1
|
Little I, Bersie S, Redente EF, McCubbrey AL, Tarling EJ. Alveolar macrophages: guardians of the alveolar lipid galaxy. Curr Opin Lipidol 2025:00041433-990000000-00114. [PMID: 40183504 DOI: 10.1097/mol.0000000000000987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/05/2025]
Abstract
PURPOSE OF REVIEW As the primary guardians at the air-surface interface, the functional profile of alveolar macrophages (AM) is wide-ranging from establishment of the alveolar niche, homeostatic maintenance of surfactant levels, to pathogen clearance and resolution and repair processes. Alveolar lipid homeostasis is disturbed in chronic lung diseases and contributes to disease pathogenesis through extracellular localization in the alveolar lumen or intracellular accumulation in AM. This review aims to provide a focused overview of the state of knowledge of AM, their ontogeny and development during health and disease, and how dysregulated AM lipids play a key role in disease processes, from initiation to resolution. RECENT FINDINGS While lipid-laden macrophages are observed across a broad spectrum of lung diseases, their occurrence has largely been considered consequential. Recent advances in lipidomic profiling of single cell types has revealed that disturbances to lipid homeostasis occur early in disease in tissue-resident cells. Comparisons between inflammatory and fibrotic injury models reveal specific alveolar macrophage subsets with different lipid utilization that contribute to the disease process. SUMMARY Understanding the intricate web of AM population seeding and development and how this niche is perturbed by lipid disturbances may help provide leverage for new interventions.
Collapse
Affiliation(s)
- Isaiah Little
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine at UCLA, University of California Los Angeles, Los Angeles, California
| | - Stephanie Bersie
- Toxicology Graduate Program, Skaggs School of Pharmacy and Pharmaceutical Sciences
| | - Elizabeth F Redente
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
- Department of Pediatrics
| | - Alexandra L McCubbrey
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
- Department of Medicine, National Jewish Health, Denver, Colorado
| | - Elizabeth J Tarling
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine at UCLA, University of California Los Angeles, Los Angeles, California
- Molecular Biology Institute
- Johnsson Comprehensive Cancer Center (JCCC), University of California Los Angeles, Los Angeles, California, USA
| |
Collapse
|
2
|
Guo YQ, Zhao HL, Zhao JM, Li SS, Meng LW, Li J, Qian YW, Li YL, Cui BY, Guo S, Li P, Li CZ. Floralozone attenuates atherosclerotic vascular injury by regulating AMPKα/SREBP-1c pathway and down-regulating miR-33-5p. Eur J Nutr 2025; 64:65. [PMID: 39775070 DOI: 10.1007/s00394-024-03578-6] [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: 10/29/2024] [Accepted: 12/20/2024] [Indexed: 01/11/2025]
Abstract
BACKGROUND Severe disruption of lipid metabolism in vivo is one of the central mechanisms in the development of atherosclerotic vascular injury (AVI). Reverse cholesterol transport (RCT) plays a pivotal role in eliminating excess cholesterol, preventing lipid deposition in the aorta, and reducing plaque formation associated with AVI. Floralozone (FL) reduces endothelial cell injury in AVI rats by regulating sphingosine-1-phosphate (S1P) expression. However, FL's potential to prevent AVI by modulating cholesterol metabolism remains unknown. METHODS In this study, network pharmacology and molecular docking predicted FL's potential targets in AVI protection. AVI rats were induced with a high-sugar, high-fat diet and vitamin D3 injection. FL intervention's effects on aortic pathology and lipid levels were assessed. The expression levels of SREBP-1c, PPARγ, ABCA1, and ABCG1 were evaluated. Raw264.7 macrophages were induced to form foam cells with ox-LDL, and FL's effects on the AMPKα/SREBP-1c pathway and miR-33-5p were investigated. RESULTS FL reduced lipid levels and SREBP-1c expression, increased HDL-C, promoted ABCA1- and ABCG1-mediated cholesterol efflux, and reduced aortic cholesterol accumulation. The AMPKα inhibitor dorsomorphin blocked FL's inhibition of intracellular cholesterol accumulation and SREBP1 down-regulation in foam cells. FL decreased miR-33-5p expression but up-regulated PPARγ, promoting ABCA1- and ABCG1-mediated cholesterol efflux. However, miR-33-5p mimic reduced FL-induced cholesterol efflux, while miR-33-5p inhibitor increased it. CONCLUSION FL may promote foam cell cholesterol efflux by modifying the AMPKα/SREBP-1c pathway and down-regulating miR-33-5p, which targets cholesterol metabolism genes (PPARγ, ABCA1, and ABCG1). These findings provide a new insight into the protective effect of FL on AVI.
Collapse
Affiliation(s)
- Ya-Qi Guo
- College of Pharmacy, Sanquan College of Xinxiang Medical University, Xinxiang, 453003, China
- The Fifth Clinical College, College of Pharmacy, Xinxiang Medical University, Xinxiang, 453003, China
| | - Hong-Lin Zhao
- The Fifth Clinical College, College of Pharmacy, Xinxiang Medical University, Xinxiang, 453003, China
| | - Jin-Ming Zhao
- The Fifth Clinical College, College of Pharmacy, Xinxiang Medical University, Xinxiang, 453003, China
| | - Shan-Shan Li
- The Fifth Clinical College, College of Pharmacy, Xinxiang Medical University, Xinxiang, 453003, China
| | - Liu-Wei Meng
- The Fifth Clinical College, College of Pharmacy, Xinxiang Medical University, Xinxiang, 453003, China
| | - Jiao Li
- The Fifth Clinical College, College of Pharmacy, Xinxiang Medical University, Xinxiang, 453003, China
| | - Yi-Wen Qian
- Department of Pharmacy, College of Basic Medicine and Forensic Medicien, Henan University of Science and Technology, Luoyang, 471000, China
| | - Yin-Lan Li
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Heilongjiang, 150040, China
| | - Bao-Yue Cui
- The Fifth Clinical College, College of Pharmacy, Xinxiang Medical University, Xinxiang, 453003, China
| | - Shuang Guo
- Hubei Key Laboratory of Diabetes and Angiopathy, Hubei University of Science and Technology, Xianning, 437100, China
| | - Peng Li
- The Fifth Clinical College, College of Pharmacy, Xinxiang Medical University, Xinxiang, 453003, China.
| | - Chang-Zheng Li
- College of Pharmacy, Sanquan College of Xinxiang Medical University, Xinxiang, 453003, China.
| |
Collapse
|
3
|
Stolz V, de Freitas e Silva R, Rica R, Zhu C, Preglej T, Hamminger P, Hainberger D, Alteneder M, Müller L, Waldherr M, Waltenberger D, Hladik A, Agerer B, Schuster M, Frey T, Krausgruber T, Knapp S, Campbell C, Schmetterer K, Trauner M, Bergthaler A, Bock C, Boucheron N, Ellmeier W. Nuclear receptor corepressor 1 controls regulatory T cell subset differentiation and effector function. eLife 2024; 13:e78738. [PMID: 39466314 PMCID: PMC11517256 DOI: 10.7554/elife.78738] [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: 03/17/2022] [Accepted: 09/25/2024] [Indexed: 10/29/2024] Open
Abstract
FOXP3+ regulatory T cells (Treg cells) are key for immune homeostasis. Here, we reveal that nuclear receptor corepressor 1 (NCOR1) controls naïve and effector Treg cell states. Upon NCOR1 deletion in T cells, effector Treg cell frequencies were elevated in mice and in in vitro-generated human Treg cells. NCOR1-deficient Treg cells failed to protect mice from severe weight loss and intestinal inflammation associated with CD4+ T cell transfer colitis, indicating impaired suppressive function. NCOR1 controls the transcriptional integrity of Treg cells, since effector gene signatures were already upregulated in naïve NCOR1-deficient Treg cells while effector NCOR1-deficient Treg cells failed to repress genes associated with naïve Treg cells. Moreover, genes related to cholesterol homeostasis including targets of liver X receptor (LXR) were dysregulated in NCOR1-deficient Treg cells. However, genetic ablation of LXRβ in T cells did not revert the effects of NCOR1 deficiency, indicating that NCOR1 controls naïve and effector Treg cell subset composition independent from its ability to repress LXRβ-induced gene expression. Thus, our study reveals that NCOR1 maintains naïve and effector Treg cell states via regulating their transcriptional integrity. We also reveal a critical role for this epigenetic regulator in supporting the suppressive functions of Treg cells in vivo.
Collapse
Affiliation(s)
- Valentina Stolz
- Medical University of Vienna, Center for Pathophysiology, Infectiology and Immunology, Institute of ImmunologyViennaAustria
| | - Rafael de Freitas e Silva
- Medical University of Vienna, Center for Pathophysiology, Infectiology and Immunology, Institute of ImmunologyViennaAustria
| | - Ramona Rica
- Medical University of Vienna, Center for Pathophysiology, Infectiology and Immunology, Institute of ImmunologyViennaAustria
| | - Ci Zhu
- Medical University of Vienna, Center for Pathophysiology, Infectiology and Immunology, Institute of ImmunologyViennaAustria
| | - Teresa Preglej
- Medical University of Vienna, Center for Pathophysiology, Infectiology and Immunology, Institute of ImmunologyViennaAustria
| | - Patricia Hamminger
- Medical University of Vienna, Center for Pathophysiology, Infectiology and Immunology, Institute of ImmunologyViennaAustria
| | - Daniela Hainberger
- Medical University of Vienna, Center for Pathophysiology, Infectiology and Immunology, Institute of ImmunologyViennaAustria
| | - Marlis Alteneder
- Medical University of Vienna, Center for Pathophysiology, Infectiology and Immunology, Institute of ImmunologyViennaAustria
| | - Lena Müller
- Medical University of Vienna, Center for Pathophysiology, Infectiology and Immunology, Institute of ImmunologyViennaAustria
| | - Monika Waldherr
- Medical University of Vienna, Center for Pathophysiology, Infectiology and Immunology, Institute of ImmunologyViennaAustria
| | - Darina Waltenberger
- Medical University of Vienna, Center for Pathophysiology, Infectiology and Immunology, Institute of ImmunologyViennaAustria
| | - Anastasiya Hladik
- Medical University of Vienna, Vienna, Department of Medicine I, Laboratory of Infection BiologyViennaAustria
| | - Benedikt Agerer
- CeMM Research Centre for Molecular Medicine of the Austrian Academy of SciencesViennaAustria
| | - Michael Schuster
- CeMM Research Centre for Molecular Medicine of the Austrian Academy of SciencesViennaAustria
| | - Tobias Frey
- Medical University of Vienna, Department of Laboratory MedicineViennaAustria
| | - Thomas Krausgruber
- CeMM Research Centre for Molecular Medicine of the Austrian Academy of SciencesViennaAustria
- Medical University of Vienna, Center for Medical Statistics, Informatics, and Intelligent Systems, Institute of Artificial IntelligenceViennaAustria
| | - Sylvia Knapp
- Medical University of Vienna, Vienna, Department of Medicine I, Laboratory of Infection BiologyViennaAustria
| | - Clarissa Campbell
- CeMM Research Centre for Molecular Medicine of the Austrian Academy of SciencesViennaAustria
| | - Klaus Schmetterer
- Medical University of Vienna, Department of Laboratory MedicineViennaAustria
| | - Michael Trauner
- Medical University of Vienna, Department of Internal Medicine III, Division of Gastroenterology and Hepatology, Hans Popper Laboratory of Molecular HepatologyViennaAustria
| | - Andreas Bergthaler
- CeMM Research Centre for Molecular Medicine of the Austrian Academy of SciencesViennaAustria
- Medical University of Vienna, Vienna, Center for Pathophysiology, Infectiology and Immunology, Institute for Hygiene and Applied ImmunologyViennaAustria
| | - Christoph Bock
- CeMM Research Centre for Molecular Medicine of the Austrian Academy of SciencesViennaAustria
- Medical University of Vienna, Center for Medical Statistics, Informatics, and Intelligent Systems, Institute of Artificial IntelligenceViennaAustria
| | - Nicole Boucheron
- Medical University of Vienna, Center for Pathophysiology, Infectiology and Immunology, Institute of ImmunologyViennaAustria
| | - Wilfried Ellmeier
- Medical University of Vienna, Center for Pathophysiology, Infectiology and Immunology, Institute of ImmunologyViennaAustria
| |
Collapse
|
4
|
Schilbert HM, Pucker B, Ries D, Viehöver P, Micic Z, Dreyer F, Beckmann K, Wittkop B, Weisshaar B, Holtgräwe D. Mapping‑by‑Sequencing Reveals Genomic Regions Associated with Seed Quality Parameters in Brassica napus. Genes (Basel) 2022; 13:genes13071131. [PMID: 35885914 PMCID: PMC9317104 DOI: 10.3390/genes13071131] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 06/15/2022] [Accepted: 06/22/2022] [Indexed: 11/21/2022] Open
Abstract
Rapeseed (Brassica napus L.) is an important oil crop and has the potential to serve as a highly productive source of protein. This protein exhibits an excellent amino acid composition and has high nutritional value for humans. Seed protein content (SPC) and seed oil content (SOC) are two complex quantitative and polygenic traits which are negatively correlated and assumed to be controlled by additive and epistatic effects. A reduction in seed glucosinolate (GSL) content is desired as GSLs cause a stringent and bitter taste. The goal here was the identification of genomic intervals relevant for seed GSL content and SPC/SOC. Mapping by sequencing (MBS) revealed 30 and 15 new and known genomic intervals associated with seed GSL content and SPC/SOC, respectively. Within these intervals, we identified known but also so far unknown putatively causal genes and sequence variants. A 4 bp insertion in the MYB28 homolog on C09 shows a significant association with a reduction in seed GSL content. This study provides insights into the genetic architecture and potential mechanisms underlying seed quality traits, which will enhance future breeding approaches in B. napus.
