zhang DW. Loss of Hepatic Surf4 Depletes Lipid Droplets in the Adrenal Cortex but Does Not Impair Adrenal Hormone Production. Front Cardiovasc Med 2021; 8:764024. [PMID: 34859075 PMCID: PMC8631933 DOI: 10.3389/fcvm.2021.764024] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 10/20/2021] [Indexed: 11/13/2022] Open
Abstract
The adrenal gland produces steroid hormones to play essential roles in regulating various physiological processes. Our previous studies showed that knockout of hepatic Surf4 (Surf4LKO) markedly reduced fasting plasma total cholesterol levels in adult mice, including low-density lipoprotein and high-density lipoprotein cholesterol. Here, we found that plasma cholesterol levels were also dramatically reduced in 4-week-old young mice and non-fasted adult mice. Circulating lipoprotein cholesterol is an important source of the substrate for the production of adrenal steroid hormones. Therefore, we investigated whether adrenal steroid hormone production was affected in Surf4LKO mice. We observed that lacking hepatic Surf4 essentially eliminated lipid droplets and significantly reduced cholesterol levels in the adrenal gland; however, plasma levels of aldosterone and corticosterone were comparable in Surf4LKO and the control mice under basal and stress conditions. Further analysis revealed that mRNA levels of genes encoding enzymes important for hormone synthesis were not altered, whereas the expression of scavenger receptor class B type I (SR-BI), low-density lipoprotein receptor (LDLR) and 3-hydroxy-3-methyl-glutaryl-CoA reductase was significantly increased in the adrenal gland of Surf4LKO mice, indicating increased de novo cholesterol biosynthesis and enhanced LDLR and SR-BI-mediated lipoprotein cholesterol uptake. We also observed that the nuclear form of SREBP2 was increased in the adrenal gland of Surf4 LKO mice. Taken together, these findings indicate that the very low levels of circulating lipoprotein cholesterol in Surf4LKO mice cause a significant reduction in adrenal cholesterol levels but do not significantly affect adrenal steroid hormone production. Reduced adrenal cholesterol levels activate SREBP2 and thus increase the expression of genes involved in cholesterol biosynthesis, which increases de novo cholesterol synthesis to compensate for the loss of circulating lipoprotein-derived cholesterol in the adrenal gland of Surf4LKO mice.
Collapse
Affiliation(s)
- Xiaole Chang
- Institute of Atherosclerosis, College of Basic Medical Sciences, Shandong First Medical University, Shandong Academy of Medical Sciences, Tai'an, China
| | - Yongfang Zhao
- Institute of Atherosclerosis, College of Basic Medical Sciences, Shandong First Medical University, Shandong Academy of Medical Sciences, Tai'an, China
| | - Shucun Qin
- Institute of Atherosclerosis, College of Basic Medical Sciences, Shandong First Medical University, Shandong Academy of Medical Sciences, Tai'an, China
| | - Hao Wang
- Institute of Atherosclerosis, College of Basic Medical Sciences, Shandong First Medical University, Shandong Academy of Medical Sciences, Tai'an, China
| | - Bingxiang Wang
- Institute of Atherosclerosis, College of Basic Medical Sciences, Shandong First Medical University, Shandong Academy of Medical Sciences, Tai'an, China
| | - Lei Zhai
- Institute of Atherosclerosis, College of Basic Medical Sciences, Shandong First Medical University, Shandong Academy of Medical Sciences, Tai'an, China
| | - Boyan Liu
- Institute of Atherosclerosis, College of Basic Medical Sciences, Shandong First Medical University, Shandong Academy of Medical Sciences, Tai'an, China
| | - Hong-Mei Gu
- Department of Pediatrics and Group on the Molecular and Cell Biology of Lipids, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Da-Wei Zhang
- Department of Pediatrics and Group on the Molecular and Cell Biology of Lipids, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
8
|
Casado ME, Huerta L, Marcos-Díaz A, Ortiz AI, Kraemer FB, Lasunción MA, Busto R, Martín-Hidalgo A. Hormone-sensitive lipase deficiency affects the expression of SR-BI, LDLr, and ABCA1 receptors/transporters involved in cellular cholesterol uptake and efflux and disturbs fertility in mouse testis. Biochim Biophys Acta Mol Cell Biol Lipids 2021; 1866:159043. [PMID: 34461308 DOI: 10.1016/j.bbalip.2021.159043] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 08/18/2021] [Accepted: 08/19/2021] [Indexed: 12/14/2022]
Abstract
Hormone-sensitive lipase (HSL) hydrolyse acylglycerols, cholesteryl and retinyl esters. HSL is a key lipase in mice testis, as HSL deficiency results in male sterility. The present work study the effects of the deficiency and lack of HSL on the localization and expression of SR-BI, LDLr, and ABCA1 receptors/transporters involved in uptake and efflux of cholesterol in mice testis, to determine the impact of HSL gene dosage on testis morphology, lipid homeostasis and fertility. The results of this work show that the lack of HSL in mice alters testis morphology and spermatogenesis, decreasing sperm counts, sperm motility and increasing the amount of Leydig cells and lipid droplets. They also show that there are differences in the localization of HSL, SR-BI, LDLr and ABCA1 in HSL+/+, HSL+/- and HSL-/- mice. The deficiency or lack of HSL has effects on protein and mRNA expression of genes involved in lipid metabolisms in mouse testis. HSL-/- testis have augmented expression of SR-BI, LDLr, ABCA1 and LXRβ, a critical sterol sensor that regulate multiple genes involved in lipid metabolism; whereas LDLr expression decreased in HSL+/- mice. Plin2, Abca1 and Ldlr mRNA levels increased; and LXRα (Nr1h3) and LXRβ (Nr1h2) decreased in testis from HSL-/- compared with HSL+/+; with no differences in Scarb1. Together these data suggest that HSL deficiency or lack in mice testis induces lipid homeostasis alterations that affect the cellular localization and expression of key receptors/transporter involved in cellular cholesterol uptake and efflux (SR-BI, LDRr, ABCA1); alters normal cellular function and impact fertility.
Collapse
Affiliation(s)
- María Emilia Casado
- Servicio de Bioquímica-Investigación, Hospital Universitario Ramón y Cajal, Instituto Ramón y Cajal de Investigación Sanitaria (IRyCIS), E-28034 Madrid, Spain; CIBER de Fisiopatología de la Obesidad y Nutrición (CIBERobn), ISCIII, Spain
| | - Lydia Huerta
- Servicio de Bioquímica-Investigación, Hospital Universitario Ramón y Cajal, Instituto Ramón y Cajal de Investigación Sanitaria (IRyCIS), E-28034 Madrid, Spain; CIBER de Fisiopatología de la Obesidad y Nutrición (CIBERobn), ISCIII, Spain
| | - Ana Marcos-Díaz
- Servicio de Bioquímica-Investigación, Hospital Universitario Ramón y Cajal, Instituto Ramón y Cajal de Investigación Sanitaria (IRyCIS), E-28034 Madrid, Spain; CIBER de Fisiopatología de la Obesidad y Nutrición (CIBERobn), ISCIII, Spain
| | - Ana Isabel Ortiz
- Unidad de Cirugía Experimental y Animalario, Hospital Universitario Ramón y Cajal, Instituto Ramón y Cajal de Investigación Sanitaria (IRyCIS), E-28034 Madrid, Spain
| | - Fredric B Kraemer
- Division of Endocrinology, Stanford University, United States of America; VA Palo Alto Health Care System, Palo Alto, CA, United States of America
| | - Miguel Angel Lasunción
- Servicio de Bioquímica-Investigación, Hospital Universitario Ramón y Cajal, Instituto Ramón y Cajal de Investigación Sanitaria (IRyCIS), E-28034 Madrid, Spain; CIBER de Fisiopatología de la Obesidad y Nutrición (CIBERobn), ISCIII, Spain
| | - Rebeca Busto
- Servicio de Bioquímica-Investigación, Hospital Universitario Ramón y Cajal, Instituto Ramón y Cajal de Investigación Sanitaria (IRyCIS), E-28034 Madrid, Spain; CIBER de Fisiopatología de la Obesidad y Nutrición (CIBERobn), ISCIII, Spain
| | - Antonia Martín-Hidalgo
- Servicio de Bioquímica-Investigación, Hospital Universitario Ramón y Cajal, Instituto Ramón y Cajal de Investigación Sanitaria (IRyCIS), E-28034 Madrid, Spain; CIBER de Fisiopatología de la Obesidad y Nutrición (CIBERobn), ISCIII, Spain.
| |
Collapse
|
9
|
Binding of liposomes composed of phosphatidylcholine to scavenger receptor class B type 1 and its modulation by phosphatidic acid in HEK293T cells. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2021; 1868:119043. [PMID: 33862056 DOI: 10.1016/j.bbamcr.2021.119043] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 04/11/2021] [Indexed: 12/29/2022]
Abstract
In this study, we developed a method to analyze liposomal binding to a cell membrane receptor using fluorescence-labeled liposomes and demonstrated that scavenger class B type 1 (SR-B1) plays a crucial role in binding of liposomes containing phosphatidylcholine (PC) to HEK293T cell membrane and phosphatidic acid (PA) can modulate it. Site-directed mutagenesis of SR-B1 revealed that S112F and T175A mutations in its ectodomain abrogated binding and endocytosis of PC liposomes in HEK293T cells. K151A and K156A mutations attenuated their binding and endocytosis too. Although the effects of mutations on binding and endocytosis were similar between PC liposomes and PC/PA and PA liposomes, SR-B1 dependency appeared to be PC > PC/PA > PA liposomes. Our data indicate that (i) nanoparticles including high-density lipoprotein (HDL), silica, and liposomes bind to a common or close site of SR-B1, and (ii) PC/PA and PA liposomes bind not only to SR-B1 but also other receptor(s) in HEK293T cells. In addition, PC/PA liposomes induced lipid droplet (LD) formation in HEK293T cells more than PC liposomes. Treatment of HEK293T cells with SR-B1 siRNA suppressed PC/PA liposome-induced LD formation. Taken together, our results demonstrate that SR-B1 plays an essential role in binding PC-containing liposomes and the subsequent induction of cellular responses, while PA can modulate them.
Collapse
|
10
|
Widjaja-Adhi MAK, Golczak M. The molecular aspects of absorption and metabolism of carotenoids and retinoids in vertebrates. Biochim Biophys Acta Mol Cell Biol Lipids 2020; 1865:158571. [PMID: 31770587 PMCID: PMC7244374 DOI: 10.1016/j.bbalip.2019.158571] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 11/04/2019] [Accepted: 11/07/2019] [Indexed: 02/08/2023]
Abstract
Vitamin A is an essential nutrient necessary for numerous basic physiological functions, including reproduction and development, immune cell differentiation and communication, as well as the perception of light. To evade the dire consequences of vitamin A deficiency, vertebrates have evolved specialized metabolic pathways that enable the absorption, transport, and storage of vitamin A acquired from dietary sources as preformed retinoids or provitamin A carotenoids. This evolutionary advantage requires a complex interplay between numerous specialized retinoid-transport proteins, receptors, and enzymes. Recent advances in molecular and structural biology resulted in a rapid expansion of our understanding of these processes at the molecular level. This progress opened new avenues for the therapeutic manipulation of retinoid homeostasis. In this review, we summarize current research related to the biochemistry of carotenoid and retinoid-processing proteins with special emphasis on the structural aspects of their physiological actions. This article is part of a Special Issue entitled Carotenoids recent advances in cell and molecular biology edited by Johannes von Lintig and Loredana Quadro.
