1
|
Petty RM, Ceresa L, Alexander E, Pham D, Sabnis N, Fudala R, Lacko AG, Krishnamoorthy RR, Gryczynski Z, Gryczynski I. Fluorescence Resonance Energy Transfer for Drug Loading Assessment in Reconstituted High-Density Lipoprotein Nanoparticles. Int J Mol Sci 2025; 26:3276. [PMID: 40244133 PMCID: PMC11989292 DOI: 10.3390/ijms26073276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Revised: 03/28/2025] [Accepted: 03/29/2025] [Indexed: 04/18/2025] Open
Abstract
Reconstituted high-density lipoprotein nanoparticles (NPs), which mimic the structure and function of endogenous human plasma HDL, hold promise as a robust drug delivery system. These nanoparticles, when loaded with appropriate agents, serve as powerful tools for targeted drug delivery. The fundamental challenge lies in controlling and estimating the actual drug load and the efficiency of drug release at the target. In this report, we present a novel approach based on enhanced Förster Resonance Energy Transfer (FRET) to assess particle load and monitor payload release. The NPs are labeled with donor molecules embedded in the lipid phase, while the spherical core volume is filled with acceptor molecules. Highly enhanced FRET efficiency to multiple acceptors in the NP core has been observed at distances significantly larger than the characteristic Förster distance (R0). To confirm that the observed changes in donor and acceptor emissions are a result of FRET, we developed a theoretical model for nonradiative energy transfer from a single donor to multiple acceptors enclosed in a spherical core volume. The load-dependent shortening of the fluorescence lifetime of the donor correlated with the presence of a negative component in the intensity decay of the acceptor clearly demonstrates that FRET can occur at a large distance comparable to the nanoparticle size (over 100 Å). Comparison of theoretical simulations with the measured intensity decays of the donor and acceptor fluorophores constitute a new method for evaluating particle load. The observed FRET efficiency depends on the number of acceptors in the core, providing a simple way to estimate the nanoparticle load efficiency. Particle disintegration and load release result in a distinct change in donor and acceptor emissions. This approach constitutes a novel strategy for assessing NP core load, monitoring NP integrity, and evaluating payload release efficiency to target cells. Significants: In the last decade, nanoparticles have emerged as a promising strategy for targeted drug delivery, with applications ranging from cancer therapy to ocular neurodegenerative disease treatments. Despite their potential, a significant issue has been the real-time monitoring of these drug delivery vehicles within biological systems. Effective strategies for monitoring NP payload loading, NP integrity, and payload release are needed to assess the quality of new drug delivery systems. In our study, we have found that FRET-enabled NPs function as an improved method for monitoring these aspects currently missing from current drug delivery efforts.
Collapse
Affiliation(s)
- R Max Petty
- Department of Pharmacology and Neuroscience, North Texas Eye Research Institute, Graduate School of Biomedical Science, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Luca Ceresa
- Department of Physics and Astronomy, Texas Christian University, Fort Worth, TX 76109, USA
| | - Emma Alexander
- Department of Physics and Astronomy, Texas Christian University, Fort Worth, TX 76109, USA
| | - Danh Pham
- Department of Physics and Astronomy, Texas Christian University, Fort Worth, TX 76109, USA
| | - Nirupama Sabnis
- Department of Physiology and Anatomy, Graduate School of Biomedical Sciences, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Rafal Fudala
- Department of Physiology and Anatomy, Graduate School of Biomedical Sciences, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Andras G Lacko
- Department of Microbiology, Immunology, and Genetics, School of Biomedical Sciences, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Raghu R Krishnamoorthy
- Department of Pharmacology and Neuroscience, North Texas Eye Research Institute, Graduate School of Biomedical Science, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Zygmunt Gryczynski
- Department of Physics and Astronomy, Texas Christian University, Fort Worth, TX 76109, USA
| | - Ignacy Gryczynski
- Department of Physics and Astronomy, Texas Christian University, Fort Worth, TX 76109, USA
| |
Collapse
|
2
|
Vanpouille C, Brichacek B, Pushkarsky T, Dubrovsky L, Fitzgerald W, Mukhamedova N, Garcia‐Hernandez S, Matthies D, Popratiloff A, Sviridov D, Margolis L, Bukrinsky M. HIV-1 Nef is carried on the surface of extracellular vesicles. J Extracell Vesicles 2024; 13:e12478. [PMID: 39016173 PMCID: PMC11252832 DOI: 10.1002/jev2.12478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 05/11/2024] [Accepted: 06/17/2024] [Indexed: 07/18/2024] Open
Abstract
Extracellular vesicles (EVs) serve as pivotal mediators of intercellular communication in both health and disease, delivering biologically active molecules from vesicle-producing cells to recipient cells. In the context of HIV infection, EVs have been shown to carry the viral protein Nef, a key pathogenic factor associated with HIV-related co-morbidities. Despite this recognition, the specific localisation of Nef within the vesicles has remained elusive. This study addresses this critical knowledge gap by investigating Nef-containing EVs. Less than 1% of the total released Nef was associated with EVs; most Nef existed as free protein released by damaged cells. Nevertheless, activity of EV-associated Nef in downregulating the major cholesterol transporter ABCA1, a critical aspect linked to the pathogenic effects of Nef, was comparable to that of free Nef present in the supernatant. Through a series of biochemical and microscopic assays, we demonstrate that the majority of EV-associated Nef molecules are localised on the external surface of the vesicles. This distinctive distribution prompts the consideration of Nef-containing EVs as potential targets for immunotherapeutic interventions aimed at preventing or treating HIV-associated co-morbidities. In conclusion, our results shed light on the localisation and functional activity of Nef within EVs, providing valuable insights for the development of targeted immunotherapies to mitigate the impact of HIV-associated co-morbidities.
Collapse
Affiliation(s)
- Christophe Vanpouille
- Section on Intercellular Interactions, Eunice Kennedy Shriver National Institute of Child Health and Human DevelopmentNational Institutes of HealthBethesdaMarylandUSA
| | - Beda Brichacek
- Department of Microbiology, Immunology and Tropical MedicineThe George Washington University School of Medicine and Health SciencesWashingtonDistrict of ColumbiaUSA
| | - Tatiana Pushkarsky
- Department of Microbiology, Immunology and Tropical MedicineThe George Washington University School of Medicine and Health SciencesWashingtonDistrict of ColumbiaUSA
| | - Larisa Dubrovsky
- Department of Microbiology, Immunology and Tropical MedicineThe George Washington University School of Medicine and Health SciencesWashingtonDistrict of ColumbiaUSA
| | - Wendy Fitzgerald
- Section on Intercellular Interactions, Eunice Kennedy Shriver National Institute of Child Health and Human DevelopmentNational Institutes of HealthBethesdaMarylandUSA
| | | | - Sofia Garcia‐Hernandez
- Nanofabrication and Imaging CenterThe George Washington UniversityWashingtonDistrict of ColumbiaUSA
| | - Doreen Matthies
- Unit on Structural Biology, Eunice Kennedy Shriver National Institute of Child Health and Human DevelopmentNational Institutes of HealthBethesdaMarylandUSA
| | - Anastas Popratiloff
- Nanofabrication and Imaging CenterThe George Washington UniversityWashingtonDistrict of ColumbiaUSA
| | - Dmitri Sviridov
- Baker Heart and Diabetes InstituteMelbourneVICAustralia
- Department of Biochemistry and Molecular BiologyMonash UniversityClaytonVICAustralia
| | - Leonid Margolis
- Section on Intercellular Interactions, Eunice Kennedy Shriver National Institute of Child Health and Human DevelopmentNational Institutes of HealthBethesdaMarylandUSA
- Faculty of Natural Sciences and MedicineIlia State UniversityTbilisiRepublic of Georgia
| | - Michael Bukrinsky
- Department of Microbiology, Immunology and Tropical MedicineThe George Washington University School of Medicine and Health SciencesWashingtonDistrict of ColumbiaUSA
| |
Collapse
|
3
|
Toner YC, Prévot G, van Leent MMT, Munitz J, Oosterwijk R, Verschuur AVD, van Elsas Y, Peric V, Maas RJF, Ranzenigo A, Morla-Folch J, Wang W, Umali M, de Dreu A, Fernandes JC, Sullivan NAT, Maier A, Mason C, Reiner T, Fayad ZA, Mulder WJM, Teunissen AJP, Pérez-Medina C. Macrophage PET imaging in mouse models of cardiovascular disease and cancer with an apolipoprotein-inspired radiotracer. NPJ IMAGING 2024; 2:12. [PMID: 38765879 PMCID: PMC11096117 DOI: 10.1038/s44303-024-00009-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 02/06/2024] [Indexed: 05/22/2024]
Abstract
Macrophages are key inflammatory mediators in many pathological conditions, including cardiovascular disease (CVD) and cancer, the leading causes of morbidity and mortality worldwide. This makes macrophage burden a valuable diagnostic marker and several strategies to monitor these cells have been reported. However, such strategies are often high-priced, non-specific, invasive, and/or not quantitative. Here, we developed a positron emission tomography (PET) radiotracer based on apolipoprotein A1 (ApoA1), the main protein component of high-density lipoprotein (HDL), which has an inherent affinity for macrophages. We radiolabeled an ApoA1-mimetic peptide (mA1) with zirconium-89 (89Zr) to generate a lipoprotein-avid PET probe (89Zr-mA1). We first characterized 89Zr-mA1's affinity for lipoproteins in vitro by size exclusion chromatography. To study 89Zr-mA1's in vivo behavior and interaction with endogenous lipoproteins, we performed extensive studies in wildtype C57BL/6 and Apoe-/- hypercholesterolemic mice. Subsequently, we used in vivo PET imaging to study macrophages in melanoma and myocardial infarction using mouse models. The tracer's cell specificity was assessed by histology and mass cytometry (CyTOF). Our data show that 89Zr-mA1 associates with lipoproteins in vitro. This is in line with our in vivo experiments, in which we observed longer 89Zr-mA1 circulation times in hypercholesterolemic mice compared to C57BL/6 controls. 89Zr-mA1 displayed a tissue distribution profile similar to ApoA1 and HDL, with high kidney and liver uptake as well as substantial signal in the bone marrow and spleen. The tracer also accumulated in tumors of melanoma-bearing mice and in the ischemic myocardium of infarcted animals. In these sites, CyTOF analyses revealed that natZr-mA1 was predominantly taken up by macrophages. Our results demonstrate that 89Zr-mA1 associates with lipoproteins and hence accumulates in macrophages in vivo. 89Zr-mA1's high uptake in these cells makes it a promising radiotracer for non-invasively and quantitatively studying conditions characterized by marked changes in macrophage burden.
Collapse
Affiliation(s)
- Yohana C. Toner
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Geoffrey Prévot
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Mandy M. T. van Leent
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Jazz Munitz
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Roderick Oosterwijk
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Laboratory of Chemical Biology, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Anna Vera D. Verschuur
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Yuri van Elsas
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Vedran Peric
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Laboratory of Chemical Biology, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Rianne J. F. Maas
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Anna Ranzenigo
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Judit Morla-Folch
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - William Wang
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Martin Umali
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Anne de Dreu
- Laboratory of Chemical Biology, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Jessica Chimene Fernandes
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Nathaniel A. T. Sullivan
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Alexander Maier
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Department of Cardiology and Angiology, Heart Center Freiburg University, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Christian Mason
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY USA
| | - Thomas Reiner
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY USA
- Department of Radiology, Weill Cornell Medical College, New York, NY USA
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY USA
| | - Zahi A. Fayad
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Willem J. M. Mulder
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
- Laboratory of Chemical Biology, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Abraham J. P. Teunissen
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Carlos Pérez-Medina
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| |
Collapse
|
4
|
Sharma A, Sharma C, Sharma L, Wal P, Mishra P, Sachdeva N, Yadav S, Vargas De-La Cruz C, Arora S, Subramaniyan V, Rawat R, Behl T, Nandave M. Targeting the vivid facets of apolipoproteins as a cardiovascular risk factor in rheumatoid arthritis. Can J Physiol Pharmacol 2024; 102:305-317. [PMID: 38334084 DOI: 10.1139/cjpp-2023-0259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2024]
Abstract
Mostly, cardiovascular diseases are blamed for casualties in rheumatoid arthritis (RA) patients. Customarily, dyslipidemia is probably the most prevalent underlying cause of untimely demise in people suffering from RA as it hastens the expansion of atherosclerosis. The engagement of inflammatory cytokines like tumor necrosis factor-α (TNF-α), interleukin-1 (IL-1), interleukin-6 (IL-6), etc., is crucial in the progression and proliferation of both RA and abnormal lipid parameters. Thus, lipid abnormalities should be monitored frequently in patients with both primary and advanced RA stages. An advanced lipid profile examination, i.e., direct role of apolipoproteins associated with various lipid molecules is a more dependable approach for better understanding of the disease and selecting suitable therapeutic targets. Therefore, studying their apolipoproteins is more relevant than assessing RA patients' altered lipid profile levels. Among the various apolipoprotein classes, Apo A1 and Apo B are primarily being focused. In addition, it also addresses how calculating Apo B:Apo A1 ratio can aid in analyzing the disease's risk. The marketed therapies available to control lipid abnormalities are associated with many other risk factors. Hence, directly targeting Apo A1 and Apo B would provide a better and safer option.