Collapse
Affiliation(s)
- Hanna Marie Schilbert
- Genetics and Genomics of Plants, CeBiTec & Faculty of Biology, Bielefeld University, Universitätsstraße 27, 33615 Bielefeld, Germany; (H.M.S.); (B.P.); (D.R.); (P.V.); (B.W.)
- Graduate School DILS, Bielefeld Institute for Bioinformatics Infrastructure (BIBI), Faculty of Technology, Bielefeld University, Universitätsstraße 27, 33615 Bielefeld, Germany
| | - Boas Pucker
- Genetics and Genomics of Plants, CeBiTec & Faculty of Biology, Bielefeld University, Universitätsstraße 27, 33615 Bielefeld, Germany; (H.M.S.); (B.P.); (D.R.); (P.V.); (B.W.)
- Plant Biotechnology and Bioinformatics, Institute of Plant Biology & Braunschweig Integrated Centre of Systems Biology (BRICS), TU Braunschweig, Mendelssohnstraße 4, 38106 Braunschweig, Germany
| | - David Ries
- Genetics and Genomics of Plants, CeBiTec & Faculty of Biology, Bielefeld University, Universitätsstraße 27, 33615 Bielefeld, Germany; (H.M.S.); (B.P.); (D.R.); (P.V.); (B.W.)
| | - Prisca Viehöver
- Genetics and Genomics of Plants, CeBiTec & Faculty of Biology, Bielefeld University, Universitätsstraße 27, 33615 Bielefeld, Germany; (H.M.S.); (B.P.); (D.R.); (P.V.); (B.W.)
| | - Zeljko Micic
- Deutsche Saatveredelung AG, Weissenburger Straße 5, 59557 Lippstadt, Germany;
| | - Felix Dreyer
- NPZ Innovation GmbH, Hohenlieth-Hof 1, 24363 Holtsee, Germany; (F.D.); (K.B.)
| | - Katrin Beckmann
- NPZ Innovation GmbH, Hohenlieth-Hof 1, 24363 Holtsee, Germany; (F.D.); (K.B.)
| | - Benjamin Wittkop
- Department of Plant Breeding, Justus Liebig University, Heinrich-Buff-Ring 26-32, 35392 Giessen, Germany;
| | - Bernd Weisshaar
- Genetics and Genomics of Plants, CeBiTec & Faculty of Biology, Bielefeld University, Universitätsstraße 27, 33615 Bielefeld, Germany; (H.M.S.); (B.P.); (D.R.); (P.V.); (B.W.)
| | - Daniela Holtgräwe
- Genetics and Genomics of Plants, CeBiTec & Faculty of Biology, Bielefeld University, Universitätsstraße 27, 33615 Bielefeld, Germany; (H.M.S.); (B.P.); (D.R.); (P.V.); (B.W.)
- Correspondence:
| |
Collapse
|
5
|
Tall AR, Wang N. New insights into cholesterol efflux via ABCA1. NATURE CARDIOVASCULAR RESEARCH 2022; 1:198-199. [PMID: 37152663 PMCID: PMC10162590 DOI: 10.1038/s44161-022-00036-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
ABCA1 promotes the efflux of cholesterol from cells to HDL and has anti-atherogenic activities. Sun and Li present cryo-EM structures of ABCA1 in the ATP-free and ATP-bound states, which reveal bound cholesterol molecules and suggest a transmembrane cholesterol-transport mechanism.
Collapse
Affiliation(s)
- Alan R. Tall
- Division of Molecular Medicine, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Nan Wang
- Division of Molecular Medicine, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
| |
Collapse
|
6
|
Structure and transport mechanism of the human cholesterol transporter ABCG1. Cell Rep 2022; 38:110298. [PMID: 35081353 DOI: 10.1016/j.celrep.2022.110298] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 12/13/2021] [Accepted: 01/04/2022] [Indexed: 12/31/2022] Open
Abstract
The reverse cholesterol transport pathway is responsible for the maintenance of human cholesterol homeostasis, an imbalance of which usually leads to atherosclerosis. As a key component of this pathway, the ATP-binding cassette transporter ABCG1 forwards cellular cholesterol to the extracellular acceptor nascent high-density lipoprotein (HDL). Here, we report a 3.26-Å cryo-electron microscopy structure of cholesterol-bound ABCG1 in an inward-facing conformation, which represents a turnover condition upon ATP binding. Structural analyses combined with functional assays reveals that a cluster of conserved hydrophobic residues, in addition to two sphingomyelins, constitute a well-defined cholesterol-binding cavity. The exit of this cavity is closed by three pairs of conserved Phe residues, which constitute a hydrophobic path for the release of cholesterol in an acceptor concentration-dependent manner. Overall, we propose an ABCG1-driven cholesterol transport cycle initiated by sphingomyelin-assisted cholesterol recruitment and accomplished by the release of cholesterol to HDL.
Collapse
|
7
|
Wilfahrt D, Philips RL, Lama J, Kizerwetter M, Shapiro MJ, McCue SA, Kennedy MM, Rajcula MJ, Zeng H, Shapiro VS. Histone deacetylase 3 represses cholesterol efflux during CD4 + T-cell activation. eLife 2021; 10:e70978. [PMID: 34854376 PMCID: PMC8639145 DOI: 10.7554/elife.70978] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 11/15/2021] [Indexed: 12/14/2022] Open
Abstract
After antigenic activation, quiescent naive CD4+ T cells alter their metabolism to proliferate. This metabolic shift increases production of nucleotides, amino acids, fatty acids, and sterols. Here, we show that histone deacetylase 3 (HDAC3) is critical for activation of murine peripheral CD4+ T cells. HDAC3-deficient CD4+ T cells failed to proliferate and blast after in vitro TCR/CD28 stimulation. Upon T-cell activation, genes involved in cholesterol biosynthesis are upregulated while genes that promote cholesterol efflux are repressed. HDAC3-deficient CD4+ T cells had reduced levels of cellular cholesterol both before and after activation. HDAC3-deficient cells upregulate cholesterol synthesis appropriately after activation, but fail to repress cholesterol efflux; notably, they overexpress cholesterol efflux transporters ABCA1 and ABCG1. Repression of these genes is the primary function for HDAC3 in peripheral CD4+ T cells, as addition of exogenous cholesterol restored proliferative capacity. Collectively, these findings demonstrate HDAC3 is essential during CD4+ T-cell activation to repress cholesterol efflux.
Collapse
Affiliation(s)
- Drew Wilfahrt
- Department of Immunology, Mayo ClinicRochesterUnited States
| | | | - Jyoti Lama
- Department of Immunology, Mayo ClinicRochesterUnited States
| | | | | | | | | | | | - Hu Zeng
- Department of Immunology, Mayo ClinicRochesterUnited States
- Division of Rheumatology, Department of Medicine, Mayo ClinicRochesterUnited States
| | | |
Collapse
|
8
|
Barna BP, Judson MA, Thomassen MJ. Inflammatory Pathways in Sarcoidosis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1304:39-52. [PMID: 34019262 DOI: 10.1007/978-3-030-68748-9_3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Concepts regarding etiology and pathophysiology of sarcoidosis have changed remarkably within the past 5 years. Sarcoidosis is now viewed as a complex multi-causation disease related to a diverse collection of external environmental or infectious signals. It is generally accepted that the cause of sarcoidosis is unknown. Moreover, concepts of the inflammatory pathway have been modified by the realization that intrinsic genetic factors and innate immunity may modify adaptive immune responses to external triggers. With those potential regulatory pathways in mind, we will attempt to discuss the current understanding of the inflammatory response in sarcoidosis with emphasis on development of pulmonary granulomatous pathology. In that context, we will emphasize that both macrophages and T lymphocytes play key roles, with sometimes overlapping cytokine production (i.e., TNFα and IFN-γ) but also with unique mediators that influence the pathologic picture. Numerous studies have shown that in a sizable number of sarcoidosis patients, granulomas spontaneously resolve, usually within 3 years. Other sarcoidosis patients, however, may develop a chronic granulomatous disease which may subsequently lead to fibrosis. This chapter will outline our current understanding of inflammatory pathways in sarcoidosis which initiate and mediate granulomatous changes or onset of pulmonary fibrosis.
Collapse
Affiliation(s)
- Barbara P Barna
- Program in Lung Cell Biology and Translational Research, Division of Pulmonary and Critical Care Medicine, East Carolina University, Greenville, NC, USA
| | - Marc A Judson
- Division of Pulmonary and Critical Care Medicine, Albany Medical College, Albany, NY, USA
| | - Mary Jane Thomassen
- Program in Lung Cell Biology and Translational Research, Division of Pulmonary and Critical Care Medicine, East Carolina University, Greenville, NC, USA.
| |
Collapse
|
9
|
Mammalian ABCG-transporters, sterols and lipids: To bind perchance to transport? Biochim Biophys Acta Mol Cell Biol Lipids 2020; 1866:158860. [PMID: 33309976 DOI: 10.1016/j.bbalip.2020.158860] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 11/15/2020] [Accepted: 12/08/2020] [Indexed: 02/08/2023]
Abstract
Members of the ATP binding cassette (ABC) transporter family perform a critical function in maintaining lipid homeostasis in cells as well as the transport of drugs. In this review, we provide an update on the ABCG-transporter subfamily member proteins, which include the homodimers ABCG1, ABCG2 and ABCG4 as well as the heterodimeric complex formed between ABCG5 and ABCG8. This review focusses on progress made in this field of research with respect to their function in health and disease and the recognised transporter substrates. We also provide an update on post-translational regulation, including by transporter substrates, and well as the involvement of microRNA as regulators of transporter expression and activity. In addition, we describe progress made in identifying structural elements that have been recognised as important for transport activity. We furthermore discuss the role of lipids such as cholesterol on the transport function of ABCG2, traditionally thought of as a drug transporter, and provide a model of potential cholesterol binding sites for ABCG2.
Collapse
|
10
|
Libby AE, Jones B, Levi M. Learning the ABCs of ATP release. J Biol Chem 2020; 295:5204-5205. [PMID: 32303644 DOI: 10.1074/jbc.h120.013321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
ATP plays important roles outside the cell, but the mechanism by which it is arrives in the extracellular environment is not clear. Dunn et al now show that decreases in cellular cholesterol levels mediated by the ABCG1 transporter increase ATP release by volume-regulated anion channels under hypotonic conditions. Importantly, these results may imply that cells that handle cholesterol differently might experience differential extracellular ATP release during hypotonicity.
Collapse
Affiliation(s)
- Andrew E Libby
- Department of Biochemistry and Molecular and Cellular Biology, Georgetown University, Washington, D. C. 20007
| | - Bryce Jones
- Department of Pharmacology and Physiology, Georgetown University, Washington, D. C. 20007
| | - Moshe Levi
- Department of Biochemistry and Molecular and Cellular Biology, Georgetown University, Washington, D. C. 20007.
| |
Collapse
|
11
|
Cheng J, Cheng A, Clifford BL, Wu X, Hedin U, Maegdefessel L, Pamir N, Sallam T, Tarling EJ, de Aguiar Vallim TQ. MicroRNA-144 Silencing Protects Against Atherosclerosis in Male, but Not Female Mice. Arterioscler Thromb Vasc Biol 2020; 40:412-425. [PMID: 31852219 PMCID: PMC7018399 DOI: 10.1161/atvbaha.119.313633] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 11/15/2019] [Indexed: 12/18/2022]
Abstract
OBJECTIVE Atherosclerosis is a leading cause of death in developed countries. MicroRNAs act as fine-tuners of gene expression and have been shown to have important roles in the pathophysiology and progression of atherosclerosis. We, and others, previously demonstrated that microRNA-144 (miR-144) functions to post-transcriptionally regulate ABCA1 (ATP binding cassette transporter A1) and plasma HDL (high-density lipoprotein) cholesterol levels. Here, we explore how miR-144 inhibition may protect against atherosclerosis. Approach and Results: We demonstrate that miR-144 silencing reduced atherosclerosis in male, but not female low-density lipoprotein receptor null (Ldlr-/-) mice. MiR-144 antagonism increased circulating HDL cholesterol levels, remodeled the HDL particle, and enhanced reverse cholesterol transport. Notably, the effects on HDL and reverse cholesterol transport were more pronounced in male mice suggesting sex-specific differences may contribute to the effects of silencing miR-144 on atherosclerosis. As a molecular mechanism, we identify the oxysterol metabolizing enzyme CYP7B1 (cytochrome P450 enzyme 7B1) as a miR-144 regulated gene in male, but not female mice. Consistent with miR-144-dependent changes in CYP7B1 activity, we show decreased levels of 27-hydroxycholesterol, a known proatherogenic sterol and the endogenous substrate for CYP7B1 in male, but not female mice. CONCLUSIONS Our data demonstrate silencing miR-144 has sex-specific effects and that treatment with antisense oligonucleotides to target miR-144 might result in enhancements in reverse cholesterol transport and oxysterol metabolism in patients with cardiovascular disease.