Collapse
Affiliation(s)
- Made Airanthi K Widjaja-Adhi
- Department of Pharmacology, School of Medicine, Case Western Reserve University, Cleveland, OH, United States of America
| | - Marcin Golczak
- Department of Pharmacology, School of Medicine, Case Western Reserve University, Cleveland, OH, United States of America; Cleveland Center for Membrane and Structural Biology, School of Medicine, Case Western Reserve University, Cleveland, OH, United States of America.
| |
Collapse
|
11
|
Transcriptome Analysis of Testis from HFD-Induced Obese Rats ( Rattus norvigicus) Indicated Predisposition for Male Infertility. Int J Mol Sci 2020; 21:ijms21186493. [PMID: 32899471 PMCID: PMC7554891 DOI: 10.3390/ijms21186493] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 08/23/2020] [Accepted: 08/27/2020] [Indexed: 12/13/2022] Open
Abstract
Obesity is a worldwide life-threatening metabolic disorder, associated with various chronic diseases, including male infertility. Obesity was induced by high fat diet (HFD), and testis RNA was used for the transcriptome analysis using RNAseq via Illumina NovaSeq 6000 System and NovaSeq 6000 Kit. Gene expression level was estimated as FPKM (Fragments Per Kilobase of transcript per Million mapped reads). Differential expressed genes (DEGs) were annotated against gene ontology (GO) and KEGG databases. More than 63.66 million reads per sample were performed with 100 bp cutoff and 6 Gb sequencing depth. Results of this study revealed that 267 GO terms (245 biological processes (BP), 14 cellular components (CC), eight molecular functions (MF)), and 89 KEGG pathways were significantly enriched. Moreover, total numbers of 136 genes were differentially expressed (107 upregulated, 29 downregulated) with |FC| ≥ 2 and bh adjusted <0.05. Interesting DEGs were detected, including obesity and lipid metabolism-related genes, immune response-related genes, cytochrome P450 genes, including aromatase were upregulated, whereas genes related to male fertility and fertilization, cell adhesion, and olfactory receptors were downregulated. The combined expression pattern of the DEGs in obese animals indicated an increase in cholesterol metabolism. Furthermore, high aromatase activity enhances the testosterone turnover into estradiol and lowers the testosterone/estradiol (T/E) ratio, which ultimately reduces fertility. In addition, downregulation of cadherens junction components genes leads to the pre-mature release of sperm from Sertoli cells resulting in the reduction of fertility. Moreover, the downregulation of olfactory receptor genes reduces the chemotaxis capacity of sperms in tracking the oocyte for fertilization, which reduces male fertility. Furthermore, various obesity molecular markers were detected in our transcriptome. The results of this study will enhance our understanding of the molecular network of obesity development, development of obesity novel molecular diagnosis markers, molecular bases of obesity-induced infertility, and the development of anti-obesity drugs.
Collapse
|
12
|
Calderón B, Huerta L, Casado ME, González-Casbas JM, Botella-Carretero JI, Martín-Hidalgo A. Morbid obesity-related changes in the expression of lipid receptors, transporters, and HSL in human sperm. J Assist Reprod Genet 2019; 36:777-786. [PMID: 30659447 PMCID: PMC6505031 DOI: 10.1007/s10815-019-01406-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 01/09/2019] [Indexed: 12/30/2022] Open
Abstract
OBJECTIVE To study the location and expression of receptors (SR-BI/CLA-1, SR-BII, and LDLr) and transporter (ABCA1) involved in uptake and efflux of cholesterol in human spermatozoa and assess whether obesity alters its location/expression and whether this could be related to infertility. DESIGN Observational study. SETTING None PATIENT(S): Ten controls and 20 obese patients. INTERVENTION(S) Anthropometric parameters. Serum and semen samples were collected. MAIN OUTCOME MEASURE(S) Spermatozoon concentration, immunolocalization, and protein expression in semen. RESULTS Spermatozoon concentration and motility was decreased in morbidly obese patients. SR-BI/CLA-1, SR-BII, LDLr, and ABCA1 are located in the spermatozoon cell membrane and the localization does not change between obese patients and controls. Control spermatozoa showed high SR-BI expression, and less expression for the rest of the receptors analyzed, indicating that SR-BI/CLA-1 is relevant in human spermatozoon cholesterol uptake/efflux. On the contrary, spermatozoa of obese patients showed less SR-BI/CLA-1 expression than controls, and more intense positive staining for SR-BII, LDLr, and ABCA1. Finally, human sperm expresses the 130- and 82-kDa hormone-sensitive lipase (HSL) isoforms. The 130-kDa isoform is expressed in the control sperm, and the expression disappears in the obese patients. CONCLUSION(S) The presence of lipid receptors/transporters and HSL in human spermatozoa suggests their role in the process of maturation/capacitation. The changes in the expression of lipid receptors/transporters and the lack of the 130-kDa HSL isoform in obese patients prevent the hydrolysis of cholesterol esters internalized by these receptors, and favor their accumulation in the cytoplasm of the spermatozoa that could contribute to lipotoxicity and infertility.
Collapse
Affiliation(s)
- Berniza Calderón
- Instituto Tecnológico Santo Domingo (INTEC), Santo Domingo, República Dominicana
- Departamento de Endocrinología y Metabolismo, Madrid, Spain
| | - Lydia Huerta
- Servicio de Bioquímica-Investigación, Madrid, Spain
| | - María Emilia Casado
- Servicio de Bioquímica-Investigación, Madrid, Spain
- CIBER de la Fisiopatología de la Obesidad y Nutrición (CIBERobn), ISCIII, Madrid, Spain
| | - José Manuel González-Casbas
- Instituto Europeo de Fertilidad y Unidad de Reproducción Asistida, Instituto Ramón y Cajal de Investigación Sanitaria (IRyCIS), Hospital Universitario Ramón y Cajal, E-28034, Madrid, Spain
| | - José Ignacio Botella-Carretero
- Departamento de Endocrinología y Metabolismo, Madrid, Spain
- CIBER de la Fisiopatología de la Obesidad y Nutrición (CIBERobn), ISCIII, Madrid, Spain
| | - Antonia Martín-Hidalgo
- Servicio de Bioquímica-Investigación, Madrid, Spain.
- CIBER de la Fisiopatología de la Obesidad y Nutrición (CIBERobn), ISCIII, Madrid, Spain.
- Department of Biochemistry-Research, Hospital Universitario Ramón y Cajal, Ctra.ColmenarViejo, Km 9.100, E-28034, Madrid, Spain.
| |
Collapse
|
13
|
Shen WJ, Azhar S, Kraemer FB. SR-B1: A Unique Multifunctional Receptor for Cholesterol Influx and Efflux. Annu Rev Physiol 2017; 80:95-116. [PMID: 29125794 DOI: 10.1146/annurev-physiol-021317-121550] [Citation(s) in RCA: 272] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The scavenger receptor, class B type 1 (SR-B1), is a multiligand membrane receptor protein that functions as a physiologically relevant high-density lipoprotein (HDL) receptor whose primary role is to mediate selective uptake or influx of HDL-derived cholesteryl esters into cells and tissues. SR-B1 also facilitates the efflux of cholesterol from peripheral tissues, including macrophages, back to liver. As a regulator of plasma membrane cholesterol content, SR-B1 promotes the uptake of lipid soluble vitamins as well as viral entry into host cells. These collective functions of SR-B1 ultimately affect programmed cell death, female fertility, platelet function, vasculature inflammation, and diet-induced atherosclerosis and myocardial infarction. SR-B1 has also been identified as a potential marker for cancer diagnosis and prognosis. Finally, the SR-B1-linked selective HDL-cholesteryl ester uptake pathway is now being evaluated as a gateway for the delivery of therapeutic and diagnostic agents. In this review, we focus on the regulation and functional significance of SR-B1 in mediating cholesterol movement into and out of cells.
Collapse
Affiliation(s)
- Wen-Jun Shen
- Division of Endocrinology, Gerontology and Metabolism, Stanford University School of Medicine, Stanford, California 94305; .,VA Palo Alto Health Care System, Palo Alto, California 94304
| | - Salman Azhar
- Division of Endocrinology, Gerontology and Metabolism, Stanford University School of Medicine, Stanford, California 94305; .,VA Palo Alto Health Care System, Palo Alto, California 94304
| | - Fredric B Kraemer
- Division of Endocrinology, Gerontology and Metabolism, Stanford University School of Medicine, Stanford, California 94305; .,VA Palo Alto Health Care System, Palo Alto, California 94304
| |
Collapse
|
14
|
Lek MT, Cruz S, Ibe NU, Beck WHJ, Bielicki JK, Weers PMM, Narayanaswami V. Swapping the N- and C-terminal domains of human apolipoprotein E3 and AI reveals insights into their structure/activity relationship. PLoS One 2017. [PMID: 28644829 PMCID: PMC5482431 DOI: 10.1371/journal.pone.0178346] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Apolipoprotein (apo) E3 and apoAI are exchangeable apolipoproteins that play a dominant role in regulating plasma lipoprotein metabolism. ApoE3 (299 residues) is composed of an N-terminal (NT) domain bearing a 4-helix bundle and a C-terminal (CT) domain bearing a series of amphipathic α-helices. ApoAI (243 residues) also comprises a highly helical NT domain and a less structured CT tail. The objective of this study was to understand their structural and functional role by generating domain swapped chimeras: apoE3-NT/apoAI-CT and apoAI-NT/apoE-CT. The bacterially overexpressed chimeras were purified by affinity chromatography and their identity confirmed by immunoblotting and mass spectrometry. Their α-helical content was comparable to that of the parent proteins. ApoE3-NT/apoAI-CT retained the denaturation profile of apoE3 NT domain, with apoAI CT tail eliciting a relatively unstructured state; its lipid binding ability improved dramatically compared to apoE3 indicative of a significant role of apoAI CT tail in lipid binding interaction. The LDL receptor interaction and ability to promote ABCA1-mediated cholesterol efflux of apoE3-NT/apoAI-CT was comparable to that of apoE3. In contrast, apoAI-NT/apoE-CT elicited an unfolding pattern and lipid binding ability that were similar to that of apoAI. As expected, DMPC/apoAI-NT/apoE-CT discoidal particles did not elicit LDLr binding ability, and promoted SR-B1 mediated cellular uptake of lipids to a limited extent. However, apoAI-NT/apoE-CT displayed an enhanced ability to promote cholesterol efflux compared to apoAI, indicative of a significant role for apoE CT domain in mediating this function. Together, these results indicate that the functional attributes of apoAI and apoE3 can be conferred on each other and that NT-CT domain interactions significantly modulate their structure and function.
Collapse
Affiliation(s)
- Mark T. Lek
- Department of Chemistry and Biochemistry, California State University Long Beach, Long Beach, California, United States of America
| | - Siobanth Cruz
- Department of Chemistry and Biochemistry, California State University Long Beach, Long Beach, California, United States of America
| | - Nnejiuwa U. Ibe
- Department of Chemistry and Biochemistry, California State University Long Beach, Long Beach, California, United States of America
| | - Wendy H. J. Beck
- Department of Chemistry and Biochemistry, California State University Long Beach, Long Beach, California, United States of America
| | - John K. Bielicki
- Donner Laboratory, Lawrence Berkeley National Laboratory, Berkeley, California, United States of America
| | - Paul M. M. Weers
- Department of Chemistry and Biochemistry, California State University Long Beach, Long Beach, California, United States of America
| | - Vasanthy Narayanaswami
- Department of Chemistry and Biochemistry, California State University Long Beach, Long Beach, California, United States of America
- * E-mail:
| |
Collapse
|
15
|
Rajora MA, Zheng G. Targeting SR-BI for Cancer Diagnostics, Imaging and Therapy. Front Pharmacol 2016; 7:326. [PMID: 27729859 PMCID: PMC5037127 DOI: 10.3389/fphar.2016.00326] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Accepted: 09/06/2016] [Indexed: 01/13/2023] Open
Abstract
Scavenger receptor class B type I (SR-BI) plays an important role in trafficking cholesteryl esters between the core of high density lipoprotein and the liver. Interestingly, this integral membrane protein receptor is also implicated in the metabolism of cholesterol by cancer cells, whereby overexpression of SR-BI has been observed in a number of tumors and cancer cell lines, including breast and prostate cancers. Consequently, SR-BI has recently gained attention as a cancer biomarker and exciting target for the direct cytosolic delivery of therapeutic agents. This brief review highlights these key developments in SR-BI-targeted cancer therapies and imaging probes. Special attention is given to the exploration of high density lipoprotein nanomimetic platforms that take advantage of upregulated SR-BI expression to facilitate targeted drug-delivery and cancer diagnostics, and promising future directions in the development of these agents.