Collapse
Affiliation(s)
- Aditi Sharma
- Department of Pharmacology, School of Pharmaceutical Sciences, Shoolini University, Solan, Himachal Pradesh, India
| | - Chakshu Sharma
- Department of Pharmacology, School of Pharmaceutical Sciences, Shoolini University, Solan, Himachal Pradesh, India
| | - Lalit Sharma
- Department of Pharmacology, School of Pharmaceutical Sciences, Shoolini University, Solan, Himachal Pradesh, India
| | - Pranay Wal
- Pranveer Singh Institute of Technology, Pharmacy, Kanpur, Uttar Pradesh, India
| | - Preeti Mishra
- Raja Balwant Singh Engineering Technical Campus, Bichpuri, Agra, India
| | - Nitin Sachdeva
- Department of Anesthesia, Mediclinic Aljowhara Hospital, Al Ain, United Arab Emirates
| | - Shivam Yadav
- School of Pharmacy, Babu Banarasi Das University, Lucknow, Uttar Pradesh, India
| | - Celia Vargas De-La Cruz
- Department of Pharmacology, Bromatology and Toxicology, Faculty of Pharmacy and Biochemistry, Universidad Nacional Mayor de San Marcos, Lima 15001, Peru
- E-Health Research Center, Universidad de Ciencias y Humanidades, Lima 15001, Peru
| | - Sandeep Arora
- Amity Institute of Pharmacy, Amity University, Noida, Uttar Pradesh, India
| | - Vetriselvan Subramaniyan
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University, Jalan Lagoon Selatan, Bandar Sunway, 47500 Selangor Darul Ehsan, Malaysia
- Centre for Transdisciplinary Research, Department of Pharmacology, Saveetha Dental College Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, Tamil Nadu 600077, India
| | - Ravi Rawat
- School of Health Sciences and Technology, University of Petroleum and Energy Studies, Bidholi, Dehradun, Uttarakhand, India
| | - Tapan Behl
- Amity School of Pharmaceutical Sciences, Amity University, Mohali, Punjab, India
| | - Mukesh Nandave
- Department of Pharmacology, Delhi Pharmaceutical Sciences and Research University, Pushp Vihar, Delhi, India
| |
Collapse
|
5
|
Solanki K, Kumar A, Khan MS, Karthikeyan S, Atre R, Zhang KY, Bezsonov E, Obukhov AG, Baig MS. Novel peptide inhibitors targeting CD40 and CD40L interaction: A potential for atherosclerosis therapy. Curr Res Struct Biol 2023; 6:100110. [PMID: 38106460 PMCID: PMC10724548 DOI: 10.1016/j.crstbi.2023.100110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 10/25/2023] [Accepted: 10/26/2023] [Indexed: 12/19/2023] Open
Abstract
Atherosclerosis is a chronic inflammatory disease characterized by plaque build-up in the arteries, leading to the obstruction of blood flow. Macrophages are the primary immune cells found in the atherosclerotic lesions and are directly involved in atherosclerosis progression. Macrophages are derived from extravasating blood monocytes. The monocytic CD40 receptor is important for monocyte recruitment on the endothelium expressing the CD40 ligand (CD40L). Thus, targeting monocyte/macrophage interaction with the endothelium by inhibiting CD40-CD40L interaction may be a promising strategy for attenuating atherosclerosis. Monoclonal antibodies have been used against this target but shows various complications. We used an array of computer-aided drug discovery tools and molecular docking approaches to design a therapeutic inhibitory peptide that could efficiently bind to the critical residues (82Y, 84D, and 86N) on the CD40 receptor essential for the receptor's binding to CD40L. The initial screen identified a parent peptide with a high binding affinity to CD40, but the peptide exhibited a positive hepatotoxicity score. We then designed several novel peptidomimetic derivatives with higher binding affinities to CD40, good physicochemical properties, and negative hepatotoxicity as compared to the parent peptide. Furthermore, we conducted molecular dynamics simulations for both the apo and complexed forms of the receptor with ligand, and screened peptides to evaluate their stability. The designed peptidomimetic derivatives are promising therapeutics targeting the CD40-CD40L interaction and may potentially be used to attenuate atherosclerosis.
Collapse
Affiliation(s)
- Kundan Solanki
- Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Simrol, Indore, 453552, India
| | - Ashutosh Kumar
- Laboratory for Structural Bioinformatics, Center for Biosystems Dynamics Research, RIKEN, Tsurumi, Yokohama, Kanagawa, Japan
| | - Mohd Shahnawaz Khan
- Department of Biochemistry, College of Science, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Subramani Karthikeyan
- Centre for Healthcare Advancement, Innovation and Research, Vellore Institute of Technology University, Chennai Campus, Chennai, 600127, India
| | - Rajat Atre
- Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Simrol, Indore, 453552, India
| | - Kam Y.J. Zhang
- Laboratory for Structural Bioinformatics, Center for Biosystems Dynamics Research, RIKEN, Tsurumi, Yokohama, Kanagawa, Japan
| | - Evgeny Bezsonov
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, 8 Baltiiskaya Street, 125315, Moscow, Russia
- Laboratory of Cellular and Molecular Pathology of Cardiovascular System, Avtsyn Research Institute of Human Morphology of Federal State Budgetary Scientific Institution “Petrovsky National Research Centre of Surgery”, 3 Tsyurupa Street, 117418, Moscow, Russia
- Department of Biology and General Genetics, I.M. Sechenov First Moscow State Medical University (Sechenov University), 8 Izmailovsky Boulevard, 105043, Moscow, Russia
| | - Alexander G. Obukhov
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Mirza S. Baig
- Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Simrol, Indore, 453552, India
| |
Collapse
|
6
|
Hafiane A, Gianopoulos I, Sorci-Thomas MG, Daskalopoulou SS. Current models of apolipoprotein A-I lipidation by adenosine triphosphate binding cassette transporter A1. Curr Opin Lipidol 2022; 33:139-145. [PMID: 34581311 DOI: 10.1097/mol.0000000000000786] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE OF REVIEW The primary cardioprotective function of high-density lipoprotein (HDL) is to remove excess cellular free cholesterol (FC) from peripheral tissues and deliver it to the liver. Here, we summarize recent research that examines apolipoprotein A-I (apoA-I) lipidation models by adenosine triphosphate binding cassette transporter A1 (ABCA1) and discuss its relevance in atherosclerotic cardiovascular disease (ASCVD). RECENT FINDINGS The first step in HDL formation involves the interaction between apoA-I and ABCA1, where ABCA1 mediates the removal of FC and phospholipids from lipid-laden macrophages to form discoidal nascent HDL (nHDL). However, there are currently no clear-cut systematic models that characterize HDL formation. A number of recent studies have investigated the importance of apoA-I C- and N-terminal domains required for optimal cholesterol efflux and nHDL production. Furthermore, functional ABCA1 is required for direct or indirect binding to apoA-I where ABCA1 dimer-monomer interconversion facilitates apoA-I lipidation from plasma membrane microdomains. Microparticles are also another lipid source for apoA-I solubilization into nHDL. SUMMARY ApoA-I and ABCA1 are key factors in macrophage-mediated cholesterol efflux and nHDL production. Understanding of the key steps in HDL formation may unlock the therapeutic potential of HDL and improve clinical management of ASCVD.
Collapse
Affiliation(s)
- Anouar Hafiane
- Division of Experimental Medicine, Department of Medicine, Faculty of Medicine, Research Institute of the McGill University Health Centre, McGill University, Montreal, Canada
| | - Ioanna Gianopoulos
- Division of Experimental Medicine, Department of Medicine, Faculty of Medicine, Research Institute of the McGill University Health Centre, McGill University, Montreal, Canada
| | - Mary G Sorci-Thomas
- Division of Endocrinology, Metabolism and Clinical Nutrition, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Stella S Daskalopoulou
- Division of Experimental Medicine, Department of Medicine, Faculty of Medicine, Research Institute of the McGill University Health Centre, McGill University, Montreal, Canada
- Division of Internal Medicine, Department of Medicine, Faculty of Medicine, McGill University Health Centre, McGill University Montreal, Montreal, Canada
| |
Collapse
|
7
|
White CR, Palgunachari M, Wolkowicz P, Anantharamaiah GM. Peptides as Therapeutic Agents for Atherosclerosis. Methods Mol Biol 2022; 2419:89-110. [PMID: 35237960 DOI: 10.1007/978-1-0716-1924-7_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
More than three decades ago, as a test for the amphipathic helix theory, an 18 amino acid residue peptide and its analogs were designed with no sequence homology to any of the exchangeable apolipoproteins. Based on the apolipoprotein A-I (the major protein component of high density lipoproteins, HDL) mimicking properties, they were termed as ApoA-I mimicking peptides. Several laboratories around the world started studying such de novo-designed peptides for their antiatherogenic properties. The present chapter describes the efforts in bringing these peptides as therapeutic agents for atherosclerosis and several lipid-mediated disorders.
Collapse
Affiliation(s)
- C Roger White
- Department of Medicine, UAB Medical Centre, Birmingham, AL, USA
| | | | - Paul Wolkowicz
- Department of Medicine, UAB Medical Centre, Birmingham, AL, USA
| | | |
Collapse
|
8
|
HDL Mimetic Peptides. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1377:141-151. [DOI: 10.1007/978-981-19-1592-5_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
9
|
Thakur R, Suri CR, Kaur IP, Rishi P. Review. Crit Rev Ther Drug Carrier Syst 2022; 40:49-100. [DOI: 10.1615/critrevtherdrugcarriersyst.2022040322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
10
|
Benitez Amaro A, Solanelles Curco A, Garcia E, Julve J, Rives J, Benitez S, Llorente Cortes V. Apolipoprotein and LRP1-Based Peptides as New Therapeutic Tools in Atherosclerosis. J Clin Med 2021; 10:jcm10163571. [PMID: 34441867 PMCID: PMC8396846 DOI: 10.3390/jcm10163571] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 08/05/2021] [Accepted: 08/08/2021] [Indexed: 12/17/2022] Open
Abstract
Apolipoprotein (Apo)-based mimetic peptides have been shown to reduce atherosclerosis. Most of the ApoC-II and ApoE mimetics exert anti-atherosclerotic effects by improving lipid profile. ApoC-II mimetics reverse hypertriglyceridemia and ApoE-based peptides such as Ac-hE18A-NH2 reduce cholesterol and triglyceride (TG) levels in humans. Conversely, other classes of ApoE and ApoA-I mimetic peptides and, more recently, ApoJ and LRP1-based peptides, exhibit several anti-atherosclerotic actions in experimental models without influencing lipoprotein profile. These other mimetic peptides display at least one atheroprotective mechanism such as providing LDL stability against mechanical modification or conferring protection against the action of lipolytic enzymes inducing LDL aggregation in the arterial intima. Other anti-atherosclerotic effects exerted by these peptides also include protection against foam cell formation and inflammation, and induction of reverse cholesterol transport. Although the underlying mechanisms of action are still poorly described, the recent findings suggest that these mimetics could confer atheroprotection by favorably influencing lipoprotein function rather than lipoprotein levels. Despite the promising results obtained with peptide mimetics, the assessment of their stability, atheroprotective efficacy and tissue targeted delivery are issues currently under progress.
Collapse
Affiliation(s)
- Aleyda Benitez Amaro
- Institute of Biomedical Research of Barcelona (IIBB), Spanish National Research Council (CSIC), 08036 Barcelona, Spain; (A.B.A.); (E.G.)
- Biomedical Research Institute Sant Pau (IIB-Sant Pau), 08041 Barcelona, Spain;
| | | | - Eduardo Garcia
- Institute of Biomedical Research of Barcelona (IIBB), Spanish National Research Council (CSIC), 08036 Barcelona, Spain; (A.B.A.); (E.G.)
- Biomedical Research Institute Sant Pau (IIB-Sant Pau), 08041 Barcelona, Spain;
| | - Josep Julve
- Metabolic Basis of Cardiovascular Risk Group, Biomedical Research Institute Sant Pau (IIB Sant Pau), 08041 Barcelona, Spain;
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28029 Madrid, Spain
| | - Jose Rives
- Biochemistry Department, Hospital de la Santa Creu i Sant Pau, 08025 Barcelona, Spain;
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Universitat Autònoma de Barcelona (UAB), Cerdanyola del Vallès, 08016 Barcelona, Spain
| | - Sonia Benitez
- Cardiovascular Biochemistry Group, Biomedical Research Institute Sant Pau (IIB Sant Pau), 08041 Barcelona, Spain
- Correspondence: (S.B.); or (V.L.C.)
| | - Vicenta Llorente Cortes
- Institute of Biomedical Research of Barcelona (IIBB), Spanish National Research Council (CSIC), 08036 Barcelona, Spain; (A.B.A.); (E.G.)
- Biomedical Research Institute Sant Pau (IIB-Sant Pau), 08041 Barcelona, Spain;
- CIBERCV, Institute of Health Carlos III, 28029 Madrid, Spain
- Correspondence: (S.B.); or (V.L.C.)
| |
Collapse
|
11
|
Wolska A, Reimund M, Sviridov DO, Amar MJ, Remaley AT. Apolipoprotein Mimetic Peptides: Potential New Therapies for Cardiovascular Diseases. Cells 2021; 10:597. [PMID: 33800446 PMCID: PMC8000854 DOI: 10.3390/cells10030597] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 03/01/2021] [Accepted: 03/02/2021] [Indexed: 12/13/2022] Open
Abstract
Since the seminal breakthrough of treating diabetic patients with insulin in the 1920s, there has been great interest in developing other proteins and their peptide mimetics as therapies for a wide variety of other medical disorders. Currently, there are at least 60 different peptides that have been approved for human use and over 150 peptides that are in various stages of clinical development. Peptides mimetic of the major proteins on lipoproteins, namely apolipoproteins, have also been developed first as tools for understanding apolipoprotein structure and more recently as potential therapeutics. In this review, we discuss the biochemistry, peptide mimetics design and clinical trials for peptides based on apoA-I, apoE and apoC-II. We primarily focus on applications of peptide mimetics related to cardiovascular diseases. We conclude with a discussion on the limitations of peptides as therapeutic agents and the challenges that need to be overcome before apolipoprotein mimetic peptides can be developed into new drugs.
Collapse
Affiliation(s)
- Anna Wolska
- Lipoprotein Metabolism Laboratory, Translational Vascular Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA; (M.R.); (D.O.S.); (M.J.A.); (A.T.R.)
| | | | | | | | | |
Collapse
|
12
|
Islam R, Sviridov DO, Drake SK, Tunyi J, Abdoulaeva G, Freeman LA, Pastor RW, Remaley AT. Incorporation of α-methylated amino acids into Apolipoprotein A-I mimetic peptides improves their helicity and cholesterol efflux potential. Biochem Biophys Res Commun 2020; 526:349-354. [PMID: 32222278 DOI: 10.1016/j.bbrc.2020.03.070] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 03/11/2020] [Indexed: 01/01/2023]
Abstract
Apolipoprotein A-I (ApoA-I) mimetic peptides are potential therapeutic agents for promoting the efflux of excess cellular cholesterol, which is dependent upon the presence of an amphipathic helix. Since α-methylated Ala enhances peptide helicity, we hypothesized that incorporating other types of α-methylated amino acids into ApoA-I mimetic peptides may also increase their helicity and cholesterol efflux potential. The last helix of apoA-I, peptide 'A' (VLESFKVSFLSALEEYTKKLNT), was used to design peptides containing a single type of α-methylated amino acid substitution (Ala/Aα, Glu/Dα, Lys/Kα, Leu/Lα), as well as a peptide containing both α-methylated Lys and Leu (6α). Depending on the specific residue, the α-helical content as measured by CD-spectroscopy and calculated hydrophobic moments were sometimes higher for peptides containing other types of α-methylated amino acids than those with α-methylated Ala. In ABCA1-transfected cells, cholesterol efflux to the peptides showed the following order of potency: 6α>Kα≈Lα≈Aα≫Dα≈A. In general, α-methylated peptides were resistant to proteolysis, but this varied depending on the type of protease and specific amino acid substitution. In summary, increased helicity and amphilicity due to α-methylated amino acid substitutions in ApoA-I mimetic peptides resulted in improved cholesterol efflux capacity and resistance to proteolysis, indicating that this modification may be useful in the future design of therapeutic ApoA-I mimetic peptides.