Collapse
Affiliation(s)
- Joan Cheng
- Department of Biological Chemistry, University of California Los Angeles, California, 90095, USA
| | - Angela Cheng
- Department of Biological Chemistry, University of California Los Angeles, California, 90095, USA
| | - Bethan L. Clifford
- Department of Medicine, University of California Los Angeles, California, 90095, USA
| | - Xiaohui Wu
- Department of Medicine, University of California Los Angeles, California, 90095, USA
| | - Ulf Hedin
- Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden
| | - Lars Maegdefessel
- Department of Medicine, Karolinska Institute, Stockholm, Sweden
- Department of Vascular and Endovascular Surgery, Klinikum rechts der Isar – Technical University Munich, Munich, Germany
| | - Nathalie Pamir
- Department of Medicine, Knight Cardiovascular Institute, Oregon Health & Sciences University, Portland, Oregon, USA
| | - Tamer Sallam
- Department of Medicine, University of California Los Angeles, California, 90095, USA
- Molecular Biology Institute, University of California Los Angeles, California, 90095, USA
| | - Elizabeth J. Tarling
- Department of Medicine, University of California Los Angeles, California, 90095, USA
- Molecular Biology Institute, University of California Los Angeles, California, 90095, USA
- Johnsson Comprehensive Cancer Center, University of California Los Angeles, California, 90095, USA
| | - Thomas Q. de Aguiar Vallim
- Department of Biological Chemistry, University of California Los Angeles, California, 90095, USA
- Department of Medicine, University of California Los Angeles, California, 90095, USA
- Molecular Biology Institute, University of California Los Angeles, California, 90095, USA
- Johnsson Comprehensive Cancer Center, University of California Los Angeles, California, 90095, USA
| |
Collapse
|
12
|
Wang D, Yang Y, Lei Y, Tzvetkov NT, Liu X, Yeung AWK, Xu S, Atanasov AG. Targeting Foam Cell Formation in Atherosclerosis: Therapeutic Potential of Natural Products. Pharmacol Rev 2019; 71:596-670. [PMID: 31554644 DOI: 10.1124/pr.118.017178] [Citation(s) in RCA: 146] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Foam cell formation and further accumulation in the subendothelial space of the vascular wall is a hallmark of atherosclerotic lesions. Targeting foam cell formation in the atherosclerotic lesions can be a promising approach to treat and prevent atherosclerosis. The formation of foam cells is determined by the balanced effects of three major interrelated biologic processes, including lipid uptake, cholesterol esterification, and cholesterol efflux. Natural products are a promising source for new lead structures. Multiple natural products and pharmaceutical agents can inhibit foam cell formation and thus exhibit antiatherosclerotic capacity by suppressing lipid uptake, cholesterol esterification, and/or promoting cholesterol ester hydrolysis and cholesterol efflux. This review summarizes recent findings on these three biologic processes and natural products with demonstrated potential to target such processes. Discussed also are potential future directions for studying the mechanisms of foam cell formation and the development of foam cell-targeted therapeutic strategies.
Collapse
Affiliation(s)
- Dongdong Wang
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China (D.W., X.L.); Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzębiec, Poland (D.W., Y.Y., Y.L., A.G.A.); Department of Pharmacognosy, University of Vienna, Vienna, Austria (A.G.A.); Institute of Clinical Chemistry, University Hospital Zurich, Schlieren, Switzerland (D.W.); Institute of Molecular Biology "Roumen Tsanev," Department of Biochemical Pharmacology and Drug Design, Bulgarian Academy of Sciences, Sofia, Bulgaria (N.T.T.); Pharmaceutical Institute, University of Bonn, Bonn, Germany (N.T.T.); Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, Rochester, New York (S.X.); Oral and Maxillofacial Radiology, Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China (A.W.K.Y.); and Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia, Bulgaria (A.G.A.)
| | - Yang Yang
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China (D.W., X.L.); Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzębiec, Poland (D.W., Y.Y., Y.L., A.G.A.); Department of Pharmacognosy, University of Vienna, Vienna, Austria (A.G.A.); Institute of Clinical Chemistry, University Hospital Zurich, Schlieren, Switzerland (D.W.); Institute of Molecular Biology "Roumen Tsanev," Department of Biochemical Pharmacology and Drug Design, Bulgarian Academy of Sciences, Sofia, Bulgaria (N.T.T.); Pharmaceutical Institute, University of Bonn, Bonn, Germany (N.T.T.); Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, Rochester, New York (S.X.); Oral and Maxillofacial Radiology, Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China (A.W.K.Y.); and Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia, Bulgaria (A.G.A.)
| | - Yingnan Lei
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China (D.W., X.L.); Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzębiec, Poland (D.W., Y.Y., Y.L., A.G.A.); Department of Pharmacognosy, University of Vienna, Vienna, Austria (A.G.A.); Institute of Clinical Chemistry, University Hospital Zurich, Schlieren, Switzerland (D.W.); Institute of Molecular Biology "Roumen Tsanev," Department of Biochemical Pharmacology and Drug Design, Bulgarian Academy of Sciences, Sofia, Bulgaria (N.T.T.); Pharmaceutical Institute, University of Bonn, Bonn, Germany (N.T.T.); Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, Rochester, New York (S.X.); Oral and Maxillofacial Radiology, Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China (A.W.K.Y.); and Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia, Bulgaria (A.G.A.)
| | - Nikolay T Tzvetkov
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China (D.W., X.L.); Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzębiec, Poland (D.W., Y.Y., Y.L., A.G.A.); Department of Pharmacognosy, University of Vienna, Vienna, Austria (A.G.A.); Institute of Clinical Chemistry, University Hospital Zurich, Schlieren, Switzerland (D.W.); Institute of Molecular Biology "Roumen Tsanev," Department of Biochemical Pharmacology and Drug Design, Bulgarian Academy of Sciences, Sofia, Bulgaria (N.T.T.); Pharmaceutical Institute, University of Bonn, Bonn, Germany (N.T.T.); Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, Rochester, New York (S.X.); Oral and Maxillofacial Radiology, Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China (A.W.K.Y.); and Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia, Bulgaria (A.G.A.)
| | - Xingde Liu
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China (D.W., X.L.); Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzębiec, Poland (D.W., Y.Y., Y.L., A.G.A.); Department of Pharmacognosy, University of Vienna, Vienna, Austria (A.G.A.); Institute of Clinical Chemistry, University Hospital Zurich, Schlieren, Switzerland (D.W.); Institute of Molecular Biology "Roumen Tsanev," Department of Biochemical Pharmacology and Drug Design, Bulgarian Academy of Sciences, Sofia, Bulgaria (N.T.T.); Pharmaceutical Institute, University of Bonn, Bonn, Germany (N.T.T.); Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, Rochester, New York (S.X.); Oral and Maxillofacial Radiology, Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China (A.W.K.Y.); and Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia, Bulgaria (A.G.A.)
| | - Andy Wai Kan Yeung
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China (D.W., X.L.); Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzębiec, Poland (D.W., Y.Y., Y.L., A.G.A.); Department of Pharmacognosy, University of Vienna, Vienna, Austria (A.G.A.); Institute of Clinical Chemistry, University Hospital Zurich, Schlieren, Switzerland (D.W.); Institute of Molecular Biology "Roumen Tsanev," Department of Biochemical Pharmacology and Drug Design, Bulgarian Academy of Sciences, Sofia, Bulgaria (N.T.T.); Pharmaceutical Institute, University of Bonn, Bonn, Germany (N.T.T.); Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, Rochester, New York (S.X.); Oral and Maxillofacial Radiology, Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China (A.W.K.Y.); and Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia, Bulgaria (A.G.A.)
| | - Suowen Xu
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China (D.W., X.L.); Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzębiec, Poland (D.W., Y.Y., Y.L., A.G.A.); Department of Pharmacognosy, University of Vienna, Vienna, Austria (A.G.A.); Institute of Clinical Chemistry, University Hospital Zurich, Schlieren, Switzerland (D.W.); Institute of Molecular Biology "Roumen Tsanev," Department of Biochemical Pharmacology and Drug Design, Bulgarian Academy of Sciences, Sofia, Bulgaria (N.T.T.); Pharmaceutical Institute, University of Bonn, Bonn, Germany (N.T.T.); Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, Rochester, New York (S.X.); Oral and Maxillofacial Radiology, Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China (A.W.K.Y.); and Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia, Bulgaria (A.G.A.)
| | - Atanas G Atanasov
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China (D.W., X.L.); Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzębiec, Poland (D.W., Y.Y., Y.L., A.G.A.); Department of Pharmacognosy, University of Vienna, Vienna, Austria (A.G.A.); Institute of Clinical Chemistry, University Hospital Zurich, Schlieren, Switzerland (D.W.); Institute of Molecular Biology "Roumen Tsanev," Department of Biochemical Pharmacology and Drug Design, Bulgarian Academy of Sciences, Sofia, Bulgaria (N.T.T.); Pharmaceutical Institute, University of Bonn, Bonn, Germany (N.T.T.); Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, Rochester, New York (S.X.); Oral and Maxillofacial Radiology, Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China (A.W.K.Y.); and Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia, Bulgaria (A.G.A.)
| |
Collapse
|
13
|
Liang Z, Chen Y, Wang L, Li D, Yang X, Ma G, Wang Y, Li Y, Zhao H, Liang Y, Niu H. CYP27A1 inhibits bladder cancer cells proliferation by regulating cholesterol homeostasis. Cell Cycle 2018; 18:34-45. [PMID: 30563407 DOI: 10.1080/15384101.2018.1558868] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
Abstract
CYP27A1, an enzyme involved in regulating cellular cholesterol homeostasis, converts cholesterol into 27-hydroxycholesterol (27-HC). The relationship between CYP27A1 and cell proliferation was studied to determine the role of CYP27A1 in bladder cancer. The expression of CYP27A1 in three bladder cancer cell lines (T24, UM-UC-3 and 5637) were assessed by qRT-PCR and Western blotting, and cells with stable CYP27A1 expression were generated by lentiviral infection. Cell proliferation was detected by MTT assays, colony formation assays and a tumor xenograft model in vitro and in vivo, and the intracellular 27-HC and cholesterol secretion levels were detected by enzyme-linked immunosorbent assays (ELISA). The results revealed that CYP27A1 expression was downregulated in androgen receptor (AR)-positive T24/UM-UC-3 cells compared with AR-negative 5637 cell. After CYP27A1 expression was restored, cell proliferation was inhibited in vitro and in vivo because much more intracellular 27-HC was produced in the CYP27A1-overexpressing cells than in the control cells. Both T24 and UM-UC-3 cells treated with 27-HC showed similar results. In addition, CYP27A1/27HC could reduce the cellular cholesterol level in both T24 and UM-UC-3 cells by upregulating ATP-binding cassette transporters G1 and A1 (ABCG1 and ABCA1) through Liver X receptors (LXRs) pathway and downregulating low-density lipoprotein receptor (LDLR) expression. These findings all suggest that CYP27A1 is a critical cholesterol sensor in bladder cancer cells that may contribute significantly to bladder cancer proliferation.
Collapse
Affiliation(s)
- Zhijuan Liang
- a Key Laboratory, Department of Urology and Andrology , Affiliated Hospital of Qingdao University , Qingdao , China
| | - Yuanbin Chen
- a Key Laboratory, Department of Urology and Andrology , Affiliated Hospital of Qingdao University , Qingdao , China
| | - Liping Wang
- a Key Laboratory, Department of Urology and Andrology , Affiliated Hospital of Qingdao University , Qingdao , China
| | - Dan Li
- a Key Laboratory, Department of Urology and Andrology , Affiliated Hospital of Qingdao University , Qingdao , China
| | - Xuecheng Yang
- b Department of Urology , Affiliated Hospital of Qingdao University , Qingdao , China
| | - Guofeng Ma
- b Department of Urology , Affiliated Hospital of Qingdao University , Qingdao , China
| | - Yonghua Wang
- b Department of Urology , Affiliated Hospital of Qingdao University , Qingdao , China
| | - Yongxin Li
- c Department of Vascular Surgery , Affiliated Hospital of Qingdao University , Qingdao , China
| | - Han Zhao
- d Department of Pathology , Affiliated Hospital of Qingdao University , Qingdao , China
| | - Ye Liang
- a Key Laboratory, Department of Urology and Andrology , Affiliated Hospital of Qingdao University , Qingdao , China
| | - Haitao Niu
- a Key Laboratory, Department of Urology and Andrology , Affiliated Hospital of Qingdao University , Qingdao , China.,b Department of Urology , Affiliated Hospital of Qingdao University , Qingdao , China
| |
Collapse
|
14
|
Moon SY, Shin SA, Oh YS, Park HH, Lee CS. Understanding the Role of the BAI Subfamily of Adhesion G Protein-Coupled Receptors (GPCRs) in Pathological and Physiological Conditions. Genes (Basel) 2018; 9:genes9120597. [PMID: 30513696 PMCID: PMC6316137 DOI: 10.3390/genes9120597] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 11/24/2018] [Accepted: 11/28/2018] [Indexed: 02/06/2023] Open
Abstract
Brain-specific angiogenesis inhibitors (BAIs) 1, 2, and 3 are members of the adhesion G protein-coupled receptors, subfamily B, which share a conserved seven-transmembrane structure and an N-terminal extracellular domain. In cell- and animal-based studies, these receptors have been shown to play diverse roles under physiological and pathological conditions. BAI1 is an engulfment receptor and performs major functions in apoptotic-cell clearance and interacts (as a pattern recognition receptor) with pathogen components. BAI1 and -3 also participate in myoblast fusion. Furthermore, BAI1–3 have been linked to tumor progression and neurological diseases. In this review, we summarize the current understanding of the functions of BAI1–3 in pathological and physiological conditions and discuss future directions in terms of the importance of BAIs as pharmacological targets in diseases.
Collapse
Affiliation(s)
- Sun Young Moon
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Gyeongsang National University, Jinju 52828, Korea.
| | - Seong-Ah Shin
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Gyeongsang National University, Jinju 52828, Korea.
| | - Yong-Seok Oh
- Department of Brain-Cognitive Sciences, Daegu-Gyeongbuk Institute of Science and Technology (DGIST), Hyeonpung-myeon, Dalseong-gun, Daegu 42988, Korea.
| | - Hyun Ho Park
- College of Pharmacy, Chung-Ang University, Seoul 06974, Korea.
| | - Chang Sup Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Gyeongsang National University, Jinju 52828, Korea.
| |
Collapse
|
15
|
Harris MT, Hussain SS, Inouye CM, Castle AM, Castle JD. Reinterpretation of the localization of the ATP binding cassette transporter ABCG1 in insulin-secreting cells and insights regarding its trafficking and function. PLoS One 2018; 13:e0198383. [PMID: 30235209 PMCID: PMC6147399 DOI: 10.1371/journal.pone.0198383] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 09/04/2018] [Indexed: 01/08/2023] Open
Abstract
The ABC transporter ABCG1 contributes to the regulation of cholesterol efflux from cells and to the distribution of cholesterol within cells. We showed previously that ABCG1 deficiency inhibits insulin secretion by pancreatic beta cells and, based on its immunolocalization to insulin granules, proposed its essential role in forming granule membranes that are enriched in cholesterol. While we confirm elsewhere that ABCG1, alongside ABCA1 and oxysterol binding protein OSBP, supports insulin granule formation, the aim here is to clarify the localization of ABCG1 within insulin-secreting cells and to provide added insight regarding ABCG1's trafficking and sites of function. We show that stably expressed GFP-tagged ABCG1 closely mimics the distribution of endogenous ABCG1 in pancreatic INS1 cells and accumulates in the trans-Golgi network (TGN), endosomal recycling compartment (ERC) and on the cell surface but not on insulin granules, early or late endosomes. Notably, ABCG1 is short-lived, and proteasomal and lysosomal inhibitors both decrease its degradation. Following blockade of protein synthesis, GFP-tagged ABCG1 first disappears from the ER and TGN and later from the ERC and plasma membrane. In addition to aiding granule formation, our findings raise the prospect that ABCG1 may act beyond the TGN to regulate activities involving the endocytic pathway, especially as the amount of transferrin receptor is increased in ABCG1-deficient cells. Thus, ABCG1 may function at multiple intracellular sites and the plasma membrane as a roving sensor and modulator of cholesterol distribution, membrane trafficking and cholesterol efflux.