Collapse
Affiliation(s)
- Maneesha A Rajora
- Princess Margaret Cancer Centre and Techna Institute, University Health NetworkToronto, ON, Canada; Institute of Biomaterials and Biomedical Engineering, University of TorontoToronto, ON, Canada
| | - Gang Zheng
- Princess Margaret Cancer Centre and Techna Institute, University Health NetworkToronto, ON, Canada; Institute of Biomaterials and Biomedical Engineering, University of TorontoToronto, ON, Canada; Department of Medical Biophysics, University of TorontoToronto, ON, Canada
| |
Collapse
|
16
|
Mutharasan RK, Foit L, Thaxton CS. High-Density Lipoproteins for Therapeutic Delivery Systems. J Mater Chem B 2016; 4:188-197. [PMID: 27069624 PMCID: PMC4825811 DOI: 10.1039/c5tb01332a] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
High-density lipoproteins (HDL) are a class of natural nanostructures found in the blood and are composed of lipids, proteins, and nucleic acids (e.g. microRNA). Their size, which appears to be well-suited for both tissue penetration/retention as well as payload delivery, long circulation half-life, avoidance of endosomal sequestration, and potential low toxicity are all excellent properties to model in a drug delivery vehicle. In this review, we consider high-density lipoproteins for therapeutic delivery systems. First we discuss the structure and function of natural HDL, describing in detail its biogenesis and transformation from immature, discoidal forms, to more mature, spherical forms. Next we consider features of HDL making them suitable vehicles for drug delivery. We then describe the use of natural HDL, discoidal HDL analogs, and spherical HDL analogs to deliver various classes of drugs, including small molecules, lipids, and oligonucleotides. We briefly consider the notion that the drug delivery vehicles themselves are therapeutic, constituting entities that exhibit "theralivery." Finally, we discuss challenges and future directions in the field.
Collapse
Affiliation(s)
- R. Kannan Mutharasan
- Feinberg Cardiovascular Research Institute, 303 E. Chicago Ave., Tarry 14-725, Chicago, IL 60611 United States
| | - Linda Foit
- Feinberg School of Medicine, Department of Urology, Northwestern University, Tarry 16-703, 303 E. Chicago Ave, Chicago, IL 60611, USA
| | - C. Shad Thaxton
- Feinberg School of Medicine, Department of Urology, Northwestern University, Tarry 16-703, 303 E. Chicago Ave, Chicago, IL 60611, USA
- Simpson Querrey Institute for BioNanotechnology, Northwestern University, 303 E. Superior St, Chicago, IL 60611, USA
- International Institute for Nanotechnology (IIN), 2145 Sheridan Road, Evanston, IL 60208, USA
- Robert H Lurie Comprehensive Cancer Center (RHLCCC), Northwestern University, Feinberg School of Medicine, 303 E Superior, Chicago, IL 60611, USA
| |
Collapse
|
17
|
Abstract
The adrenal gland is one of the prominent sites for steroid hormone synthesis. Lipoprotein-derived cholesterol esters (CEs) delivered via SR-B1 constitute the dominant source of cholesterol for steroidogenesis, particularly in rodents. Adrenocorticotropic hormone (ACTH) stimulates steroidogenesis through downstream actions on multiple components involved in steroidogenesis. Both acute and chronic ACTH treatments can modulate SR-B1 function, including its transcription, posttranscriptional stability, phosphorylation and dimerization status, as well as the interaction with other protein partners, all of which result in changes in the ability of SR-B1 to mediate HDL-CE uptake and the supply of cholesterol for conversion to steroids. Here, we provide a review of the recent findings on the regulation of adrenal SR-B1 function by ACTH.
Collapse
Affiliation(s)
- Wen-Jun Shen
- The Division of Endocrinology, Stanford University, Stanford, CA, USA
- Geriatric Research, Education and Clinical Center, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
| | - Salman Azhar
- The Division of Endocrinology, Stanford University, Stanford, CA, USA
- Geriatric Research, Education and Clinical Center, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
| | - Fredric B. Kraemer
- The Division of Endocrinology, Stanford University, Stanford, CA, USA
- Geriatric Research, Education and Clinical Center, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
- *Correspondence: Fredric B. Kraemer,
| |
Collapse
|
18
|
Hueging K, Weller R, Doepke M, Vieyres G, Todt D, Wölk B, Vondran FWR, Geffers R, Lauber C, Kaderali L, Penin F, Pietschmann T. Several Human Liver Cell Expressed Apolipoproteins Complement HCV Virus Production with Varying Efficacy Conferring Differential Specific Infectivity to Released Viruses. PLoS One 2015; 10:e0134529. [PMID: 26226615 PMCID: PMC4520612 DOI: 10.1371/journal.pone.0134529] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 07/09/2015] [Indexed: 02/08/2023] Open
Abstract
Apolipoprotein E (ApoE), an exchangeable apolipoprotein, is necessary for production of infectious Hepatitis C virus (HCV) particles. However, ApoE is not the only liver-expressed apolipoprotein and the role of other apolipoproteins for production of infectious HCV progeny is incompletely defined. Therefore, we quantified mRNA expression of human apolipoproteins in primary human hepatocytes. Subsequently, cDNAs encoding apolipoproteins were expressed in 293T/miR-122 cells to explore if they complement HCV virus production in cells that are non-permissive due to limiting endogenous levels of human apolipoproteins. Primary human hepatocytes expressed high mRNA levels of ApoA1, A2, C1, C3, E, and H. ApoA4, A5, B, D, F, J, L1, L2, L3, L4, L6, M, and O were expressed at intermediate levels, and C2, C4, and L5 were not detected. All members of the ApoA and ApoC family of lipoproteins complemented HCV virus production in HCV transfected 293T/miR-122 cells, albeit with significantly lower efficacy compared with ApoE. In contrast, ApoD expression did not support production of infectious HCV. Specific infectivity of released particles complemented with ApoA family members was significantly lower compared with ApoE. Moreover, the ratio of extracellular to intracellular infectious virus was significantly higher for ApoE compared to ApoA2 and ApoC3. Since apolipoproteins complementing HCV virus production share amphipathic alpha helices as common structural features we altered the two alpha helices of ApoC1. Helix breaking mutations in both ApoC1 helices impaired virus assembly highlighting a critical role of alpha helices in apolipoproteins supporting HCV assembly. In summary, various liver expressed apolipoproteins with amphipathic alpha helices complement HCV virus production in human non liver cells. Differences in the efficiency of virus assembly, the specific infectivity of released particles, and the ratio between extracellular and intracellular infectivity point to distinct characteristics of these apolipoproteins that influence HCV assembly and cell entry. This will guide future research to precisely pinpoint how apolipoproteins function during virus assembly and cell entry.
Collapse
Affiliation(s)
- Kathrin Hueging
- Institute of Experimental Virology, TWINCORE, Centre for Experimental and Clinical Infection Research, Hannover, Germany
| | - Romy Weller
- Institute of Experimental Virology, TWINCORE, Centre for Experimental and Clinical Infection Research, Hannover, Germany
| | - Mandy Doepke
- Institute of Experimental Virology, TWINCORE, Centre for Experimental and Clinical Infection Research, Hannover, Germany
| | - Gabrielle Vieyres
- Institute of Experimental Virology, TWINCORE, Centre for Experimental and Clinical Infection Research, Hannover, Germany
| | - Daniel Todt
- Institute of Experimental Virology, TWINCORE, Centre for Experimental and Clinical Infection Research, Hannover, Germany
| | - Benno Wölk
- Institute of Virology, Hannover Medical School, Hannover, Germany
| | - Florian W. R. Vondran
- Department of General, Visceral and Transplant Surgery, Hannover Medical School, Hannover, Germany
- Integrated Research and Treatment Center Transplantation (IFB-Tx), Hannover Medical School, Hannover, Germany
- German Centre for Infection Research (DZIF), Partner Site Hannover-Braunschweig, Hannover, Germany
| | - Robert Geffers
- Research Group Genome Analytics, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Chris Lauber
- Institute for Medical Informatics and Biometry, Medical Faculty, Technische Universität Dresden, Dresden, Germany
| | - Lars Kaderali
- Institute for Medical Informatics and Biometry, Medical Faculty, Technische Universität Dresden, Dresden, Germany
| | - François Penin
- Institut de Biologie et Chimie des Protéines, Bases Moléculaires et Structurales des Systèmes Infectieux, UMR 5086, CNRS, Labex Ecofect, University of Lyon, Lyon, France
| | - Thomas Pietschmann
- Institute of Experimental Virology, TWINCORE, Centre for Experimental and Clinical Infection Research, Hannover, Germany
- German Centre for Infection Research (DZIF), Partner Site Hannover-Braunschweig, Hannover, Germany
- * E-mail:
| |
Collapse
|
19
|
González-Pecchi V, Valdés S, Pons V, Honorato P, Martinez LO, Lamperti L, Aguayo C, Radojkovic C. Apolipoprotein A-I enhances proliferation of human endothelial progenitor cells and promotes angiogenesis through the cell surface ATP synthase. Microvasc Res 2015; 98:9-15. [PMID: 25445031 DOI: 10.1016/j.mvr.2014.11.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Revised: 10/28/2014] [Accepted: 11/02/2014] [Indexed: 12/14/2022]
|
20
|
Ecto-F1-ATPase/P2Y pathways in metabolic and vascular functions of high density lipoproteins. Atherosclerosis 2015; 238:89-100. [DOI: 10.1016/j.atherosclerosis.2014.11.017] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Revised: 11/12/2014] [Accepted: 11/13/2014] [Indexed: 12/15/2022]
|
21
|
Montecucco F, Favari E, Norata GD, Ronda N, Nofer JR, Vuilleumier N. Impact of systemic inflammation and autoimmune diseases on apoA-I and HDL plasma levels and functions. Handb Exp Pharmacol 2015; 224:455-82. [PMID: 25522998 DOI: 10.1007/978-3-319-09665-0_14] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The cholesterol of high-density lipoproteins (HDLs) and its major proteic component, apoA-I, have been widely investigated as potential predictors of acute cardiovascular (CV) events. In particular, HDL cholesterol levels were shown to be inversely and independently associated with the risk of acute CV diseases in different patient populations, including autoimmune and chronic inflammatory disorders. Some relevant and direct anti-inflammatory activities of HDL have been also recently identified targeting both immune and vascular cell subsets. These studies recently highlighted the improvement of HDL function (instead of circulating levels) as a promising treatment strategy to reduce inflammation and associated CV risk in several diseases, such as systemic lupus erythematosus and rheumatoid arthritis. In these diseases, anti-inflammatory treatments targeting HDL function might improve both disease activity and CV risk. In this narrative review, we will focus on the pathophysiological relevance of HDL and apoA-I levels/functions in different acute and chronic inflammatory pathophysiological conditions.
