1
|
Shu X, Chen Z, Wang H, Xu B, Liu L, Zhang J, Zheng X, Chen J. Cloning, phylogenetic analysis, tissue expression profiling, and functional roles of NPC1L1 in chickens, quails, and ducks. Poult Sci 2025; 104:105032. [PMID: 40106905 PMCID: PMC11964625 DOI: 10.1016/j.psj.2025.105032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 03/01/2025] [Accepted: 03/13/2025] [Indexed: 03/22/2025] Open
Abstract
The Niemann-Pick C1-Like 1 (NPC1L1) protein, primarily expressed in the epithelial cells of the small intestine, is essential for cholesterol absorption from both dietary intake and biliary secretion. Despite this conserved function across mammals, the full-length coding sequence of NPC1L1 remains uncharacterized in key avian models including chicken (Gallus gallus), quail (Coturnix japonica), and duck (Anas platyrhynchos). In this study, we successfully cloned the full NPC1L1 mRNA sequence in chicken, quail, and duck, including the entire 5' and 3' untranslated regions, utilizing rapid amplification of cDNA ends methods. Phylogenetic analysis across 12 species, comprising four avian and eight representative mammalian species, revealed that the NPC1L1 sequences in the main poultry species exhibit a high degree of similarity. Despite the phylogenetic divergence of poultry NPC1L1 sequences from their mammalian counterparts, protein sequence alignment revealed that the cholesterol-sensing peptides of NPC1L1 are conserved across all species examined in this study. These findings imply that the NPC1L1 in poultry may also play a role in cholesterol transport. Analysis of tissue gene expression profiles in chickens, quails, and ducks indicated that NPC1L1 is predominantly expressed in the duodenum, jejunum, and liver. Additionally, experiments on medium-to-cell cholesterol transit in primary intestinal epithelial cells confirmed that chicken NPC1L1 is capable of efficiently transporting cholesterol into cells. Further experiments are required to elucidate the biological function of poultry NPC1L1. In summary, this study successfully cloned the full-length sequence of NPC1L1 from chickens, quails, and ducks, and conducted a comprehensive analysis of their evolutionary history and expression patterns. This research establishes a foundation for future investigations into the role of poultry NPC1L1 in cholesterol transport.
Collapse
Affiliation(s)
- Xin Shu
- Jiangsu Key Laboratory of Sericultural Biology and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, China
| | - Ziwei Chen
- Jiangsu Key Laboratory of Sericultural Biology and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, China
| | - Hui Wang
- Jiangsu Key Laboratory of Sericultural Biology and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, China
| | - Bingjie Xu
- Jiangsu Key Laboratory of Sericultural Biology and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, China
| | - Li Liu
- Jiangsu Key Laboratory of Sericultural Biology and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, China
| | - Jilong Zhang
- Jiangsu Key Laboratory of Sericultural Biology and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, China
| | - Xiaotong Zheng
- Jiangsu Key Laboratory of Sericultural Biology and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, China; Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, Sericultural Scientific Research Center, Chinese Academy of Agricultural Sciences, Zhenjiang, China
| | - Jianfei Chen
- Jiangsu Key Laboratory of Sericultural Biology and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, China; Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, Sericultural Scientific Research Center, Chinese Academy of Agricultural Sciences, Zhenjiang, China.
| |
Collapse
|
2
|
Ferrari A, Tontonoz P. Nonvesicular cholesterol transport in physiology. J Clin Invest 2025; 135:e188127. [PMID: 40091839 PMCID: PMC11910210 DOI: 10.1172/jci188127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2025] Open
Abstract
In mammalian cells cholesterol can be synthesized endogenously or obtained exogenously through lipoprotein uptake. Plasma membrane (PM) is the primary intracellular destination for both sources of cholesterol, and maintaining appropriate membrane cholesterol levels is critical for cellular viability. The endoplasmic reticulum (ER) acts as a cellular cholesterol sensor, regulating synthesis in response to cellular needs and determining the metabolic fates of cholesterol. Upon reaching the ER, cholesterol can be esterified to facilitate its incorporation into lipoproteins and lipid droplets or converted into other molecules such as bile acids and oxysterols. In recent years, it has become clear that the intracellular redistribution of lipids, including cholesterol, is critical for the regulation of various biological processes. This Review highlights physiology and mechanisms of nonvesicular (protein-mediated) intracellular cholesterol trafficking, with a focus on the role of Aster proteins in PM to ER cholesterol transport.
Collapse
|
3
|
Le May C, Ducheix S, Cariou B, Rimbert A. From Genetic Findings to new Intestinal Molecular Targets in Lipid Metabolism. Curr Atheroscler Rep 2025; 27:26. [PMID: 39798054 DOI: 10.1007/s11883-024-01264-w] [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] [Accepted: 12/03/2024] [Indexed: 01/13/2025]
Abstract
PURPOSE OF REVIEW While lipid-lowering therapies demonstrate efficacy, many patients still contend with significant residual risk of atherosclerotic cardiovascular diseases (ASCVD). The intestine plays a pivotal role in regulating circulating lipoproteins levels, thereby exerting influence on ASCVD pathogenesis. This review underscores recent genetic findings from the last six years that delineate new biological pathways and actors in the intestine which regulate lipid-related ASCVD risk. RECENT FINDINGS Specifically, we detail the role of LIMA1 in cholesterol absorption within enterocytes, the function of PLA2G12B in the expansion and lipidation of chylomicrons, the involvement of SURF4 in lipoprotein secretion, and the discovery of a gut-derived hormone named CHOLESIN that modulates cholesterol homeostasis through GPR146 via a gut-liver crosstalk. We further discuss the potential of these newly identified genes and pathways as novel targets for pharmaceutical intervention. Newly identified genetic and intestinal molecular mechanisms offer promising opportunities for preventing and treating ASCVD, but careful evaluation and further research are needed to optimize their clinical application.
Collapse
Affiliation(s)
- Cédric Le May
- Nantes Université, CHU Nantes, CNRS, Inserm, l'institut du thorax, F-44000, Nantes, France
| | - Simon Ducheix
- Nantes Université, CHU Nantes, CNRS, Inserm, l'institut du thorax, F-44000, Nantes, France
| | - Bertrand Cariou
- Nantes Université, CHU Nantes, CNRS, Inserm, l'institut du thorax, F-44000, Nantes, France
| | - Antoine Rimbert
- Nantes Université, CHU Nantes, CNRS, Inserm, l'institut du thorax, F-44000, Nantes, France.
| |
Collapse
|
4
|
Lee YN, Wu YJ, Su CH, Wang BJ, Yang SH, Lee HI, Chou YH, Tien TY, Lin CF, Chan WH, Chung CH, Wang SW, Yeh HI. Fluorescent gold nanoclusters possess multiple actions against atherosclerosis. Redox Biol 2024; 78:103427. [PMID: 39566163 PMCID: PMC11612375 DOI: 10.1016/j.redox.2024.103427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 11/06/2024] [Accepted: 11/11/2024] [Indexed: 11/22/2024] Open
Abstract
Atherosclerosis caused major morbidity and mortality worldwide. Molecules possessing lipid-lowering and/or anti-inflammatory properties are potential druggable targets against atherosclerosis. We examined the anti-atherosclerotic effects of fluorescent gold nanoclusters (FANC), which were dihydrolipoic acid (DHLA)-capped 2-nm gold nanoparticles. We evaluated the 8-week effects of FANC in Western-type diet-fed ApoE-deficient mice by either continuous intraperitoneal delivery (20 μM, 50 μl weekly) or via drinking water (300 nM). FANC reduced aortic atheroma burden, serum total cholesterol, and oxidative stress markers malondialdehyde and 4-hydroxynonenal levels. FANC attenuated hepatic lipid deposit, with changed expression of lipid homeostasis-related genes HMGCR, SREBP, PCSK9, and LDLR in a pattern similar to mice treated with ezetimibe. FANC also inhibited intestinal cholesterol absorption, resembling the action of ezetimibe. The lipid-lowering and anti-atherosclerotic effects of FANC reappeared in Western-type diet-fed LDLr-deficient mice. FANC bound insulin receptor β (IRβ) via DHLA, leading to AKT activation. However, unlike insulin, which also bound IRβ to activate AKT to induce HO-1, activation of AKT by FANC was independent of HO-1 expression in human aortic endothelial cells (HAECs). Alternatively, FANC up-regulated NRF2, interfered the binding of KEAP1 to NRF2, and promoted KEAP1 degradation to free NRF2 for nuclear entry to induce HO-1 that suppressed the expression of ICAM-1 and VCAM-1. Consistently, FANC suppressed ox-LDL-induced enhanced attachment of THP-derived macrophages onto HAECs. In macrophages, FANC up-regulated ABCA1, and reversed ox-LDL-induced suppression of cholesterol efflux. FANC effected in vitro at nano moles. In conclusion, our findings showed novel actions and multiple mechanisms of FANC worked coherently against atherosclerosis.
Collapse
Affiliation(s)
- Yi-Nan Lee
- Cardiovascular Center, Department of Internal Medicine, and Department of Medical Research, MacKay Memorial Hospital, Taipei, 10449, Taiwan
| | - Yih-Jer Wu
- Cardiovascular Center, Department of Internal Medicine, and Department of Medical Research, MacKay Memorial Hospital, Taipei, 10449, Taiwan; Department of Medicine, MacKay Medical College, New Taipei City, 25245, Taiwan
| | - Cheng-Huang Su
- Cardiovascular Center, Department of Internal Medicine, and Department of Medical Research, MacKay Memorial Hospital, Taipei, 10449, Taiwan
| | - Bo-Jeng Wang
- Cardiovascular Center, Department of Internal Medicine, and Department of Medical Research, MacKay Memorial Hospital, Taipei, 10449, Taiwan
| | - Sheng-Hsun Yang
- Cardiovascular Center, Department of Internal Medicine, and Department of Medical Research, MacKay Memorial Hospital, Taipei, 10449, Taiwan
| | - Hsin-I Lee
- Department of Medicine, MacKay Medical College, New Taipei City, 25245, Taiwan
| | - Yen-Hung Chou
- Department of Medicine, MacKay Medical College, New Taipei City, 25245, Taiwan
| | - Ting-Yi Tien
- Cardiovascular Center, Department of Internal Medicine, and Department of Medical Research, MacKay Memorial Hospital, Taipei, 10449, Taiwan
| | - Chao-Feng Lin
- Cardiovascular Center, Department of Internal Medicine, and Department of Medical Research, MacKay Memorial Hospital, Taipei, 10449, Taiwan; Department of Medicine, MacKay Medical College, New Taipei City, 25245, Taiwan
| | - Wen-Hsiung Chan
- Department of Bioscience Technology and Center for Nanotechnology, Chung Yuan Christian University, Zhongbei Road, Zhongli District, Taoyuan City, 32023, Taiwan
| | - Ching-Hu Chung
- Department of Medicine, MacKay Medical College, New Taipei City, 25245, Taiwan
| | - Shin-Wei Wang
- Institute of Biomedical Sciences, MacKay Medical College, New Taipei City, 25245, Taiwan
| | - Hung-I Yeh
- Cardiovascular Center, Department of Internal Medicine, and Department of Medical Research, MacKay Memorial Hospital, Taipei, 10449, Taiwan.
| |
Collapse
|
5
|
Lange Y, Steck TL. How active cholesterol coordinates cell cholesterol homeostasis: Test of a hypothesis. Prog Lipid Res 2024; 96:101304. [PMID: 39491591 DOI: 10.1016/j.plipres.2024.101304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 10/23/2024] [Accepted: 11/01/2024] [Indexed: 11/05/2024]
Abstract
How do cells coordinate the diverse elements that regulate their cholesterol homeostasis? Our model postulates that membrane cholesterol forms simple complexes with bilayer phospholipids. The phospholipids in the plasma membrane are of high affinity; consequently, they are fully complexed with the sterol. This sets the resting level of plasma membrane cholesterol. Cholesterol in excess of the stoichiometric equivalence point of these complexes has high chemical activity; we refer to it as active cholesterol. It equilibrates with the low affinity phospholipids in the intracellular membranes where it serves as a negative feedback signal to a manifold of regulatory proteins that rein in ongoing cholesterol accretion. We tested the model with a review of the literature regarding fourteen homeostatic proteins in enterocytes. It provided strong albeit indirect support for the following hypothesis. Active cholesterol inhibits cholesterol uptake and biosynthesis by suppressing both the expression and the activity of the gene products activated by SREBP-2; namely, HMGCR, LDLR and NPC1L1. It also reduces free cell cholesterol by serving as the substrate for its esterification by ACAT and for the synthesis of side-chain oxysterols, 27-hydroxycholesterol in particular. The oxysterols drive cholesterol depletion by promoting the destruction of HMGCR and stimulating sterol esterification as well as the activation of LXR. The latter fosters the expression of multiple homeostatic proteins, including four transporters for which active cholesterol is the likely substrate. By nulling active cholesterol, the manifold maintains the cellular sterol at its physiologic set point.