Collapse
Affiliation(s)
- Megan T. Harris
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, Virginia, United States of America
| | - Syed Saad Hussain
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, Virginia, United States of America
| | - Candice M. Inouye
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, Virginia, United States of America
| | - Anna M. Castle
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, Virginia, United States of America
| | - J. David Castle
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, Virginia, United States of America
| |
Collapse
|
16
|
Anastasius M, Luquain-Costaz C, Kockx M, Jessup W, Kritharides L. A critical appraisal of the measurement of serum 'cholesterol efflux capacity' and its use as surrogate marker of risk of cardiovascular disease. Biochim Biophys Acta Mol Cell Biol Lipids 2018; 1863:1257-1273. [PMID: 30305243 DOI: 10.1016/j.bbalip.2018.08.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 08/02/2018] [Accepted: 08/03/2018] [Indexed: 12/15/2022]
Abstract
The 'cholesterol efflux capacity (CEC)' assay is a simple in vitro measure of the capacities of individual sera to promote the first step of the reverse cholesterol transport pathway, the delivery of cellular cholesterol to plasma HDL. This review describes the cell biology of this model and critically assesses its application as a marker of cardiovascular risk. We describe the pathways for cell cholesterol export, current cell models used in the CEC assay with their limitations and consider the contribution that measurement of serum CEC provides to our understanding of HDL function in vivo.
Collapse
Affiliation(s)
- Malcolm Anastasius
- ANZAC Research Institute, Concord Repatriation General Hospital, University of Sydney, Sydney, NSW, Australia
| | | | - Maaike Kockx
- ANZAC Research Institute, Concord Repatriation General Hospital, University of Sydney, Sydney, NSW, Australia
| | - Wendy Jessup
- ANZAC Research Institute, Concord Repatriation General Hospital, University of Sydney, Sydney, NSW, Australia
| | - Leonard Kritharides
- ANZAC Research Institute, Concord Repatriation General Hospital, University of Sydney, Sydney, NSW, Australia; Cardiology Department, Concord Repatriation General Hospital, University of Sydney, Sydney, NSW, Australia.
| |
Collapse
|
17
|
Hussain SS, Harris MT, Kreutzberger AJB, Inouye CM, Doyle CA, Castle AM, Arvan P, Castle JD. Control of insulin granule formation and function by the ABC transporters ABCG1 and ABCA1 and by oxysterol binding protein OSBP. Mol Biol Cell 2018. [PMID: 29540530 PMCID: PMC5935073 DOI: 10.1091/mbc.e17-08-0519] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
In pancreatic β-cells, insulin granule membranes are enriched in cholesterol and are both recycled and newly generated. Cholesterol’s role in supporting granule membrane formation and function is poorly understood. ATP binding cassette transporters ABCG1 and ABCA1 regulate intracellular cholesterol and are important for insulin secretion. RNAi interference–induced depletion in cultured pancreatic β-cells shows that ABCG1 is needed to stabilize newly made insulin granules against lysosomal degradation; ABCA1 is also involved but to a lesser extent. Both transporters are also required for optimum glucose-stimulated insulin secretion, likely via complementary roles. Exogenous cholesterol addition rescues knockdown-induced granule loss (ABCG1) and reduced secretion (both transporters). Another cholesterol transport protein, oxysterol binding protein (OSBP), appears to act proximally as a source of endogenous cholesterol for granule formation. Its knockdown caused similar defective stability of young granules and glucose-stimulated insulin secretion, neither of which were rescued with exogenous cholesterol. Dual knockdowns of OSBP and ABC transporters support their serial function in supplying and concentrating cholesterol for granule formation. OSBP knockdown also decreased proinsulin synthesis consistent with a proximal endoplasmic reticulum defect. Thus, membrane cholesterol distribution contributes to insulin homeostasis at production, packaging, and export levels through the actions of OSBP and ABCs G1 and A1.
Collapse
Affiliation(s)
- Syed Saad Hussain
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, VA 22908
| | - Megan T Harris
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, VA 22908
| | - Alex J B Kreutzberger
- Department of Molecular Physiology and Biological Physics, University of Virginia School of Medicine, Charlottesville, VA 22908.,Center for Membrane and Cell Physiology, University of Virginia School of Medicine, Charlottesville, VA 22908
| | - Candice M Inouye
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, VA 22908
| | - Catherine A Doyle
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, VA 22908
| | - Anna M Castle
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, VA 22908
| | - Peter Arvan
- Division of Metabolism, Endocrinology, and Diabetes, University of Michigan Medical School, Ann Arbor, MI 48105
| | - J David Castle
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, VA 22908.,Center for Membrane and Cell Physiology, University of Virginia School of Medicine, Charlottesville, VA 22908
| |
Collapse
|
18
|
Oladipo OO, Ayo JO, Ambali SF, Mohammed B, Aluwong T. Dyslipdemia induced by chronic low dose co-exposure to lead, cadmium and manganese in rats: the role of oxidative stress. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2017; 53:199-205. [PMID: 28654832 DOI: 10.1016/j.etap.2017.06.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Revised: 06/01/2017] [Accepted: 06/20/2017] [Indexed: 06/07/2023]
Abstract
Lead (Pb), cadmium (Cd) and manganese (Mn) have many potential adverse health effects in vitro and in animal models of clinical toxicity. The current study investigated the dyslipidaemic and oxidative stress effects of chronic low-dose oral exposure to Pb, Cd and Mn and the combination (Pb+Cd+Mn) in rats for 15 weeks. Chronic exposure to the metals did not significantly (P>0.05) alter serum lipid profiles. However, the atherogenic index decreased by 32.2% in the Pb+Cd+Mn group, relative to the control. The triglyceride/high-density lipoprotein cholesterol ratio decreased by 39.4% in the Pb+Cd+Mn group, relative to the control, and elevated by 81.8, 94.8 and 20.8%, relative to the Pb, Cd and Mn groups, respectively. While the serum concentrations of malondialdehyde significantly increased in the Mn and Pb+Cd+Mn groups, that of glutathione peroxidase-1 decreased in the Pb+Cd+Mn group, and metallothionein-1 and zinc concentrations markedly decreased in all the metal treatment groups. The results suggest that long-term exposure of rats to Pb+Cd+Mn may result in hypolipidaemia, mediated via oxidative stress and metal interactions. Individuals who are constantly exposed to environmentally relevant levels of the metals may be at risk of hypolipidaemia.
Collapse
Affiliation(s)
- Olusola Olalekan Oladipo
- Biochemistry Division, National Veterinary Research Institute, P.M.B. 01, Vom, Nigeria; Department of Veterinary Pharmacology & Toxicology, Ahmadu Bello University, Zaria, Nigeria.
| | - Joseph Olusegun Ayo
- Department of Veterinary Physiology, Ahmadu Bello University, Zaria, Nigeria
| | | | - Bisalla Mohammed
- Department of Veterinary Pathology, Ahmadu Bello University, Zaria, Nigeria
| | - Tanang Aluwong
- Department of Veterinary Physiology, Ahmadu Bello University, Zaria, Nigeria
| |
Collapse
|
19
|
Barna BP, McPeek M, Malur A, Fessler MB, Wingard CJ, Dobbs L, Verbanac KM, Bowling M, Judson MA, Thomassen MJ. Elevated MicroRNA-33 in Sarcoidosis and a Carbon Nanotube Model of Chronic Granulomatous Disease. Am J Respir Cell Mol Biol 2017; 54:865-71. [PMID: 26641802 DOI: 10.1165/rcmb.2015-0332oc] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
We established a murine model of multiwall carbon nanotube (MWCNT)-induced chronic granulomatous disease, which resembles human sarcoidosis pathology. At 60 days after oropharyngeal MWCNT instillation, bronchoalveolar lavage (BAL) cells from wild-type mice exhibit an M1 phenotype with elevated proinflammatory cytokines and reduced peroxisome proliferator-activated receptor γ (PPARγ)-characteristics also present in human sarcoidosis. Based upon MWCNT-associated PPARγ deficiency, we hypothesized that the PPARγ target gene, ATP-binding cassette (ABC) G1, a lipid transporter with antiinflammatory properties, might also be repressed. Results after MWCNT instillation indicated significantly repressed ABCG1, but, surprisingly, lipid transporter ABCA1 was also repressed, suggesting a possible second pathway. Exploration of potential regulators revealed that microRNA (miR)-33, a lipid transporter regulator, was strikingly elevated (13.9 fold) in BAL cells from MWCNT-instilled mice but not sham control mice. Elevated miR-33 was also detected in murine granulomatous lung tissue. In vitro studies confirmed that lentivirus-miR-33 overexpression repressed both ABCA1 and ABCG1 (but not PPARγ) in cultured murine alveolar macrophages. BAL cells of patients with sarcoidosis also displayed elevated miR-33 together with reduced ABCA1 and ABCG1 messenger RNA and protein compared with healthy control subjects. Moreover, miR-33 was elevated within sarcoidosis granulomatous tissue. The findings suggest that alveolar macrophage miR-33 is up-regulated by proinflammatory cytokines and may perpetuate chronic inflammatory granulomatous disease by repressing antiinflammatory functions of ABCA1 and ABCG1 lipid transporters. The results also suggest two possible pathways for transporter dysregulation in granulomatous disease-one associated with intrinsic PPARγ status and the other with miR-33 up-regulation triggered by environmental challenges, such as MWCNT.
Collapse
Affiliation(s)
- Barbara P Barna
- 1 Division of Pulmonary, Critical Care, and Sleep Medicine, East Carolina University, Greenville, North Carolina
| | - Matthew McPeek
- 1 Division of Pulmonary, Critical Care, and Sleep Medicine, East Carolina University, Greenville, North Carolina
| | - Anagha Malur
- 1 Division of Pulmonary, Critical Care, and Sleep Medicine, East Carolina University, Greenville, North Carolina
| | - Michael B Fessler
- 2 Immunity, Inflammation, and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina
| | | | | | | | - Mark Bowling
- 1 Division of Pulmonary, Critical Care, and Sleep Medicine, East Carolina University, Greenville, North Carolina
| | - Marc A Judson
- 6 Division of Pulmonary and Critical Care, Medical College of Albany, Albany, New York
| | - Mary Jane Thomassen
- 1 Division of Pulmonary, Critical Care, and Sleep Medicine, East Carolina University, Greenville, North Carolina
| |
Collapse
|
20
|
de Aguiar Vallim TQ, Lee E, Merriott DJ, Goulbourne CN, Cheng J, Cheng A, Gonen A, Allen RM, Palladino END, Ford DA, Wang T, Baldán Á, Tarling EJ. ABCG1 regulates pulmonary surfactant metabolism in mice and men. J Lipid Res 2017; 58:941-954. [PMID: 28264879 DOI: 10.1194/jlr.m075101] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 03/03/2017] [Indexed: 12/27/2022] Open
Abstract
Idiopathic pulmonary alveolar proteinosis (PAP) is a rare lung disease characterized by accumulation of surfactant. Surfactant synthesis and secretion are restricted to epithelial type 2 (T2) pneumocytes (also called T2 cells). Clearance of surfactant is dependent upon T2 cells and macrophages. ABCG1 is highly expressed in both T2 cells and macrophages. ABCG1-deficient mice accumulate surfactant, lamellar body-loaded T2 cells, lipid-loaded macrophages, B-1 lymphocytes, and immunoglobulins, clearly demonstrating that ABCG1 has a critical role in pulmonary homeostasis. We identify a variant in the ABCG1 promoter in patients with PAP that results in impaired activation of ABCG1 by the liver X receptor α, suggesting that ABCG1 basal expression and/or induction in response to sterol/lipid loading is essential for normal lung function. We generated mice lacking ABCG1 specifically in either T2 cells or macrophages to determine the relative contribution of these cell types on surfactant lipid homeostasis. These results establish a critical role for T2 cell ABCG1 in controlling surfactant and overall lipid homeostasis in the lung and in the pathogenesis of human lung disease.