Collapse
Affiliation(s)
- Fabrizio Montecucco
- Division of Laboratory Medicine, Department of Genetics and Laboratory Medicine, Geneva University Hospitals, 4 rue Gabrielle Perret-Gentil, 1211, Geneva, Switzerland
| | | | | | | | | | | |
Collapse
|
22
|
DeAngelis AM, Roy-O'Reilly M, Rodriguez A. Genetic alterations affecting cholesterol metabolism and human fertility. Biol Reprod 2014; 91:117. [PMID: 25122065 DOI: 10.1095/biolreprod.114.119883] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
Single nucleotide polymorphisms (SNPs) represent genetic variations among individuals in a population. In medicine, these small variations in the DNA sequence may significantly impact an individual's response to certain drugs or influence the risk of developing certain diseases. In the field of reproductive medicine, a significant amount of research has been devoted to identifying polymorphisms which may impact steroidogenesis and fertility. This review discusses current understanding of the effects of genetic variations in cholesterol metabolic pathways on human fertility that bridge novel linkages between cholesterol metabolism and reproductive health. For example, the role of the low-density lipoprotein receptor (LDLR) in cellular metabolism and human reproduction has been well studied, whereas there is now an emerging body of research on the role of the high-density lipoprotein (HDL) receptor scavenger receptor class B type I (SR-BI) in human lipid metabolism and female reproduction. Identifying and understanding how polymorphisms in the SCARB1 gene or other genes related to lipid metabolism impact human physiology is essential and will play a major role in the development of personalized medicine for improved diagnosis and treatment of infertility.
Collapse
Affiliation(s)
| | | | - Annabelle Rodriguez
- Center for Vascular Biology, University of Connecticut Health Center, Farmington, Connecticut
| |
Collapse
|
23
|
Shen WJ, Hu J, Hu Z, Kraemer FB, Azhar S. Scavenger receptor class B type I (SR-BI): a versatile receptor with multiple functions and actions. Metabolism 2014; 63:875-86. [PMID: 24854385 PMCID: PMC8078058 DOI: 10.1016/j.metabol.2014.03.011] [Citation(s) in RCA: 101] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Revised: 03/12/2014] [Accepted: 03/18/2014] [Indexed: 11/16/2022]
Abstract
Scavenger receptor class B type I (SR-BI), is a physiologically relevant HDL receptor that mediates selective uptake of lipoprotein (HDL)-derived cholesteryl ester (CE) in vitro and in vivo. Mammalian SR-BI is a 509-amino acid, ~82 kDa glycoprotein that contains N- and C-terminal cytoplasmic domains, two-transmembrane domains, as well as a large extracellular domain containing 5-6 cysteine residues and multiple sites for N-linked glycosylation. The size and structural characteristics of SR-BI, however, vary considerably among lower vertebrates and insects. Recently, significant progress has been made in understanding the molecular mechanisms involved in the posttranscriptional/posttranslational regulation of SR-BI in a tissue specific manner. The purpose of this review is to summarize the current body of knowledge about the events and molecules connected with the posttranscriptional/posttranslational regulation of SR-BI and to update the molecular and functional characteristics of the insect SR-BI orthologs.
Collapse
MESH Headings
- Animals
- Biological Transport
- Gene Expression Regulation
- Glycosylation
- Humans
- Insect Proteins/chemistry
- Insect Proteins/genetics
- Insect Proteins/metabolism
- Lipoproteins, HDL/chemistry
- Lipoproteins, HDL/genetics
- Lipoproteins, HDL/metabolism
- Liver/metabolism
- Organ Specificity
- Protein Conformation
- Protein Processing, Post-Translational
- Receptors, Lipoprotein/chemistry
- Receptors, Lipoprotein/genetics
- Receptors, Lipoprotein/metabolism
- Scavenger Receptors, Class B/chemistry
- Scavenger Receptors, Class B/genetics
- Scavenger Receptors, Class B/metabolism
- Species Specificity
Collapse
Affiliation(s)
- Wen-Jun Shen
- Geriatric Research, Education and Clinical Center, VA Palo Alto Health Care System, Palo Alto, California 94304; Division of Endocrinology, Stanford University, Stanford, California 94305
| | - Jie Hu
- Geriatric Research, Education and Clinical Center, VA Palo Alto Health Care System, Palo Alto, California 94304
| | - Zhigang Hu
- Geriatric Research, Education and Clinical Center, VA Palo Alto Health Care System, Palo Alto, California 94304; Division of Endocrinology, Stanford University, Stanford, California 94305
| | - Fredric B Kraemer
- Geriatric Research, Education and Clinical Center, VA Palo Alto Health Care System, Palo Alto, California 94304; Division of Endocrinology, Stanford University, Stanford, California 94305
| | - Salman Azhar
- Geriatric Research, Education and Clinical Center, VA Palo Alto Health Care System, Palo Alto, California 94304; Division of Gastroenterology and Hepatology, Stanford University, Stanford, California 94305.
| |
Collapse
|
24
|
Pécheur EI. Lipoprotein receptors and lipid enzymes in hepatitis C virus entry and early steps of infection. SCIENTIFICA 2012; 2012:709853. [PMID: 24278733 PMCID: PMC3820461 DOI: 10.6064/2012/709853] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/10/2012] [Accepted: 10/31/2012] [Indexed: 06/02/2023]
Abstract
Viruses are obligate intracellular agents that depend on host cells for successful propagation, hijacking cellular machineries to their own profit. The molecular interplay between host factors and invading viruses is a continuous coevolutionary process that determines viral host range and pathogenesis. The hepatitis C virus (HCV) is a strictly human pathogen, causing chronic liver injuries accompanied by lipid disorders. Upon infection, in addition to protein-protein and protein-RNA interactions usual for such a positive-strand RNA virus, HCV relies on protein-lipid interactions at multiple steps of its life cycle to establish persistent infection, making use of hepatic lipid pathways. This paper focuses on lipoproteins in HCV entry and on receptors and enzymes involved in lipid metabolism that HCV exploits to enter hepatocytes.
Collapse
Affiliation(s)
- Eve-Isabelle Pécheur
- Department of Mechanisms of Chronic Hepatitis B and C, Centre de Recherche en Cancérologie de Lyon, 69008 Lyon, France
- Inserm U1052/CNRS UMR 5286, CRCL, Université de Lyon, 151 Cours Albert Thomas, 69424 Lyon Cedex 03, France
| |
Collapse
|
25
|
Corbin IR, Ng KK, Ding L, Jurisicova A, Zheng G. Near-infrared fluorescent imaging of metastatic ovarian cancer using folate receptor-targeted high-density lipoprotein nanocarriers. Nanomedicine (Lond) 2012; 8:875-90. [PMID: 23067398 DOI: 10.2217/nnm.12.137] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
AIM The targeting efficiency of folate receptor-α (FR-α)-targeted high-density lipoprotein nanoparticles (HDL NPs) was evaluated in a syngeneic mouse model of ovarian cancer. MATERIALS & METHODS Folic acid was conjugated to the surface of fluorescent-labeled HDL NPs. In vivo tumor targeting of folic acid-HDL NPs and HDL NPs were evaluated in mice with metastatic ovarian cancer following intravenous or intraperitoneal (ip.) administration. RESULTS & DISCUSSION Intravenous FR-α-targeted HDL resulted in high uptake of the fluorescent nanoparticle in host liver and spleen. The ip. injection of fluorescent HDL produced moderate fluorescence throughout the abdomen. Conversely, animals receiving the ip. FR-α-targeted HDL showed a high fluorescence signal in ovarian tumors, surpassing that seen in all of the host tissues. CONCLUSION The authors' findings demonstrate that the combination of local-regional ip. administration and FR-α-directed nanoparticles provides an enhanced approach to selectively targeting ovarian cancer cells for drug treatment.
Collapse
Affiliation(s)
- Ian R Corbin
- Advanced Imaging Research Center, University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390, USA.
| | | | | | | | | |
Collapse
|
26
|
Scavenger receptor class B type I and the hypervariable region-1 of hepatitis C virus in cell entry and neutralisation. Expert Rev Mol Med 2011; 13:e13. [PMID: 21489334 DOI: 10.1017/s1462399411001785] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Hepatitis C virus (HCV) infection is a leading cause of chronic liver disease worldwide and represents a major public health problem. Viral attachment and entry - the first encounter of the virus with the host cell - are major targets of neutralising immune responses. Thus, a detailed understanding of the HCV entry process offers interesting opportunities for the development of novel therapeutic strategies. Different cellular or soluble host factors mediate HCV entry, and considerable progress has been made in recent years to decipher how they induce HCV attachment, internalisation and membrane fusion. Among these factors, the scavenger receptor class B type I (SR-BI/SCARB1) is essential for HCV replication in vitro, through its interaction with the HCV E1E2 surface glycoproteins and, more particularly, the HVR1 segment located in the E2 protein. SR-BI is an interesting receptor because HCV, whose replication cycle intersects with lipoprotein metabolism, seems to exploit some aspects of its physiological functions, such as cholesterol transfer from high-density lipoprotein (HDL), during cell entry. SR-BI is also involved in neutralisation attenuation and therefore could be an important target for therapeutic intervention. Recent results suggest that it should be possible to identify inhibitors of the interaction of HCV with SR-BI that do not impair its important physiological properties, as discussed in this review.
Collapse
|
27
|
Nieland TJ, Xu S, Penman M, Krieger M. Negatively cooperative binding of high-density lipoprotein to the HDL receptor SR-BI. Biochemistry 2011; 50:1818-30. [PMID: 21254782 PMCID: PMC3065119 DOI: 10.1021/bi101657j] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Scavenger receptor class B, type I (SR-BI), is a high-density lipoprotein (HDL) receptor, which also binds low-density lipoprotein (LDL), and mediates the cellular selective uptake of cholesteryl esters from lipoproteins. SR-BI also is a coreceptor for hepatitis C virus and a signaling receptor that regulates cell metabolism. Many investigators have reported that lipoproteins bind to SR-BI via a single class of independent (not interacting), high-affinity binding sites (one site model). We have reinvestigated the ligand concentration dependence of (125)I-HDL binding to SR-BI and SR-BI-mediated specific uptake of [(3)H]CE from [(3)H]CE-HDL using an expanded range of ligand concentrations (<1 μg of protein/mL, lower than previously reported). Scatchard and nonlinear least-squares model fitting analyses of the binding and uptake data were both inconsistent with a single class of independent binding sites binding univalent lipoprotein ligands. The data are best fit by models in which SR-BI has either two independent classes of binding sites or one class of sites exhibiting negative cooperativity due to either classic allostery or ensemble effects ("lattice model"). Similar results were observed for LDL. Application of the "infinite dilution" dissociation rate method established that the binding of (125)I-HDL to SR-BI at 4 °C exhibits negative cooperativity. The unexpected complexity of the interactions of lipoproteins with SR-BI should be taken into account when interpreting the results of experiments that explore the mechanism(s) by which SR-BI mediates ligand binding, lipid transport, and cell signaling.