Collapse
Affiliation(s)
- Yvonne Lange
- Department of Pathology, Rush University Medical Center, Chicago, IL 60612, United States of America.
| | - Theodore L Steck
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL 60637, United States of America
| |
Collapse
|
6
|
Jiao B, Wang B, Liu B, Zhao J, Zhang Y. Potential impact of ezetimibe on patients with NAFLD/NASH: a meta-analysis of randomized controlled trials. Front Endocrinol (Lausanne) 2024; 15:1468476. [PMID: 39439571 PMCID: PMC11493694 DOI: 10.3389/fendo.2024.1468476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 09/20/2024] [Indexed: 10/25/2024] Open
Abstract
Background Non-alcoholic fatty liver disease (NAFLD) is now the most common cause of chronic liver disease. Studies have found that ezetimibe may be utilized as a supplemental treatment for NAFLD. Additionally, many clinical trials reported the potential impacts of ezetimibe on patients with NAFLD, although some conclusions remain controversial. Therefore, this study aimed to evaluate the effects of ezetimibe on patients with NAFLD. Method Online search was conducted across databases including PubMed, Embase, Scopus, Web of Science, Cochrane Library, Wanfang, VIP, and CNKI to retrieve all relevant controlled studies on the treatment of NAFLD with ezetimibe from the inception of the databases until April 2024. This meta-analysis comprised 10 randomized controlled trials (RCTs). Statistical analysis was conducted using the Meta package in R v4.3.2. Results A total of ten RCTs were included in this study, encompassing 578 patients (290 in the ezetimibe group and 288 in the control group) diagnosed with NAFLD/non-alcoholic steatohepatitis (NASH). The results indicated that ezetimibe significantly reduced levels of aspartate aminotransferase (P < 0.01), glutamyl transferase (γ-GT) (P < 0.01), total cholesterol (P < 0.01), low-density lipoprotein cholesterol (P < 0.01), high-sensitivity C-reactive protein (P < 0.01), and interleukin-6 (P < 0.01), and markedly increased levels of glycated hemoglobin (P = 0.02). Conclusions Ezetimibe may partially improve transaminase levels and positively impact liver function in patients with NAFLD/NASH. Systematic Review Registration https://www.crd.york.ac.uk/PROSPERO/, identifier CRD42023461467.
Collapse
|
7
|
El Din Taha HS, Kandil H, Badran HM, Farag N, Khamis H, Nasr G, Samy M, Abdrabou M, Abuelezz M, Shaker MM. 2024 Egyptian consensus statement on the role of non-statin therapies for LDL cholesterol lowering in different patient risk categories. Egypt Heart J 2024; 76:131. [PMID: 39302613 DOI: 10.1186/s43044-024-00562-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 09/15/2024] [Indexed: 09/22/2024] Open
Abstract
BACKGROUND The new millennium has witnessed increased understanding of cardiovascular (CV) risk factors and improvement in atherosclerotic cardiovascular disease (ASCVD) management. The role of LDL cholesterol and other atherogenic lipid particles in the development of atherosclerosis is now beyond doubt. MAIN BODY Statins have been widely used and recommended in guidelines for preventing and managing ischemic events. However, statins have side effects, and many patients do not achieve their low-density lipoprotein cholesterol (LDL-C) goals. In recent years, non-statin lipid-lowering agents have gained increasing use as adjuncts to statins or as alternatives in patients who cannot tolerate statins. This consensus proposes a simple approach for initiating non-statin lipid-lowering therapy and provides evidence-based recommendations. Our key advancements include the identification of patients at extreme risk for CV events, the consideration of initial combination therapy of statin and ezetimibe in very high-risk and extreme-risk groups and the extended use of bempedoic acid in patients not reaching LDL-C targets especially in resource-limited settings. CONCLUSIONS Overall, this consensus statement provides valuable insights into the expanding field of non-statin therapies and offers practical recommendations to enhance CV care, specifically focusing on improving LDL-C control in Egypt. While these recommendations hold promise, further research and real-world data are needed for validation and refinement.
Collapse
Affiliation(s)
| | | | | | - Nabil Farag
- Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | | | - Gamila Nasr
- Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Mina Samy
- Faculty of Medicine, Cairo University, Cairo, Egypt
| | | | | | | |
Collapse
|
8
|
Ferrari A, Whang E, Xiao X, Kennelly JP, Romartinez-Alonso B, Mack JJ, Weston T, Chen K, Kim Y, Tol MJ, Bideyan L, Nguyen A, Gao Y, Cui L, Bedard AH, Sandhu J, Lee SD, Fairall L, Williams KJ, Song W, Munguia P, Russell RA, Martin MG, Jung ME, Jiang H, Schwabe JW, Young SG, Tontonoz P. Aster-dependent nonvesicular transport facilitates dietary cholesterol uptake. Science 2023; 382:eadf0966. [PMID: 37943936 PMCID: PMC11073449 DOI: 10.1126/science.adf0966] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 09/27/2023] [Indexed: 11/12/2023]
Abstract
Intestinal absorption is an important contributor to systemic cholesterol homeostasis. Niemann-Pick C1 Like 1 (NPC1L1) assists in the initial step of dietary cholesterol uptake, but how cholesterol moves downstream of NPC1L1 is unknown. We show that Aster-B and Aster-C are critical for nonvesicular cholesterol movement in enterocytes. Loss of NPC1L1 diminishes accessible plasma membrane (PM) cholesterol and abolishes Aster recruitment to the intestinal brush border. Enterocytes lacking Asters accumulate PM cholesterol and show endoplasmic reticulum cholesterol depletion. Aster-deficient mice have impaired cholesterol absorption and are protected against diet-induced hypercholesterolemia. Finally, the Aster pathway can be targeted with a small-molecule inhibitor to manipulate cholesterol uptake. These findings identify the Aster pathway as a physiologically important and pharmacologically tractable node in dietary lipid absorption.
Collapse
Affiliation(s)
- Alessandra Ferrari
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles; Los Angeles, CA 90095, USA
- Department of Biological Chemistry, University of California, Los Angeles; Los Angeles, CA 90095, USA
| | - Emily Whang
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles; Los Angeles, CA 90095, USA
- Department of Biological Chemistry, University of California, Los Angeles; Los Angeles, CA 90095, USA
- Pediatric Gastroenterology, Hepatology, and Nutrition, David Geffen School of Medicine, University of California, Los Angeles; Los Angeles, CA 90095, USA
| | - Xu Xiao
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles; Los Angeles, CA 90095, USA
- Department of Biological Chemistry, University of California, Los Angeles; Los Angeles, CA 90095, USA
| | - John P. Kennelly
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles; Los Angeles, CA 90095, USA
- Department of Biological Chemistry, University of California, Los Angeles; Los Angeles, CA 90095, USA
| | | | - Julia J. Mack
- Department of Medicine, Division of Cardiology, University of California, Los Angeles; Los Angeles, CA, 90095, USA
| | - Thomas Weston
- Department of Medicine, Division of Cardiology, University of California, Los Angeles; Los Angeles, CA, 90095, USA
- Department of Human Genetics, University of California, Los Angeles; Los Angeles, CA 90095, USA
| | - Kai Chen
- Department of Chemistry, The University of Hong Kong, Hong Kong, 999077, China
- School of Molecular Sciences, The University of Western Australia, 35 Stirling Hwy, Crawley WA 6009, Australia
| | - Youngjae Kim
- Department of Chemistry, University of California, Los Angeles; Los Angeles, CA 90095, USA
| | - Marcus J. Tol
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles; Los Angeles, CA 90095, USA
- Department of Biological Chemistry, University of California, Los Angeles; Los Angeles, CA 90095, USA
| | - Lara Bideyan
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles; Los Angeles, CA 90095, USA
- Department of Biological Chemistry, University of California, Los Angeles; Los Angeles, CA 90095, USA
| | - Alexander Nguyen
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles; Los Angeles, CA 90095, USA
- Department of Biological Chemistry, University of California, Los Angeles; Los Angeles, CA 90095, USA
- Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine David Geffen School of Medicine, University of California, Los Angeles; Los Angeles, CA 90095, USA
| | - Yajing Gao
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles; Los Angeles, CA 90095, USA
- Department of Biological Chemistry, University of California, Los Angeles; Los Angeles, CA 90095, USA
| | - Liujuan Cui
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles; Los Angeles, CA 90095, USA
- Department of Biological Chemistry, University of California, Los Angeles; Los Angeles, CA 90095, USA
| | - Alexander H. Bedard
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles; Los Angeles, CA 90095, USA
- Department of Biological Chemistry, University of California, Los Angeles; Los Angeles, CA 90095, USA
| | - Jaspreet Sandhu
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles; Los Angeles, CA 90095, USA
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Stephen D. Lee
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles; Los Angeles, CA 90095, USA
- Department of Biological Chemistry, University of California, Los Angeles; Los Angeles, CA 90095, USA
| | - Louise Fairall
- Institute for Structural and Chemical Biology, University of Leicester, Leicester LE1 7RH, UK
| | - Kevin J. Williams
- Department of Biological Chemistry, University of California, Los Angeles; Los Angeles, CA 90095, USA
- UCLA Lipidomics Core, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Wenxin Song
- Department of Medicine, Division of Cardiology, University of California, Los Angeles; Los Angeles, CA, 90095, USA
- Department of Human Genetics, University of California, Los Angeles; Los Angeles, CA 90095, USA
| | - Priscilla Munguia
- Department of Medicine, Division of Cardiology, University of California, Los Angeles; Los Angeles, CA, 90095, USA
- Department of Human Genetics, University of California, Los Angeles; Los Angeles, CA 90095, USA
| | - Robert A. Russell
- National Deuteration Facility, Australian Nuclear Science and Technology Organisation, Lucas Heights, Australia
| | - Martin G. Martin
- Pediatric Gastroenterology, Hepatology, and Nutrition, David Geffen School of Medicine, University of California, Los Angeles; Los Angeles, CA 90095, USA
| | - Michael E. Jung
- Department of Chemistry, University of California, Los Angeles; Los Angeles, CA 90095, USA
| | - Haibo Jiang
- Department of Chemistry, The University of Hong Kong, Hong Kong, 999077, China
- School of Molecular Sciences, The University of Western Australia, 35 Stirling Hwy, Crawley WA 6009, Australia
| | - John W.R. Schwabe
- Institute for Structural and Chemical Biology, University of Leicester, Leicester LE1 7RH, UK
| | - Stephen G. Young
- Department of Medicine, Division of Cardiology, University of California, Los Angeles; Los Angeles, CA, 90095, USA
- Department of Human Genetics, University of California, Los Angeles; Los Angeles, CA 90095, USA
| | - Peter Tontonoz
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles; Los Angeles, CA 90095, USA
- Department of Biological Chemistry, University of California, Los Angeles; Los Angeles, CA 90095, USA
| |
Collapse
|
9
|
Xiao J, Dong LW, Liu S, Meng FH, Xie C, Lu XY, Zhang WJ, Luo J, Song BL. Bile acids-mediated intracellular cholesterol transport promotes intestinal cholesterol absorption and NPC1L1 recycling. Nat Commun 2023; 14:6469. [PMID: 37833289 PMCID: PMC10575946 DOI: 10.1038/s41467-023-42179-5] [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: 11/07/2022] [Accepted: 10/03/2023] [Indexed: 10/15/2023] Open
Abstract
Niemann-Pick C1-like 1 (NPC1L1) is essential for intestinal cholesterol absorption. Together with the cholesterol-rich and Flotillin-positive membrane microdomain, NPC1L1 is internalized via clathrin-mediated endocytosis and transported to endocytic recycling compartment (ERC). When ERC cholesterol level decreases, NPC1L1 interacts with LIMA1 and moves back to plasma membrane. However, how cholesterol leaves ERC is unknown. Here, we find that, in male mice, intracellular bile acids facilitate cholesterol transport to other organelles, such as endoplasmic reticulum, in a non-micellar fashion. When cholesterol level in ERC is decreased by bile acids, the NPC1L1 carboxyl terminus that previously interacts with the cholesterol-rich membranes via the A1272LAL residues dissociates from membrane, exposing the Q1277KR motif for LIMA1 recruitment. Then NPC1L1 moves back to plasma membrane. This study demonstrates an intracellular cholesterol transport function of bile acids and explains how the substantial amount of cholesterol in NPC1L1-positive compartments is unloaded in enterocytes during cholesterol absorption.