Collapse
Affiliation(s)
- Thomas Q de Aguiar Vallim
- Department of Medicine, University of California Los Angeles, Los Angeles, CA 90095.,Department of Biological Chemistry, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095.,Molecular Biology Institute, University of California Los Angeles, Los Angeles, CA 90095.,Johnson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA 90095
| | - Elinor Lee
- Department of Medicine, University of California Los Angeles, Los Angeles, CA 90095.,Division of Pulmonary and Critical Care Medicine, University of California Los Angeles, Los Angeles, CA 90095
| | - David J Merriott
- Department of Biological Chemistry, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095
| | | | - Joan Cheng
- Department of Biological Chemistry, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095
| | - Angela Cheng
- Department of Biological Chemistry, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095
| | - Ayelet Gonen
- Department of Medicine, University of California San Diego, La Jolla, CA 92093
| | - Ryan M Allen
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University, St. Louis, MO 63104
| | - Elisa N D Palladino
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University, St. Louis, MO 63104.,Center for Cardiovascular Research, School of Medicine, Saint Louis University, St. Louis, MO 63104
| | - David A Ford
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University, St. Louis, MO 63104.,Center for Cardiovascular Research, School of Medicine, Saint Louis University, St. Louis, MO 63104
| | - Tisha Wang
- Department of Medicine, University of California Los Angeles, Los Angeles, CA 90095.,Division of Pulmonary and Critical Care Medicine, University of California Los Angeles, Los Angeles, CA 90095
| | - Ángel Baldán
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University, St. Louis, MO 63104
| | - Elizabeth J Tarling
- Department of Medicine, University of California Los Angeles, Los Angeles, CA 90095 .,Molecular Biology Institute, University of California Los Angeles, Los Angeles, CA 90095.,Johnson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA 90095
| |
Collapse
|
21
|
El Asmar Z, Terrand J, Jenty M, Host L, Mlih M, Zerr A, Justiniano H, Matz RL, Boudier C, Scholler E, Garnier JM, Bertaccini D, Thiersé D, Schaeffer C, Van Dorsselaer A, Herz J, Bruban V, Boucher P. Convergent Signaling Pathways Controlled by LRP1 (Receptor-related Protein 1) Cytoplasmic and Extracellular Domains Limit Cellular Cholesterol Accumulation. J Biol Chem 2016; 291:5116-27. [PMID: 26792864 DOI: 10.1074/jbc.m116.714485] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Indexed: 11/06/2022] Open
Abstract
The low density lipoprotein receptor-related protein 1 (LRP1) is a ubiquitously expressed cell surface receptor that protects from intracellular cholesterol accumulation. However, the underlying mechanisms are unknown. Here we show that the extracellular (α) chain of LRP1 mediates TGFβ-induced enhancement of Wnt5a, which limits intracellular cholesterol accumulation by inhibiting cholesterol biosynthesis and by promoting cholesterol export. Moreover, we demonstrate that the cytoplasmic (β) chain of LRP1 suffices to limit cholesterol accumulation in LRP1(-/-) cells. Through binding of Erk2 to the second of its carboxyl-terminal NPXY motifs, LRP1 β-chain positively regulates the expression of ATP binding cassette transporter A1 (ABCA1) and of neutral cholesterol ester hydrolase (NCEH1). These results highlight the unexpected functions of LRP1 and the canonical Wnt5a pathway and new therapeutic potential in cholesterol-associated disorders including cardiovascular diseases.
Collapse
Affiliation(s)
- Zeina El Asmar
- From the CNRS, UMR 7213, University of Strasbourg, 67401 Illkirch, France
| | - Jérome Terrand
- From the CNRS, UMR 7213, University of Strasbourg, 67401 Illkirch, France
| | - Marion Jenty
- From the CNRS, UMR 7213, University of Strasbourg, 67401 Illkirch, France
| | - Lionel Host
- From the CNRS, UMR 7213, University of Strasbourg, 67401 Illkirch, France
| | - Mohamed Mlih
- From the CNRS, UMR 7213, University of Strasbourg, 67401 Illkirch, France
| | - Aurélie Zerr
- From the CNRS, UMR 7213, University of Strasbourg, 67401 Illkirch, France
| | - Hélène Justiniano
- From the CNRS, UMR 7213, University of Strasbourg, 67401 Illkirch, France
| | - Rachel L Matz
- From the CNRS, UMR 7213, University of Strasbourg, 67401 Illkirch, France
| | - Christian Boudier
- From the CNRS, UMR 7213, University of Strasbourg, 67401 Illkirch, France
| | - Estelle Scholler
- From the CNRS, UMR 7213, University of Strasbourg, 67401 Illkirch, France
| | - Jean-Marie Garnier
- IGBMC (Institut de Génétique et de Biologie Moléculaire et Cellulaire), INSERM 964/CNRS UMR 7104, University of Strasbourg, 67401 Illkirch, France
| | - Diego Bertaccini
- CNRS, UMR 7178, University of Strasbourg, 67087 Strasbourg, France, and
| | - Danièle Thiersé
- CNRS, UMR 7178, University of Strasbourg, 67087 Strasbourg, France, and
| | | | | | - Joachim Herz
- Department of Molecular Genetics and Center for Translational Neurodegeneration Research, UT Southwestern Medical Center, Dallas, Texas 75390
| | - Véronique Bruban
- From the CNRS, UMR 7213, University of Strasbourg, 67401 Illkirch, France,
| | - Philippe Boucher
- From the CNRS, UMR 7213, University of Strasbourg, 67401 Illkirch, France,
| |
Collapse
|
22
|
Lee SD, Tontonoz P. Liver X receptors at the intersection of lipid metabolism and atherogenesis. Atherosclerosis 2015; 242:29-36. [PMID: 26164157 PMCID: PMC4546914 DOI: 10.1016/j.atherosclerosis.2015.06.042] [Citation(s) in RCA: 109] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Revised: 06/19/2015] [Accepted: 06/22/2015] [Indexed: 12/14/2022]
Affiliation(s)
- Stephen D Lee
- Howard Hughes Medical Institute, Department of Pathology and Laboratory Medicine, University of California, Los Angeles, CA 90095, USA
| | - Peter Tontonoz
- Howard Hughes Medical Institute, Department of Pathology and Laboratory Medicine, University of California, Los Angeles, CA 90095, USA.
| |
Collapse
|
23
|
Gadella BM, Boerke A. An update on post-ejaculatory remodeling of the sperm surface before mammalian fertilization. Theriogenology 2015; 85:113-24. [PMID: 26320574 DOI: 10.1016/j.theriogenology.2015.07.018] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Revised: 07/07/2015] [Accepted: 07/12/2015] [Indexed: 11/17/2022]
Abstract
The fusion of a sperm with an oocyte to form new life is a highly regulated event. The activation-also termed capacitation-of the sperm cell is one of the key preparative steps required for this process. Ejaculated sperm has to make a journey through the female uterus and oviduct before it can approach the oocyte. The oocyte at that moment also has become prepared to facilitate monospermic fertilization and block immediately thereafter the chance for polyspermic fertilization. Interestingly, ejaculated sperm is not properly capacitated and consequently is not yet able to fertilize the oocyte. During the capacitation process, the formation of competent lipid-protein domains on the sperm head enables sperm-cumulus and zona pellucida interactions. This sperm binding allows the onset for a cascade reaction ultimately resulting in oocyte-sperm fusion. Many different lipids and proteins from the sperm surface are involved in this process. Sperm surface processing already starts when sperm are liberated from the seminiferous tubules and is followed by epididymal maturation where the sperm cell surface is modified and loaded with proteins to ensure it is prepared for its fertilization task. Although cauda epididymal sperm can fertilize the oocyte IVF, they are coated with so-called decapacitation factors during ejaculation. The seminal plasma-induced stabilization of the sperm surface permits the sperm transit through the cervix and uterus but prevents sperm capacitation and thus inhibits fertilization. For IVF purposes, sperm are washed out of seminal plasma and activated to get rid of decapacitation factors. Only after capacitation, the sperm can fertilize the oocyte. In recent years, IVF has become a widely used tool to achieve successful fertilization in both the veterinary field and human medicine. Although IVF procedures are very successful, scientific knowledge is still far from complete when identifying all the molecular players and processes during the first stages the fusion of two gametes into a new life. A concise overview in the current understanding of the process of capacitation and the sperm surface changes is provided. The gaps in knowledge of these prefertilization processes are critically discussed.
Collapse
Affiliation(s)
- B M Gadella
- Department of Farm Animal Health, Faculty of Veterinary Medicine, Utrecht University, The Netherlands; Department of Biochemistry & Cell Biology, Faculty of Veterinary Medicine, Utrecht University, The Netherlands.
| | - A Boerke
- Department of Farm Animal Health, Faculty of Veterinary Medicine, Utrecht University, The Netherlands; Department of Biochemistry & Cell Biology, Faculty of Veterinary Medicine, Utrecht University, The Netherlands
| |
Collapse
|
24
|
Gulati S, Balderes D, Kim C, Guo ZA, Wilcox L, Area-Gomez E, Snider J, Wolinski H, Stagljar I, Granato JT, Ruggles KV, DeGiorgis JA, Kohlwein SD, Schon EA, Sturley SL. ATP-binding cassette transporters and sterol O-acyltransferases interact at membrane microdomains to modulate sterol uptake and esterification. FASEB J 2015. [PMID: 26220175 DOI: 10.1096/fj.14-264796] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
A key component of eukaryotic lipid homeostasis is the esterification of sterols with fatty acids by sterol O-acyltransferases (SOATs). The esterification reactions are allosterically activated by their sterol substrates, the majority of which accumulate at the plasma membrane. We demonstrate that in yeast, sterol transport from the plasma membrane to the site of esterification is associated with the physical interaction of the major SOAT, acyl-coenzyme A:cholesterol acyltransferase (ACAT)-related enzyme (Are)2p, with 2 plasma membrane ATP-binding cassette (ABC) transporters: Aus1p and Pdr11p. Are2p, Aus1p, and Pdr11p, unlike the minor acyltransferase, Are1p, colocalize to sterol and sphingolipid-enriched, detergent-resistant microdomains (DRMs). Deletion of either ABC transporter results in Are2p relocalization to detergent-soluble membrane domains and a significant decrease (53-36%) in esterification of exogenous sterol. Similarly, in murine tissues, the SOAT1/Acat1 enzyme and activity localize to DRMs. This subcellular localization is diminished upon deletion of murine ABC transporters, such as Abcg1, which itself is DRM associated. We propose that the close proximity of sterol esterification and transport proteins to each other combined with their residence in lipid-enriched membrane microdomains facilitates rapid, high-capacity sterol transport and esterification, obviating any requirement for soluble intermediary proteins.
Collapse
Affiliation(s)
- Sonia Gulati
- *Institute of Human Nutrition, Department of Neurology, **Department of Genetics and Development, and Department of Pediatrics, Columbia University Medical Center, New York, New York, USA; Department of Biological Sciences and Department of Chemistry, Columbia University, New York, New York, USA; Donnelly Center for Cellular and Biomolecular Research, Toronto, Ontario, Canada; Institute of Molecular Biosciences, BioTechMed Graz, University of Graz, Graz, Austria; Department of Biology, Providence College, Providence, Rhode Island, USA; and Marine Biological Laboratory, Woods Hole, Massachusetts, USA
| | - Dina Balderes
- *Institute of Human Nutrition, Department of Neurology, **Department of Genetics and Development, and Department of Pediatrics, Columbia University Medical Center, New York, New York, USA; Department of Biological Sciences and Department of Chemistry, Columbia University, New York, New York, USA; Donnelly Center for Cellular and Biomolecular Research, Toronto, Ontario, Canada; Institute of Molecular Biosciences, BioTechMed Graz, University of Graz, Graz, Austria; Department of Biology, Providence College, Providence, Rhode Island, USA; and Marine Biological Laboratory, Woods Hole, Massachusetts, USA
| | - Christine Kim
- *Institute of Human Nutrition, Department of Neurology, **Department of Genetics and Development, and Department of Pediatrics, Columbia University Medical Center, New York, New York, USA; Department of Biological Sciences and Department of Chemistry, Columbia University, New York, New York, USA; Donnelly Center for Cellular and Biomolecular Research, Toronto, Ontario, Canada; Institute of Molecular Biosciences, BioTechMed Graz, University of Graz, Graz, Austria; Department of Biology, Providence College, Providence, Rhode Island, USA; and Marine Biological Laboratory, Woods Hole, Massachusetts, USA
| | - Zhongmin A Guo
- *Institute of Human Nutrition, Department of Neurology, **Department of Genetics and Development, and Department of Pediatrics, Columbia University Medical Center, New York, New York, USA; Department of Biological Sciences and Department of Chemistry, Columbia University, New York, New York, USA; Donnelly Center for Cellular and Biomolecular Research, Toronto, Ontario, Canada; Institute of Molecular Biosciences, BioTechMed Graz, University of Graz, Graz, Austria; Department of Biology, Providence College, Providence, Rhode Island, USA; and Marine Biological Laboratory, Woods Hole, Massachusetts, USA
| | - Lisa Wilcox
- *Institute of Human Nutrition, Department of Neurology, **Department of Genetics and Development, and Department of Pediatrics, Columbia University Medical Center, New York, New York, USA; Department of Biological Sciences and Department of Chemistry, Columbia University, New York, New York, USA; Donnelly Center for Cellular and Biomolecular Research, Toronto, Ontario, Canada; Institute of Molecular Biosciences, BioTechMed Graz, University of Graz, Graz, Austria; Department of Biology, Providence College, Providence, Rhode Island, USA; and Marine Biological Laboratory, Woods Hole, Massachusetts, USA
| | - Estela Area-Gomez
- *Institute of Human Nutrition, Department of Neurology, **Department of Genetics and Development, and Department of Pediatrics, Columbia University Medical Center, New York, New York, USA; Department of Biological Sciences and Department of Chemistry, Columbia University, New York, New York, USA; Donnelly Center for Cellular and Biomolecular Research, Toronto, Ontario, Canada; Institute of Molecular Biosciences, BioTechMed Graz, University of Graz, Graz, Austria; Department of Biology, Providence College, Providence, Rhode Island, USA; and Marine Biological Laboratory, Woods Hole, Massachusetts, USA
| | - Jamie Snider
- *Institute of Human Nutrition, Department of Neurology, **Department of Genetics and Development, and Department of Pediatrics, Columbia University Medical Center, New York, New York, USA; Department of Biological Sciences and Department of Chemistry, Columbia University, New York, New York, USA; Donnelly Center for Cellular and Biomolecular Research, Toronto, Ontario, Canada; Institute of Molecular Biosciences, BioTechMed Graz, University of Graz, Graz, Austria; Department of Biology, Providence College, Providence, Rhode Island, USA; and Marine Biological Laboratory, Woods Hole, Massachusetts, USA
| | - Heimo Wolinski