Collapse
Affiliation(s)
- Thomas J.F. Nieland
- Department of Biology, Massachusetts Institute of Technology, Room 68-483, 77 Massachusetts Avenue, Cambridge, MA 02139
- Broad Institute of Harvard and MIT, 7 Cambridge Center, Cambridge, MA 02142
| | - Shangzhe Xu
- Department of Biology, Massachusetts Institute of Technology, Room 68-483, 77 Massachusetts Avenue, Cambridge, MA 02139
| | - Marsha Penman
- Department of Biology, Massachusetts Institute of Technology, Room 68-483, 77 Massachusetts Avenue, Cambridge, MA 02139
| | - Monty Krieger
- Department of Biology, Massachusetts Institute of Technology, Room 68-483, 77 Massachusetts Avenue, Cambridge, MA 02139
| |
Collapse
|
28
|
Nieland TJF, Xu S, Penman M, Krieger M. Negatively cooperative binding of high-density lipoprotein to the HDL receptor SR-BI. Biochemistry 2011. [PMID: 21254782 DOI: 10.1021/bi1011657j] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Scavenger receptor class B, type I (SR-BI), is a high-density lipoprotein (HDL) receptor, which also binds low-density lipoprotein (LDL), and mediates the cellular selective uptake of cholesteryl esters from lipoproteins. SR-BI also is a coreceptor for hepatitis C virus and a signaling receptor that regulates cell metabolism. Many investigators have reported that lipoproteins bind to SR-BI via a single class of independent (not interacting), high-affinity binding sites (one site model). We have reinvestigated the ligand concentration dependence of (125)I-HDL binding to SR-BI and SR-BI-mediated specific uptake of [(3)H]CE from [(3)H]CE-HDL using an expanded range of ligand concentrations (<1 μg of protein/mL, lower than previously reported). Scatchard and nonlinear least-squares model fitting analyses of the binding and uptake data were both inconsistent with a single class of independent binding sites binding univalent lipoprotein ligands. The data are best fit by models in which SR-BI has either two independent classes of binding sites or one class of sites exhibiting negative cooperativity due to either classic allostery or ensemble effects ("lattice model"). Similar results were observed for LDL. Application of the "infinite dilution" dissociation rate method established that the binding of (125)I-HDL to SR-BI at 4 °C exhibits negative cooperativity. The unexpected complexity of the interactions of lipoproteins with SR-BI should be taken into account when interpreting the results of experiments that explore the mechanism(s) by which SR-BI mediates ligand binding, lipid transport, and cell signaling.
Collapse
Affiliation(s)
- Thomas J F Nieland
- Department of Biology, Massachusetts Institute of Technology, Room 68-483, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | | | | | | |
Collapse
|
29
|
Abstract
High density lipoprotein (HDL) possesses important anti-atherogenic properties and this review addresses the molecular mechanisms underlying these functions. The structures and cholesterol transport abilities of HDL particles are determined by the properties of their exchangeable apolipoprotein (apo) components. ApoA-I and apoE, which are the best characterized in structural terms, contain a series of amphipathic alpha-helical repeats. The helices located in the amino-terminal two-thirds of the molecule adopt a helix bundle structure while the carboxy-terminal segment forms a separately folded, relatively disorganized, domain. The latter domain initiates lipid binding and this interaction induces changes in conformation; the alpha-helix content increases and the amino-terminal helix bundle can open subsequently. These conformational changes alter the abilities of apoA-I and apoE to function as ligands for their receptors. The apoA-I and apoE molecules possess detergent-like properties and they can solubilize vesicular phospholipid to create discoidal HDL particles with hydrodynamic diameters of ~10 nm. In the case of apoA-I, such a particle is stabilized by two protein molecules arranged in an anti-parallel, double-belt, conformation around the edge of the disc. The abilities of apoA-I and apoE to solubilize phospholipid and stabilize HDL particles enable these proteins to be partners with ABCA1 in mediating efflux of cellular phospholipid and cholesterol, and the biogenesis of HDL particles. ApoA-I-containing nascent HDL particles play a critical role in cholesterol transport in the circulation whereas apoE-containing HDL particles mediate cholesterol transport in the brain. The mechanisms by which HDL particles are remodeled by lipases and lipid transfer proteins, and interact with SR-BI to deliver cholesterol to cells, are reviewed.
Collapse
Affiliation(s)
- Sissel Lund-Katz
- The Children’s Hospital of Philadelphia, Philadelphia, PA 19104-4318, USA
| | | |
Collapse
|
30
|
de Souza JA, Vindis C, Nègre-Salvayre A, Rye KA, Couturier M, Therond P, Chantepie S, Salvayre R, Chapman MJ, Kontush A. Small, dense HDL 3 particles attenuate apoptosis in endothelial cells: pivotal role of apolipoprotein A-I. J Cell Mol Med 2009; 14:608-20. [PMID: 19243471 PMCID: PMC3823460 DOI: 10.1111/j.1582-4934.2009.00713.x] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Plasma high-density lipoproteins (HDLs) protect endothelial cells against apoptosis induced by oxidized low-density lipoprotein (oxLDL). The specific component(s) of HDLs implicated in such cytoprotection remain(s) to be identified. Human microvascular endothelial cells (HMEC-1) were incubated with mildly oxLDL in the presence or absence of each of five physicochemically distinct HDL subpopulations fractionated from normolipidemic human plasma (n= 7) by isopycnic density gradient ultracentrifugation. All HDL subfractions protected HMEC-1 against oxLDL-induced primary apoptosis as revealed by nucleic acid staining, annexin V binding, quantitative DNA fragmentation, inhibition of caspase-3 activity and reduction of cytoplasmic release of cytochrome c and apoptosis-inducing factor. Small, dense HDL 3c displayed twofold superior intrinsic cytoprotective activity (as determined by mitochondrial dehydrogenase activity) relative to large, light HDL 2b on a per particle basis (P < 0.05). Equally, all HDL subfractions attenuated intracellular generation of reactive oxygen species (ROS); such anti-oxidative activity diminished from HDL 3c to HDL 2b. The HDL protein moiety, in which apolipoprotein A-I (apoA-I) predominated, accounted for ∼70% of HDL anti-apoptotic activity. Furthermore, HDL reconstituted with apoA-I, cholesterol and phospholipid potently protected HMEC-1 from apoptosis. By contrast, modification of the content of sphingosine-1-phosphate in HDL did not significantly alter cytoprotection. We conclude that small, dense, lipid-poor HDL 3 potently protects endothelial cells from primary apoptosis and intracellular ROS generation induced by mildly oxLDL, and that apoA-I is pivotal to such protection.
Collapse
Affiliation(s)
- Juliana A de Souza
- Dyslipoproteinemia and Atherosclerosis Research Unit (UMR939), National Institute for Health and Medical Research (INSERM), Hôpital de la Pitié, Paris, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
de Haan W, Out R, Berbée JFP, van der Hoogt CC, van Dijk KW, van Berkel TJC, Romijn JA, Jukema JW, Havekes LM, Rensen PCN. Apolipoprotein CI inhibits scavenger receptor BI and increases plasma HDL levels in vivo. Biochem Biophys Res Commun 2008; 377:1294-8. [PMID: 18992221 DOI: 10.1016/j.bbrc.2008.10.147] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2008] [Accepted: 10/30/2008] [Indexed: 10/21/2022]
Abstract
Apolipoprotein CI (apoCI) has been suggested to influence HDL metabolism by activation of LCAT and inhibition of HL and CETP. However, the effect of apoCI on scavenger receptor BI (SR-BI)-mediated uptake of HDL-cholesteryl esters (CE), as well as the net effect of apoCI on HDL metabolism in vivo is unknown. Therefore, we evaluated the effect of apoCI on the SR-BI-mediated uptake of HDL-CE in vitro and determined the net effect of apoCI on HDL metabolism in mice. Enrichment of HDL with apoCI dose-dependently decreased the SR-BI-dependent association of [(3)H]CE-labeled HDL with primary murine hepatocytes, similar to the established SR-BI-inhibitors apoCIII and oxLDL. ApoCI deficiency in mice gene dose-dependently decreased HDL-cholesterol levels. Adenovirus-mediated expression of human apoCI in mice increased HDL levels at a low dose and increased the HDL particle size at higher doses. We conclude that apoCI is a novel inhibitor of SR-BI in vitro and increases HDL levels in vivo.
Collapse
Affiliation(s)
- Willeke de Haan
- Dept. of General Internal Medicine, Leiden University Medical Center, P.O. Box 9600, Albinusdreef 2, 2300 RC Leiden, The Netherlands
| | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Van Eck M, Hoekstra M, Out R, Bos IST, Kruijt JK, Hildebrand RB, Van Berkel TJC. Scavenger receptor BI facilitates the metabolism of VLDL lipoproteins in vivo. J Lipid Res 2007; 49:136-46. [PMID: 17954936 DOI: 10.1194/jlr.m700355-jlr200] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Scavenger receptor class B type I (SR-BI) functions as an HDL receptor that promotes the selective uptake of cholesteryl esters (CEs). The physiological role of SR-BI in VLDL metabolism, however, is largely unknown. SR-BI deficiency resulted in elevated VLDL cholesterol levels, both on chow diet and upon challenge with high-cholesterol diets. To specifically elucidate the role of SR-BI in VLDL metabolism, the plasma clearance and hepatic uptake of (125)I-beta-VLDL were studied in SR-BI(+/+) and SR-BI(-/-) mice. At 20 min after injection, 66 +/- 2% of the injected dose was taken up by the liver in SR-BI(+/+) mice, as compared with only 22 +/- 4% (P = 0.0007) in SR-BI(-/-) mice. In vitro studies established that the B(max) of (125)I-beta-VLDL binding was reduced from 469 +/- 30 ng/mg in SR-BI(+/+) hepatocytes to 305 +/- 20 ng/mg (P = 0.01) in SR-BI(-/-) hepatocytes. Both in vivo and in vitro, limited to no selective uptake of CEs from beta-VLDL was found. Interestingly, HDL effectively competed for the association of beta-VLDL in the presence as well as in the absence of SR-BI, indicating a second common recognition site. In conclusion, SR-BI plays an important physiological role in the metabolism of VLDL (remnants).
Collapse
Affiliation(s)
- Miranda Van Eck
- Divison of Biopharmaceutics, Leiden/Amsterdam Center for Drug Research, Gorlaeus Laboratories, Leiden University, Leiden, The Netherlands.
| | | | | | | | | | | | | |
Collapse
|
33
|
Orlowski S, Coméra C, Tercé F, Collet X. Lipid rafts: dream or reality for cholesterol transporters? EUROPEAN BIOPHYSICS JOURNAL: EBJ 2007; 36:869-85. [PMID: 17576551 DOI: 10.1007/s00249-007-0193-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2007] [Revised: 05/11/2007] [Accepted: 05/15/2007] [Indexed: 01/12/2023]
Abstract
As a key constituent of the cell membranes, cholesterol is an endogenous component of mammalian cells of primary importance, and is thus subjected to highly regulated homeostasis at the cellular level as well as at the level of the whole body. This regulation requires adapted mechanisms favoring the handling of cholesterol in aqueous compartments, as well as its transfer into or out of membranes, involving membrane proteins. A membrane exhibits functional properties largely depending on its lipid composition and on its structural organization, which very often involves cholesterol-rich microdomains. Then there is the appealing possibility that cholesterol may regulate its own transmembrane transport at a purely functional level, independently of any transcriptional regulation based on cholesterol-sensitive nuclear factors controling the expression level of lipid transport proteins. Indeed, the main cholesterol "transporters" presently believed to mediate for instance the intestinal absorption of cholesterol, that are SR-BI, NPC1L1, ABCA1, ABCG1, ABCG5/G8 and even P-glycoprotein, all present privileged functional relationships with membrane cholesterol-containing microdomains. In particular, they all more or less clearly induce membrane disorganization, supposed to facilitate cholesterol exchanges with the close aqueous medium. The actual lipid substrates handled by these transporters are not yet unambiguously determined, but they likely concern the components of membrane microdomains. Conversely, raft alterations may provide specific modulations of the transporter activities, as well as they can induce indirect effects via local perturbations of the membrane. Finally, these cholesterol transporters undergo regulated intracellular trafficking, with presumably some relationships to rafts which remain to be clarified.