Collapse
Affiliation(s)
- Jian Xiao
- College of Life Sciences, Taikang Center for Life and Medical Sciences, Taikang Medical School, Hubei Key Laboratory of Cell Homeostasis, Wuhan University, Wuhan, China
| | - Le-Wei Dong
- College of Life Sciences, Taikang Center for Life and Medical Sciences, Taikang Medical School, Hubei Key Laboratory of Cell Homeostasis, Wuhan University, Wuhan, China
| | - Shuai Liu
- College of Life Sciences, Taikang Center for Life and Medical Sciences, Taikang Medical School, Hubei Key Laboratory of Cell Homeostasis, Wuhan University, Wuhan, China
- Heart Center, First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, Xinjiang, China
| | - Fan-Hua Meng
- College of Life Sciences, Taikang Center for Life and Medical Sciences, Taikang Medical School, Hubei Key Laboratory of Cell Homeostasis, Wuhan University, Wuhan, China
- Heart Center, First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, Xinjiang, China
- Affiliated Hospital of Jining Medical University, Jining, 272007, Shandong, China
| | - Chang Xie
- College of Life Sciences, Taikang Center for Life and Medical Sciences, Taikang Medical School, Hubei Key Laboratory of Cell Homeostasis, Wuhan University, Wuhan, China
| | - Xiao-Yi Lu
- College of Life Sciences, Taikang Center for Life and Medical Sciences, Taikang Medical School, Hubei Key Laboratory of Cell Homeostasis, Wuhan University, Wuhan, China
| | - Weiping J Zhang
- Department of Pathophysiology, Naval Medical University, Shanghai, China
| | - Jie Luo
- College of Life Sciences, Taikang Center for Life and Medical Sciences, Taikang Medical School, Hubei Key Laboratory of Cell Homeostasis, Wuhan University, Wuhan, China
| | - Bao-Liang Song
- College of Life Sciences, Taikang Center for Life and Medical Sciences, Taikang Medical School, Hubei Key Laboratory of Cell Homeostasis, Wuhan University, Wuhan, China.
| |
Collapse
|
10
|
Qusairy Z, Gangloff A, Leung SOA. Dysregulation of Cholesterol Homeostasis in Ovarian Cancer. Curr Oncol 2023; 30:8386-8400. [PMID: 37754524 PMCID: PMC10527727 DOI: 10.3390/curroncol30090609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 07/27/2023] [Accepted: 08/01/2023] [Indexed: 09/28/2023] Open
Abstract
Cholesterol plays an essential role in maintaining the rigidity of cell membranes and signal transduction. Various investigations confirmed empirically that the dysregulation of cholesterol homeostasis positively correlates with tumor progression. More specifically, recent studies suggested the distinct role of cholesterol in ovarian cancer cell proliferation, metastasis and chemoresistance. In this review, we summarize the current findings that suggest the contribution of cholesterol homeostasis dysregulation to ovarian cancer progression and resistance to anti-cancer agents. We also discuss the therapeutic implications of cholesterol-lowering drugs in ovarian cancer.
Collapse
Affiliation(s)
- Zahraa Qusairy
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada
| | - Anne Gangloff
- CHU de Québec-Université Laval Research Center, Québec City, QC G1V 4G2, Canada;
- Faculty of Medicine, Laval University, Québec City, QC G1V 0A6, Canada
| | - Shuk On Annie Leung
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, McGill University Health Centre, McGill University, Montreal, QC H4A 3J1, Canada
| |
Collapse
|
11
|
Ferrari A, Whang E, Xiao X, Kennelly JP, Romartinez-Alonso B, Mack JJ, Weston T, Chen K, Kim Y, Tol MJ, Bideyan L, Nguyen A, Gao Y, Cui L, Bedard AH, Sandhu J, Lee SD, Fairall L, Williams KJ, Song W, Munguia P, Russell RA, Martin MG, Jung ME, Jiang H, Schwabe JWR, Young SG, Tontonoz P. Aster-dependent non-vesicular transport facilitates dietary cholesterol uptake. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.07.548168. [PMID: 37503112 PMCID: PMC10369906 DOI: 10.1101/2023.07.07.548168] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Intestinal cholesterol absorption is an important contributor to systemic cholesterol homeostasis. Niemann-Pick C1 Like 1 (NPC1L1), the target of the drug ezetimibe (EZ), assists in the initial step of dietary cholesterol uptake. However, how cholesterol moves downstream of NPC1L1 is unknown. Here we show that Aster-B and Aster-C are critical for non-vesicular cholesterol movement in enterocytes, bridging NPC1L1 at the plasma membrane (PM) and ACAT2 in the endoplasmic reticulum (ER). Loss of NPC1L1 diminishes accessible PM cholesterol in enterocytes and abolishes Aster recruitment to the intestinal brush border. Enterocytes lacking Asters accumulate cholesterol at the PM and display evidence of ER cholesterol depletion, including decreased cholesterol ester stores and activation of the SREBP-2 transcriptional pathway. Aster-deficient mice have impaired cholesterol absorption and are protected against diet-induced hypercholesterolemia. Finally, we show that the Aster pathway can be targeted with a small molecule inhibitor to manipulate dietary cholesterol uptake. These findings identify the Aster pathway as a physiologically important and pharmacologically tractable node in dietary lipid absorption. One-Sentence Summary Identification of a targetable pathway for regulation of dietary cholesterol absorption.
Collapse
|
12
|
Mo P, Chen H, Jiang X, Hu F, Zhang F, Shan G, Chen W, Li S, Xu G. Effect of hepatic NPC1L1 on cholesterol gallstone disease and its mechanism. Heliyon 2023; 9:e15757. [PMID: 37159680 PMCID: PMC10163659 DOI: 10.1016/j.heliyon.2023.e15757] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 04/18/2023] [Accepted: 04/20/2023] [Indexed: 05/11/2023] Open
Abstract
Cholesterol gallstone disease (CGD) is associated with bile cholesterol supersaturation. The Niemann-Pick C1-like 1 (NPC1L1), the inhibitory target of ezetimibe (EZE), is a critical sterol transporter of cholesterol absorption. Intestinal NPC1L1 facilitates the absorption of cholesterol, whereas hepatic NPC1L1 promotes cholesterol uptake by hepatocytes and reduces bile cholesterol supersaturation. The potential of hepatic NPC1L1 to prevent CGD has yet to be established due to its absence in the mice model. In this study, we generated mice expressing hepatic NPC1L1 using adeno-associated virus (AAV) gene delivery. The biliary cholesterol saturations and gallstone formations were explored under chow diet and lithogenic diet (LD) with or without EZE treatment. The long-term (8-week) LD-fed AAV-mNPC1L1 mice exhibited no significant differences in biliary cholesterol saturation and gallstone formation compared to WT mice. EZE effectively prevented CGD in both WT and AAV-mNPC1L1 mice. Mechanistically, prolonged LD feeding induced the degradation of hepatic NPC1L1, whereas short-term (2-week) LD feeding preserved the expression of hepatic NPC1L1. In conclusion, our findings suggest that hepatic NPC1L1 is unable to prevent CGD, whereas EZE functions as an efficient bile cholesterol desaturator during CGD development.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Guoqiang Xu
- Corresponding author. Department of Gastroenterology, the First Affiliated Hospital, College of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou 310006, Zhejiang, China.
| |
Collapse
|
13
|
Wu X, Ma XH, Lin J, Yang X, Shi JH, Xie Z, Chen YX, Zhang WJ. Fluorescent visualization and evaluation of NPC1L1-mediated vesicular endocytosis during intestinal cholesterol absorption in mice. LIFE METABOLISM 2023; 2:load011. [PMID: 39872735 PMCID: PMC11749127 DOI: 10.1093/lifemeta/load011] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 03/10/2023] [Accepted: 03/16/2023] [Indexed: 01/30/2025]
Abstract
Excessive cholesterol absorption from intestinal lumen contributes to the pathogenesis of hypercholesterolemia, which is an independent risk factor for atherosclerotic cardiovascular disease. Niemann-Pick C1-like 1 (NPC1L1) is a major membrane protein responsible for cholesterol absorption, in which the physiological role of vesicular endocytosis is still controversial, and it lacks a feasible tool to visualize and evaluate the endocytosis of NPC1L1 vesicles in vivo. Here, we genetically labeled endogenous NPC1L1 protein with EGFP in a knock-in mouse model, and demonstrated fluorescent visualization and evaluation of the endocytic vesicles of NPC1L1-cago during intestinal cholesterol absorption. The homozygous NPC1L1-EGFP mice have normal NPC1L1 expression pattern as well as cholesterol homeostasis on chow or high-cholesterol diets. The fluorescence of NPC1L1-EGFP fusion protein localizes at the brush border membrane of small intestine, and EGFP-positive vesicles is visualized beneath the membrane as early as 5 min post oral gavage of cholesterol. Of note, the vesicles colocalize with the early endosomal marker early endosome antigen 1 (EEA1) and the filipin-stained free cholesterol. Pretreatment with NPC1L1 inhibitor ezetimibe inhibits the formation of these cholesterol-induced endocytic vesicles. Our data support the notion that NPC1L1-mediated cholesterol absorption is a vesicular endocytic process. NPC1L1-EGFP mice are a useful model for visualizing cellular NPC1L1-cargo vesicle itineraries and for evaluating NPC1L1 activity in vivo in response to diverse pharmacological agents and nutrients.
Collapse
Affiliation(s)
- Xiaojing Wu
- Department of Pathophysiology, Naval Medical University, Shanghai 200433, China
- NHC Key Laboratory of Hormones and Development, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin 300134, China
| | - Xian-Hua Ma
- Department of Pathophysiology, Naval Medical University, Shanghai 200433, China
| | - Jie Lin
- Department of Pathophysiology, Naval Medical University, Shanghai 200433, China
| | - Xiaohang Yang
- NHC Key Laboratory of Hormones and Development, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin 300134, China
| | - Jian-Hui Shi
- Department of Pathophysiology, Naval Medical University, Shanghai 200433, China
| | - Zhifang Xie
- Ministry of Education Shanghai Key Laboratory of Children’s Environmental Health, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| | - Yu-Xia Chen
- Department of Pathophysiology, Naval Medical University, Shanghai 200433, China
| | - Weiping J Zhang
- Department of Pathophysiology, Naval Medical University, Shanghai 200433, China
- NHC Key Laboratory of Hormones and Development, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin 300134, China
| |
Collapse
|
14
|
Rosenhouse-Dantsker A, Gazgalis D, Logothetis DE. PI(4,5)P 2 and Cholesterol: Synthesis, Regulation, and Functions. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1422:3-59. [PMID: 36988876 DOI: 10.1007/978-3-031-21547-6_1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 03/30/2023]
Abstract
Phosphatidylinositol 4,5-bisphosphate (PI(4,5)P2) is the most abundant membrane phosphoinositide and cholesterol is an essential component of the plasma membrane (PM). Both lipids play key roles in a variety of cellular functions including as signaling molecules and major regulators of protein function. This chapter provides an overview of these two important lipids. Starting from a brief description of their structure, synthesis, and regulation, the chapter continues to describe the primary functions and signaling processes in which PI(4,5)P2 and cholesterol are involved. While PI(4,5)P2 and cholesterol can act independently, they often act in concert or affect each other's impact. The chapters in this volume on "Cholesterol and PI(4,5)P2 in Vital Biological Functions: From Coexistence to Crosstalk" focus on the emerging relationship between cholesterol and PI(4,5)P2 in a variety of biological systems and processes. In this chapter, the next section provides examples from the ion channel field demonstrating that PI(4,5)P2 and cholesterol can act via common mechanisms. The chapter ends with a discussion of future directions.