Collapse
Affiliation(s)
- Rafique Islam
- Lipoprotein Metabolism Laboratory, Translational Vascular Medicine Branch, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Denis O Sviridov
- Lipoprotein Metabolism Laboratory, Translational Vascular Medicine Branch, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, 20892, USA.
| | - Steven K Drake
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Jude Tunyi
- Laboratory of Computational Biology National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Galina Abdoulaeva
- Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, 20993, USA
| | - Lita A Freeman
- Lipoprotein Metabolism Laboratory, Translational Vascular Medicine Branch, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Richard W Pastor
- Laboratory of Computational Biology National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Alan T Remaley
- Lipoprotein Metabolism Laboratory, Translational Vascular Medicine Branch, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| |
Collapse
|
13
|
Fawaz MV, Kim SY, Li D, Ming R, Xia Z, Olsen K, Pogozheva ID, Tesmer JJG, Schwendeman A. Phospholipid Component Defines Pharmacokinetic and Pharmacodynamic Properties of Synthetic High-Density Lipoproteins. J Pharmacol Exp Ther 2020; 372:193-204. [PMID: 31776208 PMCID: PMC6978696 DOI: 10.1124/jpet.119.257568] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 11/18/2019] [Indexed: 12/14/2022] Open
Abstract
Synthetic high-density lipoprotein (sHDL) nanoparticles composed of apolipoprotein A-I mimetic peptide and phospholipids have been shown to reduce atherosclerosis in animal models. Cholesterol is mobilized from atheroma macrophages by sHDL into the blood compartment and delivered to the liver for elimination. Historically, sHDL drug discovery efforts were focused on optimizing peptide sequences for interaction with cholesterol cellular transporters rather than understanding how both sHDL components, peptide and lipid, influence its pharmacokinetic and pharmacodynamic profiles. We designed two sets of sHDL having either identical phospholipid but variable peptide sequences with different plasma stability or identical peptide and phospholipids with variable fatty acid chain length and saturation. We found that sHDL prepared with proteolytically stable 22A-P peptide had 2-fold longer circulation half-time relative to the less stable 22A peptide. Yet, longer half-life did not translate into any improvement in cholesterol mobilization. In contrast, sHDL with variable phospholipid compositions showed significant differences in phospholipid PK, with distearoyl phosphatidylcholine-based sHDL demonstrating the longest half-life of 6.0 hours relative to 1.0 hour for palmitoyl-oleoyl phosphatidylcholine-based sHDL. This increase in half-life corresponded to an approx. 6.5-fold increase in the area under the curve for the mobilized cholesterol. Therefore, the phospholipid component in sHDL plays a major role in cholesterol mobilization in vivo and should not be overlooked in the design of future sHDL. SIGNIFICANCE STATEMENT: The phospholipid composition in sHDL plays a critical role in determining half-life and cholesterol mobilization in vivo.
Collapse
Affiliation(s)
- Maria V Fawaz
- Departments of Medicinal Chemistry (M.V.F., I.D.P.) and Pharmaceutical Sciences (S.Y.K., D.L., R.M., Z.X., K.O., A.S.), College of Pharmacy, and Biointerfaces Institute (A.S.), University of Michigan, Ann Arbor, Michigan; and Department of Biological Sciences, Purdue University, West Lafayette, Indiana (J.J.G.T.)
| | - Sang Yeop Kim
- Departments of Medicinal Chemistry (M.V.F., I.D.P.) and Pharmaceutical Sciences (S.Y.K., D.L., R.M., Z.X., K.O., A.S.), College of Pharmacy, and Biointerfaces Institute (A.S.), University of Michigan, Ann Arbor, Michigan; and Department of Biological Sciences, Purdue University, West Lafayette, Indiana (J.J.G.T.)
| | - Dan Li
- Departments of Medicinal Chemistry (M.V.F., I.D.P.) and Pharmaceutical Sciences (S.Y.K., D.L., R.M., Z.X., K.O., A.S.), College of Pharmacy, and Biointerfaces Institute (A.S.), University of Michigan, Ann Arbor, Michigan; and Department of Biological Sciences, Purdue University, West Lafayette, Indiana (J.J.G.T.)
| | - Ran Ming
- Departments of Medicinal Chemistry (M.V.F., I.D.P.) and Pharmaceutical Sciences (S.Y.K., D.L., R.M., Z.X., K.O., A.S.), College of Pharmacy, and Biointerfaces Institute (A.S.), University of Michigan, Ann Arbor, Michigan; and Department of Biological Sciences, Purdue University, West Lafayette, Indiana (J.J.G.T.)
| | - Ziyun Xia
- Departments of Medicinal Chemistry (M.V.F., I.D.P.) and Pharmaceutical Sciences (S.Y.K., D.L., R.M., Z.X., K.O., A.S.), College of Pharmacy, and Biointerfaces Institute (A.S.), University of Michigan, Ann Arbor, Michigan; and Department of Biological Sciences, Purdue University, West Lafayette, Indiana (J.J.G.T.)
| | - Karl Olsen
- Departments of Medicinal Chemistry (M.V.F., I.D.P.) and Pharmaceutical Sciences (S.Y.K., D.L., R.M., Z.X., K.O., A.S.), College of Pharmacy, and Biointerfaces Institute (A.S.), University of Michigan, Ann Arbor, Michigan; and Department of Biological Sciences, Purdue University, West Lafayette, Indiana (J.J.G.T.)
| | - Irina D Pogozheva
- Departments of Medicinal Chemistry (M.V.F., I.D.P.) and Pharmaceutical Sciences (S.Y.K., D.L., R.M., Z.X., K.O., A.S.), College of Pharmacy, and Biointerfaces Institute (A.S.), University of Michigan, Ann Arbor, Michigan; and Department of Biological Sciences, Purdue University, West Lafayette, Indiana (J.J.G.T.)
| | - John J G Tesmer
- Departments of Medicinal Chemistry (M.V.F., I.D.P.) and Pharmaceutical Sciences (S.Y.K., D.L., R.M., Z.X., K.O., A.S.), College of Pharmacy, and Biointerfaces Institute (A.S.), University of Michigan, Ann Arbor, Michigan; and Department of Biological Sciences, Purdue University, West Lafayette, Indiana (J.J.G.T.)
| | - Anna Schwendeman
- Departments of Medicinal Chemistry (M.V.F., I.D.P.) and Pharmaceutical Sciences (S.Y.K., D.L., R.M., Z.X., K.O., A.S.), College of Pharmacy, and Biointerfaces Institute (A.S.), University of Michigan, Ann Arbor, Michigan; and Department of Biological Sciences, Purdue University, West Lafayette, Indiana (J.J.G.T.)
| |
Collapse
|
14
|
First eight residues of apolipoprotein A-I mediate the C-terminus control of helical bundle unfolding and its lipidation. PLoS One 2020; 15:e0221915. [PMID: 31945064 PMCID: PMC6964839 DOI: 10.1371/journal.pone.0221915] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 12/30/2019] [Indexed: 11/23/2022] Open
Abstract
The crystal structure of a C-terminal deletion of apolipoprotein A-I (apoA1) shows a large helical bundle structure in the amino half of the protein, from residues 8 to 115. Using site directed mutagenesis, guanidine or thermal denaturation, cell free liposome clearance, and cellular ABCA1-mediated cholesterol efflux assays, we demonstrate that apoA1 lipidation can occur when the thermodynamic barrier to this bundle unfolding is lowered. The absence of the C-terminus renders the bundle harder to unfold resulting in loss of apoA1 lipidation that can be reversed by point mutations, such as Trp8Ala, and by truncations as short as 8 residues in the amino terminus, both of which facilitate helical bundle unfolding. Locking the bundle via a disulfide bond leads to loss of apoA1 lipidation. We propose a model in which the C-terminus acts on the N-terminus to destabilize this helical bundle. Upon lipid binding to the C-terminus, Trp8 is displaced from its interaction with Phe57, Arg61, Leu64, Val67, Phe71, and Trp72 to destabilize the bundle. However, when the C-terminus is deleted, Trp8 cannot be displaced, the bundle cannot unfold, and apoA1 cannot be lipidated.
Collapse
|
15
|
Schultz ML, Fawaz MV, Azaria RD, Hollon TC, Liu EA, Kunkel TJ, Halseth TA, Krus KL, Ming R, Morin EE, McLoughlin HS, Bushart DD, Paulson HL, Shakkottai VG, Orringer DA, Schwendeman AS, Lieberman AP. Synthetic high-density lipoprotein nanoparticles for the treatment of Niemann-Pick diseases. BMC Med 2019; 17:200. [PMID: 31711490 PMCID: PMC6849328 DOI: 10.1186/s12916-019-1423-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Accepted: 09/10/2019] [Indexed: 01/23/2023] Open
Abstract
BACKGROUND Niemann-Pick disease type C is a fatal and progressive neurodegenerative disorder characterized by the accumulation of unesterified cholesterol in late endosomes and lysosomes. We sought to develop new therapeutics for this disorder by harnessing the body's endogenous cholesterol scavenging particle, high-density lipoprotein (HDL). METHODS Here we design, optimize, and define the mechanism of action of synthetic HDL (sHDL) nanoparticles. RESULTS We demonstrate a dose-dependent rescue of cholesterol storage that is sensitive to sHDL lipid and peptide composition, enabling the identification of compounds with a range of therapeutic potency. Peripheral administration of sHDL to Npc1 I1061T homozygous mice mobilizes cholesterol, reduces serum bilirubin, reduces liver macrophage size, and corrects body weight deficits. Additionally, a single intraventricular injection into adult Npc1 I1061T brains significantly reduces cholesterol storage in Purkinje neurons. Since endogenous HDL is also a carrier of sphingomyelin, we tested the same sHDL formulation in the sphingomyelin storage disease Niemann-Pick type A. Utilizing stimulated Raman scattering microscopy to detect endogenous unlabeled lipids, we show significant rescue of Niemann-Pick type A lipid storage. CONCLUSIONS Together, our data establish that sHDL nanoparticles are a potential new therapeutic avenue for Niemann-Pick diseases.
Collapse
Affiliation(s)
- Mark L Schultz
- Department of Pathology, University of Michigan Medical School, 3510 MSRB1, 1150 W. Medical Center Dr., Ann Arbor, MI, 48109, USA
| | - Maria V Fawaz
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Ruth D Azaria
- Department of Pathology, University of Michigan Medical School, 3510 MSRB1, 1150 W. Medical Center Dr., Ann Arbor, MI, 48109, USA
| | - Todd C Hollon
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Elaine A Liu
- Department of Pathology, University of Michigan Medical School, 3510 MSRB1, 1150 W. Medical Center Dr., Ann Arbor, MI, 48109, USA
- Cellular and Molecular Biology Graduate Program, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
- Medical Scientist Training Program, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Thaddeus J Kunkel
- Department of Pathology, University of Michigan Medical School, 3510 MSRB1, 1150 W. Medical Center Dr., Ann Arbor, MI, 48109, USA
| | - Troy A Halseth
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Kelsey L Krus
- Department of Pathology, University of Michigan Medical School, 3510 MSRB1, 1150 W. Medical Center Dr., Ann Arbor, MI, 48109, USA
| | - Ran Ming
- Department of Pharmaceutical Sciences, University of Michigan College of Pharmacy, B20-102W NCRC, 2800 Plymouth Road, Ann Arbor, MI, 48109, USA
| | - Emily E Morin
- Department of Pharmaceutical Sciences, University of Michigan College of Pharmacy, B20-102W NCRC, 2800 Plymouth Road, Ann Arbor, MI, 48109, USA
| | - Hayley S McLoughlin
- Department of Neurology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - David D Bushart
- Department of Neurology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Henry L Paulson
- Department of Neurology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Vikram G Shakkottai
- Department of Neurology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Daniel A Orringer
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Anna S Schwendeman
- Department of Pharmaceutical Sciences, University of Michigan College of Pharmacy, B20-102W NCRC, 2800 Plymouth Road, Ann Arbor, MI, 48109, USA.
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, 48109, USA.
| | - Andrew P Lieberman
- Department of Pathology, University of Michigan Medical School, 3510 MSRB1, 1150 W. Medical Center Dr., Ann Arbor, MI, 48109, USA.
| |
Collapse
|
16
|
Kornmueller K, Vidakovic I, Prassl R. Artificial High Density Lipoprotein Nanoparticles in Cardiovascular Research. Molecules 2019; 24:E2829. [PMID: 31382521 PMCID: PMC6695986 DOI: 10.3390/molecules24152829] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 07/31/2019] [Accepted: 08/01/2019] [Indexed: 02/07/2023] Open
Abstract
Lipoproteins are endogenous nanoparticles which are the major transporter of fats and cholesterol in the human body. They play a key role in the regulatory mechanisms of cardiovascular events. Lipoproteins can be modified and manipulated to act as drug delivery systems or nanocarriers for contrast agents. In particular, high density lipoproteins (HDL), which are the smallest class of lipoproteins, can be synthetically engineered either as nascent HDL nanodiscs or spherical HDL nanoparticles. Reconstituted HDL (rHDL) particles are formed by self-assembly of various lipids and apolipoprotein AI (apo-AI). A variety of substances including drugs, nucleic acids, signal emitting molecules, or dyes can be loaded, making them efficient nanocarriers for therapeutic applications or medical diagnostics. This review provides an overview about synthesis techniques, physicochemical properties of rHDL nanoparticles, and structural determinants for rHDL function. We discuss recent developments utilizing either apo-AI or apo-AI mimetic peptides for the design of pharmaceutical rHDL formulations. Advantages, limitations, challenges, and prospects for clinical translation are evaluated with a special focus on promising strategies for the treatment and diagnosis of atherosclerosis and cardiovascular diseases.