- *Institute of Human Nutrition, Department of Neurology, **Department of Genetics and Development, and Department of Pediatrics, Columbia University Medical Center, New York, New York, USA; Department of Biological Sciences and Department of Chemistry, Columbia University, New York, New York, USA; Donnelly Center for Cellular and Biomolecular Research, Toronto, Ontario, Canada; Institute of Molecular Biosciences, BioTechMed Graz, University of Graz, Graz, Austria; Department of Biology, Providence College, Providence, Rhode Island, USA; and Marine Biological Laboratory, Woods Hole, Massachusetts, USA
| | - Igor Stagljar
- *Institute of Human Nutrition, Department of Neurology, **Department of Genetics and Development, and Department of Pediatrics, Columbia University Medical Center, New York, New York, USA; Department of Biological Sciences and Department of Chemistry, Columbia University, New York, New York, USA; Donnelly Center for Cellular and Biomolecular Research, Toronto, Ontario, Canada; Institute of Molecular Biosciences, BioTechMed Graz, University of Graz, Graz, Austria; Department of Biology, Providence College, Providence, Rhode Island, USA; and Marine Biological Laboratory, Woods Hole, Massachusetts, USA
| | - Juliana T Granato
- *Institute of Human Nutrition, Department of Neurology, **Department of Genetics and Development, and Department of Pediatrics, Columbia University Medical Center, New York, New York, USA; Department of Biological Sciences and Department of Chemistry, Columbia University, New York, New York, USA; Donnelly Center for Cellular and Biomolecular Research, Toronto, Ontario, Canada; Institute of Molecular Biosciences, BioTechMed Graz, University of Graz, Graz, Austria; Department of Biology, Providence College, Providence, Rhode Island, USA; and Marine Biological Laboratory, Woods Hole, Massachusetts, USA
| | - Kelly V Ruggles
- *Institute of Human Nutrition, Department of Neurology, **Department of Genetics and Development, and Department of Pediatrics, Columbia University Medical Center, New York, New York, USA; Department of Biological Sciences and Department of Chemistry, Columbia University, New York, New York, USA; Donnelly Center for Cellular and Biomolecular Research, Toronto, Ontario, Canada; Institute of Molecular Biosciences, BioTechMed Graz, University of Graz, Graz, Austria; Department of Biology, Providence College, Providence, Rhode Island, USA; and Marine Biological Laboratory, Woods Hole, Massachusetts, USA
| | - Joseph A DeGiorgis
- *Institute of Human Nutrition, Department of Neurology, **Department of Genetics and Development, and Department of Pediatrics, Columbia University Medical Center, New York, New York, USA; Department of Biological Sciences and Department of Chemistry, Columbia University, New York, New York, USA; Donnelly Center for Cellular and Biomolecular Research, Toronto, Ontario, Canada; Institute of Molecular Biosciences, BioTechMed Graz, University of Graz, Graz, Austria; Department of Biology, Providence College, Providence, Rhode Island, USA; and Marine Biological Laboratory, Woods Hole, Massachusetts, USA
| | - Sepp D Kohlwein
- *Institute of Human Nutrition, Department of Neurology, **Department of Genetics and Development, and Department of Pediatrics, Columbia University Medical Center, New York, New York, USA; Department of Biological Sciences and Department of Chemistry, Columbia University, New York, New York, USA; Donnelly Center for Cellular and Biomolecular Research, Toronto, Ontario, Canada; Institute of Molecular Biosciences, BioTechMed Graz, University of Graz, Graz, Austria; Department of Biology, Providence College, Providence, Rhode Island, USA; and Marine Biological Laboratory, Woods Hole, Massachusetts, USA
| | - Eric A Schon
- *Institute of Human Nutrition, Department of Neurology, **Department of Genetics and Development, and Department of Pediatrics, Columbia University Medical Center, New York, New York, USA; Department of Biological Sciences and Department of Chemistry, Columbia University, New York, New York, USA; Donnelly Center for Cellular and Biomolecular Research, Toronto, Ontario, Canada; Institute of Molecular Biosciences, BioTechMed Graz, University of Graz, Graz, Austria; Department of Biology, Providence College, Providence, Rhode Island, USA; and Marine Biological Laboratory, Woods Hole, Massachusetts, USA
| | - Stephen L Sturley
- *Institute of Human Nutrition, Department of Neurology, **Department of Genetics and Development, and Department of Pediatrics, Columbia University Medical Center, New York, New York, USA; Department of Biological Sciences and Department of Chemistry, Columbia University, New York, New York, USA; Donnelly Center for Cellular and Biomolecular Research, Toronto, Ontario, Canada; Institute of Molecular Biosciences, BioTechMed Graz, University of Graz, Graz, Austria; Department of Biology, Providence College, Providence, Rhode Island, USA; and Marine Biological Laboratory, Woods Hole, Massachusetts, USA
| |
Collapse
|
25
|
Fond AM, Lee CS, Schulman IG, Kiss RS, Ravichandran KS. Apoptotic cells trigger a membrane-initiated pathway to increase ABCA1. J Clin Invest 2015; 125:2748-58. [PMID: 26075824 DOI: 10.1172/jci80300] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Accepted: 05/12/2015] [Indexed: 01/15/2023] Open
Abstract
Macrophages clear millions of apoptotic cells daily and, during this process, take up large quantities of cholesterol. The membrane transporter ABCA1 is a key player in cholesterol efflux from macrophages and has been shown via human genetic studies to provide protection against cardiovascular disease. How the apoptotic cell clearance process is linked to macrophage ABCA1 expression is not known. Here, we identified a plasma membrane-initiated signaling pathway that drives a rapid upregulation of ABCA1 mRNA and protein. This pathway involves the phagocytic receptor brain-specific angiogenesis inhibitor 1 (BAI1), which recognizes phosphatidylserine on apoptotic cells, and the intracellular signaling intermediates engulfment cell motility 1 (ELMO1) and Rac1, as ABCA1 induction was attenuated in primary macrophages from mice lacking these molecules. Moreover, this apoptotic cell-initiated pathway functioned independently of the liver X receptor (LXR) sterol-sensing machinery that is known to regulate ABCA1 expression and cholesterol efflux. When placed on a high-fat diet, mice lacking BAI1 had increased numbers of apoptotic cells in their aortic roots, which correlated with altered lipid profiles. In contrast, macrophages from engineered mice with transgenic BAI1 overexpression showed greater ABCA1 induction in response to apoptotic cells compared with those from control animals. Collectively, these data identify a membrane-initiated pathway that is triggered by apoptotic cells to enhance ABCA1 within engulfing phagocytes and with functional consequences in vivo.
Collapse
|
26
|
Favari E, Chroni A, Tietge UJF, Zanotti I, Escolà-Gil JC, Bernini F. Cholesterol efflux and reverse cholesterol transport. Handb Exp Pharmacol 2015; 224:181-206. [PMID: 25522988 DOI: 10.1007/978-3-319-09665-0_4] [Citation(s) in RCA: 103] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Both alterations of lipid/lipoprotein metabolism and inflammatory events contribute to the formation of the atherosclerotic plaque, characterized by the accumulation of abnormal amounts of cholesterol and macrophages in the artery wall. Reverse cholesterol transport (RCT) may counteract the pathogenic events leading to the formation and development of atheroma, by promoting the high-density lipoprotein (HDL)-mediated removal of cholesterol from the artery wall. Recent in vivo studies established the inverse relationship between RCT efficiency and atherosclerotic cardiovascular diseases (CVD), thus suggesting that the promotion of this process may represent a novel strategy to reduce atherosclerotic plaque burden and subsequent cardiovascular events. HDL plays a primary role in all stages of RCT: (1) cholesterol efflux, where these lipoproteins remove excess cholesterol from cells; (2) lipoprotein remodeling, where HDL undergo structural modifications with possible impact on their function; and (3) hepatic lipid uptake, where HDL releases cholesterol to the liver, for the final excretion into bile and feces. Although the inverse association between HDL plasma levels and CVD risk has been postulated for years, recently this concept has been challenged by studies reporting that HDL antiatherogenic functions may be independent of their plasma levels. Therefore, assessment of HDL function, evaluated as the capacity to promote cell cholesterol efflux may offer a better prediction of CVD than HDL levels alone. Consistent with this idea, it has been recently demonstrated that the evaluation of serum cholesterol efflux capacity (CEC) is a predictor of atherosclerosis extent in humans.
Collapse
Affiliation(s)
- Elda Favari
- Department of Pharmacy, University of Parma, Parco Area delle Scienze 27/A, 43124, Parma, Italy
| | | | | | | | | | | |
Collapse
|
27
|
Pfeiffer L, Wahl S, Pilling LC, Reischl E, Sandling JK, Kunze S, Holdt LM, Kretschmer A, Schramm K, Adamski J, Klopp N, Illig T, Hedman ÅK, Roden M, Hernandez DG, Singleton AB, Thasler WE, Grallert H, Gieger C, Herder C, Teupser D, Meisinger C, Spector TD, Kronenberg F, Prokisch H, Melzer D, Peters A, Deloukas P, Ferrucci L, Waldenberger M. DNA methylation of lipid-related genes affects blood lipid levels. ACTA ACUST UNITED AC 2015; 8:334-42. [PMID: 25583993 DOI: 10.1161/circgenetics.114.000804] [Citation(s) in RCA: 140] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2014] [Accepted: 12/16/2014] [Indexed: 01/03/2023]
Abstract
BACKGROUND Epigenetic mechanisms might be involved in the regulation of interindividual lipid level variability and thus may contribute to the cardiovascular risk profile. The aim of this study was to investigate the association between genome-wide DNA methylation and blood lipid levels high-density lipoprotein cholesterol, low-density lipoprotein cholesterol, triglycerides, and total cholesterol. Observed DNA methylation changes were also further analyzed to examine their relationship with previous hospitalized myocardial infarction. METHODS AND RESULTS Genome-wide DNA methylation patterns were determined in whole blood samples of 1776 subjects of the Cooperative Health Research in the Region of Augsburg F4 cohort using the Infinium HumanMethylation450 BeadChip (Illumina). Ten novel lipid-related CpG sites annotated to various genes including ABCG1, MIR33B/SREBF1, and TNIP1 were identified. CpG cg06500161, located in ABCG1, was associated in opposite directions with both high-density lipoprotein cholesterol (β coefficient=-0.049; P=8.26E-17) and triglyceride levels (β=0.070; P=1.21E-27). Eight associations were confirmed by replication in the Cooperative Health Research in the Region of Augsburg F3 study (n=499) and in the Invecchiare in Chianti, Aging in the Chianti Area study (n=472). Associations between triglyceride levels and SREBF1 and ABCG1 were also found in adipose tissue of the Multiple Tissue Human Expression Resource cohort (n=634). Expression analysis revealed an association between ABCG1 methylation and lipid levels that might be partly mediated by ABCG1 expression. DNA methylation of ABCG1 might also play a role in previous hospitalized myocardial infarction (odds ratio, 1.15; 95% confidence interval=1.06-1.25). CONCLUSIONS Epigenetic modifications of the newly identified loci might regulate disturbed blood lipid levels and thus contribute to the development of complex lipid-related diseases.
Collapse
|
28
|
Kardassis D, Gafencu A, Zannis VI, Davalos A. Regulation of HDL genes: transcriptional, posttranscriptional, and posttranslational. Handb Exp Pharmacol 2015; 224:113-179. [PMID: 25522987 DOI: 10.1007/978-3-319-09665-0_3] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
HDL regulation is exerted at multiple levels including regulation at the level of transcription initiation by transcription factors and signal transduction cascades; regulation at the posttranscriptional level by microRNAs and other noncoding RNAs which bind to the coding or noncoding regions of HDL genes regulating mRNA stability and translation; as well as regulation at the posttranslational level by protein modifications, intracellular trafficking, and degradation. The above mechanisms have drastic effects on several HDL-mediated processes including HDL biogenesis, remodeling, cholesterol efflux and uptake, as well as atheroprotective functions on the cells of the arterial wall. The emphasis is on mechanisms that operate in physiologically relevant tissues such as the liver (which accounts for 80% of the total HDL-C levels in the plasma), the macrophages, the adrenals, and the endothelium. Transcription factors that have a significant impact on HDL regulation such as hormone nuclear receptors and hepatocyte nuclear factors are extensively discussed both in terms of gene promoter recognition and regulation but also in terms of their impact on plasma HDL levels as was revealed by knockout studies. Understanding the different modes of regulation of this complex lipoprotein may provide useful insights for the development of novel HDL-raising therapies that could be used to fight against atherosclerosis which is the underlying cause of coronary heart disease.
Collapse
Affiliation(s)
- Dimitris Kardassis
- Department of Biochemistry, University of Crete Medical School and Institute of Molecular Biology and Biotechnology, Foundation of Research and Technology of Hellas, Heraklion, Crete, 71110, Greece,
| | | | | | | |
Collapse
|
29
|
Baldan A, Gonen A, Choung C, Que X, Marquart TJ, Hernandez I, Bjorkhem I, Ford DA, Witztum JL, Tarling EJ. ABCG1 is required for pulmonary B-1 B cell and natural antibody homeostasis. THE JOURNAL OF IMMUNOLOGY 2014; 193:5637-48. [PMID: 25339664 DOI: 10.4049/jimmunol.1400606] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Many metabolic diseases, including atherosclerosis, type 2 diabetes, pulmonary alveolar proteinosis, and obesity, have a chronic inflammatory component involving both innate and adaptive immunity. Mice lacking the ATP-binding cassette transporter G1 (ABCG1) develop chronic inflammation in the lungs, which is associated with the lipid accumulation (cholesterol, cholesterol ester, and phospholipid) and cholesterol crystal deposition that are characteristic of atherosclerotic lesions and pulmonary alveolar proteinosis. In this article, we demonstrate that specific lipids, likely oxidized phospholipids and/or sterols, elicit a lung-specific immune response in Abcg1(-/-) mice. Loss of ABCG1 results in increased levels of specific oxysterols, phosphatidylcholines, and oxidized phospholipids, including 1-palmitoyl-2-(5'-oxovaleroyl)-sn-glycero-3-phosphocholine, in the lungs. Further, we identify a niche-specific increase in natural Ab (NAb)-secreting B-1 B cells in response to this lipid accumulation that is paralleled by increased titers of IgM, IgA, and IgG against oxidation-specific epitopes, such as those on oxidized low-density lipoprotein and malondialdehyde-modified low-density lipoprotein. Finally, we identify a cytokine/chemokine signature that is reflective of increased B cell activation, Ab secretion, and homing. Collectively, these data demonstrate that the accumulation of lipids in Abcg1(-/-) mice induces the specific expansion and localization of B-1 B cells, which secrete NAbs that may help to protect against the development of atherosclerosis. Indeed, despite chronic lipid accumulation and inflammation, hyperlipidemic mice lacking ABCG1 develop smaller atherosclerotic lesions compared with controls. These data also suggest that Abcg1(-/-) mice may represent a new model in which to study the protective functions of B-1 B cells/NAbs and suggest novel targets for pharmacologic intervention and treatment of disease.