Collapse
Affiliation(s)
- Stéphane Orlowski
- SB2SM/IBTS and URA 2096 CNRS, CEA, Centre de Saclay, 91191, Gif-sur-Yvette cedex, France.
| | | | | | | |
Collapse
|
34
|
Tréguier M, Moreau M, Sposito A, Chapman MJ, Huby T. LDL particle subspecies are distinct in their capacity to mediate free cholesterol efflux via the SR-BI/Cla-1 receptor. Biochim Biophys Acta Mol Cell Biol Lipids 2006; 1771:129-38. [PMID: 17240192 DOI: 10.1016/j.bbalip.2006.12.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2006] [Revised: 11/25/2006] [Accepted: 12/14/2006] [Indexed: 10/23/2022]
Abstract
The human scavenger receptor SR-BI/Cla-1 promotes efflux of free cholesterol from cells to both high-density and low-density lipoproteins (HDL, LDL). SR-BI/Cla-1-mediated cholesterol efflux to HDL is dependent on particle size, lipid content and apolipoprotein conformation; in contrast, the capacity of LDL subspecies to accept cellular cholesterol via this receptor is indeterminate. Cholesterol efflux assays were performed with CHO cells stably transfected with Cla-1 cDNA. Expression of Cla-1 in CHO cells induced elevation in total cholesterol efflux to plasma, LDL and HDL. Such Cla-1-specific efflux was abrogated by addition of anti-Cla-1 antibody. LDL were fractionated into five subspecies either on the basis of hydrated density or size. Among LDL subfractions, small dense LDL (sdLDL) were 1.5-to 3-fold less active acceptors for Cla-1-mediated cellular cholesterol efflux. Equally, sdLDL markedly reduced Cla-1-specific cholesterol efflux to large buoyant LDL in a dose-dependent manner. Conversely, sdLDL did not influence efflux to HDL(2). These findings provide evidence that LDL particles are heterogeneous in their capacity to promote Cla-1-mediated cholesterol efflux. Relative to HDL(2), large buoyant LDL may constitute physiologically-relevant acceptors for cholesterol efflux via Cla-1.
Collapse
Affiliation(s)
- Morgan Tréguier
- INSERM U551, Dyslipoproteinemia and Atherosclerosis Research Unit, Hôpital de la Pitié, 83 Boulevard de l'Hôpital, 75651 Paris Cedex 13, France
| | | | | | | | | |
Collapse
|
35
|
Kleveland EJ, Syvertsen BL, Ruyter B, Vegusdal A, Jørgensen SM, Gjøen T. Characterization of scavenger receptor class B, type I in Atlantic salmon (Salmo salar L.). Lipids 2006; 41:1017-27. [PMID: 17263301 DOI: 10.1007/s11745-006-5052-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
The scavenger receptor class B, type I (SR-BI) is an important player in regulation of mammalian lipid homeostasis. We therefore wanted to study this receptor in Atlantic salmon (Salmo salar L.), which requires a diet with particular high lipid content. We have for the first time cloned and characterized SR-BI from a salmonid fish. The predicted 494 amino acid protein contained two transmembrane domains, several putative N-glycosylation sites, and showed 72% sequence identity with the predicted homolog from zebrafish. SR-BI expression was analyzed by reverse transcription Real-Time PCR in several tissues, and a high relative expression in salmon midgut was detected, which may suggest that SR-BI has a role in uptake of lipids from the diet. We also expressed a construct of salmon myc-tagged SR-BI in salmon TO cells and HeLa cells, which gave a protein of approximately 80 kDa on reducing SDS-PAGE using an antibody against the myc-epitope. Immunofluorescence microscopy analyses of the salmon SR-BI protein in transiently transfected HeLa cells revealed staining in the cell periphery and in some intracellular membranes, but not in the nucleus, which indicated that the salmon protein may be a functional membrane protein. We also observed a high degree of co-localization using an anti-peptide SR-BI antiserum. We found that 20 microg mL(-1) insulin up-regulated the SR-BI mRNA levels in primary cultures of salmon hepatocytes relative to untreated cells. Oleic acid, EPA, DHA, or dexamethasone did not affect the relative expression of SR-BI in this liver model system. In conclusion, the salmon SR-BI cDNA encoded a protein with several features common to those of mammalian species. SR-BI gene expression was high in the intestine, which leads us to propose that SR-BI may contribute to the uptake of lipids from the diet.
Collapse
Affiliation(s)
- E J Kleveland
- Department of Pharmaceutical Biosciences, School of Pharmacy, University of Oslo, Oslo, Norway
| | | | | | | | | | | |
Collapse
|
36
|
Zhou W, Scott SA, Shelton SB, Crutcher KA. Cathepsin D-mediated proteolysis of apolipoprotein E: possible role in Alzheimer's disease. Neuroscience 2006; 143:689-701. [PMID: 16997486 DOI: 10.1016/j.neuroscience.2006.08.019] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2006] [Revised: 06/12/2006] [Accepted: 08/09/2006] [Indexed: 10/24/2022]
Abstract
Proteolysis of apolipoprotein E (apoE) may be involved in the pathogenesis of Alzheimer's disease (AD). We previously identified aspartic protease(s) as possibly contributing to the proteolysis of apoE in human brain homogenates. The current study used biochemical and immunohistochemical methods to examine whether cathepsin D (catD) and cathepsin E (catE), candidate aspartic proteases, may be involved in apoE proteolysis. CatD was found to proteolyze both lipid-free recombinant full-length human apoE and lipidated human plasma full-length apoE (apoE4/dipalmitoylphosphatidylcholine-reconstituted discs). CatE was found to proteolyze lipid-free recombinant human apoE to a much greater extent than lipidated apoE. This proteolysis, as well as proteolysis of human apoE added to brain homogenates from apoE-deficient mice, was inhibited by pepstatin A (an aspartic protease inhibitor), but not by phenylmethanesulfonyl fluoride (a serine protease inhibitor). The major apoE fragment obtained with catD included the receptor-binding domain and had an apparent molecular weight similar to that found in human brain homogenates. There was little immunoreactivity for catE in AD brain tissue sections. In contrast, qualitative and quantitative analyses of immunostained sections of the frontal cortex revealed that catD and apoE are colocalized in a subset of predominantly dense-core neuritic plaques and in some neurofibrillary tangles. A positive correlation was observed between estimated duration of illness and the percentage of apoE-positive plaques that were also catD-positive. These results suggest that aspartic proteases, catD in particular, may be involved in proteolysis of apoE and perhaps contribute to the generation of apoE fragments previously implicated in AD pathology.
Collapse
Affiliation(s)
- W Zhou
- Department of Neurosurgery, University of Cincinnati College of Medicine, 231 Albert Sabin Way, Cincinnati, OH 45267-0515, USA
| | | | | | | |
Collapse
|
37
|
Ajees AA, Anantharamaiah GM, Mishra VK, Hussain MM, Murthy HMK. Crystal structure of human apolipoprotein A-I: insights into its protective effect against cardiovascular diseases. Proc Natl Acad Sci U S A 2006; 103:2126-31. [PMID: 16452169 PMCID: PMC1413691 DOI: 10.1073/pnas.0506877103] [Citation(s) in RCA: 160] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Despite three decades of extensive studies on human apolipoprotein A-I (apoA-I), the major protein component in high-density lipoproteins, the molecular basis for its antiatherogenic function is elusive, in part because of lack of a structure of the full-length protein. We describe here the crystal structure of lipid-free apoA-I at 2.4 A. The structure shows that apoA-I is comprised of an N-terminal four-helix bundle and two C-terminal helices. The N-terminal domain plays a prominent role in maintaining its lipid-free conformation, indicating that mutants with truncations in this region form inadequate models for explaining functional properties of apoA-I. A model for transformation of the lipid-free conformation to the high-density lipoprotein-bound form follows from an analysis of solvent-accessible hydrophobic patches on the surface of the structure and their proximity to the hydrophobic core of the four-helix bundle. The crystal structure of human apoA-I displays a hitherto-unobserved array of positively and negatively charged areas on the surface. Positioning of the charged surface patches relative to hydrophobic regions near the C terminus of the protein offers insights into its interaction with cell-surface components of the reverse cholesterol transport pathway and antiatherogenic properties of this protein. This structure provides a much-needed structural template for exploration of molecular mechanisms by which human apoA-I ameliorates atherosclerosis and inflammatory diseases.
Collapse
Affiliation(s)
| | - G. M. Anantharamaiah
- Atherosclerosis Research Unit and Departments of Medicine, Biochemistry, and Molecular Genetics, University of Alabama, 1530 3rd Avenue South, Birmingham, AL 35294; and
| | - Vinod K. Mishra
- Atherosclerosis Research Unit and Departments of Medicine, Biochemistry, and Molecular Genetics, University of Alabama, 1530 3rd Avenue South, Birmingham, AL 35294; and
| | - M. Mahmood Hussain
- Departments of Anatomy, Cell Biology, and Pediatrics, State University of New York Downstate Medical Center, Brooklyn, NY 11203
| | - H. M. Krishna Murthy
- *Center for Biophysical Sciences and Engineering and
- To whom correspondence should be addressed. E-mail:
| |
Collapse
|
38
|
Klerkx AHEM, El Harchaoui K, van der Steeg WA, Boekholdt SM, Stroes ESG, Kastelein JJP, Kuivenhoven JA. Cholesteryl ester transfer protein (CETP) inhibition beyond raising high-density lipoprotein cholesterol levels: pathways by which modulation of CETP activity may alter atherogenesis. Arterioscler Thromb Vasc Biol 2006; 26:706-15. [PMID: 16439711 DOI: 10.1161/01.atv.0000205595.19612.c9] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Raising high-density lipoprotein cholesterol (HDL-C) is a promising strategy in the struggle to prevent cardiovascular disease, and cholesteryl ester transfer protein (CETP) inhibitors have been developed to accomplish this. The first results are encouraging, and, in fact, in rabbits, inhibition of CETP reduces atherosclerosis. Because human data regarding the reduction of atheroma burden require more time, the biochemical mechanisms underlying the putative atheroprotection of CETP inhibitors are currently dissected, and several pathways have emerged. First, CETP inhibition increases HDL-C and reduces low-density lipoprotein cholesterol (LDL-C) levels consistent with CETP lipid transfer activity and its role in reverse cholesterol transport (RCT). This coincides with putative beneficial increases in both HDL and LDL size. However, many aspects regarding the impact of CETP inhibition on the RCT pathway remain elusive, in particular whether the first step concerning cholesterol efflux from peripheral tissues to HDL is influenced. Moreover, the relevance of scavenger receptor BI and consequently the central role of HDL in human RCT is still unclear. Second, CETP inhibition was shown recently to increase antioxidant enzymes associated with HDL, in turn associated with decreased oxidation of LDL. Atheroprotection in man is currently anticipated based on the improvement of these biochemical parameters known to influence atherosclerosis, but final confirmation regarding the impact of CETP inhibition on cardiovascular outcome will have to come from trials evaluating clinical end points.