Collapse
Affiliation(s)
- Karin Kornmueller
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Biophysics, Medical University of Graz, Neue Stiftingtalstraße 6/IV, 8010 Graz, Austria
| | - Ivan Vidakovic
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Biophysics, Medical University of Graz, Neue Stiftingtalstraße 6/IV, 8010 Graz, Austria
| | - Ruth Prassl
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Biophysics, Medical University of Graz, Neue Stiftingtalstraße 6/IV, 8010 Graz, Austria.
| |
Collapse
|
17
|
ABCA1 Agonist Mimetic Peptide CS-6253 Induces Microparticles Release From Different Cell Types by ABCA1-Efflux-Dependent Mechanism. Can J Cardiol 2019; 35:770-781. [PMID: 31151713 DOI: 10.1016/j.cjca.2019.02.018] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 02/18/2019] [Accepted: 02/18/2019] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND Small peptides based on the C-terminal domain of apo E have recently been proposed as ATP-binding cassette transporter A1 (ABCA1) agonist with therapeutic potential. Previous work has shown that a novel synthetic peptide, CS-6253, acts synergistically with apolipoprotein A-I or alone to generate high-density lipoprotein (HDL) particles; we have also shown that cells can release microparticles (50-350 nm in apparent diameter) in an ABCA1- and apolipoprotein A-I-dependent manner. The purpose of this study was to explore the ability of a novel synthetic peptide CS-6253 to induce microparticle release from various cell lines in the process of HDL biogenesis. METHODS The effects of CS-6253 on microparticle formation through the ABCA1 transporter were examined in vitro using cell-based systems and pharmacologic manipulations. RESULTS In cell-based systems combined with fast performance liquid chromatography and nano-sight-tracking analysis, we show that ABCA1 and CS-6253 mediate and increase the production of microparticles containing cholesterol. CS-6253 in baby hamster kidney cells not expressing ABCA1 (baby hamster kidney mock cells) did not alter cholesterol removal across the plasma membrane in the absence of ABCA1 expression even at high concentrations. We report that CS-6253 is not cytotoxic. CONCLUSIONS The present study shows that CS-6253 generates cholesterol containing microparticles with size heterogeneity (100-350 nm) in an ABCA1-dependent manner. We show that microparticles contribute to cell cholesterol efflux from monocyte-macrophage cells. At high doses, CS-6253 is not able to extract cholesterol from cells not expressing ABCA1, indicating that CS-6253 requires ABCA1 cooperation for cholesterol mobilization. We conclude that CS-6253 is an ABCA1 agonist peptide that promotes cellular cholesterol efflux through HDL biogenesis and microparticle formation.
Collapse
|
18
|
|
19
|
Yu XH, Zhang DW, Zheng XL, Tang CK. Cholesterol transport system: An integrated cholesterol transport model involved in atherosclerosis. Prog Lipid Res 2018; 73:65-91. [PMID: 30528667 DOI: 10.1016/j.plipres.2018.12.002] [Citation(s) in RCA: 148] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 10/30/2018] [Accepted: 12/01/2018] [Indexed: 02/07/2023]
Abstract
Atherosclerosis, the pathological basis of most cardiovascular disease (CVD), is closely associated with cholesterol accumulation in the arterial intima. Excessive cholesterol is removed by the reverse cholesterol transport (RCT) pathway, representing a major antiatherogenic mechanism. In addition to the RCT, other pathways are required for maintaining the whole-body cholesterol homeostasis. Thus, we propose a working model of integrated cholesterol transport, termed the cholesterol transport system (CTS), to describe body cholesterol metabolism. The novel model not only involves the classical view of RCT but also contains other steps, such as cholesterol absorption in the small intestine, low-density lipoprotein uptake by the liver, and transintestinal cholesterol excretion. Extensive studies have shown that dysfunctional CTS is one of the major causes for hypercholesterolemia and atherosclerosis. Currently, several drugs are available to improve the CTS efficiently. There are also several therapeutic approaches that have entered into clinical trials and shown considerable promise for decreasing the risk of CVD. In recent years, a variety of novel findings reveal the molecular mechanisms for the CTS and its role in the development of atherosclerosis, thereby providing novel insights into the understanding of whole-body cholesterol transport and metabolism. In this review, we summarize the latest advances in this area with an emphasis on the therapeutic potential of targeting the CTS in CVD patients.
Collapse
Affiliation(s)
- Xiao-Hua Yu
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Medical Research Experiment Center, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan 421001, China
| | - Da-Wei Zhang
- Department of Pediatrics and Group on the Molecular and Cell Biology of Lipids, University of Alberta, Alberta, Canada
| | - Xi-Long Zheng
- Department of Biochemistry and Molecular Biology, Libin Cardiovascular Institute of Alberta, Cumming School of Medicine, University of Calgary, Health Sciences Center, 3330 Hospital Dr NW, Calgary, Alberta T2N 4N1, Canada
| | - Chao-Ke Tang
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Medical Research Experiment Center, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan 421001, China.
| |
Collapse
|
20
|
Tsujita M, Wolska A, Gutmann DAP, Remaley AT. Reconstituted Discoidal High-Density Lipoproteins: Bioinspired Nanodiscs with Many Unexpected Applications. Curr Atheroscler Rep 2018; 20:59. [PMID: 30397748 DOI: 10.1007/s11883-018-0759-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
PURPOSE OF REVIEW Summarize the initial discovery of discoidal high-density lipoprotein (HDL) in human plasma and review more recent innovations that span the use of reconstituted nanodisc HDL for membrane protein characterization to its use as a drug carrier and a novel therapeutic agent for cardiovascular disease. RECENT FINDINGS Using a wide variety of biophysical techniques, the structure and composition of endogenous discoidal HDL have now largely been solved. This has led to the development of new methods for the in vitro reconstitution of nanodisc HDL, which have proven to have a wide variety of biomedical applications. Nanodisc HDL has been used as a platform for mimicking the plasma membrane for the reconstitution and investigation of the structures of several plasma membrane proteins, such as cytochrome P450s and ABC transporters. Nanodisc HDL has also been designed as drug carriers to transport amphipathic, as well as hydrophobic small molecules, and has potential therapeutic applications for several diseases. Finally, nanodisc HDL itself like native discoidal HDL can mediate cholesterol efflux from cells and are currently being tested in late-stage clinical trials for cardiovascular disease. The discovery of the characterization of native discoidal HDL has inspired a new field of synthetic nanodisc HDL, which has offered a growing number of unanticipated biomedical applications.
Collapse
Affiliation(s)
- Maki Tsujita
- Department of Biochemistry, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, 467-8601, Japan.
| | - Anna Wolska
- Lipoprotein Metabolism Laboratory, Translational Vascular Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | | | - Alan T Remaley
- Lipoprotein Metabolism Laboratory, Translational Vascular Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| |
Collapse
|
21
|
Sasamura A, Akazawa S, Haraguchi A, Horie I, Ando T, Abiru N, Takei H, Nittono H, Une M, Kurosawa T, Murai T, Naruse H, Nakayama T, Kotani K, Remaley AT, Kawakami A. Late-onset Cerebrotendinous Xanthomatosis with a Novel Mutation in the CYP27A1 Gene. INTERNAL MEDICINE (TOKYO, JAPAN) 2018. [PMID: 29434128 DOI: 10.2169/internalmedicine.0120‐17] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Cerebrotendinous xanthomatosis (CTX) is a rare, autosomal recessive, inborn disruption in bile acid synthesis characterized by severe systemic xanthomas, cataracts and neurological injuries occurring before adolescence without elevation of the serum cholesterol or triglyceride levels. CTX is caused by a deficiency of the mitochondrial enzyme sterol 27-hydroxylase, which is encoded by the CYP27A1 gene. We herein report a 50-year-old Japanese woman with late-onset CTX who had no relevant symptoms before the development of bilateral Achilles tendon xanthomas in middle age. A genetic analysis revealed a compound heterozygous mutation in the CYP27A1 gene with a previously known missense mutation (NM_000784.3:c.1421 G>A) and a novel frame shift mutation of NM_000784.3:c.1342_1343insCACC.
Collapse
Affiliation(s)
- Akari Sasamura
- Department of Endocrinology and Metabolism, Nagasaki University Hospital, Japan
| | - Satoru Akazawa
- Department of Endocrinology and Metabolism, Nagasaki University Hospital, Japan
| | - Ai Haraguchi
- Department of Endocrinology and Metabolism, Nagasaki University Hospital, Japan
| | - Ichiro Horie
- Department of Endocrinology and Metabolism, Nagasaki University Hospital, Japan
| | - Takao Ando
- Department of Endocrinology and Metabolism, Nagasaki University Hospital, Japan
| | - Norio Abiru
- Department of Endocrinology and Metabolism, Nagasaki University Hospital, Japan
| | | | | | - Mizuho Une
- Faculty of Pharmaceutical Science, Hiroshima International University, Japan
| | - Takao Kurosawa
- School of Pharmaceutical Sciences, Health Sciences University of Hokkaido, Japan
| | - Tsuyoshi Murai
- School of Pharmaceutical Sciences, Health Sciences University of Hokkaido, Japan
| | - Hiromu Naruse
- Division of Laboratory Medicine, Department of Pathology and Microbiology, Nihon University School of Medicine, Japan.,Health Science Research Institute, Inc., Japan
| | - Tomohiro Nakayama
- Division of Laboratory Medicine, Department of Pathology and Microbiology, Nihon University School of Medicine, Japan.,Division of Companion Diagnostics, Department of Pathology of Microbiology, Nihon University School of Medicine, Japan
| | - Kazuhiko Kotani
- Division of Community and Family Medicine, Jichi Medical University, Japan
| | - Alan T Remaley
- Lipoprotein Metabolism Section, National Institutes of Health, USA
| | - Atsushi Kawakami
- Department of Endocrinology and Metabolism, Nagasaki University Hospital, Japan
| |
Collapse
|
22
|
Sasamura A, Akazawa S, Haraguchi A, Horie I, Ando T, Abiru N, Takei H, Nittono H, Une M, Kurosawa T, Murai T, Naruse H, Nakayama T, Kotani K, Remaley AT, Kawakami A. Late-onset Cerebrotendinous Xanthomatosis with a Novel Mutation in the CYP27A1 Gene. Intern Med 2018; 57:1611-1616. [PMID: 29434128 PMCID: PMC6028668 DOI: 10.2169/internalmedicine.0120-17] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Cerebrotendinous xanthomatosis (CTX) is a rare, autosomal recessive, inborn disruption in bile acid synthesis characterized by severe systemic xanthomas, cataracts and neurological injuries occurring before adolescence without elevation of the serum cholesterol or triglyceride levels. CTX is caused by a deficiency of the mitochondrial enzyme sterol 27-hydroxylase, which is encoded by the CYP27A1 gene. We herein report a 50-year-old Japanese woman with late-onset CTX who had no relevant symptoms before the development of bilateral Achilles tendon xanthomas in middle age. A genetic analysis revealed a compound heterozygous mutation in the CYP27A1 gene with a previously known missense mutation (NM_000784.3:c.1421 G>A) and a novel frame shift mutation of NM_000784.3:c.1342_1343insCACC.
Collapse
Affiliation(s)
- Akari Sasamura
- Department of Endocrinology and Metabolism, Nagasaki University Hospital, Japan
| | - Satoru Akazawa
- Department of Endocrinology and Metabolism, Nagasaki University Hospital, Japan
| | - Ai Haraguchi
- Department of Endocrinology and Metabolism, Nagasaki University Hospital, Japan
| | - Ichiro Horie
- Department of Endocrinology and Metabolism, Nagasaki University Hospital, Japan
| | - Takao Ando
- Department of Endocrinology and Metabolism, Nagasaki University Hospital, Japan
| | - Norio Abiru
- Department of Endocrinology and Metabolism, Nagasaki University Hospital, Japan
| | | | | | - Mizuho Une
- Faculty of Pharmaceutical Science, Hiroshima International University, Japan
| | - Takao Kurosawa
- School of Pharmaceutical Sciences, Health Sciences University of Hokkaido, Japan
| | - Tsuyoshi Murai
- School of Pharmaceutical Sciences, Health Sciences University of Hokkaido, Japan
| | - Hiromu Naruse
- Division of Laboratory Medicine, Department of Pathology and Microbiology, Nihon University School of Medicine, Japan
- Health Science Research Institute, Inc., Japan
| | - Tomohiro Nakayama
- Division of Laboratory Medicine, Department of Pathology and Microbiology, Nihon University School of Medicine, Japan
- Division of Companion Diagnostics, Department of Pathology of Microbiology, Nihon University School of Medicine, Japan
| | - Kazuhiko Kotani
- Division of Community and Family Medicine, Jichi Medical University, Japan
| | - Alan T Remaley
- Lipoprotein Metabolism Section, National Institutes of Health, USA
| | - Atsushi Kawakami
- Department of Endocrinology and Metabolism, Nagasaki University Hospital, Japan
| |
Collapse
|
23
|
Molecular dynamics simulations of lipid nanodiscs. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2018; 1860:2094-2107. [PMID: 29729280 DOI: 10.1016/j.bbamem.2018.04.015] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/31/2017] [Revised: 04/27/2018] [Accepted: 04/28/2018] [Indexed: 01/02/2023]
Abstract
A lipid nanodisc is a discoidal lipid bilayer stabilized by proteins, peptides, or polymers on its edge. Nanodiscs have two important connections to structural biology. The first is associated with high-density lipoprotein (HDL), a particle with a variety of functionalities including lipid transport. Nascent HDL (nHDL) is a nanodisc stabilized by Apolipoprotein A-I (APOA1). Determining the structure of APOA1 and its mimetic peptides in nanodiscs is crucial to understanding pathologies related to HDL maturation and designing effective therapies. Secondly, nanodiscs offer non-detergent membrane-mimicking environments and greatly facilitate structural studies of membrane proteins. Although seemingly similar, natural and synthetic nanodiscs are different in that nHDL is heterogeneous in size, due to APOA1 elasticity, and gradually matures to become spherical. Synthetic nanodiscs, in contrast, should be homogenous, stable, and size-tunable. This report reviews previous molecular dynamics (MD) simulation studies of nanodiscs and illustrates convergence and accuracy issues using results from new multi-microsecond atomistic MD simulations. These new simulations reveal that APOA1 helices take 10-20 μs to rearrange on the nanodisc, while peptides take 2 μs to migrate from the disc surfaces to the edge. These systems can also become kinetically trapped depending on the initial conditions. For example, APOA1 was trapped in a biologically irrelevant conformation for the duration of a 10 μs trajectory; the peptides were similarly trapped for 5 μs. It therefore remains essential to validate MD simulations of these systems with experiments due to convergence and accuracy issues. This article is part of a Special Issue entitled: Emergence of Complex Behavior in Biomembranes edited by Marjorie Longo.