Collapse
Affiliation(s)
- Angel Baldan
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095; Edward A. Doisy Department of Biochemistry and Molecular Biology, St. Louis University, St. Louis, MO 63104
| | - Ayelet Gonen
- Department of Medicine, University of California San Diego, La Jolla, CA 92093
| | - Christina Choung
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095
| | - Xuchu Que
- Department of Medicine, University of California San Diego, La Jolla, CA 92093
| | - Tyler J Marquart
- Edward A. Doisy Department of Biochemistry and Molecular Biology, St. Louis University, St. Louis, MO 63104
| | - Irene Hernandez
- Instituto de Investigaciones Biomédicas "Alberto Sols" Consejo Superior de Investigaciones Cientificas - Universidad Autonoma de Madrid, Madrid 28006; Unidad Asociada de Biomedicina IIBM-Universidad de Las Palmas de Gran Canaria, Las Palmas 35016, Spain; and
| | | | - David A Ford
- Edward A. Doisy Department of Biochemistry and Molecular Biology, St. Louis University, St. Louis, MO 63104
| | - Joseph L Witztum
- Department of Medicine, University of California San Diego, La Jolla, CA 92093
| | - Elizabeth J Tarling
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095;
| |
Collapse
|
30
|
Engel T, Fobker M, Buchmann J, Kannenberg F, Rust S, Nofer JR, Schürmann A, Seedorf U. 3β,5α,6β-Cholestanetriol and 25-hydroxycholesterol accumulate in ATP-binding cassette transporter G1 (ABCG1)-deficiency. Atherosclerosis 2014; 235:122-9. [PMID: 24833118 DOI: 10.1016/j.atherosclerosis.2014.04.023] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Revised: 04/08/2014] [Accepted: 04/17/2014] [Indexed: 10/25/2022]
Abstract
OBJECTIVE Oxysterols are oxidized derivatives of sterols that have cytotoxic effects and are potent regulators of diverse cellular functions. Efficient oxysterol removal by the sub-family G member 1 of the ATP-binding cassette transporters (ABCG1) is essential for cell survival and control of cellular processes. However, the specific role of ABCG1 in the transport of various oxysterol species has been not systematically investigated to date. Here, we examined the involvement of ABCG1 in the oxysterol metabolism by studying oxysterol tissue levels in a mouse model of Abcg1-deficiency. METHODS AND RESULTS Analysis of lung tissue of Abcg1(-/-) mice on a standard diet revealed that 3β,5α,6β-cholestanetriol (CT) and 25-hydroxycholesterol (HC) accumulated at more than 100-fold higher levels in comparison to wild-type mice. 24S-HC and 27-HC levels were also elevated, but were minor constituents. A radiolabeled assay employing regulable ABCG1-expressing HeLa cell lines revealed that 25-HC export to albumin was dependent on functional ABCG1 expression and could be blocked by an excess of unlabeled 25-HC or 27-HC. In this cell line, 25-HC at low doses triggered mitochondrial membrane potential, and reactive oxygen species production, which are both indirect indicators of cellular energy expenditure. CONCLUSION Our results suggest that 25-HC and CT are physiologic substrates for ABCG1. Excessive accumulation of these oxysterols may explain the increased rate of cell death and the inflammatory phenotype observed in Abcg1-deficient animals and cells.
Collapse
Affiliation(s)
- Thomas Engel
- Leibniz-Institute for Arteriosclerosis Research at The Westphalian Wilhelms-University, 48149 Muenster, Germany.
| | - Manfred Fobker
- Center for Laboratory Medicine, University Hospital Muenster, 48149 Muenster, Germany
| | - Jana Buchmann
- German Institute of Human Nutrition, Department of Experimental Diabetology, 14558 Potsdam-Rehbruecke, Germany
| | - Frank Kannenberg
- Center for Laboratory Medicine, University Hospital Muenster, 48149 Muenster, Germany
| | - Stephan Rust
- Leibniz-Institute for Arteriosclerosis Research at The Westphalian Wilhelms-University, 48149 Muenster, Germany
| | - Jerzy-Roch Nofer
- Center for Laboratory Medicine, University Hospital Muenster, 48149 Muenster, Germany
| | - Annette Schürmann
- German Institute of Human Nutrition, Department of Experimental Diabetology, 14558 Potsdam-Rehbruecke, Germany
| | - Udo Seedorf
- Leibniz-Institute for Arteriosclerosis Research at The Westphalian Wilhelms-University, 48149 Muenster, Germany
| |
Collapse
|
31
|
Zakrzewicz A. LDL is out again - through the crosstalk of two atheroprotective mechanisms: ABCA1 of macrophages and NO˙ from endothelial cells. Acta Physiol (Oxf) 2014; 210:708-9. [PMID: 24629160 DOI: 10.1111/apha.12252] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Affiliation(s)
- A. Zakrzewicz
- Charité-Universitätsmedizin Berlin; Institut für Physiologie; Berlin Germany
| |
Collapse
|
32
|
Validation of homogeneous assays for HDL-cholesterol using fresh samples from healthy and diseased subjects. Atherosclerosis 2014; 233:253-9. [PMID: 24529153 DOI: 10.1016/j.atherosclerosis.2013.12.033] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Revised: 12/09/2013] [Accepted: 12/13/2013] [Indexed: 11/23/2022]
Abstract
BACKGROUND High-density lipoprotein-cholesterol (HDL-C) is a negative risk factor for cardiovascular events. Although several homogeneous HDL-C assays are available, their accuracy has not been validated, particularly in subjects with disease. We aimed to clarify whether HDL-C concentrations measured by homogeneous assays [HDL-C (H)] agree with those determined by the reference measurement procedures [HDL-C (RMP)] using ultracentrifugation and precipitation with heparin-manganese reagent in fresh clinical samples. METHODS HDL-C concentrations in samples from 48 healthy subjects and 119 subjects with disease were determined using 12 homogeneous assays and RMPs. RESULTS All reagents showed excellent intra- and inter-assay CVs (<2.23%) for two pooled sera. Furthermore, the mean bias was within ± 1.0% in nine reagents using samples from healthy subjects and in eight reagents using samples from subjects with disease. In a single HDL-C (H) determination, the total error requirement of the National Cholesterol Education Program (95% of results < 13%) was fulfilled in nine reagents using samples from healthy subjects and six reagents in those from subjects with disease. Error component analysis revealed that only one reagent exceeded ± 10% total error in samples from healthy subjects, whereas four reagents exceeded this error in samples from subjects with disease. Correlations between HDL-C (H) and HDL-C (RMP) revealed that the slopes were within 1.00 ± 0.06 in six reagents in healthy subjects, and eight reagents in subjects with disease. CONCLUSIONS Except for three reagents, HDL-C (H) agrees well with HDL-C (RMP) in subjects with common disease, but not in those with extremely low HDL-C or abnormal HDL composition.
Collapse
|
33
|
Mo ZC, Xiao J, Tang SL, Ouyang XP, He PP, Lv YC, Long ZF, Yao F, Tan YL, Xie W, Zhang M, Liu D, Tian GP, Tang DP, Zheng XL, Zhao GJ, Tang CK. Advanced Oxidation Protein Products Exacerbates Lipid Accumulation and Atherosclerosis Through Downregulation of ATP-Binding Cassette Transporter A1 and G1 Expression in Apolipoprotein E Knockout Mice. Circ J 2014; 78:2760-70. [DOI: 10.1253/circj.cj-14-0193] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Zhong-Cheng Mo
- Institute of Cardiovascular Research, Key Laboratory for Atherosclerology of Hunan Province, Life Science Research Center, University of South China
- Department of Histology and Embryology, University of South China
| | - Ji Xiao
- Department of Anesthesiology, the Second Affiliated Hospital of University of South China
| | - Shi-Lin Tang
- Department of Intensive Care Unit, the First Affiliated Hospital of University of South China
| | - Xin-Ping Ouyang
- Institute of Cardiovascular Research, Key Laboratory for Atherosclerology of Hunan Province, Life Science Research Center, University of South China
| | - Ping-ping He
- Institute of Cardiovascular Research, Key Laboratory for Atherosclerology of Hunan Province, Life Science Research Center, University of South China
| | - Yun-cheng Lv
- Institute of Cardiovascular Research, Key Laboratory for Atherosclerology of Hunan Province, Life Science Research Center, University of South China
| | - Zhi-feng Long
- Department of Histology and Embryology, University of South China
| | - Feng Yao
- Institute of Cardiovascular Research, Key Laboratory for Atherosclerology of Hunan Province, Life Science Research Center, University of South China
| | - Yu-lin Tan
- Institute of Cardiovascular Research, Key Laboratory for Atherosclerology of Hunan Province, Life Science Research Center, University of South China
| | - Wei Xie
- Institute of Cardiovascular Research, Key Laboratory for Atherosclerology of Hunan Province, Life Science Research Center, University of South China
| | - Min Zhang
- Institute of Cardiovascular Research, Key Laboratory for Atherosclerology of Hunan Province, Life Science Research Center, University of South China
| | - Dan Liu
- Institute of Cardiovascular Research, Key Laboratory for Atherosclerology of Hunan Province, Life Science Research Center, University of South China
| | - Guo-Ping Tian
- Department of Cardiovascular Medicine, Second Affiliated Hospital of University of South China
| | - Deng-Pei Tang
- Department of Biochemistry, University of Saskatchewan
| | - Xi-Long Zheng
- Department of Biochemistry and Molecular Biology, The Libin Cardiovascular Institute of Alberta, The University of Calgary, Health Sciences Center
| | - Guo-jun Zhao
- Institute of Cardiovascular Research, Key Laboratory for Atherosclerology of Hunan Province, Life Science Research Center, University of South China
| | - Chao-ke Tang
- Institute of Cardiovascular Research, Key Laboratory for Atherosclerology of Hunan Province, Life Science Research Center, University of South China
| |
Collapse
|
34
|
Freeman SR, Jin X, Anzinger JJ, Xu Q, Purushothaman S, Fessler MB, Addadi L, Kruth HS. ABCG1-mediated generation of extracellular cholesterol microdomains. J Lipid Res 2013; 55:115-27. [PMID: 24212237 DOI: 10.1194/jlr.m044552] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Previous studies have demonstrated that the ATP-binding cassette transporters (ABC)A1 and ABCG1 function in many aspects of cholesterol efflux from macrophages. In this current study, we continued our investigation of extracellular cholesterol microdomains that form during enrichment of macrophages with cholesterol. Human monocyte-derived macrophages and mouse bone marrow-derived macrophages, differentiated with macrophage colony-stimulating factor (M-CSF) or granulocyte macrophage colony-stimulation factor (GM-CSF), were incubated with acetylated LDL (AcLDL) to allow for cholesterol enrichment and processing. We utilized an anti-cholesterol microdomain monoclonal antibody to reveal pools of unesterified cholesterol, which were found both in the extracellular matrix and associated with the cell surface, that we show function in reverse cholesterol transport. Coincubation of AcLDL with 50 μg/ml apoA-I eliminated all extracellular and cell surface-associated cholesterol microdomains, while coincubation with the same concentration of HDL only removed extracellular matrix-associated cholesterol microdomains. Only at an HDL concentration of 200 µg/ml did HDL eliminate the cholesterol microdomains that were cell-surface associated. The deposition of cholesterol microdomains was inhibited by probucol, but it was increased by the liver X receptor (LXR) agonist TO901317, which upregulates ABCA1 and ABCG1. Extracellular cholesterol microdomains did not develop when ABCG1-deficient mouse bone marrow-derived macrophages were enriched with cholesterol. Our findings show that generation of extracellular cholesterol microdomains is mediated by ABCG1 and that reverse cholesterol transport occurs not only at the cell surface but also within the extracellular space.
Collapse
Affiliation(s)
- Sebastian R Freeman
- Section of Experimental Atherosclerosis, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Cerda A, Issa MH, Genvigir FDV, Rohde CB, Cavalli SA, Bertolami MC, Faludi AA, Hirata MH, Hirata RDC. Atorvastatin and hormone therapy influence expression of ABCA1, APOA1 and SCARB1 in mononuclear cells from hypercholesterolemic postmenopausal women. J Steroid Biochem Mol Biol 2013; 138:403-9. [PMID: 24007717 DOI: 10.1016/j.jsbmb.2013.08.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2013] [Revised: 08/22/2013] [Accepted: 08/26/2013] [Indexed: 11/15/2022]
Abstract
BACKGROUND Reverse cholesterol transport (RCT) has been inversely related to atherosclerosis and cardiovascular risk. The influence of menopause in the RCT process is poorly understood and the effects of cholesterol-lowering interventions, including statins and hormone therapy (HT), on genes controlling the RCT in postmenopausal women are also unknown. METHODS The effects on serum lipids and expression profile of genes involved in RCT - APOA1, ABCA1, ABCG1, SCARB1 and LXRA - were evaluated by TaqMan(®) quantitative PCR in peripheral blood mononuclear cells (PBMC) from 87 postmenopausal hypercholesterolemic women treated with atorvastatin (AT, n=17), estrogen or estrogen plus progestin (HT, n=34) and estrogen or estrogen plus progestin associated with atorvastatin (HT+AT, n=36). RESULTS Atorvastatin and HT treatments reduced the mRNA levels of APOA1 and SCARB1, respectively, whereas ABCA1 expression was reduced after all treatments. Although the expression of LXRA, an important transcription factor controlling the expression of genes involved in RCT, was not modified after any treatment, it was correlated with ABCA1, APOA1 and SCARB1 RNAm values before and after treatments, however no correlation with ABCG1 was observed. In a linear regression analysis, HT was related to an increase in apoAI levels after treatment when compared to atorvastatin and, moreover, higher SCARB1 and ABCA1 basal expression were also associated with decreased apoAI levels after treatments. CONCLUSION ABCA1 mRNA levels are decreased by atorvastatin and HT, however these treatments have a differential effect on APOA1 and SCARB1 expression in PBMC from postmenopausal women. Basal ABCA1 and SCARB1 expression profile could be helpful markers in predicting the effect of atorvastatin and HT on RCT, according to the changes in apoAI levels in this sample population.