Collapse
Affiliation(s)
- Anke H E M Klerkx
- Department of Experimental Vascular Medicine, Academic Medical Center, Amsterdam, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
39
|
Truong TQ, Aubin D, Bourgeois P, Falstrault L, Brissette L. Opposite effect of caveolin-1 in the metabolism of high-density and low-density lipoproteins. Biochim Biophys Acta Mol Cell Biol Lipids 2006; 1761:24-36. [PMID: 16443388 DOI: 10.1016/j.bbalip.2005.12.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2005] [Revised: 12/06/2005] [Accepted: 12/12/2005] [Indexed: 11/21/2022]
Abstract
Receptors of the scavenger class B family were reported to be localized in caveolae, the cell surface microdomains rich in free cholesterol and glycosphyngolipids, which are characterized by the presence of caveolin-1. Parenchymal hepatic and hepatoma HepG2 cells express very low levels of caveolin-1. In the present study, stable transformants of HepG2 cells expressing caveolin-1 were generated to address the effect of caveolin-1 on receptor activity. Compared to normal cells, these cells show higher (125)I-bovine serum albumin (BSA) uptake and cholesterol efflux, two indicators of functional caveolae. By immunoprecipitation, cell fractionation and confocal analyses, we found that caveolin-1 is well colocalized with the cluster of differentiation-36 (CD36) and the low-density lipoprotein (LDL) receptor (LDLr) but to a lesser extent with the scavenger receptor class B type I (SR-BI) in HepG2 cells expressing caveolin-1. However, caveolin-1 expression favors the dimerization of SR-BI. Two clones of cells expressing caveolin-1 were investigated for their lipoprotein metabolism activity. Compared to normal cells, these cells show a 71-144% increase in (125)I-LDL degradation. The analysis of the cholesteryl esters (CE)-selective uptake (CE association minus protein association) revealed that the expression of caveolin-1 in HepG2 cells decreases by 59%-73% LDL-CE selective uptake and increases high-density lipoprotein (HDL)-CE selective uptake by 44%-66%. We conclude that the expression of caveolin-1 in HepG2 cells moves the balance of LDL degradation/CE selective uptake towards degradation and favors HDL-CE selective uptake. Thus, in the normal hepatic parenchymal situation where caveolin-1 is poorly expressed, LDL-CE selective uptake is the preferred pathway.
Collapse
Affiliation(s)
- To Quyen Truong
- Département des Sciences Biologiques, Université du Québec à Montréal, C.P. 8888, Succursale Centre-ville, Montréal, Québec, Canada H3C 3P8.
| | | | | | | | | |
Collapse
|
40
|
Ren X, Zhao L, Sivashanmugam A, Miao Y, Korando L, Yang Z, Reardon CA, Getz GS, Brouillette CG, Jerome WG, Wang J. Engineering mouse apolipoprotein A-I into a monomeric, active protein useful for structural determination. Biochemistry 2006; 44:14907-19. [PMID: 16274238 DOI: 10.1021/bi0508385] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Apolipoprotein AI (apoAI), the major protein component of HDL, is one of the best predictors of coronary artery disease (CAD), with high apoAI and HDL levels being correlated with low occurrences of CAD. The primary function of apoAI is to recruit phospholipid and cholesterol for assembly of HDL particles. Like other exchangeable apolipoproteins, lipid-free apoAI forms a mixture of different oligomers even at 1.0 mg/mL. This self-association property of the exchangeable apolipoproteins is closely associated with the lipoprotein-binding activity of this protein family. It is unclear if the self-association property of apolipoprotein is required for its lipoprotein-binding activity. We developed a novel method for engineering an oligomeric protein to a monomeric, biologically active protein. Using this method, we generated a monomeric mouse apoAI mutant that is active. This mutant contains the first 216 residues of mouse apoAI and replaces six hydrophobic residues with either polar or smaller hydrophobic residues at the defined positions (V118A/A119S/L121Q/T191S/T195S/T199S). Cross-linking results show that this mutant is greater than 90% monomeric at 8 mg/mL. CD, DSC, and NMR results indicate that the mutant maintains an identical secondary, tertiary structure and stability as those of the wild-type mouse apoAI. Lipid-binding assays suggest that the mutant shares an equal lipoprotein-binding activity as that of the wild-type apoAI. In addition, both the monomeric mutant and the wild-type protein make nearly identical rHDL particles. With this monomeric mouse apoAI, high-quality NMR data has been collected, allowing for the NMR structural determination of lipid-free apoAI. On the basis of these results, we conclude that this apoAI mutant is a monomeric, active apoAI useful for structural determination.
Collapse
Affiliation(s)
- Xuefeng Ren
- Department of Biochemistry and Molecular Biology, School of Medicine, Wayne State University, Detroit, Michigan 48201, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Stratman NC, Castle CK, Taylor BM, Epps DE, Melchior GW, Carter DB. Isoform-specific interactions of human apolipoprotein E to an intermediate conformation of human Alzheimer amyloid-beta peptide. Chem Phys Lipids 2005; 137:52-61. [PMID: 16140289 DOI: 10.1016/j.chemphyslip.2005.06.005] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2005] [Revised: 06/22/2005] [Accepted: 06/29/2005] [Indexed: 10/25/2022]
Abstract
Brain plaque deposits of amyloid-beta peptide (Abeta) is a pathological hallmark of Alzheimer's disease (AD) and apolipoprotein E (apoE) is thought to be involved in its deposition. One hypothesis for the role of apoE in the pathogenesis of AD is that apoE may be involved in deposition or clearance of Abeta by direct protein-to-protein interaction. Lipidated apoE4 bound preferentially to an intermediate aggregated form of Abeta and formed two- to three-fold more binding complexes than isoforms apoE2 or apoE3. The interaction was detected by a sandwich ELISA with capture antibodies specific for the N-terminus of apoE, whereas the interaction was not recognized with a C-terminal antibody. The observations indicate that the C-terminus of apoE4 interacts with the intermediate form of Abeta. The differential risk of AD related to apoE genotype may be the result of an enhanced capacity of apoE4 binding to an intermediate aggregated form of Abeta.
Collapse
|
42
|
Huard K, Bourgeois P, Rhainds D, Falstrault L, Cohn JS, Brissette L. Apolipoproteins C-II and C-III inhibit selective uptake of low- and high-density lipoprotein cholesteryl esters in HepG2 cells. Int J Biochem Cell Biol 2005; 37:1308-18. [PMID: 15778093 DOI: 10.1016/j.biocel.2005.01.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2004] [Accepted: 01/18/2005] [Indexed: 01/26/2023]
Abstract
Plasma low- and high-density lipoproteins (LDL and HDL) are cleared from the circulation by specific receptors and are either totally degraded or their cholesteryl esters (CE) are selectively delivered to cells by receptors such as the scavenger receptor class B type I (SR-BI). The aim of the present study was to define the effect of apoC-II and apoC-III on the uptake of LDL and HDL by HepG2 cells. Stable transformants were obtained with sense or antisense strategies that secrete 47-294% the normal level of apoC-II or 60-200% that of apoC-III. Different levels of secreted apoC-II or apoC-III had little effect on LDL and HDL protein degradation by HepG2 cells. However, compared to controls, cells under-expressing apoC-II showed a 160% higher capacity to selectively take up HDL-CE, while cells under-expressing apoC-III demonstrated 70 and 160% higher capacity to take up CE from LDL and HDL, respectively. In experiments conducted with exogenously added apoC-II or apoC-III, no significant effect was observed on lipoprotein-protein association/degradation; however, LDL-CE and HDL-CE selective uptake was significantly reduced in a dose-dependent manner. These results indicate that apoC-II and apoC-III inhibit CE-selective uptake.
Collapse
Affiliation(s)
- Karine Huard
- Département des Sciences Biologiques, Université du Québec à Montréal, C.P. 8888, Succ. Centre-ville, Montréal, Que., Canada H3C 3P8
| | | | | | | | | | | |
Collapse
|
43
|
Domingo N, Mastellone I, Grès S, Marin V, Lorec AM, Tosini F, Grosclaude J, Farnarier C, Chanussot F. The endothelial cholesterol efflux is promoted by the high-density lipoprotein anionic peptide factor. Metabolism 2005; 54:1087-94. [PMID: 16092060 DOI: 10.1016/j.metabol.2005.03.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The prevention of atherosclerosis depends on the high-density lipoprotein (HDL) capacity to stimulate the efflux of unesterified cholesterol (UC). We tested here the effects of 2 HDL apolipoproteins, apo A-I and the 7-kd anionic peptide factor (APF), on the UC efflux by human endothelial ECV 304 cells in culture. Apolipoprotein A-I (10 micromol/L) or APF (3.5 micromol/L) in lipid-free forms or small particles (13 nm with apo A-I or 19 nm with APF) were incubated in the presence of [4-14C]UC. The phosphatidylcholines (PCs) were present either at a low level (0.35 mmol/L with apo A-I or 0.20 mmol/L with APF) or at a high level (1 mmol/L with apo A-I). We also tested either large 53-nm bile lipoprotein complex-like particles (3.5 micromol/L APF [13 microg/500 microL]) with a high PC level (0.65 mmol/L) or a 9-residue synthetic peptide (13 microg/500 microL), derived from the NH2-terminal domain of HDL3-APF, in a lipid-free or low-lipidated (0.20 mmol/L PCs) form. A control was developed in absence of the added compounds. A rapid [4-14C]UC efflux mediated by APF added in free form or in 19-nm complexes was 2.2- to 2.3-fold higher than that mediated by apo A-I in free form or in 13-nm particles (P < .05). The level of this high APF-related efflux was comparable with that obtained with the 12-nm native HDLs (10 micromol/L apo A-I) or free PCs (1 mmol/L). The increase in the UC efflux was much more limited (1.4-fold) in the presence of the 53-nm APF/high-PC particles, but it was higher than that mediated by apo A-I. In addition, the efflux mediated by the synthetic peptide, in lipid-free or low-lipidated form, constituted the major part of that related to the full-length APF. Thus, all these particles are very active HDL components, able to act as cholesterol acceptors. Interestingly, we further showed a new anti-atherogenic property of APF as well as its metabolic importance and clinical relevance. By its involvement in the first step of the reverse cholesterol transport, APF could reduce the risk of cardiovascular disease.
Collapse
Affiliation(s)
- Nicole Domingo
- Department of UMR 476 INSERM/1260 INRA, Faculty of Medicine, 27 bd Jean Moulin, 13385 Marseille, France
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Asztalos BF, de la Llera-Moya M, Dallal GE, Horvath KV, Schaefer EJ, Rothblat GH. Differential effects of HDL subpopulations on cellular ABCA1- and SR-BI-mediated cholesterol efflux. J Lipid Res 2005; 46:2246-53. [PMID: 16061948 DOI: 10.1194/jlr.m500187-jlr200] [Citation(s) in RCA: 188] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Our objective was to evaluate the associations of individual apolipoprotein A-I (apoA-I)-containing HDL subpopulation levels with ABCA1- and scavenger receptor class B type I (SR-BI)-mediated cellular cholesterol efflux. HDL subpopulations were measured by nondenaturing two-dimensional gel electrophoresis from 105 male subjects selected with various levels of apoA-I in pre-beta-1, alpha-1, and alpha-3 HDL particles. ApoB-containing lipoprotein-depleted serum was incubated with [(3)H]cholesterol-labeled cells to measure efflux. The difference in efflux between control and ABCA1-upregulated J774 macrophages was taken as a measure of ABCA1-mediated efflux. SR-BI-mediated efflux was determined using cholesterol-labeled Fu5AH hepatoma cells. Fractional efflux values obtained from these two cell systems were correlated with the levels of individual HDL subpopulations. A multivariate analysis showed that two HDL subspecies correlated significantly with ABCA1-mediated efflux: small, lipid-poor pre-beta-1 particles (P=0.0022) and intermediate-sized alpha-2 particles (P=0.0477). With regard to SR-BI-mediated efflux, multivariate analysis revealed significant correlations with alpha-2 (P=0.0004), alpha-1 (P=0.0030), pre-beta-1 (P=0.0056), and alpha-3 (P=0.0127) HDL particles. These data demonstrate that the small, lipid-poor pre-beta-1 HDL has the strongest association with ABCA1-mediated cholesterol even in the presence of all other HDL subpopulations. Cholesterol efflux via the SR-BI pathway is associated with several HDL subpopulations with different apolipoprotein composition, lipid content, and size.