Collapse
|
24
|
Islam RM, Pourmousa M, Sviridov D, Gordon SM, Neufeld EB, Freeman LA, Perrin BS, Pastor RW, Remaley AT. Structural properties of apolipoprotein A-I mimetic peptides that promote ABCA1-dependent cholesterol efflux. Sci Rep 2018; 8:2956. [PMID: 29440748 PMCID: PMC5811490 DOI: 10.1038/s41598-018-20965-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 01/29/2018] [Indexed: 01/05/2023] Open
Abstract
Peptides mimicking the major protein of highdensity lipoprotein (HDL), apolipoprotein A-I (apoA-I), are promising therapeutics for cardiovascular diseases. Similar to apoA-I, their atheroprotective property is attributed to their ability to form discoidal HDL-like particles by extracting cellular cholesterol and phospholipids from lipid microdomains created by the ABCA1 transporter in a process called cholesterol efflux. The structural features of peptides that enable cholesterol efflux are not well understood. Herein, four synthetic amphipathic peptides denoted ELK, which only contain Glu, Leu, Lys, and sometimes Ala, and which have a wide range of net charges and hydrophobicities, were examined for cholesterol efflux. Experiments show that ELKs with a net neutral charge and a hydrophobic face that subtends an angle of at least 140° are optimal for cholesterol efflux. All-atom molecular dynamics simulations show that peptides that are effective in promoting cholesterol efflux stabilize HDL nanodiscs formed by these peptides by the orderly covering of the hydrophobic acyl chains on the edge of the disc. In contrast to apoA-I, which forms an anti-parallel double belt around the HDL, active peptides assemble in a mostly anti-parallel “picket fence” arrangement. These results shed light on the efflux ability of apoA-I mimetics and inform the future design of such therapeutics.
Collapse
Affiliation(s)
- Rafique M Islam
- School of Systems Biology, George Mason University, Fairfax, VA, 22030, USA.,Cardiovascular and Pulmonary Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Mohsen Pourmousa
- Laboratory of Computational Biology, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Denis Sviridov
- Cardiovascular and Pulmonary Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Scott M Gordon
- Cardiovascular and Pulmonary Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Edward B Neufeld
- Cardiovascular and Pulmonary Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Lita A Freeman
- Cardiovascular and Pulmonary Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - B Scott Perrin
- Laboratory of Computational Biology, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Richard W Pastor
- Laboratory of Computational Biology, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, 20892, USA.
| | - Alan T Remaley
- Cardiovascular and Pulmonary Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
25
|
Abstract
Releasing sterols to the extracellular milieu is an important part of sterol homeostasis in cells and in the body. ATP-binding cassette transporter A1 (ABCA1) plays an essential role in cellular phospholipid and sterol release to lipid-free or lipid-poor apolipoprotein A-I (apoA-I), the major apolipoprotein in high-density lipoprotein (HDL), and constitutes the first step in the formation of nascent HDL. Loss-of-function mutations in the ABCA1 gene lead to a rare disease known as Tangier disease that causes severe deficiency in plasma HDL level. Mammalian cells receive exogenous cholesterol mainly from low-density lipoprotein. In addition, they synthesize cholesterol endogenously, as well as multiple precursor sterols that are sterol intermediates en route to be converted to cholesterol. HDL contains phospholipids, cholesterol, and precursor sterols, and ABCA1 has an ability to release phospholipids and various sterol molecules. Recent studies using model cell lines showed that ABCA1 prefers to use sterols newly synthesized endogenously as its preferred substrate, rather than cholesterol derived from LDL or cholesterol being recycled within the cells. Here, we describe several methods at the cell culture level to monitor ABCA1-dependent release of sterol molecules to apoA-I present at the cell exterior. Sterol release can be assessed by using a simple colorimetric enzymatic assay, and/or by monitoring the radioactivities of radiolabeled cholesterol incorporated into the cells, and/or of sterols biosynthesized from radioactive acetate, and/or by using gas chromatography-mass spectrometry analysis of various sterols present in medium and in cells. We also discuss the pros and cons of these methods. Together, these methods allow researchers to detect the release not only of cholesterol but also of other sterols present in minor quantities.
Collapse
Affiliation(s)
- Yoshio Yamauchi
- Department of Biochemistry II, Nagoya University Graduate School of Medicine, 65 Tsurumai, Showa-ku, Nagoya, 466-8550, Japan. .,Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo, 113-8657, Japan.
| | - Shinji Yokoyama
- Nutritional Health Science Research Center, and Department of Food and Nutritional Sciences, Chubu University, 1200 Matsumotocho, Kasugai, 487-8501, Japan
| | - Ta-Yuan Chang
- Department of Biochemistry, Geisel School of Medicine at Dartmouth, 7200 Vail Bldg. Room 304, Hanover, NH, 03755, USA.
| |
Collapse
|
26
|
Phillips MC. Is ABCA1 a lipid transfer protein? J Lipid Res 2018; 59:749-763. [PMID: 29305383 DOI: 10.1194/jlr.r082313] [Citation(s) in RCA: 123] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 01/02/2018] [Indexed: 12/16/2022] Open
Abstract
ABCA1 functions as a lipid transporter because it mediates the transfer of cellular phospholipid (PL) and free (unesterified) cholesterol (FC) to apoA-I and related proteins present in the extracellular medium. ABCA1 is a membrane PL translocase and its enzymatic activity leads to transfer of PL molecules from the cytoplasmic leaflet to the exofacial leaflet of a cell plasma membrane (PM). The presence of active ABCA1 in the PM promotes binding of apoA-I to the cell surface. About 10% of this bound apoA-I interacts directly with ABCA1 and stabilizes the transporter. Most of the pool of cell surface-associated apoA-I is bound to lipid domains in the PM that are created by the activity of ABCA1. The amphipathic α-helices in apoA-I confer detergent-like properties on the protein enabling it to solubilize PL and FC in these membrane domains to create a heterogeneous population of discoidal nascent HDL particles. This review focuses on current understanding of the structure-function relationships of human ABCA1 and the molecular mechanisms underlying HDL particle production.
Collapse
Affiliation(s)
- Michael C Phillips
- Division of Translational Medicine and Human Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-5158
| |
Collapse
|
27
|
Relapsing-remitting multiple sclerosis patients display an altered lipoprotein profile with dysfunctional HDL. Sci Rep 2017; 7:43410. [PMID: 28230201 PMCID: PMC5322497 DOI: 10.1038/srep43410] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Accepted: 01/24/2017] [Indexed: 01/08/2023] Open
Abstract
Lipoproteins modulate innate and adaptive immune responses. In the chronic inflammatory disease multiple sclerosis (MS), reports on lipoprotein level alterations are inconsistent and it is unclear whether lipoprotein function is affected. Using nuclear magnetic resonance (NMR) spectroscopy, we analysed the lipoprotein profile of relapsing-remitting (RR) MS patients, progressive MS patients and healthy controls (HC). We observed smaller LDL in RRMS patients compared to healthy controls and to progressive MS patients. Furthermore, low-BMI (BMI ≤ 23 kg/m2) RRMS patients show increased levels of small HDL (sHDL), accompanied by larger, triglyceride (TG)-rich VLDL, and a higher lipoprotein insulin resistance (LP-IR) index. These alterations coincide with a reduced serum capacity to accept cholesterol via ATP-binding cassette (ABC) transporter G1, an impaired ability of HDL3 to suppress inflammatory activity of human monocytes, and modifications of HDL3’s main protein component ApoA-I. In summary, lipoprotein levels and function are altered in RRMS patients, especially in low-BMI patients, which may contribute to disease progression in these patients.
Collapse
|
28
|
Recio C, Maione F, Iqbal AJ, Mascolo N, De Feo V. The Potential Therapeutic Application of Peptides and Peptidomimetics in Cardiovascular Disease. Front Pharmacol 2017; 7:526. [PMID: 28111551 PMCID: PMC5216031 DOI: 10.3389/fphar.2016.00526] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Accepted: 12/19/2016] [Indexed: 12/11/2022] Open
Abstract
Cardiovascular disease (CVD) remains a leading cause of mortality and morbidity worldwide. Numerous therapies are currently under investigation to improve pathological cardiovascular complications, but yet, there have been very few new medications approved for intervention/treatment. Therefore, new approaches to treat CVD are urgently required. Attempts to prevent vascular complications usually involve amelioration of contributing risk factors and underlying processes such as inflammation, obesity, hyperglycaemia, or hypercholesterolemia. Historically, the development of peptides as therapeutic agents has been avoided by the Pharmaceutical industry due to their low stability, size, rate of degradation, and poor delivery. However, more recently, resurgence has taken place in developing peptides and their mimetics for therapeutic intervention. As a result, increased attention has been placed upon using peptides that mimic the function of mediators involved in pathologic processes during vascular damage. This review will provide an overview on novel targets and experimental therapeutic approaches based on peptidomimetics for modulation in CVD. We aim to specifically examine apolipoprotein A-I (apoA-I) and apoE mimetic peptides and their role in cholesterol transport during atherosclerosis, suppressors of cytokine signaling (SOCS)1-derived peptides and annexin-A1 as potent inhibitors of inflammation, incretin mimetics and their function in glucose-insulin tolerance, among others. With improvements in technology and synthesis platforms the future looks promising for the development of novel peptides and mimetics for therapeutic use. However, within the area of CVD much more work is required to identify and improve our understanding of peptide structure, interaction, and function in order to select the best targets to take forward for treatment.
Collapse
Affiliation(s)
- Carlota Recio
- Sir William Dunn School of Pathology, University of Oxford Oxford, UK
| | - Francesco Maione
- Department of Pharmacy, University of Naples Federico II Naples, Italy
| | - Asif J Iqbal
- Sir William Dunn School of Pathology, University of Oxford Oxford, UK
| | - Nicola Mascolo
- Department of Pharmacy, University of Naples Federico II Naples, Italy
| | - Vincenzo De Feo
- Department of Pharmacy, University of Salerno Salerno, Italy
| |
Collapse
|
29
|
McMahon M, Grossman J, Chen W, Hahn BH. Inflammation and the pathogenesis of atherosclerosis in systemic lupus erythematosus. Lupus 2016. [DOI: 10.1177/0961203306071668] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Atherosclerosis is a complicated inflammatory process characterized by the interactions of numerous different moieties including lipids, enzymes, endothelial cells, cytokines, chemokines, leukocytes, adhesion molecules, complement and antibodies. As in the pathogenesis of many lupus disease processes, the increased risk of atherosclerosis seen in systemic lupus erythematosus (SLE) is likely due to the complex interplay of many of these inflammatory mediators. Expanding our understanding of the pathogenesis of atherosclerosis in SLE is critical if we are to improve the quality of care and reduce mortality in this vulnerable population.
Collapse
Affiliation(s)
- M McMahon
- David Geffen School of Medicine at The University of California Los Angeles, Division of Rheumatology, Department of Medicine, Los Angeles, California, USA
| | - J Grossman
- David Geffen School of Medicine at The University of California Los Angeles, Division of Rheumatology, Department of Medicine, Los Angeles, California, USA
| | - W Chen
- David Geffen School of Medicine at The University of California Los Angeles, Division of Rheumatology, Department of Medicine, Los Angeles, California, USA
| | - BH Hahn
- David Geffen School of Medicine at The University of California Los Angeles, Division of Rheumatology, Department of Medicine, Los Angeles, California, USA
| |
Collapse
|
30
|
Gulshan K, Brubaker G, Conger H, Wang S, Zhang R, Hazen SL, Smith JD. PI(4,5)P2 Is Translocated by ABCA1 to the Cell Surface Where It Mediates Apolipoprotein A1 Binding and Nascent HDL Assembly. Circ Res 2016; 119:827-38. [PMID: 27514935 DOI: 10.1161/circresaha.116.308856] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Accepted: 08/11/2016] [Indexed: 12/23/2022]
Abstract
RATIONALE The molecular mechanism by which ATP-binding cassette transporter A1 (ABCA1) mediates cellular binding of apolipoprotein A-I (apoA1) and nascent high-density lipoprotein (HDL) assembly is not well understood. OBJECTIVE To determine the cell surface lipid that mediates apoA1 binding to ABCA1-expressing cells and the role it plays in nascent HDL assembly. METHODS AND RESULTS Using multiple biochemical and biophysical methods, we found that apoA1 binds specifically to phosphatidylinositol (4,5) bis-phosphate (PIP2). Flow cytometry and PIP2 reporter-binding assays demonstrated that ABCA1 led to PIP2 redistribution from the inner to the outer leaflet of the plasma membrane. Enzymatic cleavage of cell surface PIP2 or decreased cellular PIP2 by knockdown of phosphatidylinositol-5-phosphate 4-kinase impaired apoA1 binding and cholesterol efflux to apoA1. PIP2 also increased the spontaneous solubilization of phospholipid liposomes by apoA1. Using site-directed mutagenesis, we found that ABCA1's PIP2 and phosphatidylserine translocase activities are independent from each other. Furthermore, we discovered that PIP2 is effluxed from cells to apoA1, where it is associated with HDL in plasma, and that PIP2 on HDL is taken up by target cells in a scavenger receptor-BI-dependent manner. Mouse plasma PIP2 levels are apoA1 gene dosage-dependent and are >1 μM in apoA1 transgenic mice. CONCLUSIONS ABCA1 has PIP2 floppase activity, which increases cell surface PIP2 levels that mediate apoA1 binding and lipid efflux during nascent HDL assembly. We found that PIP2 itself is effluxed to apoA1 and it circulates on plasma HDL, where it can be taken up via the HDL receptor scavenger receptor-BI.