Collapse
Affiliation(s)
| | - Dimitris Gazgalis
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Bouvé College of Health Sciences, Northeastern University, Boston, MA, USA
| | - Diomedes E Logothetis
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Bouvé College of Health Sciences, Northeastern University, Boston, MA, USA
| |
Collapse
|
15
|
Duan Y, Gong K, Xu S, Zhang F, Meng X, Han J. Regulation of cholesterol homeostasis in health and diseases: from mechanisms to targeted therapeutics. Signal Transduct Target Ther 2022; 7:265. [PMID: 35918332 PMCID: PMC9344793 DOI: 10.1038/s41392-022-01125-5] [Citation(s) in RCA: 168] [Impact Index Per Article: 56.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 07/04/2022] [Accepted: 07/12/2022] [Indexed: 12/13/2022] Open
Abstract
Disturbed cholesterol homeostasis plays critical roles in the development of multiple diseases, such as cardiovascular diseases (CVD), neurodegenerative diseases and cancers, particularly the CVD in which the accumulation of lipids (mainly the cholesteryl esters) within macrophage/foam cells underneath the endothelial layer drives the formation of atherosclerotic lesions eventually. More and more studies have shown that lowering cholesterol level, especially low-density lipoprotein cholesterol level, protects cardiovascular system and prevents cardiovascular events effectively. Maintaining cholesterol homeostasis is determined by cholesterol biosynthesis, uptake, efflux, transport, storage, utilization, and/or excretion. All the processes should be precisely controlled by the multiple regulatory pathways. Based on the regulation of cholesterol homeostasis, many interventions have been developed to lower cholesterol by inhibiting cholesterol biosynthesis and uptake or enhancing cholesterol utilization and excretion. Herein, we summarize the historical review and research events, the current understandings of the molecular pathways playing key roles in regulating cholesterol homeostasis, and the cholesterol-lowering interventions in clinics or in preclinical studies as well as new cholesterol-lowering targets and their clinical advances. More importantly, we review and discuss the benefits of those interventions for the treatment of multiple diseases including atherosclerotic cardiovascular diseases, obesity, diabetes, nonalcoholic fatty liver disease, cancer, neurodegenerative diseases, osteoporosis and virus infection.
Collapse
Affiliation(s)
- Yajun Duan
- Department of Cardiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.,Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Ke Gong
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Suowen Xu
- Department of Cardiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Feng Zhang
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Xianshe Meng
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Jihong Han
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China. .,College of Life Sciences, Key Laboratory of Bioactive Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China.
| |
Collapse
|
16
|
Sphingolipids and Cholesterol. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1372:1-14. [DOI: 10.1007/978-981-19-0394-6_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
17
|
Huang WW, Hong BH, Bai KK, Tan R, Yang T, Sun JP, Yi RZ, Wu H. Cis- and Trans-Palmitoleic Acid Isomers Regulate Cholesterol Metabolism in Different Ways. Front Pharmacol 2020; 11:602115. [PMID: 33363473 PMCID: PMC7753117 DOI: 10.3389/fphar.2020.602115] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 10/23/2020] [Indexed: 11/13/2022] Open
Abstract
Hypercholesterolemia is a preventable risk factor for atherosclerosis and cardiovascular disease. However, the mechanisms whereby cis-palmitoleic acid (cPOA) and trans-palmitoleic acid (tPOA) promote cholesterol homeostasis and ameliorate hypercholesterolemia remain elusive. To investigate the effects of cPOA and tPOA on cholesterol metabolism and its mechanisms, we induced hypercholesterolemia in mice using a high-fat diet and then intragastrically administered cPOA or tPOA once daily for 4 weeks. tPOA administration reduced serum cholesterol, low-density lipoprotein, high-density lipoprotein, and hepatic free cholesterol and total bile acids (TBAs). Conversely, cPOA had no effect on these parameters except for TBAs. Histological examination of the liver, however, revealed that cPOA ameliorated hepatic steatosis more effectively than tPOA. tPOA significantly reduced the expression of 3-hydroxy-3-methyl glutaryl coenzyme reductase (HMGCR), LXRα, and intestinal Niemann-Pick C1-Like 1 (NPC1L1) and increased cholesterol 7-alpha hydroxylase (CYP7A1) in the liver, whereas cPOA reduced the expression of HMGCR and CYP7A1 in the liver and had no effect on intestinal NPC1L1. In summary, our results suggest that cPOA and tPOA reduce cholesterol synthesis by decreasing HMGCR levels. Furthermore, tPOA, but not cPOA, inhibited intestinal cholesterol absorption by downregulating NPC1L1. Both high-dose tPOA and cPOA may promote the conversion of cholesterol into bile acids by upregulating CYP7A1. tPOA and cPOA prevent hypercholesterolemia via distinct mechanisms.
Collapse
Affiliation(s)
- Wen-Wen Huang
- Third Institute of Oceanography, Ministry of Natural Resources, Xiamen, China.,Department of Pharmacology, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Bi-Hong Hong
- Third Institute of Oceanography, Ministry of Natural Resources, Xiamen, China
| | - Kai-Kai Bai
- Third Institute of Oceanography, Ministry of Natural Resources, Xiamen, China
| | - Ran Tan
- Third Institute of Oceanography, Ministry of Natural Resources, Xiamen, China
| | - Ting Yang
- Fujian Provincial Key Laboratory on Hematology, Department of Hematology, Fujian Medical University Union Hospital, Fujian Institute of Hematology, Fuzhou, China
| | - Ji-Peng Sun
- Zhejiang Marine Development Research Institute, Zhoushan, China
| | - Rui-Zao Yi
- Third Institute of Oceanography, Ministry of Natural Resources, Xiamen, China
| | - Hao Wu
- Department of Pharmacology, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
18
|
Sun Y, Tang Y, Hou X, Wang H, Huang L, Wen J, Niu H, Zeng W, Bai Y. Novel Lactobacillus reuteri HI120 Affects Lipid Metabolism in C57BL/6 Obese Mice. Front Vet Sci 2020; 7:560241. [PMID: 33195535 PMCID: PMC7592399 DOI: 10.3389/fvets.2020.560241] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 09/02/2020] [Indexed: 12/24/2022] Open
Abstract
Intestinal probiotics are a primary focus area of current medical research. Probiotics such as bifidobacteria and lactobacilli can positively impact obesity and other metabolic diseases by directly or indirectly affecting lipid metabolism. However, the precise mechanisms of these effects remain unclear. In our previous work, the novel strain Lactobacillus reuteri HI120 was isolated and identified. HI120 expresses high levels of linoleic isomerase, resulting in the production of large amounts of conjugated linoleic acid (CLA) when mixed with linoleic acid (LA). As HI120 can efficiently transform LA into CLA, the effect of HI120 on the lipid metabolism in C57BL/6 obese mice was studied and the underlying molecular mechanism was explored in vitro. The results revealed no significant change in the diet, body weight, and serum triglyceride levels in mice. However, serum cholesterol levels were significantly decreased. The underlying mechanism may involve a CLA-mediated reduction in the gene expression levels of NPC1L1, SREBP-2, and HMG-CR, resulting in reduced cholesterol synthesis and absorption. Thus, HI120 can be developed as a potential probiotic formulation. After oral administration, LA from certain food sources can be converted into CLA in the human intestine to contribute to the prevention and treatment of obesity and hyperlipidemia.
Collapse
Affiliation(s)
- Ye Sun
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Department of General Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yanqing Tang
- Department of Cell Biology, School of Basic Medicine, Southern Medical University, Guangzhou, China
| | - Xufeng Hou
- Department of Cell Biology, School of Basic Medicine, Southern Medical University, Guangzhou, China
| | - Hesong Wang
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Liuying Huang
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Junjie Wen
- Guangzhou Weisengene Biological Technology Co., Ltd, Guangzhou, China
| | - Hongxin Niu
- Department of General Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Weisen Zeng
- Department of Cell Biology, School of Basic Medicine, Southern Medical University, Guangzhou, China
| | - Yang Bai
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
19
|
Nakano T, Inoue I, Takenaka Y, Ito R, Kotani N, Sato S, Nakano Y, Hirasaki M, Shimada A, Murakoshi T. Ezetimibe impairs transcellular lipid trafficking and induces large lipid droplet formation in intestinal absorptive epithelial cells. Biochim Biophys Acta Mol Cell Biol Lipids 2020; 1865:158808. [PMID: 32860884 DOI: 10.1016/j.bbalip.2020.158808] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 07/30/2020] [Accepted: 08/18/2020] [Indexed: 01/26/2023]
Abstract
Ezetimibe inhibits Niemann-Pick C1-like 1 (NPC1L1) protein, which mediates intracellular cholesterol trafficking from the brush border membrane to the endoplasmic reticulum, where chylomicron assembly takes place in enterocytes or in the intestinal absorptive epithelial cells. Cholesterol is a minor lipid constituent of chylomicrons; however, whether or not a shortage of cholesterol attenuates chylomicron assembly is unknown. The aim of this study was to examine the effect of ezetimibe, a potent NPC1L1 inhibitor, on trans-epithelial lipid transport, and chylomicron assembly and secretion in enterocytes. Caco-2 cells, an absorptive epithelial model, grown onto culture inserts were given lipid micelles from the apical side, and chylomicron-like triacylglycerol-rich lipoprotein secreted basolaterally were analyzed after a 24-h incubation period in the presence of ezetimibe up to 50 μM. The secretion of lipoprotein and apolipoprotein B48 were reduced by adding ezetimibe (30% and 34%, respectively). Although ezetimibe allowed the cells to take up cholesterol normally, the esterification was abolished. Meanwhile, oleic acid esterification was unaffected. Moreover, ezetimibe activated sterol regulatory element-binding protein 2 by approximately 1.5-fold. These results suggest that ezetimibe limited cellular cholesterol mobilization required for lipoprotein assembly. In such conditions, large lipid droplet formation in Caco-2 cells and the enterocytes of mice were induced, implying that unprocessed triacylglycerol was sheltered in these compartments. Although ezetimibe did not reduce the post-prandial lipid surge appreciably in triolein-infused mice, the results of the present study indicated that pharmacological actions of ezetimibe may participate in a novel regulatory mechanism for the efficient chylomicron assembly and secretion.