Collapse
Affiliation(s)
- Bela F Asztalos
- Lipid Metabolism Laboratory, Jean Mayer U.S. Department of Agriculture Human Nutrition Research Center on Aging at Tufts University, Boston, MA, USA.
| | | | | | | | | | | |
Collapse
|
45
|
Van Eck M, Pennings M, Hoekstra M, Out R, Van Berkel TJ. Scavenger receptor BI and ATP-binding cassette transporter A1 in reverse cholesterol transport and atherosclerosis. Curr Opin Lipidol 2005; 16:307-15. [PMID: 15891392 DOI: 10.1097/01.mol.0000169351.28019.04] [Citation(s) in RCA: 128] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW The appearance of scavenger receptor class B type I (SR-BI) and ATP-binding cassette transporter A1 (ABCA1) in macrophages and liver implicates these transporters in different stages of reverse cholesterol transport. This review focuses on the role of SR-BI and ABCA1 in reverse cholesterol transport in the context of atherosclerotic lesion development. RECENT FINDINGS Recent studies indicate that hepatic expression of ABCA1 and SR-BI is important for the generation of nascent HDL and the delivery of HDL cholesteryl esters to the liver, respectively. Although macrophage SR-BI and ABCA1 do not contribute significantly to circulating HDL levels, the perpetual cycle of HDL lipidation and delipidation by the liver ensures the availability of acceptors for cholesterol efflux that maintain cholesterol homeostasis in arterial macrophages, thereby reducing atherogenesis. In addition to its established role in the selective uptake of HDL cholesteryl esters, there is now evidence that hepatic SR-BI facilitates postprandial lipid metabolism, and that hepatic secretion of VLDL is dependent on ABCA1-mediated nascent HDL formation. Thus, remnant and HDL metabolism are more intimately intertwined in hepatic lipid metabolism than has previously been appreciated. SUMMARY Recent advances in the understanding of the role of ABCA1 and SR-BI in HDL metabolism and their atheroprotective properties indicate the significant potential of modulating ABCA1 and SR-BI expression in both arterial wall macrophages and the liver for the treatment of atherosclerotic coronary artery disease.
Collapse
Affiliation(s)
- Miranda Van Eck
- Division of Biopharmaceutics, Leiden/Amsterdam Center for Drug Research, Gorlaeus Laboratories, Leiden University, Leiden, The Netherlands.
| | | | | | | | | |
Collapse
|
46
|
Greenow K, Pearce NJ, Ramji DP. The key role of apolipoprotein E in atherosclerosis. J Mol Med (Berl) 2005; 83:329-42. [PMID: 15827760 DOI: 10.1007/s00109-004-0631-3] [Citation(s) in RCA: 167] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2004] [Accepted: 11/08/2004] [Indexed: 01/17/2023]
Abstract
Apolipoprotein E is a multifunctional protein that is synthesized by the liver and several peripheral tissues and cell types, including macrophages. The protein is involved in the efficient hepatic uptake of lipoprotein particles, stimulation of cholesterol efflux from macrophage foam cells in the atherosclerotic lesion, and the regulation of immune and inflammatory responses. Apolipoprotein E deficiency in mice leads to the development of atherosclerosis and re-expression of the protein reduces the extent of the disease. This review presents evidence for the potent anti-atherogenic action of apolipoprotein E and describes our current understanding of its multiple functions and regulation by factors implicated in the pathogenesis of cardiovascular disease.
Collapse
Affiliation(s)
- Kirsty Greenow
- Cardiff School of Biosciences, Cardiff University, Museum Avenue, P.O. Box 911, Cardiff CF10 3US, Wales, UK
| | | | | |
Collapse
|
47
|
Rhainds D, Bourgeois P, Bourret G, Huard K, Falstrault L, Brissette L. Localization and regulation of SR-BI in membrane rafts of HepG2 cells. J Cell Sci 2005; 117:3095-105. [PMID: 15226391 DOI: 10.1242/jcs.01182] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
The scavenger receptor class B, type I (SR-BI) mediates cholesteryl esters (CE) selective uptake from low density lipoprotein (LDL) and high-density lipoprotein (HDL) particles. In a number of tissues expressing caveolin, SR-BI is localized in caveolae. We show using detergent-free sucrose gradients that SR-BI is found in membrane rafts devoid of caveolin-1 in the human hepatoma HepG2 cell. Perturbation of the structure of HepG2 cell membrane rafts with cholesterol oxidase or sphingomyelinase decreased LDL-CE association due to selective uptake by 60%, while HDL3-CE selective uptake was increased 2.3-fold by cholesterol oxidase but was not affected by sphingomyelinase. Sequestration of membrane cholesterol with filipin III decreased LDL-CE selective uptake by 25%, while it had no effect on HDL3-CE selective uptake. Extraction of cell membrane cholesterol with beta-cyclodextrin increased LDL- and HDL3-CE selective uptake by 1.6-fold and 3-fold, respectively. We found that CE-selective uptake from both HDL and LDL occurs by a pathway involving retro-endocytosis in HepG2 cells. An analysis of the effect of SR-BI level on the expression of critical lipid sensor and lipid binding proteins was conducted with stable transformants of HepG2 cell overexpressing SR-BI. We found that liver-type fatty acid binding protein expression level is higher in SR-BI-overexpressing cells and that caveolin-1 and sterol response element binding protein-2 levels are reduced. Thus, in this hepatic cell model, SR-BI is associated with membrane rafts devoid of caveolin and its expression affects intracellular lipid binding and lipid sensor proteins. SR-BI-dependent LDL- and HDL-CE selective uptake are affected differently by the integrity of membrane rafts, but both occur by a retroendocytic pathway in HepG2 cells.
Collapse
Affiliation(s)
- David Rhainds
- Département des Sciences Biologiques, Université du Québec à Montréal, 1200 Saint-Alexandre, H3B 3H5, Canada
| | | | | | | | | | | |
Collapse
|
48
|
Zhang B, Fan P, Shimoji E, Xu H, Takeuchi K, Bian C, Saku K. Inhibition of Cholesteryl Ester Transfer Protein Activity by JTT-705 Increases Apolipoprotein E–Containing High-Density Lipoprotein and Favorably Affects the Function and Enzyme Composition of High-Density Lipoprotein in Rabbits. Arterioscler Thromb Vasc Biol 2004; 24:1910-5. [PMID: 15331428 DOI: 10.1161/01.atv.0000143389.00252.bc] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background—
Inhibition of cholesteryl ester transfer protein (CETP) is an efficient way to increase high-density lipoprotein (HDL) levels in humans. We investigated the effects of the inhibition of CETP activity by a CETP inhibitor, JTT-705, on the function and composition of HDL particles.
Methods and Results—
Japanese white rabbits were fed either normal rabbit chow LRC-4 (n=10) or a food admixture of LRC-4 and 0.75% JTT-705 (n=10) for 7 months. JTT-705 significantly inhibited CETP activities, increased HDL cholesterol (HDL-C) levels and the ratio of HDL
2
-C/HDL
3
-C, and decreased the fractional esterification rate of cholesterol in HDL, indicating preferentially increased large HDL particles. Treatment with JTT-705 increased all of the 3 charge-based HDL subfractions as determined by capillary isotachophoresis: fast-migrating, intermediate-migrating, and slow-migrating HDL. The percentage of slow HDL, ie, apolipoprotein E (apoE)-containing HDL and levels of apoE in HDL fraction, was also increased. JTT-705 treatment increased serum paraoxonase activity and HDL-associated platelet-activating factor acetylhydrolase activity, but decreased the plasma lysophosphatidylcholine concentration.
Conclusion—
Inhibition of CETP activity by JTT-705 not only increased the quantity of HDL, including HDL-C levels and charge-based HDL subfractions, but also favorably affected the size distribution of HDL subpopulations and the apolipoprotein and enzyme composition of HDL in rabbits.
Collapse
Affiliation(s)
- Bo Zhang
- Department of Cardiology, Fukuoka University School of Medicine, 7-45-1 Nanakuma Jonan-ku, Fukuoka 814-0180, Japan.
| | | | | | | | | | | | | |
Collapse
|
49
|
Dashti N, Datta G, Manchekar M, Chaddha M, Anantharamaiah GM. Model class A and class L peptides increase the production of apoA-I-containing lipoproteins in HepG2 cells. J Lipid Res 2004; 45:1919-28. [PMID: 15292373 DOI: 10.1194/jlr.m400251-jlr200] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Class A peptides inhibit atherosclerosis and protect cells from class L peptide-mediated lysis. Because the cytolytic process is concentration dependent, we hypothesized that at certain concentrations both classes of peptides exert similar effect(s) on cells. To test this hypothesis, we studied the effects of a class L peptide (18L = GIKKFLGSIWKFIKAFVG) and a class A peptide, 18A-Pro-18A (18A = DWLKAFYDKVAEKLKEAF) (37pA), on apolipoprotein and lipoprotein production in HepG2 cells. Secretion of (35)S-labeled apolipoprotein A-I (apoA-I) was stimulated by both 18L (110%) and 37pA (135%) at 10 and 20 nM of peptides, respectively. Both peptides enhanced the secretion of (3)H-labeled phospholipids by 140% and (14)C-labeled HDL-cholesterol (HDL-C) by 35% but had no significant effect on the total cholesterol mass or secretion. These results indicate that class L and class A peptides cause redistribution of cholesterol among lipoproteins in favor of HDL-C. Both peptides remodeled apoA-I-containing particles forming prebeta- as well as alpha-HDL. This study suggests that increased secretion of phospholipids and apoA-I and the formation of prebeta-HDL particles might contribute to the antiatherogenic properties of these peptides.
Collapse
Affiliation(s)
- Nassrin Dashti
- Department of Medicine, Biochemistry, and Molecular Genetics, and Atherosclerosis Research Unit, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| | | | | | | | | |
Collapse
|
50
|
Saito H, Lund-Katz S, Phillips MC. Contributions of domain structure and lipid interaction to the functionality of exchangeable human apolipoproteins. Prog Lipid Res 2004; 43:350-80. [PMID: 15234552 DOI: 10.1016/j.plipres.2004.05.002] [Citation(s) in RCA: 167] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Exchangeable apolipoproteins function in lipid transport as structural components of lipoprotein particles, cofactors for enzymes and ligands for cell-surface receptors. Recent findings with apoA-I and apoE suggest that the tertiary structures of these two members of the human exchangeable apolipoprotein gene family are related. Characteristically, these proteins contain a series of proline-punctuated, 11- or 22-amino acid, amphipathic alpha-helical repeats that can adopt a helix bundle conformation in the lipid-free state. The amino- and carboxyl-terminal regions form separate domains with the latter being primarily responsible for lipid binding. Interaction with lipid induces changes in the conformation of the amino-terminal domain leading to alterations in function; for example, opening of the amino-terminal four-helix bundle in apolipoprotein E upon lipid binding is associated with enhanced receptor-binding activity. The concept of a two-domain structure for the larger exchangeable apolipoproteins is providing new molecular insights into how these apolipoproteins interact with lipids and other proteins, such as receptors. The ways in which structural changes induced by lipid interaction modulate the functionality of these apolipoproteins are reviewed.
Collapse
Affiliation(s)
- Hiroyuki Saito
- Lipid Research Group, The Children's Hospital of Philadelphia, Abramson Research Center, Suite 1102, 3615 Civic Center Boulevard, University of Pennsylvania School of Medicine, Philadelphia, 19104-4318, USA
| | | | | |
Collapse
|