Collapse
Affiliation(s)
- Kailash Gulshan
- From the Department of Cellular and Molecular Medicine (K.G., G.B., H.C., S.W., R.Z., S.L.H., J.D.S.) and Department of Cardiovascular Medicine (S.L.H., J.D.S.), Cleveland Clinic, OH.
| | - Gregory Brubaker
- From the Department of Cellular and Molecular Medicine (K.G., G.B., H.C., S.W., R.Z., S.L.H., J.D.S.) and Department of Cardiovascular Medicine (S.L.H., J.D.S.), Cleveland Clinic, OH
| | - Heather Conger
- From the Department of Cellular and Molecular Medicine (K.G., G.B., H.C., S.W., R.Z., S.L.H., J.D.S.) and Department of Cardiovascular Medicine (S.L.H., J.D.S.), Cleveland Clinic, OH
| | - Shuhui Wang
- From the Department of Cellular and Molecular Medicine (K.G., G.B., H.C., S.W., R.Z., S.L.H., J.D.S.) and Department of Cardiovascular Medicine (S.L.H., J.D.S.), Cleveland Clinic, OH
| | - Renliang Zhang
- From the Department of Cellular and Molecular Medicine (K.G., G.B., H.C., S.W., R.Z., S.L.H., J.D.S.) and Department of Cardiovascular Medicine (S.L.H., J.D.S.), Cleveland Clinic, OH
| | - Stanley L Hazen
- From the Department of Cellular and Molecular Medicine (K.G., G.B., H.C., S.W., R.Z., S.L.H., J.D.S.) and Department of Cardiovascular Medicine (S.L.H., J.D.S.), Cleveland Clinic, OH
| | - Jonathan D Smith
- From the Department of Cellular and Molecular Medicine (K.G., G.B., H.C., S.W., R.Z., S.L.H., J.D.S.) and Department of Cardiovascular Medicine (S.L.H., J.D.S.), Cleveland Clinic, OH.
| |
Collapse
|
31
|
Taylor MJ, Sanjanwala AR, Morin EE, Rowland-Fisher E, Anderson K, Schwendeman A, Rainey WE. Synthetic High-Density Lipoprotein (sHDL) Inhibits Steroid Production in HAC15 Adrenal Cells. Endocrinology 2016; 157:3122-9. [PMID: 27253994 PMCID: PMC4967112 DOI: 10.1210/en.2014-1663] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Accepted: 05/26/2016] [Indexed: 11/19/2022]
Abstract
High density lipoprotein (HDL) transported cholesterol represents one of the sources of substrate for adrenal steroid production. Synthetic HDL (sHDL) particles represent a new therapeutic option to reduce atherosclerotic plaque burden by increasing cholesterol efflux from macrophage cells. The effects of the sHDL particles on steroidogenic cells have not been explored. sHDL, specifically ETC-642, was studied in HAC15 adrenocortical cells. Cells were treated with sHDL, forskolin, 22R-hydroxycholesterol, or pregnenolone. Experiments included time and concentration response curves, followed by steroid assay. Quantitative real-time RT-PCR was used to study mRNA of 3-hydroxy-3-methyl-glutaryl-coenzyme A reductase, lanosterol 14-α-methylase, cholesterol side-chain cleavage enzyme, and steroid acute regulatory protein. Cholesterol assay was performed using cell culture media and cell lipid extracts from a dose response experiment. sHDL significantly inhibited production of cortisol. Inhibition occurred in a concentration- and time-dependent manner and in a concentration range of 3μM-50μM. Forskolin (10μM) stimulated cortisol production was also inhibited. Incubation with 22R-hydroxycholesterol (10μM) and pregnenolone (10μM) increased cortisol production, which was unaffected by sHDL treatment. sHDL increased transcript levels for the rate-limiting cholesterol biosynthetic enzyme, 3-hydroxy-3-methyl-glutaryl-coenzyme A reductase. Extracellular cholesterol assayed in culture media showed a positive correlation with increasing concentration of sHDL, whereas intracellular cholesterol decreased after treatment with sHDL. The current study suggests that sHDL inhibits HAC15 adrenal cell steroid production by efflux of cholesterol, leading to an overall decrease in steroid production and an adaptive rise in adrenal cholesterol biosynthesis.
Collapse
Affiliation(s)
- Matthew J Taylor
- Departments of Molecular and Integrative Physiology and Internal Medicine (M.J.T., A.R.S., K.A.,W.E.R.)., University of Michigan, Ann Arbor, Michigan 48109; Medical College of Georgia (A.R.S.), Georgia Regents University, Augusta, Georgia 30912; Department of Pharmaceutical Science (E.E.M., A.S.), Biointerfaces Institute, University of Michigan, Ann Arbor, Michigan 48109; and Department of Medicinal Chemistry (E.R.-F.), College of Pharmacy, University of Michigan, Ann Arbor, Michigan 48109
| | - Aalok R Sanjanwala
- Departments of Molecular and Integrative Physiology and Internal Medicine (M.J.T., A.R.S., K.A.,W.E.R.)., University of Michigan, Ann Arbor, Michigan 48109; Medical College of Georgia (A.R.S.), Georgia Regents University, Augusta, Georgia 30912; Department of Pharmaceutical Science (E.E.M., A.S.), Biointerfaces Institute, University of Michigan, Ann Arbor, Michigan 48109; and Department of Medicinal Chemistry (E.R.-F.), College of Pharmacy, University of Michigan, Ann Arbor, Michigan 48109
| | - Emily E Morin
- Departments of Molecular and Integrative Physiology and Internal Medicine (M.J.T., A.R.S., K.A.,W.E.R.)., University of Michigan, Ann Arbor, Michigan 48109; Medical College of Georgia (A.R.S.), Georgia Regents University, Augusta, Georgia 30912; Department of Pharmaceutical Science (E.E.M., A.S.), Biointerfaces Institute, University of Michigan, Ann Arbor, Michigan 48109; and Department of Medicinal Chemistry (E.R.-F.), College of Pharmacy, University of Michigan, Ann Arbor, Michigan 48109
| | - Elizabeth Rowland-Fisher
- Departments of Molecular and Integrative Physiology and Internal Medicine (M.J.T., A.R.S., K.A.,W.E.R.)., University of Michigan, Ann Arbor, Michigan 48109; Medical College of Georgia (A.R.S.), Georgia Regents University, Augusta, Georgia 30912; Department of Pharmaceutical Science (E.E.M., A.S.), Biointerfaces Institute, University of Michigan, Ann Arbor, Michigan 48109; and Department of Medicinal Chemistry (E.R.-F.), College of Pharmacy, University of Michigan, Ann Arbor, Michigan 48109
| | - Kyle Anderson
- Departments of Molecular and Integrative Physiology and Internal Medicine (M.J.T., A.R.S., K.A.,W.E.R.)., University of Michigan, Ann Arbor, Michigan 48109; Medical College of Georgia (A.R.S.), Georgia Regents University, Augusta, Georgia 30912; Department of Pharmaceutical Science (E.E.M., A.S.), Biointerfaces Institute, University of Michigan, Ann Arbor, Michigan 48109; and Department of Medicinal Chemistry (E.R.-F.), College of Pharmacy, University of Michigan, Ann Arbor, Michigan 48109
| | - Anna Schwendeman
- Departments of Molecular and Integrative Physiology and Internal Medicine (M.J.T., A.R.S., K.A.,W.E.R.)., University of Michigan, Ann Arbor, Michigan 48109; Medical College of Georgia (A.R.S.), Georgia Regents University, Augusta, Georgia 30912; Department of Pharmaceutical Science (E.E.M., A.S.), Biointerfaces Institute, University of Michigan, Ann Arbor, Michigan 48109; and Department of Medicinal Chemistry (E.R.-F.), College of Pharmacy, University of Michigan, Ann Arbor, Michigan 48109
| | - William E Rainey
- Departments of Molecular and Integrative Physiology and Internal Medicine (M.J.T., A.R.S., K.A.,W.E.R.)., University of Michigan, Ann Arbor, Michigan 48109; Medical College of Georgia (A.R.S.), Georgia Regents University, Augusta, Georgia 30912; Department of Pharmaceutical Science (E.E.M., A.S.), Biointerfaces Institute, University of Michigan, Ann Arbor, Michigan 48109; and Department of Medicinal Chemistry (E.R.-F.), College of Pharmacy, University of Michigan, Ann Arbor, Michigan 48109
| |
Collapse
|
32
|
Bocharov AV, Wu T, Baranova IN, Birukova AA, Sviridov D, Vishnyakova TG, Remaley AT, Eggerman TL, Patterson AP, Birukov KG. Synthetic Amphipathic Helical Peptides Targeting CD36 Attenuate Lipopolysaccharide-Induced Inflammation and Acute Lung Injury. THE JOURNAL OF IMMUNOLOGY 2016; 197:611-9. [PMID: 27316682 DOI: 10.4049/jimmunol.1401028] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Accepted: 04/28/2016] [Indexed: 01/07/2023]
Abstract
Synthetic amphipathic helical peptides (SAHPs) designed as apolipoprotein A-I mimetics are known to bind to class B scavenger receptors (SR-Bs), SR-BI, SR-BII, and CD36, receptors that mediate lipid transport and facilitate pathogen recognition. In this study, we evaluated SAHPs, selected for targeting human CD36, by their ability to attenuate LPS-induced inflammation, endothelial barrier dysfunction, and acute lung injury (ALI). L37pA, which targets CD36 and SR-BI equally, inhibited LPS-induced IL-8 secretion and barrier dysfunction in cultured endothelial cells while reducing lung neutrophil infiltration by 40% in a mouse model of LPS-induced ALI. A panel of 20 SAHPs was tested in HEK293 cell lines stably transfected with various SR-Bs to identify SAHPs with preferential selectivity toward CD36. Among several SAHPs targeting both SR-BI/BII and CD36 receptors, ELK-B acted predominantly through CD36. Compared with L37pA, 5A, and ELK SAHPs, ELK-B was most effective in reducing the pulmonary barrier dysfunction, neutrophil migration into the lung, and lung inflammation induced by LPS. We conclude that SAHPs with relative selectivity toward CD36 are more potent at inhibiting acute pulmonary inflammation and dysfunction. These data indicate that therapeutic strategies using SAHPs targeting CD36, but not necessarily mimicking all apolipoprotein A-I functions, may be considered a possible new treatment approach for inflammation-induced ALI and pulmonary edema.
Collapse
Affiliation(s)
- Alexander V Bocharov
- Department of Laboratory Medicine, Clinical Center, National Institutes of Health, Bethesda, MD 20892; National Heart, Lung, and Blood Institute, Bethesda, MD 20892;
| | - Tinghuai Wu
- Lung Injury Center, The University of Chicago, Chicago, IL 60637
| | - Irina N Baranova
- Department of Laboratory Medicine, Clinical Center, National Institutes of Health, Bethesda, MD 20892
| | - Anna A Birukova
- Lung Injury Center, The University of Chicago, Chicago, IL 60637
| | - Denis Sviridov
- National Institute of Diabetes, Digestive, and Kidney Diseases, Bethesda, MD 20892; and
| | - Tatyana G Vishnyakova
- Department of Laboratory Medicine, Clinical Center, National Institutes of Health, Bethesda, MD 20892
| | - Alan T Remaley
- National Heart, Lung, and Blood Institute, Bethesda, MD 20892
| | - Thomas L Eggerman
- Department of Laboratory Medicine, Clinical Center, National Institutes of Health, Bethesda, MD 20892; National Institute of Diabetes, Digestive, and Kidney Diseases, Bethesda, MD 20892; and
| | - Amy P Patterson
- Department of Laboratory Medicine, Clinical Center, National Institutes of Health, Bethesda, MD 20892; Office of Science Policy, Office of the Director, National Institutes of Health, Bethesda, MD 20892
| | | |
Collapse
|
33
|
Sviridov DO, Drake SK, Freeman LA, Remaley AT. Amphipathic polyproline peptides stimulate cholesterol efflux by the ABCA1 transporter. Biochem Biophys Res Commun 2016; 471:560-5. [PMID: 26879139 PMCID: PMC4819318 DOI: 10.1016/j.bbrc.2016.02.032] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 02/09/2016] [Indexed: 12/24/2022]
Abstract
ApoA-I mimetics are short synthetic peptides that contain an amphipathic α-helix and stimulate cholesterol efflux by the ABCA1 transporter in a detergent-like extraction mechanism. We investigated the use of amphipathic peptides with a polypro helix for stimulating cholesterol efflux by ABCA1. Polypro peptides were synthesized with modified prolines, containing either a hydrophobic phenyl group (Prop) or a polar N-acetylgalactosamine (Prog) attached to the pyrrolidine ring and were designated as either PP-2, 3, 4, or 5, depending on the number of 3 amino acid repeat units (Prop-Prog-Prop). Based on molecular modeling, these peptides were predicted to be relatively rigid and to bind to a phospholipid bilayer. By CD spectroscopy, PP peptides formed a Type-II polypro helix in an aqueous solution. PP-2 was inactive in promoting cholesterol efflux, but peptides with more than 2 repeat units were active. PP-4 showed a similar Vmax as a much longer amphipathic α-helical peptide, containing 37 amino acids, but had a Km that was approximately 20-fold lower. PP peptides were specific in that they did not stimulate cholesterol efflux from cells not expressing ABCA1 and were also non-cytotoxic. Addition of PP-3, 4 and 5 to serum promoted the formation of smaller size HDL species (7 nM) and increased its capacity for ABCA1-dependent cholesterol efflux by approximately 20-35% (p < 0.05). Because of their relatively small size and increased potency, amphipathic peptides with a polypro helix may represent an alternative structural motif for the development of apoA-I mimetic peptides.
Collapse
Affiliation(s)
- D O Sviridov
- Lipoprotein Metabolism Section, Cardiopulmonary Branch, NHLBI, National Institutes of Health, Bethesda, MD, USA.
| | - S K Drake
- Critical Care Medicine Department, NIH Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - L A Freeman
- Lipoprotein Metabolism Section, Cardiopulmonary Branch, NHLBI, National Institutes of Health, Bethesda, MD, USA
| | - A T Remaley
- Lipoprotein Metabolism Section, Cardiopulmonary Branch, NHLBI, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
34
|
Lacko AG, Sabnis NA, Nagarajan B, McConathy WJ. HDL as a drug and nucleic acid delivery vehicle. Front Pharmacol 2015; 6:247. [PMID: 26578957 PMCID: PMC4620406 DOI: 10.3389/fphar.2015.00247] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Accepted: 10/12/2015] [Indexed: 01/13/2023] Open
Abstract
This review is intended to evaluate the research findings and potential clinical applications of drug transport systems, developed based on the concepts of the structure/function and physiological role(s) of high density lipoprotein type nanoparticles. These macromolecules provide targeted transport of cholesteryl esters (a highly lipophilic payload) in their natural/physiological environment. The ability to accommodate highly water insoluble constituents in their core regions enables High density lipoproteins (HDL) type nanoparticles to effectively transport hydrophobic drugs subsequent to systemic administration. Even though the application of reconstituted HDL in the treatment of a number of diseases is reviewed, the primary focus is on the application of HDL type drug delivery agents in cancer chemotherapy. The use of both native and synthetic HDL as drug delivery agents is compared to evaluate their respective potentials for commercial and clinical development. The current status and future perspectives for HDL type nanoparticles are discussed, including current obstacles and future applications in therapeutics.