Collapse
Affiliation(s)
- Takanari Nakano
- Department of Biochemistry, Faculty of Medicine, Saitama Medical University, Saitama, Japan.
| | - Ikuo Inoue
- Department of Diabetes and Endocrinology, Faculty of Medicine, Saitama Medical University, Saitama, Japan
| | - Yasuhiro Takenaka
- Department of Diabetes and Endocrinology, Faculty of Medicine, Saitama Medical University, Saitama, Japan; Department of Physiology, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
| | - Rina Ito
- Department of Biochemistry, Faculty of Medicine, Saitama Medical University, Saitama, Japan
| | - Norihiro Kotani
- Department of Biochemistry, Faculty of Medicine, Saitama Medical University, Saitama, Japan
| | - Sawako Sato
- Department of Diabetes and Endocrinology, Faculty of Medicine, Saitama Medical University, Saitama, Japan
| | - Yuka Nakano
- Department of Diabetes and Endocrinology, Faculty of Medicine, Saitama Medical University, Saitama, Japan
| | - Masataka Hirasaki
- Division of Developmental Biology, Research Center for Genomic Medicine, Faculty of Medicine, Saitama Medical University, Saitama, Japan
| | - Akira Shimada
- Department of Diabetes and Endocrinology, Faculty of Medicine, Saitama Medical University, Saitama, Japan
| | - Takayuki Murakoshi
- Department of Biochemistry, Faculty of Medicine, Saitama Medical University, Saitama, Japan
| |
Collapse
|
20
|
Kiourtzidis M, Kühn J, Schutkowski A, Baur AC, Hirche F, Stangl GI. Inhibition of Niemann-Pick C1-like protein 1 by ezetimibe reduces uptake of deuterium-labeled vitamin D in mice. J Steroid Biochem Mol Biol 2020; 197:105504. [PMID: 31682937 DOI: 10.1016/j.jsbmb.2019.105504] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 09/22/2019] [Accepted: 10/22/2019] [Indexed: 12/17/2022]
Abstract
For a long time, orally ingested vitamin D was assumed to enter the body exclusively via simple passive diffusion. Recent data from in vitro experiments have described Niemann-Pick C1-like protein 1 (Npc1l1) as an important sterol transporter for vitamin D absorption. However, short-term applications of ezetimibe, which inhibits Npc1l1, were not associated with reduced vitamin D uptake in animals and humans. The current study aimed to elucidate the effect of long-term inhibition of Npc1l1 by ezetimibe on the uptake and storage of orally administered triple deuterated vitamin D3 (vitamin D3-d3). Therefore, 30 male wild-type mice were randomly assigned into three groups and received diets with 25 μg/kg of vitamin D3-d3 that contained 0 (control group), 50 or 100 mg/kg ezetimibe for six weeks. Mice fed diets with 50 or 100 mg/kg ezetimibe had lower circulating levels of cholesterol than control mice (-12 %, -15 %, P < 0.01). In contrast, the concentrations of 7-dehydrocholesterol in serum (P < 0.001) and liver (P < 0.05) were higher in mice treated with ezetimibe than in control mice, indicating an increased sterol synthesis to compensate for cholesterol reduction. Long-term application of ezetimibe significantly reduced the concentrations of vitamin D3-d3 in the serum and tissues of mice. The magnitude of vitamin D3 reduction was comparable between the two ezetimibe groups. In comparison to the control group, mice treated with ezetimibe had lower concentrations of deuterated vitamin D3 compared with the control group in serum (62 %, P < 0.001), liver (79 %, P < 0.001), kidney (54 %, P < 0.001), adipose tissues (55 %, P < 0.001) and muscle (41 %, P < 0.001). Surprisingly, the serum concentration of deuterated 25-hydroxyvitamin D3 was higher in the group fed 100 mg/kg ezetimibe than in the control group (P < 0.05). The protein expression of the vitamin D hydroxylases Cyp2r1, Cyp27a1, Cyp3a11, Cyp24a1 and Cyp2j3 in liver and Cyp27b1 and Cyp24a1 in kidney remained largely unaffected by ezetimibe. To conclude, Npc1l1 appears to be crucial for the uptake of orally ingested vitamin D because long-term inhibition of Npc1l1 by ezetimibe strongly reduced the levels of deuterium-labeled vitamin D in the body; the observed rise in deuterated 25-hydroxyvitamin D3 in serum of these mice can not be explained by the expression levels of the key enzymes involved in vitamin D hydroxylation.
Collapse
Affiliation(s)
- Mikis Kiourtzidis
- Institute of Agricultural and Nutritional Sciences, Martin Luther University Halle-Wittenberg, Germany.
| | - Julia Kühn
- Institute of Agricultural and Nutritional Sciences, Martin Luther University Halle-Wittenberg, Germany.
| | - Alexandra Schutkowski
- Institute of Agricultural and Nutritional Sciences, Martin Luther University Halle-Wittenberg, Germany.
| | - Anja C Baur
- Institute of Agricultural and Nutritional Sciences, Martin Luther University Halle-Wittenberg, Germany; Competence Cluster of Cardiovascular Health and Nutrition (nutriCARD), Halle-Jena-Leipzig, Germany.
| | - Frank Hirche
- Institute of Agricultural and Nutritional Sciences, Martin Luther University Halle-Wittenberg, Germany.
| | - Gabriele I Stangl
- Institute of Agricultural and Nutritional Sciences, Martin Luther University Halle-Wittenberg, Germany; Competence Cluster of Cardiovascular Health and Nutrition (nutriCARD), Halle-Jena-Leipzig, Germany.
| |
Collapse
|
21
|
Mechanisms and regulation of cholesterol homeostasis. Nat Rev Mol Cell Biol 2019; 21:225-245. [DOI: 10.1038/s41580-019-0190-7] [Citation(s) in RCA: 450] [Impact Index Per Article: 75.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/24/2019] [Indexed: 12/14/2022]
|
22
|
Ahmed O, Littmann K, Gustafsson U, Pramfalk C, Öörni K, Larsson L, Minniti ME, Sahlin S, Camejo G, Parini P, Eriksson M. Ezetimibe in Combination With Simvastatin Reduces Remnant Cholesterol Without Affecting Biliary Lipid Concentrations in Gallstone Patients. J Am Heart Assoc 2019; 7:e009876. [PMID: 30561264 PMCID: PMC6405603 DOI: 10.1161/jaha.118.009876] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Background In randomized trials (SHARP [Study of Heart and Renal Protection], IMPROVE‐IT [Improved Reduction of Outcomes: Vytorin Efficacy International Trial]), combination of statin and ezetimibe resulted in additional reduction of cardiovascular events. The reduction was greater in patients with type 2 diabetes mellitus (T2DM), where elevated remnant cholesterol and high cardiovascular disease risk is characteristic. To evaluate possible causes behind these results, 40 patients eligible for cholecystectomy, randomized to simvastatin, ezetimibe, combined treatment (simvastatin+ezetimibe), or placebo treatment during 4 weeks before surgery, were studied. Methods and Results Fasting blood samples were taken before treatment start and at the end (just before surgery). Bile samples and liver biopsies were collected during surgery. Hepatic gene expression levels were assessed with qPCR. Lipoprotein, apolipoprotein levels, and content of cholesterol, cholesteryl ester, and triglycerides were measured after lipoprotein fractionation. Lipoprotein subclasses were analyzed by nuclear magnetic resonance. Apolipoprotein affinity for human arterial proteoglycans (PG) was measured. Biomarkers of cholesterol biosynthesis and intestinal absorption and bile lipid composition were analyzed using mass spectrometry. Combined treatment caused a statistically significant decrease in plasma remnant particles and apolipoprotein B (ApoB)/lipoprotein content of cholesterol, cholesteryl esters, and triglycerides. All treatments reduced ApoB‐lipoprotein PG binding. Simvastatin and combined treatment modified the composition of lipoproteins. Changes in biomarkers of cholesterol synthesis and absorption and bile acid synthesis were as expected. No adverse events were found. Conclusions Combined treatment caused atheroprotective changes on ApoB‐lipoproteins, remnant particles, bile components, and in ApoB‐lipoprotein affinity for arterial PG. These effects might explain the decrease of cardiovascular events seen in the SHARP and IMPROVE‐IT trials. Clinical Trial Registration URL: www.clinicaltrialsregister.eu. Unique identifier: 2006‐004839‐30).
Collapse
Affiliation(s)
- Osman Ahmed
- 1 Division of Clinical Chemistry Department of Laboratory Medicine Karolinska Institutet Stockholm Sweden.,2 Department of Biochemistry Faculty of Medicine Khartoum University Khartoum Sudan
| | - Karin Littmann
- 1 Division of Clinical Chemistry Department of Laboratory Medicine Karolinska Institutet Stockholm Sweden.,3 Function Area Clinical Chemistry Karolinska University Laboratory Function Karolinska University Hospital Stockholm Sweden
| | - Ulf Gustafsson
- 5 Department of Surgery Karolinska Institutet at Danderyd Hospital Stockholm Sweden
| | - Camilla Pramfalk
- 1 Division of Clinical Chemistry Department of Laboratory Medicine Karolinska Institutet Stockholm Sweden
| | | | - Lilian Larsson
- 1 Division of Clinical Chemistry Department of Laboratory Medicine Karolinska Institutet Stockholm Sweden
| | - Mirko E Minniti
- 1 Division of Clinical Chemistry Department of Laboratory Medicine Karolinska Institutet Stockholm Sweden
| | - Staffan Sahlin
- 5 Department of Surgery Karolinska Institutet at Danderyd Hospital Stockholm Sweden
| | - German Camejo
- 1 Division of Clinical Chemistry Department of Laboratory Medicine Karolinska Institutet Stockholm Sweden
| | - Paolo Parini
- 1 Division of Clinical Chemistry Department of Laboratory Medicine Karolinska Institutet Stockholm Sweden.,4 Patient Area Endocrinology and Nephrology, Inflammation and Infection Theme Karolinska University Hospital Stockholm Sweden.,7 Metabolism Unit Department of Medicine Karolinska Institutet at Karolinska University Hospital Huddinge Stockholm Sweden
| | - Mats Eriksson
- 4 Patient Area Endocrinology and Nephrology, Inflammation and Infection Theme Karolinska University Hospital Stockholm Sweden.,7 Metabolism Unit Department of Medicine Karolinska Institutet at Karolinska University Hospital Huddinge Stockholm Sweden
| |
Collapse
|
23
|
Chen L, Chen XW, Huang X, Song BL, Wang Y, Wang Y. Regulation of glucose and lipid metabolism in health and disease. SCIENCE CHINA-LIFE SCIENCES 2019; 62:1420-1458. [PMID: 31686320 DOI: 10.1007/s11427-019-1563-3] [Citation(s) in RCA: 180] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 10/15/2019] [Indexed: 02/08/2023]
Abstract
Glucose and fatty acids are the major sources of energy for human body. Cholesterol, the most abundant sterol in mammals, is a key component of cell membranes although it does not generate ATP. The metabolisms of glucose, fatty acids and cholesterol are often intertwined and regulated. For example, glucose can be converted to fatty acids and cholesterol through de novo lipid biosynthesis pathways. Excessive lipids are secreted in lipoproteins or stored in lipid droplets. The metabolites of glucose and lipids are dynamically transported intercellularly and intracellularly, and then converted to other molecules in specific compartments. The disorders of glucose and lipid metabolism result in severe diseases including cardiovascular disease, diabetes and fatty liver. This review summarizes the major metabolic aspects of glucose and lipid, and their regulations in the context of physiology and diseases.