Collapse
Affiliation(s)
- Alvaro Cerda
- School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo, Brazil.
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Tarling EJ, de Aguiar Vallim TQ, Edwards PA. Role of ABC transporters in lipid transport and human disease. Trends Endocrinol Metab 2013; 24:342-50. [PMID: 23415156 PMCID: PMC3659191 DOI: 10.1016/j.tem.2013.01.006] [Citation(s) in RCA: 214] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2012] [Revised: 01/16/2013] [Accepted: 01/18/2013] [Indexed: 12/28/2022]
Abstract
Almost half of the 48 human ATP-binding cassette (ABC) transporter proteins are thought to facilitate the ATP-dependent translocation of lipids or lipid-related compounds. Such substrates include cholesterol, plant sterols, bile acids, phospholipids, and sphingolipids. Mutations in a substantial number of the 48 human ABC transporters have been linked to human disease. Indeed the finding that 12 diseases have been associated with abnormal lipid transport and/or homeostasis demonstrates the importance of this family of transporters in cell physiology. This review highlights the role of ABC transporters in lipid transport and movement, in addition to discussing their roles in cellular homeostasis and inherited disorders.
Collapse
Affiliation(s)
- Elizabeth J Tarling
- Department of Biological Chemistry, David Geffen School of Medicine at the University of California Los Angeles (UCLA), Los Angeles, CA 90095, USA.
| | | | | |
Collapse
|
37
|
Luquain-Costaz C, Lefai E, Arnal-Levron M, Markina D, Sakaï S, Euthine V, Makino A, Guichardant M, Yamashita S, Kobayashi T, Lagarde M, Moulin P, Delton-Vandenbroucke I. Bis(monoacylglycero)phosphate accumulation in macrophages induces intracellular cholesterol redistribution, attenuates liver-X receptor/ATP-Binding cassette transporter A1/ATP-binding cassette transporter G1 pathway, and impairs cholesterol efflux. Arterioscler Thromb Vasc Biol 2013; 33:1803-11. [PMID: 23788762 DOI: 10.1161/atvbaha.113.301857] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Endosomal signature phospholipid bis(monoacylglycero)phosphate (BMP) has been involved in the regulation of cellular cholesterol homeostasis. Accumulation of BMP is a hallmark of lipid storage disorders and was recently reported as a noticeable feature of oxidized low-density lipoprotein-laden macrophages. This study was designed to delineate the consequences of macrophage BMP accumulation on intracellular cholesterol distribution, metabolism, and efflux and to unravel the underlying molecular mechanisms. APPROACH AND RESULTS We have developed an experimental design to specifically increase BMP content in RAW 264.7 macrophages. After BMP accumulation, cell cholesterol distribution was markedly altered, despite no change in low-density lipoprotein uptake and hydrolysis, cholesterol esterification, or total cell cholesterol content. The expression of cholesterol-regulated genes sterol regulatory element-binding protein 2 and hydroxymethylglutaryl-coenzyme A reductase was decreased by 40%, indicative of an increase of endoplasmic reticulum-associated cholesterol. Cholesterol delivery to plasma membrane was reduced as evidenced by the 20% decrease of efflux by cyclodextrin. Functionally, BMP accumulation reduced cholesterol efflux to both apolipoprotein A1 and high-density lipoprotein by 40% and correlated with a 40% decrease in mRNA contents of ATP-binding cassette transporter A1, ATP-binding cassette transporter G1, and liver-X receptor α and β. Foam cell formation induced by oxidized low-density lipoprotein exposure was exacerbated in BMP-enriched cells. CONCLUSIONS The present work shows for the first time a strong functional link between BMP and cholesterol-regulating genes involved in both intracellular metabolism and efflux. We propose that accumulation of cellular BMP might contribute to the deregulation of cholesterol homeostasis in atheromatous macrophages.
Collapse
Affiliation(s)
- Céline Luquain-Costaz
- Université de Lyon, UMR 1060 Inserm, CarMeN, Institut National des Sciences Appliquées-Lyon, Villeurbanne, France
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Campos-Salinas J, León-Guerrero D, González-Rey E, Delgado M, Castanys S, Pérez-Victoria JM, Gamarro F. LABCG2, a new ABC transporter implicated in phosphatidylserine exposure, is involved in the infectivity and pathogenicity of Leishmania. PLoS Negl Trop Dis 2013; 7:e2179. [PMID: 23638200 PMCID: PMC3636091 DOI: 10.1371/journal.pntd.0002179] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2012] [Accepted: 03/15/2013] [Indexed: 12/18/2022] Open
Abstract
Leishmaniasis is a neglected disease produced by the intracellular protozoan parasite Leishmania. In the present study, we show that LABCG2, a new ATP-binding cassette half-transporter (ABCG subfamily) from Leishmania, is involved in parasite virulence. Down-regulation of LABCG2 function upon expression of an inactive mutant version of this half-transporter (LABCG2(K/M)) is shown to reduce the translocation of short-chain analogues of phosphatidylserine (PS). This dominant-negative phenotype is specific for the headgroup of the phospholipid, as the movement of phospholipid analogues of phosphatidylcholine, phosphatidylethanolamine or sphingomyelin is not affected. In addition, promastigotes expressing LABCG2(K/M) expose less endogenous PS in the stationary phase than control parasites. Transient exposure of PS at the outer leaflet of the plasma membrane is known to be one of the mechanisms used by Leishmania to infect macrophages and to silence their immune response. Stationary phase/metacyclic promastigotes expressing LABCG2(K/M) are less infective for macrophages and show decreased pathogenesis in a mouse model of cutaneous leishmaniasis. Thus, mice infected with parasites expressing LABCG2(K/M) did not develop any lesion and showed significantly lower inflammation and parasite burden than mice infected with control parasites. Our results indicate that LABCG2 function is required for the externalization of PS in Leishmania promastigotes, a process that is involved in the virulence of the parasite.
Collapse
Affiliation(s)
- Jenny Campos-Salinas
- Instituto de Parasitología y Biomedicina “López-Neyra”, CSIC, (IPBLN-CSIC), Parque Tecnológico de Ciencias de la Salud, Armilla, Granada, Spain
| | - David León-Guerrero
- Instituto de Parasitología y Biomedicina “López-Neyra”, CSIC, (IPBLN-CSIC), Parque Tecnológico de Ciencias de la Salud, Armilla, Granada, Spain
| | - Elena González-Rey
- Instituto de Parasitología y Biomedicina “López-Neyra”, CSIC, (IPBLN-CSIC), Parque Tecnológico de Ciencias de la Salud, Armilla, Granada, Spain
| | - Mario Delgado
- Instituto de Parasitología y Biomedicina “López-Neyra”, CSIC, (IPBLN-CSIC), Parque Tecnológico de Ciencias de la Salud, Armilla, Granada, Spain
| | - Santiago Castanys
- Instituto de Parasitología y Biomedicina “López-Neyra”, CSIC, (IPBLN-CSIC), Parque Tecnológico de Ciencias de la Salud, Armilla, Granada, Spain
| | - José M. Pérez-Victoria
- Instituto de Parasitología y Biomedicina “López-Neyra”, CSIC, (IPBLN-CSIC), Parque Tecnológico de Ciencias de la Salud, Armilla, Granada, Spain
| | - Francisco Gamarro
- Instituto de Parasitología y Biomedicina “López-Neyra”, CSIC, (IPBLN-CSIC), Parque Tecnológico de Ciencias de la Salud, Armilla, Granada, Spain
- * E-mail:
| |
Collapse
|
39
|
Impaired cholesterol efflux in senescent macrophages promotes age-related macular degeneration. Cell Metab 2013; 17:549-61. [PMID: 23562078 PMCID: PMC3640261 DOI: 10.1016/j.cmet.2013.03.009] [Citation(s) in RCA: 213] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2012] [Revised: 01/21/2013] [Accepted: 03/18/2013] [Indexed: 01/03/2023]
Abstract
Pathologic angiogenesis mediated by abnormally polarized macrophages plays a central role in common age-associated diseases such as atherosclerosis, cancer, and macular degeneration. Here we demonstrate that abnormal polarization in older macrophages is caused by programmatic changes that lead to reduced expression of ATP binding cassette transporter ABCA1. Downregulation of ABCA1 by microRNA-33 impairs the ability of macrophages to effectively efflux intracellular cholesterol, which in turn leads to higher levels of free cholesterol within senescent macrophages. Elevated intracellular lipid polarizes older macrophages to an abnormal, alternatively activated phenotype that promotes pathologic vascular proliferation. Mice deficient for Abca1, but not Abcg1, demonstrate an accelerated aging phenotype, whereas restoration of cholesterol efflux using LXR agonists or miR-33 inhibitors reverses it. Monocytes from older humans with age-related macular degeneration showed similar changes. These findings provide an avenue for therapeutic modulation of macrophage function in common age-related diseases.
Collapse
|
40
|
Abstract
PURPOSE OF REVIEW To offer a comprehensive review on the role of ABCG1 in cellular sterol homeostasis. RECENT FINDINGS Early studies with Abcg1 mice indicated that ABCG1 was crucial for tissue lipid homeostasis, especially in the lung. More recent studies have demonstrated that loss of ABCG1 has wide-ranging consequences and impacts lymphocyte and stem cell proliferation, endothelial cell function, macrophage foam cell formation, as well as insulin secretion from pancreatic β cells. Recent studies have also demonstrated that ABCG1 functions as an intracellular lipid transporter, localizes to intracellular vesicles/endosomes, and that the transmembrane domains are sufficient for localization and transport function. SUMMARY ABCG1 plays a crucial role in maintaining intracellular sterol and lipid homeostasis. Loss of this transporter has significant, cell-type-specific consequences ranging from effects on cellular proliferation, to surfactant production and/or insulin secretion. Elucidation of the mechanisms by which ABCG1 affects intracellular sterol flux/movement should provide important information that may link ABCG1 to diseases of dysregulated tissue lipid homeostasis.
Collapse
Affiliation(s)
- Elizabeth J Tarling
- Departments of Biological Chemistry and Medicine, David Geffen School of Medicine at UCLA, University of California, Los Angeles, CA 90095-1737, USA.
| |
Collapse
|
41
|
Characterization of palmitoylation of ATP binding cassette transporter G1: effect on protein trafficking and function. Biochim Biophys Acta Mol Cell Biol Lipids 2013; 1831:1067-78. [PMID: 23388354 DOI: 10.1016/j.bbalip.2013.01.019] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2012] [Revised: 01/14/2013] [Accepted: 01/25/2013] [Indexed: 12/12/2022]
Abstract
ATP-binding cassette transporter G1 (ABCG1) mediates cholesterol efflux onto lipidated apolipoprotein A-I and HDL and plays a role in various important physiological functions. However, the mechanism by which ABCG1 mediates cholesterol translocation is unclear. Protein palmitoylation regulates many functions of proteins such as ABCA1. Here we investigated if ABCG1 is palmitoylated and the subsequent effects on ABCG1-mediated cholesterol efflux. We demonstrated that ABCG1 is palmitoylated in both human embryonic kidney 293 cells and in mouse macrophage, J774. Five cysteine residues located at positions 26, 150, 311, 390 and 402 in the NH2-terminal cytoplasmic region of ABCG1 were palmitoylated. Removal of palmitoylation at Cys311 by mutating the residue to Ala (C311A) or Ser significantly decreased ABCG1-mediated cholesterol efflux. On the other hand, removal of palmitoylation at sites 26, 150, 390 and 402 had no significant effect. We further demonstrated that mutations of Cys311 affected ABCG1 trafficking from the endoplasmic reticulum. Therefore, our data suggest that palmitoylation plays a critical role in ABCG1-mediated cholesterol efflux through the regulation of trafficking.
Collapse
|
42
|
Witwer KW. XenomiRs and miRNA homeostasis in health and disease: evidence that diet and dietary miRNAs directly and indirectly influence circulating miRNA profiles. RNA Biol 2012; 9:1147-54. [PMID: 22951590 PMCID: PMC3579881 DOI: 10.4161/rna.21619] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Contributions of dietary miRNAs to circulating small RNA profiles would have profound implications for interpretation of miRNA biomarker studies: presumptive disease-specific markers might instead indicate responses to disease-associated quantitative or qualitative dietary alteration. This examination weighs the evidence for a 2-fold hypothesis: first, that ingested biological matter contributes directly to the miRNA complement of body compartments; and second, that these diet-derived exogenous miRNAs (or "xenomiRs") affect total miRNA profiles as part of a circulating miRNA homeostasis that is altered in many diseases. Homeostasis of high-density lipoprotein (HDL), a known miRNA carrier-provides a model as a proposed component of broader miRNA homeostasis. Further research into the dietary xenomiR hypothesis is needed to ensure rigor in the search for truly disease-specific miRNA biomarkers.
Collapse
Affiliation(s)
- Kenneth W Witwer
- Department of Molecular and Comparative Pathobiology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|