Collapse
Affiliation(s)
- Andras G Lacko
- Department of Integrative Physiology and Anatomy, University of North Texas Health Science Center , Fort Worth, TX, USA ; Department of Pediatrics, University of North Texas Health Science Center , Fort Worth, TX, USA
| | - Nirupama A Sabnis
- Department of Integrative Physiology and Anatomy, University of North Texas Health Science Center , Fort Worth, TX, USA ; Department of Pediatrics, University of North Texas Health Science Center , Fort Worth, TX, USA
| | - Bhavani Nagarajan
- Department of Integrative Physiology and Anatomy, University of North Texas Health Science Center , Fort Worth, TX, USA
| | | |
Collapse
|
35
|
Hafiane A, Bielicki JK, Johansson JO, Genest J. Novel Apo E-Derived ABCA1 Agonist Peptide (CS-6253) Promotes Reverse Cholesterol Transport and Induces Formation of preβ-1 HDL In Vitro. PLoS One 2015. [PMID: 26207756 PMCID: PMC4514675 DOI: 10.1371/journal.pone.0131997] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Apolipoprotein (apo) mimetic peptides replicate some aspects of HDL function. We have previously reported the effects of compound ATI-5261 on its ability to replicate many functions of native apo A-I in the process of HDL biogenesis. ATI-5261 induced muscle toxicity in wild type C57Bl/6 mice, increased CPK, ALT and AST and increase in triglyceride (Tg) levels. Aromatic phenylalanine residues on the non-polar face of ATI-5261, together with positively charged arginine residues at the lipid-water interface were responsible for these effects. This information was used to create a novel analog (CS-6253) that was non-toxic. We evaluated this peptide designed from the carboxyl terminus of apo E, in its ability to mimic apo A-I functionality. Our data shows that the lipidated particles generated by incubating cells overexpressing ABCA1 with lipid free CS-6253 enhances the rate of ABCA1 lipid efflux with high affinity interactions with native ABCA1 oligomeric forms and plasma membrane micro-domains. Interaction between ABCA1 and lipid free CS-6253 resulted in formation of nascent HDL-CS-6253 particles that are actively remodeled in plasma. Mature HDL-CS-6253 particles deliver cholesterol to liver cells via SR-BI in-vitro. CS-6253 significantly increases cholesterol efflux in murine macrophages and in human THP-1 macrophage-derived foam cells expressing ABCA1. Addition of CS-6253 to plasma dose-dependently displaced apo A-I from α-HDL particles and led to de novo formation of preβ-1 HDL that stimulates ABCA1 dependent cholesterol efflux efficiently. When incubated with human plasma CS-6253 was also found to bind with HDL and LDL and promoted the transfer of cholesterol from HDL to LDL predominantly. Our data shows that CS-6253 mimics apo A-I in its ability to promote ABCA1-mediated formation of nascent HDL particles, and enhances formation of preβ-1 HDL with increase in the cycling of apo A-I between the preβ and α-HDL particles in-vitro. These mechanisms are potentially anti-atherogenic.
Collapse
Affiliation(s)
- Anouar Hafiane
- Cardiovascular Research Laboratories Laboratory, Research Institute of the McGill University Health Centre, Montréal, Québec H4A 3J1, Canada
| | - John K. Bielicki
- Lawrence Berkeley National Laboratory, Donner Laboratory, MS1-267, Berkeley, CA, United States of America
| | | | - Jacques Genest
- Cardiovascular Research Laboratories Laboratory, Research Institute of the McGill University Health Centre, Montréal, Québec H4A 3J1, Canada
- * E-mail:
| |
Collapse
|
36
|
Al Gadban MM, Alwan MM, Smith KJ, Hammad SM. Accelerated vascular disease in systemic lupus erythematosus: role of macrophage. Clin Immunol 2015; 157:133-44. [PMID: 25638414 PMCID: PMC4410070 DOI: 10.1016/j.clim.2015.01.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Revised: 01/07/2015] [Accepted: 01/08/2015] [Indexed: 02/07/2023]
Abstract
Atherosclerosis is a chronic inflammatory condition that is considered a major cause of death worldwide. Striking phenomena of atherosclerosis associated with systemic lupus erythematosus (SLE) is its high incidence in young patients. Macrophages are heterogeneous cells that differentiate from hematopoietic progenitors and reside in different tissues to preserve tissue integrity. Macrophages scavenge modified lipids and play a major role in the development of atherosclerosis. When activated, macrophages secret inflammatory cytokines. This activation triggers apoptosis of cells in the vicinity of macrophages. As such, macrophages play a significant role in tissue remodeling including atherosclerotic plaque formation and rupture. In spite of studies carried on identifying the role of macrophages in atherosclerosis, this role has not been studied thoroughly in SLE-associated atherosclerosis. In this review, we address factors released by macrophages as well as extrinsic factors that may control macrophage behavior and their effect on accelerated development of atherosclerosis in SLE.
Collapse
Affiliation(s)
- Mohammed M Al Gadban
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Mohamed M Alwan
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Kent J Smith
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Samar M Hammad
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, USA.
| |
Collapse
|
37
|
Amar MJA, Sakurai T, Sakurai-Ikuta A, Sviridov D, Freeman L, Ahsan L, Remaley AT. A novel apolipoprotein C-II mimetic peptide that activates lipoprotein lipase and decreases serum triglycerides in apolipoprotein E-knockout mice. J Pharmacol Exp Ther 2015; 352:227-35. [PMID: 25395590 PMCID: PMC4293430 DOI: 10.1124/jpet.114.220418] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Accepted: 11/11/2014] [Indexed: 12/16/2022] Open
Abstract
Apolipoprotein A-I (apoA-I) mimetic peptides are currently being developed as possible new agents for the treatment of cardiovascular disease based on their ability to promote cholesterol efflux and their other beneficial antiatherogenic properties. Many of these peptides, however, have been reported to cause transient hypertriglyceridemia due to inhibition of lipolysis by lipoprotein lipase (LPL). We describe a novel bihelical amphipathic peptide (C-II-a) that contains an amphipathic helix (18A) for binding to lipoproteins and stimulating cholesterol efflux as well as a motif based on the last helix of apolipoprotein C-II (apoC-II) that activates lipolysis by LPL. The C-II-a peptide promoted cholesterol efflux from ATP-binding cassette transporter ABCA1-transfected BHK cells similar to apoA-I mimetic peptides. Furthermore, it was shown in vitro to be comparable to the full-length apoC-II protein in activating lipolysis by LPL. When added to serum from a patient with apoC-II deficiency, it restored normal levels of LPL-induced lipolysis and also enhanced lipolysis in serum from patients with type IV and V hypertriglyceridemia. Intravenous injection of C-II-a (30 mg/kg) in apolipoprotein E-knockout mice resulted in a significant reduction of plasma cholesterol and triglycerides of 38 ± 6% and 85 ± 7%, respectively, at 4 hours. When coinjected with the 5A peptide (60 mg/kg), the C-II-a (30 mg/kg) peptide was found to completely block the hypertriglyceridemic effect of the 5A peptide in C57Bl/6 mice. In summary, C-II-a is a novel peptide based on apoC-II, which promotes cholesterol efflux and lipolysis and may therefore be useful for the treatment of apoC-II deficiency and other forms of hypertriglyceridemia.
Collapse
Affiliation(s)
- Marcelo J A Amar
- Lipoprotein Metabolism Section, Cardio-Pulmonary Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Toshihiro Sakurai
- Lipoprotein Metabolism Section, Cardio-Pulmonary Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Akiko Sakurai-Ikuta
- Lipoprotein Metabolism Section, Cardio-Pulmonary Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Denis Sviridov
- Lipoprotein Metabolism Section, Cardio-Pulmonary Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Lita Freeman
- Lipoprotein Metabolism Section, Cardio-Pulmonary Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Lusana Ahsan
- Lipoprotein Metabolism Section, Cardio-Pulmonary Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Alan T Remaley
- Lipoprotein Metabolism Section, Cardio-Pulmonary Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
38
|
Abstract
A wealth of evidence indicates that plasma levels of high-density lipoprotein cholesterol (HDL-C) are inversely related to the risk of cardiovascular disease (CVD). Consequently, HDL-C has been considered a target for therapy in order to reduce the residual CVD burden that remains significant, even after application of current state-of-the-art medical interventions. In recent years, however, a number of clinical trials of therapeutic strategies that increase HDL-C levels failed to show the anticipated beneficial effect on CVD outcomes. As a result, attention has begun to shift toward strategies to improve HDL functionality, rather than levels of HDL-C per se. ApoA-I, the major protein component of HDL, is considered to play an important role in many of the antiatherogenic functions of HDL, most notably reverse cholesterol transport (RCT), and several therapies have been developed to mimic apoA-I function, including administration of apoA-I, mutated variants of apoA-I, and apoA-I mimetic peptides. Based on the potential anti-inflammatory effects, apoA-I mimetics hold promise not only as anti-atherosclerotic therapy but also in other therapeutic areas.
Collapse
Affiliation(s)
- R M Stoekenbroek
- Department of Vascular Medicine, Academic Medical Center, 22660, 1100 DD, Amsterdam, The Netherlands
| | | | | |
Collapse
|
39
|
Neufeld EB, O'Brien K, Walts AD, Stonik JA, Malide D, Combs CA, Remaley AT. The Human ABCG1 Transporter Mobilizes Plasma Membrane and Late Endosomal Non-Sphingomyelin-Associated-Cholesterol for Efflux and Esterification. BIOLOGY 2014; 3:866-91. [PMID: 25485894 PMCID: PMC4280515 DOI: 10.3390/biology3040866] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Revised: 11/22/2014] [Accepted: 11/26/2014] [Indexed: 11/16/2022]
Abstract
We have previously shown that GFP-tagged human ABCG1 on the plasma membrane (PM) and in late endosomes (LE) mobilizes sterol on both sides of the membrane lipid bilayer, thereby increasing cellular cholesterol efflux to lipid surfaces. In the present study, we examined ABCG1-induced changes in membrane cholesterol distribution, organization, and mobility. ABCG1-GFP expression increased the amount of mobile, non-sphingomyelin(SM)-associated cholesterol at the PM and LE, but not the amount of SM-associated-cholesterol or SM. ABCG1-mobilized non-SM-associated-cholesterol rapidly cycled between the PM and LE and effluxed from the PM to extracellular acceptors, or, relocated to intracellular sites of esterification. ABCG1 increased detergent-soluble pools of PM and LE cholesterol, generated detergent-resistant, non-SM-associated PM cholesterol, and increased resistance to both amphotericin B-induced (cholesterol-mediated) and lysenin-induced (SM-mediated) cytolysis, consistent with altered organization of both PM cholesterol and SM. ABCG1 itself resided in detergent-soluble membrane domains. We propose that PM and LE ABCG1 residing at the phase boundary between ordered (Lo) and disordered (Ld) membrane lipid domains alters SM and cholesterol organization thereby increasing cholesterol flux between Lo and Ld, and hence, the amount of cholesterol available for removal by acceptors on either side of the membrane bilayer for either efflux or esterification.
Collapse
Affiliation(s)
- Edward B Neufeld
- Lipoprotein Metabolism Section, Cardiovascular and Pulmonary Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Katherine O'Brien
- Lipid Trafficking Core, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Avram D Walts
- Lipid Trafficking Core, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| | - John A Stonik
- Lipoprotein Metabolism Section, Cardiovascular and Pulmonary Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Daniela Malide
- NHLBI Light Microscopy Core Facility, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Christian A Combs
- NHLBI Light Microscopy Core Facility, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Alan T Remaley
- Lipoprotein Metabolism Section, Cardiovascular and Pulmonary Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
40
|
Cellular Localization and Trafficking of the Human ABCG1 Transporter. BIOLOGY 2014; 3:781-800. [PMID: 25405320 PMCID: PMC4280511 DOI: 10.3390/biology3040781] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Revised: 10/23/2014] [Accepted: 10/28/2014] [Indexed: 11/17/2022]
Abstract
We have developed a suitable heterologous cell expression system to study the localization, trafficking, and site(s) of function of the human ABCG1 transporter. Increased plasma membrane (PM) and late endosomal (LE) cholesterol generated by ABCG1 was removed by lipoproteins and liposomes, but not apoA-I. Delivery of ABCG1 to the PM and LE was required for ABCG1-mediated cellular cholesterol efflux. ABCG1 LEs frequently contacted the PM, providing a collisional mechanism for transfer of ABCG1-mobilized cholesterol, similar to ABCG1-mediated PM cholesterol efflux to lipoproteins. ABCG1-mobilized LE cholesterol also trafficked to the PM by a non-vesicular pathway. Transfer of ABCG1-mobilized cholesterol from the cytoplasmic face of LEs to the PM and concomitant removal of cholesterol from the outer leaflet of the PM bilayer by extracellular acceptors suggests that ABCG1 mobilizes cholesterol on both sides of the lipid bilayer for removal by acceptors. ABCG1 increased uptake of HDL into LEs, consistent with a potential ABCG1-mediated cholesterol efflux pathway involving HDL resecretion. Thus, ABCG1 at the PM mobilizes PM cholesterol and ABCG1 in LE/LYS generates mobile pools of cholesterol that can traffic by both vesicular and non-vesicular pathways to the PM where it can also be transferred to extracellular acceptors with a lipid surface.
Collapse
|
41
|
Uehara Y, Saku K. High-density lipoprotein and atherosclerosis: Roles of lipid transporters. World J Cardiol 2014; 6:1049-1059. [PMID: 25349649 PMCID: PMC4209431 DOI: 10.4330/wjc.v6.i10.1049] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2014] [Revised: 02/10/2014] [Accepted: 08/27/2014] [Indexed: 02/06/2023] Open
Abstract
Various previous studies have found a negative correlation between the risk of cardiovascular events and serum high-density lipoprotein (HDL) cholesterol levels. The reverse cholesterol transport, a pathway of cholesterol from peripheral tissue to liver which has several potent antiatherogenic properties. For instance, the particles of HDL mediate to transport cholesterol from cells in arterial tissues, particularly from atherosclerotic plaques, to the liver. Both ATP-binding cassette transporters (ABC) A1 and ABCG1 are membrane cholesterol transporters and have been implicated in mediating cholesterol effluxes from cells in the presence of HDL and apolipoprotein A-I, a major protein constituent of HDL. Previous studies demonstrated that ABCA1 and ABCG1 or the interaction between ABCA1 and ABCG1 exerted antiatherosclerotic effects. As a therapeutic approach for increasing HDL cholesterol levels, much focus has been placed on increasing HDL cholesterol levels as well as enhancing HDL biochemical functions. HDL therapies that use injections of reconstituted HDL, apoA-I mimetics, or full-length apoA-I have shown dramatic effectiveness. In particular, a novel apoA-I mimetic peptide, Fukuoka University ApoA-I Mimetic Peptide, effectively removes cholesterol via specific ABCA1 and other transporters, such as ABCG1, and has an antiatherosclerotic effect by enhancing the biological functions of HDL without changing circulating HDL cholesterol levels. Thus, HDL-targeting therapy has significant atheroprotective potential, as it uses lipid transporter-targeting agents, and may prove to be a therapeutic tool for atherosclerotic cardiovascular diseases.