Collapse
Affiliation(s)
- Ligong Chen
- School of Pharmaceutical Sciences, Beijing Advanced Innovation Center for Structural Biology, Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Tsinghua University, Beijing, 100084, China.
| | - Xiao-Wei Chen
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, China.
| | - Xun Huang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Bao-Liang Song
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, 430072, China.
| | - Yan Wang
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, 430072, China.
| | - Yiguo Wang
- MOE Key Laboratory of Bioinformatics, Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China.
| |
Collapse
|
24
|
Thang SK, Chen PY, Gao WY, Wu MJ, Pan MH, Yen JH. Xanthohumol Suppresses NPC1L1 Gene Expression through Downregulation of HNF-4α and Inhibits Cholesterol Uptake in Caco-2 Cells. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2019; 67:11119-11128. [PMID: 31525874 DOI: 10.1021/acs.jafc.9b05221] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Xanthohumol (Xan) is a prenylated chalcone mainly found in hops; it has been demonstrated to function against hypercholesterolemia, hyperlipidemia, and atherosclerosis. In this study, we focused on the hypocholesterolemic effect of Xan on cholesterol uptake and the underlying molecular mechanisms of Xan in human intestinal Caco-2 cells. The microarray data showed that Niemann-Pick C1-like 1 (NPC1L1), an essential transporter for dietary cholesterol absorption, was significantly downregulated in Xan-treated Caco-2 cells. We demonstrated that Xan (10 and 20 μM) suppressed the mRNA and protein expression of NPC1L1 by 0.65 ± 0.12-fold and 0.54 ± 0.15-fold and 0.72 ± 0.04-fold and 0.44 ± 0.12-fold, respectively, compared to that of the vehicle-treated Caco-2 cells. Moreover, Xan (10 and 20 μM) significantly inhibited cholesterol uptake by approximately 12 and 32% in Caco-2 cells. NPC1L1 promoter activity was significantly suppressed by Xan, and a DNA element within the NPC1L1 promoter involved in Xan-mediated NPC1L1 reduction located between the -120 and -20 positions was identified. Moreover, Xan markedly decreased the mRNA and protein levels of hepatocyte nuclear factor 4α (HNF-4α), a critical activator of NPC1L1 transcription, and subsequently attenuated HNF-4α/NPC1L1 promoter complex formation, resulting in the suppression of NPC1L1 gene expression. Finally, we demonstrated that Xan markedly abolished lovastatin-induced NPC1L1 overexpression in Caco-2 cells. These findings reveal that Xan suppresses NPC1L1 expression via downregulation of HNF-4α and exerts inhibitory effects on cholesterol uptake in the intestinal Caco-2 cells. Our findings suggest Xan could serve as a potential cholesterol-lowering agent and supplement for statin therapy.
Collapse
Affiliation(s)
- Sang Kim Thang
- Institute of Medical Sciences , Tzu Chi University , Hualien 970 , Taiwan
| | - Pei-Yi Chen
- Center of Medical Genetics , Hualien Tzu Chi Hospital, Buddhist Tzu Chi Foundation , Hualien 970 , Taiwan
| | - Wan-Yun Gao
- Department of Molecular Biology and Human Genetics , Tzu Chi University , Hualien 970 , Taiwan
| | - Ming-Jiuan Wu
- Department of Biotechnology , Chia-Nan University of Pharmacy and Science , Tainan 717 , Taiwan
| | - Min-Hsiung Pan
- Institute of Food Science and Technology , National Taiwan University , Taipei 10617 , Taiwan
| | - Jui-Hung Yen
- Institute of Medical Sciences , Tzu Chi University , Hualien 970 , Taiwan
- Department of Molecular Biology and Human Genetics , Tzu Chi University , Hualien 970 , Taiwan
| |
Collapse
|
25
|
Lee Y, Han CY, Bae M, Park YK, Lee JY. Egg phospholipids exert an inhibitory effect on intestinal cholesterol absorption in mice. Nutr Res Pract 2019; 13:295-301. [PMID: 31388405 PMCID: PMC6669065 DOI: 10.4162/nrp.2019.13.4.295] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 04/04/2019] [Accepted: 05/09/2019] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND/OBJECTIVES Consumption of cholesterol-rich foods, such as eggs, has a minimal effect on circulating cholesterol levels in healthy humans. To gain insight, we investigated whether phospholipids rich in eggs (EPL) interfere with intestinal cholesterol absorption in vivo. MATERIALS/METHODS To investigate the acute effect of EPL on intestinal cholesterol absorption, male C57BL/6J mice were orally administered with 6, 11, or 19 mg of EPL for three days. We also tested the effect of chronic EPL consumption on cholesterol metabolism in the small intestine and the liver in mice with diet-induced hypercholesterolemia. Male C57BL/6J mice were fed a high fat/high cholesterol (HF/HC; 35% fat, 0.25% cholesterol, w/w) diet for 4 weeks to induce hypercholesterolemia, and subsequently the mice were either fed 0, 0.4 or 0.8% (w/w) of EPL for 6 weeks. RESULTS Intestinal cholesterol absorption was significantly decreased by the highest dose of acute EPL administration compared to control. Chronic EPL supplementation did not significantly alter intestinal cholesterol absorption nor plasma levels of total cholesterol and low-density lipoprotein cholesterol. In the small intestine and the liver, EPL supplementation minimally altered the expression of genes which regulate cellular cholesterol levels. CONCLUSION Although chronic EPL consumption was not able to counteract hypercholesterolemia in HF/HC-fed mice, acute EPL administration decreased intestinal cholesterol absorption. This study provides in vivo evidence that acute administration of PLs in eggs prevent cholesterol absorption in the intestine, suggesting a mechanism for a minimal effect of egg consumption on circulating cholesterol levels.
Collapse
Affiliation(s)
- Yoojin Lee
- Department of Nutritional Sciences, University of Connecticut, Storrs, Connecticut 06269, USA
| | - Catherine Y Han
- Department of Nutritional Sciences, University of Connecticut, Storrs, Connecticut 06269, USA
| | - Minkyung Bae
- Department of Nutritional Sciences, University of Connecticut, Storrs, Connecticut 06269, USA
| | - Young-Ki Park
- Department of Nutritional Sciences, University of Connecticut, Storrs, Connecticut 06269, USA
| | - Ji-Young Lee
- Department of Nutritional Sciences, University of Connecticut, Storrs, Connecticut 06269, USA.,Department of Food and Nutrition, Kyung Hee University, Seoul 02447, South Korea
| |
Collapse
|
26
|
Duangjai A, Ontawong A, Srimaroeng C. Siamese neem flower extract suppresses cholesterol absorption by interfering NPC1L1 and micellar property in vitro and in intestinal Caco-2 cells. Res Pharm Sci 2019; 14:190-200. [PMID: 31160896 PMCID: PMC6540922 DOI: 10.4103/1735-5362.258485] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Siamese neem (Azadirachta indica A. Juss var. siamensis Valeton) (A. indica) leaf extract, a traditional ayurvedic medicine, has been reported to exhibit antipyretic, antibacterial, antidyslipidemic, and antihyperglycemia effects. This study investigated the mechanism of hypocholesterolemic effect of methanolic extract of Siamese neem flowers in in vitro studies and in Caco-2 cells. Pancreatic cholesterol esterase and 3-hydroxy 3-methylglutaryl-CoA (HMG-CoA) reductase activities were assessed. Cholesterol micelle formation was prepared for in vitro cholesterol physicochemical property analyses, micelle size and solubility, and transport of cholesterol into the Caco-2 cells. The expression of niemann-pick C1 like 1 (NPC1L1), and its major regulator, peroxisome proliferator-activated receptor δ (PPARδ), were determined by western blot and real time polymerase chain reaction, respectively. A. indica flower extract inhibited pancreatic cholesterol esterase activity and increased cholesterol micelles size. Uptake of cholesterol into Caco-2 cells was inhibited by A. indica flower extract in a dose-dependent manner. In addition, A. indica extract inhibited HMG-CoA reductase activity, resulting in low level of intracellular cholesterol accumulation, together with increased cytosolic NPC1L1 protein expression and decreased PPARδ gene expression. In conclusion, A. indica flower extract has cholesterol-lowering effects by inhibiting intestinal cholesterol absorption, interfering micellar cholesterol formation, and attenuating cholesterol synthesis. As such, A. indica flower extract has potential for developing into nutraceutical product for prevention of hypocholesterolemia.
Collapse
Affiliation(s)
- Acharaporn Duangjai
- Division of Physiology, School of Medical Sciences, University of Phayao, Phayao, Thailand
| | - Atcharaporn Ontawong
- Division of Physiology, School of Medical Sciences, University of Phayao, Phayao, Thailand.,Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Chutima Srimaroeng
- Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| |
Collapse
|
27
|
Li R, Liu Y, Shi J, Yu Y, Lu H, Yu L, Liu Y, Zhang F. Diosgenin regulates cholesterol metabolism in hypercholesterolemic rats by inhibiting NPC1L1 and enhancing ABCG5 and ABCG8. Biochim Biophys Acta Mol Cell Biol Lipids 2019; 1864:1124-1133. [PMID: 31054325 DOI: 10.1016/j.bbalip.2019.04.010] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 04/20/2019] [Accepted: 04/26/2019] [Indexed: 12/11/2022]
Abstract
Hypercholesterolemia is a preventable risk factor for atherosclerosis and cardiovascular disease. However, the mechanisms of diosgenin (DG) that promote cholesterol homeostasis and alleviate hypercholesterolemia remain elusive. To investigate the effects and molecular mechanisms of the promotion of cholesterol metabolism by DG, a rat model of hypercholesterolemia was induced by providing a high-fat diet for 4 weeks. After 4 weeks, the rats were intragastrically administered high-dose DG (0.3 g/kg/d), low-dose DG (0.15 g/kg/d) or simvastatin (4 mg/kg/d) once a day for 8 weeks. The serum and hepatic cholesterol were tested, the mRNA and protein expression levels of Niemann-Pick C1-Like 1 (NPC1L1), liver X receptor-α (LXR-α) and the ATP-binding cassette G5/G8 (ABCG5/G8) transporters were measured. The results indicate that DG could reduce body weight, decrease the serum total cholesterol, triglyceride, low-density lipoprotein cholesterol, liver total cholesterol and free cholesterol levels compared to those in the controls. Simultaneously, liver tissue pathological morphology analyses revealed that DG could attenuate hepatic steatosis compared to that in the high-fat diet group. Further investigation demonstrated that DG significantly decreased the expression of NPC1L1 and LXR-α in the intestine and markedly increased the expression of ABCG5/G8 in the liver and intestine. Compared to the high-fat diet group, the rats in the DG-treated groups ameliorated hypercholesterolemia in a dose- and time-dependent manner. These data suggest that DG may not only inhibit intestinal cholesterol absorption by downregulating NPC1L1 but also enhance cholesterol excretion by increasing the expression of ABCG5/G8. DG could be a new candidate for the prevention of hypercholesterolemia.
Collapse
Affiliation(s)
- Ruoqi Li
- Department of Neurology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, China
| | - Yi Liu
- Department of Neurology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, China
| | - Jingjing Shi
- Department of Neurology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, China
| | - Yantong Yu
- Department of Neurology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, China
| | - Haifei Lu
- Department of Neurology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, China
| | - Lu Yu
- Department of Neurology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, China
| | - Yanqiang Liu
- Department of Neurology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, China
| | - Fengxia Zhang
- Department of Neurology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, China.
| |
Collapse
|
28
|
Liu Y, Chen D, Li J, Xia D, Yu M, Tao J, Zhang X, Li L, Gan Y. NPC1L1-Targeted Cholesterol-Grafted Poly(β-Amino Ester)/pDNA Complexes for Oral Gene Delivery. Adv Healthc Mater 2019; 8:e1800934. [PMID: 30773830 DOI: 10.1002/adhm.201800934] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 01/15/2019] [Indexed: 12/12/2022]
Abstract
Gene vectors for oral delivery encounter harsh conditions throughout the gastrointestinal tract, and the continuous peristaltic activity can quickly remove the vectors, leading to inefficient intestinal permeation. Therefore, vectors have demanding property requirements, such as stability under various pH and, more importantly, efficient uptake in different intestinal segments. In this study, a functional polymer, cholesterol-grafted poly(β-amino ester) (poly[hexamethylene diacrylate-β-(5-amino-1-pentanol)] (CH-PHP)), is synthesized and electrostatically interacted with plasmid DNA to form a CH-PHP/DNA complex (CPNC). This complex is designed to target the Niemann-Pick C1-like receptor, a cholesterol receptor, to improve oral gene delivery efficacy. With the presence of cholesterol, CH-PHP shows mitigated cytotoxicity, enhanced enzyme resistance, and improved gene condensing ability. CPNC further contributes to ≈43.1- and 2.3-fold increases in luciferase expression in Caco-2 cells compared with PNC and Lipo 2000/DNA complexes, respectively. In addition, the in vivo transfection efficacy of CPNC is ≈4.1-, 2.1-, and 1.6-fold higher than that of Lipo 2000/DNA complexes in rat duodenum, jejunum, and ileum, respectively. Therefore, CPNC may be a promising delivery vector for gene delivery, and using a cholesterol-specific endocytic pathway can be a novel approach to achieve efficient oral gene transfection.