Collapse
Affiliation(s)
- Yoshinari Uehara
- Yoshinari Uehara, Keijiro Saku, Department of Cardiology, Fukuoka University School of Medicine, Fukuoka 814-0180, Japan
| | - Keijiro Saku
- Yoshinari Uehara, Keijiro Saku, Department of Cardiology, Fukuoka University School of Medicine, Fukuoka 814-0180, Japan
| |
Collapse
|
42
|
White CR, Garber DW, Anantharamaiah GM. Anti-inflammatory and cholesterol-reducing properties of apolipoprotein mimetics: a review. J Lipid Res 2014; 55:2007-21. [PMID: 25157031 DOI: 10.1194/jlr.r051367] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Reduced levels of HDL cholesterol (HDL-C) are a strong independent predictor of coronary artery disease (CAD) risk. The major anti-atherogenic function of HDL is to mediate reverse cholesterol transport. This response is highly dependent on apoA-I and apoE, protein components of HDL. Randomized clinical trials have assessed effects of several classes of drugs on plasma cholesterol levels in CAD patients. Agents including cholestyramine, fibrates, niacin, and statins significantly lower LDL cholesterol (LDL-C) and induce modest increases in HDL-C, but tolerance issues and undesirable side effects are common. Additionally, residual risk may be present in patients with persistently low HDL-C and other complications despite a reduction in LDL-C. These observations have fueled interest in the development of new pharmacotherapies that positively impact circulating lipoproteins. The goal of this review is to discuss the therapeutic potential of synthetic apolipoprotein mimetic peptides. These include apoA-I mimetic peptides that have undergone initial clinical assessment. We also discuss newer apoE mimetics that mediate the clearance of atherogenic lipids from the circulation and possess anti-inflammatory properties. One of these (AEM-28) has recently been given orphan drug status and is undergoing clinical trials.
Collapse
Affiliation(s)
- C Roger White
- Department of Medicine, Divisions of Cardiovascular Disease, Gerontology, Geriatric Medicine University of Alabama at Birmingham, Birmingham, AL
| | - David W Garber
- Palliative Care, University of Alabama at Birmingham, Birmingham, AL
| | - G M Anantharamaiah
- Palliative Care, University of Alabama at Birmingham, Birmingham, AL Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL
| |
Collapse
|
43
|
Hafiane A, Bielicki JK, Johansson JO, Genest J. Apolipoprotein E derived HDL mimetic peptide ATI-5261 promotes nascent HDL formation and reverse cholesterol transport in vitro. Biochim Biophys Acta Mol Cell Biol Lipids 2014; 1842:1498-512. [PMID: 25091998 DOI: 10.1016/j.bbalip.2014.07.018] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2014] [Revised: 07/17/2014] [Accepted: 07/24/2014] [Indexed: 01/24/2023]
Abstract
Modulation of the reverse cholesterol transport (RCT) pathway may provide a therapeutic target for the prevention and treatment of atherosclerotic cardiovascular disease (CVD). In the present study, we evaluated a novel 26-amino acid apolipoprotein mimetic peptide (ATI-5261) designed from the carboxyl terminal of apoE, in its ability to mimic apoA-I functionality in RCT in vitro. Our data shows that nascent HDL-like (nHDL) particles generated by incubating cells over-expressing ABCA1 with ATI-5261 increase the rate of specific ABCA1 dependent lipid efflux, with high affinity interactions with ABCA1. We also show that these nHDL particles interact with membrane micro-domains in a manner similar to nHDL apoA-I. These nHDL particles then interact with the ABCG1 transporter and are remodeled by plasma HDL-modulating enzymes. Finally, we show that these mature HDL-like particles are taken up by SR-BI for cholesterol delivery to liver cells. This ATI-5621-mediated process mimics apoA-I and may provide a means to prevent cholesterol accumulation in the artery wall. In this study, we propose an integrative physiology approach of HDL biogenesis with the synthetic peptide ATI-5261. These experiments provide new insights for potential therapeutic use of apolipoprotein mimetic peptides.
Collapse
Affiliation(s)
- Anouar Hafiane
- Cardiovascular Genetics Laboratory, Cardiology Division, McGill University Health Centre/Royal Victoria Hospital, Montréal, Québec H3A 1A1, Canada
| | - John K Bielicki
- Lawrence Berkeley National Laboratory, Donner Laboratory, MS1-267 Berkeley, CA, USA
| | | | - Jacques Genest
- Cardiovascular Genetics Laboratory, Cardiology Division, McGill University Health Centre/Royal Victoria Hospital, Montréal, Québec H3A 1A1, Canada.
| |
Collapse
|
44
|
Abstract
High-density lipoproteins (HDL) are a target for drug development because of their proposed anti-atherogenic properties. In this review, we will briefly discuss the currently established drugs for increasing HDL-C, namely niacin and fibrates, and some of their limitations. Next, we will focus on novel alternative therapies that are currently being developed for raising HDL-C, such as CETP inhibitors. Finally, we will conclude with a review of novel drugs that are being developed for modulating the function of HDL based on HDL mimetics. Gaps in our knowledge and the challenges that will have to be overcome for these new HDL based therapies will also be discussed.
Collapse
Affiliation(s)
- Alan T Remaley
- National Heart, Lung and Blood Institute, NIH, 10 Center Drive, Bldg. 10, Rm. 2C-433, Bethesda, MD, USA
| | - Giuseppe D Norata
- Department of Pharmacological Sciences, Università degli Studi di Milano, Milano, Italy Center for the Study of Atherosclerosis, Società Italiana Studio Aterosclerosi, Ospedale Bassini, Cinisello Balsamo, Italy The Blizard Institute, Centre for Diabetes, Barts and The London School of Medicine & Dentistry, Queen Mary University, London, UK
| | - Alberico L Catapano
- Department of Pharmacological Sciences, Università degli Studi di Milano, Milano, Italy IRCCS Multimedica, Milan, Italy
| |
Collapse
|
45
|
Karavia EA, Zvintzou E, Petropoulou PI, Xepapadaki E, Constantinou C, Kypreos KE. HDL quality and functionality: what can proteins and genes predict? Expert Rev Cardiovasc Ther 2014; 12:521-32. [DOI: 10.1586/14779072.2014.896741] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
46
|
Tran TN, Kosaraju MG, Tamamizu-Kato S, Akintunde O, Zheng Y, Bielicki JK, Pinkerton K, Uchida K, Lee YY, Narayanaswami V. Acrolein modification impairs key functional features of rat apolipoprotein E: identification of modified sites by mass spectrometry. Biochemistry 2014; 53:361-75. [PMID: 24325674 DOI: 10.1021/bi401404u] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Apolipoprotein E (apoE), an antiatherogenic apolipoprotein, plays a significant role in the metabolism of lipoproteins. It lowers plasma lipid levels by acting as a ligand for the low-density lipoprotein receptor (LDLr) family of proteins, in addition to playing a role in promoting macrophage cholesterol efflux in atherosclerotic lesions. The objective of this study is to examine the effect of acrolein modification on the structure and function of rat apoE and to determine the sites and nature of modification by mass spectrometry. Acrolein is a highly reactive aldehyde, which is generated endogenously as one of the products of lipid peroxidation and is present in the environment in pollutants such as tobacco smoke and heated oils. In initial studies, acrolein-modified apoE was identified by immunoprecipitation using an acrolein-lysine specific antibody in the plasma of 10-week old male rats that were exposed to filtered air (FA) or low doses of environmental tobacco smoke (ETS). While both groups displayed acrolein-modified apoE in the lipoprotein fraction, the ETS group had higher levels in the lipid-free fraction compared with the FA group. This observation provided the rationale to further investigate the effect of acrolein modification on rat apoE at a molecular level. Treatment of recombinant rat apoE with a 10-fold molar excess of acrolein resulted in (i) a significant decrease in lipid-binding and cholesterol efflux abilities, (ii) impairment in the LDLr- and heparin-binding capabilities, and (iii) significant alterations in the overall stability of the protein. The disruption in the functional abilities is attributed directly or indirectly to acrolein modification yielding an aldimine adduct at K149 and K155 (+38); a propanal adduct at K135 and K138 (+56); an N(ε)-(3-methylpyridinium)lysine (MP-lysine) at K64, K67, and K254 (+76), and an N(ε)-(3-formyl-3,4-dehydropiperidino)lysine (FDP-lysine) derivative at position K68 (+94), as determined by matrix-assisted laser desorption/ionization-time of flight/time of flight mass spectrometry (MALDI-TOF/TOF MS). The loss of function may also be attributed to alterations in the overall fold of the protein as noted by changes in the guanidine HCl-induced unfolding pattern and to protein cross-linking. Overall, disruption of the structural and functional integrity of apoE by oxidative modification of essential lysine residues by acrolein is expected to affect its role in maintaining plasma cholesterol homeostasis and lead to dysregulation in lipid metabolism.
Collapse
Affiliation(s)
- Tuyen N Tran
- Department of Chemistry & Biochemistry, California State University Long Beach , Long Beach, California 90840, United States
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Retention of α-helical structure by HDL mimetic peptide ATI-5261 upon extensive dilution represents an important determinant for stimulating ABCA1 cholesterol efflux with high efficiency. Biochem Biophys Res Commun 2013; 441:71-6. [DOI: 10.1016/j.bbrc.2013.10.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2013] [Accepted: 10/05/2013] [Indexed: 11/20/2022]
|
48
|
Leman LJ, Maryanoff BE, Ghadiri MR. Molecules that mimic apolipoprotein A-I: potential agents for treating atherosclerosis. J Med Chem 2013; 57:2169-96. [PMID: 24168751 DOI: 10.1021/jm4005847] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Certain amphipathic α-helical peptides can functionally mimic many of the properties of full-length apolipoproteins, thereby offering an approach to modulate high-density lipoprotein (HDL) for combating atherosclerosis. In this Perspective, we summarize the key findings and advances over the past 25 years in the development of peptides that mimic apolipoproteins, especially apolipoprotein A-I (apoA-I). This assemblage of information provides a reasonably clear picture of the state of the art in the apolipoprotein mimetic field, an appreciation of the potential for such agents in pharmacotherapy, and a sense of the opportunities for optimizing the functional properties of HDL.
Collapse
Affiliation(s)
- Luke J Leman
- Department of Chemistry and The Skaggs Institute for Chemical Biology, The Scripps Research Institute , 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| | | | | |
Collapse
|
49
|
Nagao K, Hata M, Tanaka K, Takechi Y, Nguyen D, Dhanasekaran P, Lund-Katz S, Phillips MC, Saito H. The roles of C-terminal helices of human apolipoprotein A-I in formation of high-density lipoprotein particles. Biochim Biophys Acta Mol Cell Biol Lipids 2013; 1841:80-7. [PMID: 24120703 DOI: 10.1016/j.bbalip.2013.10.005] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2013] [Revised: 09/07/2013] [Accepted: 10/01/2013] [Indexed: 12/20/2022]
Abstract
Apolipoprotein A-I (apoA-I) accepts cholesterol and phospholipids from ATP-binding cassette transporter A1 (ABCA1)-expressing cells to form high-density lipoprotein (HDL). Human apoA-I has two tertiary structural domains and the C-terminal domain (approximately amino acids 190-243) plays a key role in lipid binding. Although the high lipid affinity region of the C-terminal domain of apoA-I (residues 223-243) is essential for the HDL formation, the function of low lipid affinity region (residues 191-220) remains unclear. To evaluate the role of residues 191-220, we analyzed the structure, lipid binding properties, and HDL formation activity of Δ191-220 apoA-I, in comparison to wild-type and Δ223-243 apoA-I. Although deletion of residues 191-220 has a slight effect on the tertiary structure of apoA-I, the Δ191-220 variant showed intermediate behavior between wild-type and Δ223-243 regarding the formation of hydrophobic sites and lipid interaction through the C-terminal domain. Physicochemical analysis demonstrated that defective lipid binding of Δ191-220 apoA-I is due to the decreased ability to form α-helix structure which provides the energetic source for lipid binding. In addition, the ability to form HDL particles in vitro and induce cholesterol efflux from ABCA1-expressing cells of Δ191-220 apoA-I was also intermediate between wild-type and Δ223-243 apoA-I. These results suggest that despite possessing low lipid affinity, residues 191-220 play a role in enhancing the ability of apoA-I to bind to and solubilize lipids by forming α-helix upon lipid interaction. Our results demonstrate that the combination of low lipid affinity region and high lipid affinity region of apoA-I is required for efficient ABCA1-dependent HDL formation.
Collapse
Affiliation(s)
- Kohjiro Nagao
- Institute of Health Biosciences, The University of Tokushima, 1-78-1 Shomachi, Tokushima 770-8505, Japan; Graduate School of Pharmaceutical Sciences, The University of Tokushima, 1-78-1 Shomachi, Tokushima 770-8505, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Ditiatkovski M, D’Souza W, Kesani R, Chin-Dusting J, de Haan JB, Remaley A, Sviridov D. An apolipoprotein A-I mimetic peptide designed with a reductionist approach stimulates reverse cholesterol transport and reduces atherosclerosis in mice. PLoS One 2013; 8:e68802. [PMID: 23874769 PMCID: PMC3706315 DOI: 10.1371/journal.pone.0068802] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2013] [Accepted: 06/01/2013] [Indexed: 01/19/2023] Open
Abstract
Apolipoprotein A-I (apoA-I) mimetic peptides are considered a promising novel therapeutic approach to prevent and/or treat atherosclerosis. An apoA-I mimetic peptide ELK-2A2K2E was designed with a reductionist approach and has shown exceptional activity in supporting cholesterol efflux but modest anti-inflammatory and anti-oxidant properties in vitro. In this study we compared these in vitro properties with the capacity of this peptide to modify rates of reverse cholesterol transport and development of atherosclerosis in mouse models. The peptide enhanced the rate of reverse cholesterol transport in C57BL/6 mice and reduced atherosclerosis in Apoe(-/-) mice receiving a high fat diet. The peptide modestly reduced the size of the plaques in aortic arch, but was highly active in reducing vascular inflammation and oxidation. Administration of the peptide to Apoe(-/-) mice on a high fat diet reduced the levels of total, high density lipoprotein and non-high density lipoprotein cholesterol and triglycerides. It increased the proportion of smaller HDL particles in plasma at the expense of larger HDL particles, and increased the capacity of the plasma to support cholesterol efflux. Thus, ELK-2A2K2E peptide reduced atherosclerosis in Apoe(-/-) mice, however, the functional activity profile after chronic in vivo administration was different from that found in acute in vitro studies.
Collapse
Affiliation(s)
| | - Wilissa D’Souza
- Baker Heart and Diabetes Institute, Melbourne, Vic., Australia
| | - Rajitha Kesani
- Baker Heart and Diabetes Institute, Melbourne, Vic., Australia
| | | | - Judy B. de Haan
- Baker Heart and Diabetes Institute, Melbourne, Vic., Australia
| | - Alan Remaley
- Lipoprotein Section, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Dmitri Sviridov
- Baker Heart and Diabetes Institute, Melbourne, Vic., Australia
- * E-mail:
| |
Collapse
|