Collapse
Affiliation(s)
- Yuan Liu
- Department of ChemistryShanghai University Shanghai 200444 P. R. China
- Shanghai Institute of Materia MedicaChinese Academy of Sciences No. 501 Haike Road Shanghai 201203 P. R. China
| | - Dan Chen
- Shanghai Institute of Materia MedicaChinese Academy of Sciences No. 501 Haike Road Shanghai 201203 P. R. China
| | - Jialin Li
- Shanghai Institute of Materia MedicaChinese Academy of Sciences No. 501 Haike Road Shanghai 201203 P. R. China
- School of PharmacyShanghai University of Traditional Chinese Medicine Shanghai 201203 P. R. China
| | - Dengning Xia
- Shanghai Institute of Materia MedicaChinese Academy of Sciences No. 501 Haike Road Shanghai 201203 P. R. China
| | - Miaorong Yu
- Shanghai Institute of Materia MedicaChinese Academy of Sciences No. 501 Haike Road Shanghai 201203 P. R. China
| | - Jinsong Tao
- Shanghai Institute of Materia MedicaChinese Academy of Sciences No. 501 Haike Road Shanghai 201203 P. R. China
| | - Xinxin Zhang
- Shanghai Institute of Materia MedicaChinese Academy of Sciences No. 501 Haike Road Shanghai 201203 P. R. China
| | - Li Li
- Department of ChemistryShanghai University Shanghai 200444 P. R. China
| | - Yong Gan
- Shanghai Institute of Materia MedicaChinese Academy of Sciences No. 501 Haike Road Shanghai 201203 P. R. China
| |
Collapse
|
29
|
Kim YC, Byun S, Seok S, Guo G, Xu HE, Kemper B, Kemper JK. Small Heterodimer Partner and Fibroblast Growth Factor 19 Inhibit Expression of NPC1L1 in Mouse Intestine and Cholesterol Absorption. Gastroenterology 2019; 156:1052-1065. [PMID: 30521806 PMCID: PMC6409196 DOI: 10.1053/j.gastro.2018.11.061] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 11/08/2018] [Indexed: 12/13/2022]
Abstract
BACKGROUND & AIMS The nuclear receptor subfamily 0 group B member 2 (NR0B2, also called SHP) is expressed at high levels in the liver and intestine. Postprandial fibroblast growth factor 19 (human FGF19, mouse FGF15) signaling increases the transcriptional activity of SHP. We studied the functions of SHP and FGF19 in the intestines of mice, including their regulation of expression of the cholesterol transporter NPC1L1 )NPC1-like intracellular cholesterol transporter 1) and cholesterol absorption. METHODS We performed histologic and biochemical analyses of intestinal tissues from C57BL/6 and SHP-knockout mice and performed RNA-sequencing analyses to identify genes regulated by SHP. The effects of fasting and refeeding on intestinal expression of NPC1L1 were examined in C57BL/6, SHP-knockout, and FGF15-knockout mice. Mice were given FGF19 daily for 1 week; fractional cholesterol absorption, cholesterol and bile acid (BA) levels, and composition of BAs were measured. Intestinal organoids were generated from C57BL/6 and SHP-knockout mice, and cholesterol uptake was measured. Luciferase reporter assays were performed with HT29 cells. RESULTS We found that the genes that regulate lipid and ion transport in intestine, including NPC1L1, were up-regulated and that cholesterol absorption was increased in SHP-knockout mice compared with C57BL/6 mice. Expression of NPC1L1 was reduced in C57BL/6 mice after refeeding after fasting but not in SHP-knockout or FGF15-knockout mice. SHP-knockout mice had altered BA composition compared with C57BL/6 mice. FGF19 injection reduced expression of NPC1L1, decreased cholesterol absorption, and increased levels of hydrophilic BAs, including tauro-α- and -β-muricholic acids; these changes were not observed in SHP-knockout mice. SREBF2 (sterol regulatory element binding transcription factor 2), which regulates cholesterol, activated transcription of NPC1L1. FGF19 signaling led to phosphorylation of SHP, which inhibited SREBF2 activity. CONCLUSIONS Postprandial FGF19 and SHP inhibit SREBF2, which leads to repression of intestinal NPC1L1 expression and cholesterol absorption. Strategies to increase FGF19 signaling to activate SHP might be developed for treatment of hypercholesterolemia.
Collapse
Affiliation(s)
- Young-Chae Kim
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, Illinois.
| | - Sangwon Byun
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Sunmi Seok
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Grace Guo
- Department of Pharmacology and Toxicology, School of Pharmacy, Rutgers University, Piscataway, New Jersey
| | - H Eric Xu
- Laboratory of Structure Sciences, Van Andel Research Institute, Grand Rapids, Michigan
| | - Byron Kemper
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Jongsook Kim Kemper
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, Illinois.
| |
Collapse
|
30
|
Han S, Zhang W, Zhang R, Jiao J, Fu C, Tong X, Zhang W, Qin L. Cereal fiber improves blood cholesterol profiles and modulates intestinal cholesterol metabolism in C57BL/6 mice fed a high-fat, high-cholesterol diet. Food Nutr Res 2019; 63:1591. [PMID: 30863273 PMCID: PMC6403461 DOI: 10.29219/fnr.v63.1591] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 12/24/2018] [Accepted: 12/29/2018] [Indexed: 01/06/2023] Open
Abstract
Background Dietary intake of cereal fiber has been reported to benefit lipid metabolism through multiple mechanisms. The present study aimed to discover the potential mechanisms by which cereal fiber could modify the intestinal cholesterol metabolism. Design Male C57BL/6 mice were fed a reference chow (RC) diet; high-fat, high-cholesterol (HFC) diet; HFC plus oat fiber diet; or HFC plus wheat bran fiber diet for 24 weeks. Serum lipids were measured by enzymatic methods. Western blot was used to determine the protein expressions involved in intestinal cholesterol metabolism. Results Our results showed that HFC-induced elevations of serum triglycerides, total cholesterol, and low-density lipoprotein cholesterol were normalized in both groups that received cereal fiber. At the protein level, compared with the HFC diet group, the two cereal fibers, especially the oat fiber, significantly increased the protein expression of peroxisome proliferator-activated receptor alpha, liver X receptor alpha, sterol regulatory element-binding protein (SREBP) 2, low-density lipoprotein receptor, adenosine triphosphate (ATP)-binding cassette A1, and ATP-binding cassette G1, while decreasing the protein expression of Niemann-Pick C1-like protein 1, SREBP-1, fatty acid synthase, and acetyl-coenzyme A carboxylase, which were involved in intestinal cholesterol metabolism. Conclusion Taken together, increased intake of cereal fiber improved blood cholesterol profiles and increased the intestinal cholesterol efflux and cholesterol clearance in C57BL/6 mice fed a HFC diet. Oat fiber had a stronger effect than wheat bran fiber on cholesterol metabolism by modulating the PPARα, LXRα, and SREBP signaling pathways.
Collapse
Affiliation(s)
- Shufen Han
- Department of Nutrition and Food Hygiene, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Disease, School of Public Health, Soochow University, Suzhou, China
| | - Wei Zhang
- Department of Nutrition and Food Hygiene, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Disease, School of Public Health, Soochow University, Suzhou, China.,Suzhou Maternal and Child Health Care and Family Planning Service Center, Suzhou, China
| | - Ru Zhang
- Department of Nutrition and Food Hygiene, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Disease, School of Public Health, Soochow University, Suzhou, China
| | - Jun Jiao
- Department of Nutrition and Food Hygiene, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Disease, School of Public Health, Soochow University, Suzhou, China
| | - Chunling Fu
- Department of Nutrition and Food Hygiene, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Disease, School of Public Health, Soochow University, Suzhou, China
| | - Xing Tong
- Department of Nutrition and Food Hygiene, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Disease, School of Public Health, Soochow University, Suzhou, China
| | | | - Liqiang Qin
- Department of Nutrition and Food Hygiene, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Disease, School of Public Health, Soochow University, Suzhou, China
| |
Collapse
|
31
|
Nakano T, Inoue I, Murakoshi T. A Newly Integrated Model for Intestinal Cholesterol Absorption and Efflux Reappraises How Plant Sterol Intake Reduces Circulating Cholesterol Levels. Nutrients 2019; 11:nu11020310. [PMID: 30717222 PMCID: PMC6412963 DOI: 10.3390/nu11020310] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 01/28/2019] [Accepted: 01/29/2019] [Indexed: 12/27/2022] Open
Abstract
Cholesterol homeostasis is maintained through a balance of de novo synthesis, intestinal absorption, and excretion from the gut. The small intestine contributes to cholesterol homeostasis by absorbing and excreting it, the latter of which is referred to as trans-intestinal cholesterol efflux (TICE). Because the excretion efficiency of endogenous cholesterol is inversely associated with the development of atherosclerosis, TICE provides an attractive therapeutic target. Thus, elucidation of the mechanism is warranted. We have shown that intestinal cholesterol absorption and TICE are inversely correlated in intestinal perfusion experiments in mice. In this review, we summarized 28 paired data sets for absorption efficiency and fecal neutral sterol excretion, a surrogate marker of TICE, obtained from 13 available publications in a figure, demonstrating the inverse correlation were nearly consistent with the assumption. We then offer a bidirectional flux model that accommodates absorption and TICE occurring in the same segment. In this model, the brush border membrane (BBM) of intestinal epithelial cells stands as the dividing ridge for cholesterol fluxes, making the opposite fluxes competitive and being coordinated by shared BBM-localized transporters, ATP-binding cassette G5/G8 and Niemann-Pick C1-like 1. Furthermore, the idea is applied to address how excess plant sterol/stanol (PS) intake reduces circulating cholesterol level, because the mechanism is still unclear. We propose that unabsorbable PS repeatedly shuttles between the BBM and lumen and promotes concomitant cholesterol efflux. Additionally, PSs, which are chemically analogous to cholesterol, may disturb the trafficking machineries that transport cholesterol to the cell interior.
Collapse
Affiliation(s)
- Takanari Nakano
- Department of Biochemistry, Faculty of Medicine, Saitama Medical University, Saitama 350-0495, Japan.
| | - Ikuo Inoue
- Department of Diabetes and Endocrinology, Faculty of Medicine, Saitama Medical University, Saitama 350-0495, Japan.
| | - Takayuki Murakoshi
- Department of Biochemistry, Faculty of Medicine, Saitama Medical University, Saitama 350-0495, Japan.
| |
Collapse
|
32
|
Nihei W, Nagafuku M, Hayamizu H, Odagiri Y, Tamura Y, Kikuchi Y, Veillon L, Kanoh H, Inamori KI, Arai K, Kabayama K, Fukase K, Inokuchi JI. NPC1L1-dependent intestinal cholesterol absorption requires ganglioside GM3 in membrane microdomains. J Lipid Res 2018; 59:2181-2187. [PMID: 30242108 DOI: 10.1194/jlr.m089201] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2018] [Revised: 09/14/2018] [Indexed: 12/13/2022] Open
Abstract
Intestinal cholesterol absorption is a key regulator of systemic cholesterol homeostasis. Excessive dietary cholesterol and its intestinal uptake lead to hypercholesterolemia, a major risk factor for cardiovascular disease. Intestinal cholesterol uptake is mediated by Niemann-Pick C1-like 1 (NPC1L1), a transmembrane protein localized in membrane microdomains (lipid rafts) enriched in gangliosides and cholesterol. The roles of gangliosides, such as monosialodihexosylganglioside (GM3) and its synthesizing enzyme GM3 synthase (GM3S), in NPC1L1-dependent cholesterol uptake have not been examined previously. Here, we examined NPC1L1-dependent cholesterol uptake in a cell model as well as in wild-type and apoE-deficient mice fed normal or high-cholesterol diets. We showed that NPC1L1-dependent cholesterol uptake was impaired in GM3S-deficient cells and that GM3S deficiency promoted resistance to hypercholesterolemia in both wild-type and apoE-deficient mice fed the high-cholesterol but not the normal diet. Our findings suggest that GM3 and related gangliosides are essential for NPC1L1-mediated intestinal cholesterol absorption and are potential targets for hypercholesterolemia therapy.
Collapse
Affiliation(s)
- Wataru Nihei
- Division of Glycopathology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Masakazu Nagafuku
- Division of Glycopathology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Hirotaka Hayamizu
- Division of Glycopathology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Yuta Odagiri
- Division of Glycopathology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Yumi Tamura
- Division of Glycopathology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Yui Kikuchi
- Division of Glycopathology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Lucas Veillon
- Division of Glycopathology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai, Japan.,Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Hirotaka Kanoh
- Division of Glycopathology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Kei-Ichiro Inamori
- Division of Glycopathology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Kenta Arai
- Laboratory of Natural Product Chemistry, Department of Chemistry, Osaka University, Toyonaka, Japan
| | - Kazuya Kabayama
- Laboratory of Natural Product Chemistry, Department of Chemistry, Osaka University, Toyonaka, Japan
| | - Koichi Fukase
- Laboratory of Natural Product Chemistry, Department of Chemistry, Osaka University, Toyonaka, Japan
| | - Jin-Ichi Inokuchi
- Division of Glycopathology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| |
Collapse
|
33
|
Zhang YY, Fu ZY, Wei J, Qi W, Baituola G, Luo J, Meng YJ, Guo SY, Yin H, Jiang SY, Li YF, Miao HH, Liu Y, Wang Y, Li BL, Ma YT, Song BL. A LIMA1 variant promotes low plasma LDL cholesterol and decreases intestinal cholesterol absorption. Science 2018; 360:1087-1092. [DOI: 10.1126/science.aao6575] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2017] [Revised: 02/07/2018] [Accepted: 04/17/2018] [Indexed: 12/19/2022]
Abstract
A high concentration of low-density lipoprotein cholesterol (LDL-C) is a major risk factor for cardiovascular disease. Although LDL-C levels vary among humans and are heritable, the genetic factors affecting LDL-C are not fully characterized. We identified a rare frameshift variant in the LIMA1 (also known as EPLIN or SREBP3) gene from a Chinese family of Kazakh ethnicity with inherited low LDL-C and reduced cholesterol absorption. In a mouse model, LIMA1 was mainly expressed in the small intestine and localized on the brush border membrane. LIMA1 bridged NPC1L1, an essential protein for cholesterol absorption, to a transportation complex containing myosin Vb and facilitated cholesterol uptake. Similar to the human phenotype, Lima1-deficient mice displayed reduced cholesterol absorption and were resistant to diet-induced hypercholesterolemia. Through our study of both mice and humans, we identify LIMA1 as a key protein regulating intestinal cholesterol absorption.
Collapse
|
34
|
Abstract
Vitamin K (VK) is an essential cofactor for the post-translational conversion of peptide-bound glutamate to γ-carboxyglutamate. The resultant vitamin K-dependent proteins are known or postulated to possess a variety of biological functions, chiefly in the maintenance of hemostasis. The vitamin K cycle is a cellular pathway that drives γ-carboxylation and recycling of VK via γ-carboxyglutamyl carboxylase (GGCX) and vitamin K epoxide reductase (VKOR), respectively. In this review, we show how novel molecular biological approaches are providing new insights into the pathophysiological mechanisms caused by rare mutations of both GGCX and VKOR. We also discuss how other protein regulators influence the intermediary metabolism of VK, first through intestinal absorption and second through a pathway that converts some dietary phylloquinone to menadione, which is prenylated to menaquinone-4 (MK-4) in target tissues by UBIAD1. The contribution of MK-4 synthesis to VK functions is yet to be revealed.
Collapse
Affiliation(s)
- Martin J Shearer
- Centre for Haemostasis and Thrombosis, Guy's and St Thomas' NHS Foundation Trust, London SE1 7EH, United Kingdom;
| | - Toshio Okano
- Department of Hygienic Sciences, Kobe Pharmaceutical University, Kobe 658-8558 Japan;
| |
Collapse
|
35
|
Ahangari N, Ghayour Mobarhan M, Sahebkar A, Pasdar A. Molecular aspects of hypercholesterolemia treatment: current perspectives and hopes. Ann Med 2018; 50:303-311. [PMID: 29578362 DOI: 10.1080/07853890.2018.1457795] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Hypercholesterolemia is a pathological condition which has been reported in 39% of the worlds' adult population. We aimed to review molecular aspects of current and novel therapeutic approaches based on low-density lipoprotein cholesterol lowering strategies. Pathogenic mutations in the LDLR, ApoB, PCSK9 and LDLRAP genes cause deficient clearance of circulating low-density lipoprotein cholesterol particles via hepatic LDL receptor. This leads to increased plasma LDL cholesterol levels from birth, which can cause LDL depositions in the arterial walls. Ultimately, it progresses to atherosclerosis and an increased risk of premature cardiovascular diseases. Currently, statins, Ezetimibe, Bile acid sequestrants and PCSK9 inhibitors are the main therapeutic agents for the treatment of hypercholesterolemia. Moreover, novel RNA-based therapy had a strong impact on therapeutic strategies in recent decades. Additional development in understanding of the molecular basis of hypercholesterolemia will provide opportunities for the development of targeted therapy in the near future. Key Messages The most common genes involved in hypercholesterolemia are LDLR, PCSK9 and ApoB. Pharmacogenetic effects are typically constrained to pathways closely related to the pharmacodynamics and pharmacokinetics. Change in lifestyle and diet along with treatment of the underlying disease and drug therapy are the current therapeutic strategies.
Collapse
Affiliation(s)
- Najmeh Ahangari
- a Departement of Modern Sciences and Technologies, Faculty of Medicine , Mashhad University of Medical Sciences , Mashhad , Iran
| | - Majid Ghayour Mobarhan
- b Metabolic Syndrome Research Centre, School of Medicine , Mashhad University of Medical Sciences , Mashhad , Iran
| | - Amirhossein Sahebkar
- c Biotechnology Research Center , Pharmaceutical Technology Institute, Mashhad University of Medical Sciences , Mashhad , Iran.,d Neurogenic Inflammation Research Center , Mashhad University of Medical Sciences , Mashhad , Iran
| | - Alireza Pasdar
- e Medical Genetics Research Centre, Faculty of Medicine , Mashhad University of Medical Sciences , Mashhad , Iran.,f Division of Applied Medicine, Medical School , University of Aberdeen , Foresterhill , Aberdeen , UK
| |
Collapse
|
36
|
Engelking LJ, Cantoria MJ, Xu Y, Liang G. Developmental and extrahepatic physiological functions of SREBP pathway genes in mice. Semin Cell Dev Biol 2017; 81:98-109. [PMID: 28736205 DOI: 10.1016/j.semcdb.2017.07.011] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Accepted: 07/07/2017] [Indexed: 12/17/2022]
Abstract
Sterol regulatory element-binding proteins (SREBPs), master transcriptional regulators of cholesterol and fatty acid synthesis, have been found to contribute to a diverse array of cellular processes. In this review, we focus on genetically engineered mice in which the activities of six components of the SREBP gene pathway, namely SREBP-1, SREBP-2, Scap, Insig-1, Insig-2, or Site-1 protease have been altered through gene knockout or transgenic approaches. In addition to the expected impacts on lipid metabolism, manipulation of these genes in mice is found to affect a wide array of developmental and physiologic processes ranging from interferon signaling in macrophages to synaptic transmission in the brain. The findings reviewed herein provide a blueprint to guide future studies defining the complex interactions between lipid biology and the physiologic processes of many distinct organ systems.
Collapse
Affiliation(s)
- Luke J Engelking
- Departments of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| | - Mary Jo Cantoria
- Departments of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Yanchao Xu
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Guosheng Liang
- Departments of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
37
|
Wang YJ, Bian Y, Luo J, Lu M, Xiong Y, Guo SY, Yin HY, Lin X, Li Q, Chang CCY, Chang TY, Li BL, Song BL. Cholesterol and fatty acids regulate cysteine ubiquitylation of ACAT2 through competitive oxidation. Nat Cell Biol 2017; 19:808-819. [PMID: 28604676 PMCID: PMC5518634 DOI: 10.1038/ncb3551] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 05/10/2017] [Indexed: 12/26/2022]
Abstract
Ubiquitin linkage to cysteine is an unconventional modification targeting protein for degradation. However, the physiological regulation of cysteine ubiquitylation is still mysterious. Here we found that ACAT2, a cellular enzyme converting cholesterol and fatty acid to cholesteryl esters, was ubiquitylated on Cys277 for degradation when the lipid level was low. gp78-Insigs catalysed Lys48-linked polyubiquitylation on this Cys277. A high concentration of cholesterol and fatty acid, however, induced cellular reactive oxygen species (ROS) that oxidized Cys277, resulting in ACAT2 stabilization and subsequently elevated cholesteryl esters. Furthermore, ACAT2 knockout mice were more susceptible to high-fat diet-associated insulin resistance. By contrast, expression of a constitutively stable form of ACAT2 (C277A) resulted in higher insulin sensitivity. Together, these data indicate that lipid-induced stabilization of ACAT2 ameliorates lipotoxicity from excessive cholesterol and fatty acid. This unconventional cysteine ubiquitylation of ACAT2 constitutes an important mechanism for sensing lipid-overload-induced ROS and fine-tuning lipid homeostasis.
Collapse
Affiliation(s)
- Yong-Jian Wang
- The State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai 200031, China
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou, Jiangsu, China
| | - Yan Bian
- The State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai 200031, China
| | - Jie Luo
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, the Institute for Advanced Studies, Wuhan University, Wuhan 430072, China
| | - Ming Lu
- The State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai 200031, China
| | - Ying Xiong
- The State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai 200031, China
| | - Shu-Yuan Guo
- Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Hui-Yong Yin
- Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Xu Lin
- Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Qin Li
- The State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai 200031, China
| | - Catherine CY Chang
- Department of Biochemistry, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA
| | - Ta-Yuan Chang
- Department of Biochemistry, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA
| | - Bo-Liang Li
- The State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai 200031, China
| | - Bao-Liang Song
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, the Institute for Advanced Studies, Wuhan University, Wuhan 430072, China
| |
Collapse
|
38
|
Abstract
Vitamin K is a cofactor for γ-glutamyl carboxylase, which catalyzes the posttranslational conversion of specific glutamyl residues to γ-carboxyglutamyl residues in a variety of vitamin K-dependent proteins (VKDPs) involved in blood coagulation, bone and cartilage metabolism, signal transduction, and cell proliferation. Despite the great advances in the genetic, structural, and functional studies of VKDPs as well as the enzymes identified as part of the vitamin K cycle which enable it to be repeatedly recycled within the cells, little is known of the identity and roles of key regulators of vitamin K metabolism in mammals and humans. This review focuses on new insights into the molecular mechanisms underlying the intestinal absorption and in vivo tissue conversion of vitamin K1 to menaquinone-4 (MK-4) with special emphasis on two major advances in the studies of intestinal vitamin K transporters in enterocytes and a tissue MK-4 biosynthetic enzyme UbiA prenyltransferase domain-containing protein 1 (UBIAD1), which participates in the in vivo conversion of a fraction of dietary vitamin K1 to MK-4 in mammals and humans, although it remains uncertain whether UBIAD1 functions as a key regulator of intracellular cholesterol metabolism, bladder and prostate tumor cell progression, vascular integrity, and protection from oxidative stress.
Collapse
|
39
|
Xie C, Gong XM, Luo J, Li BL, Song BL. AAV9-NPC1 significantly ameliorates Purkinje cell death and behavioral abnormalities in mouse NPC disease. J Lipid Res 2017; 58:512-518. [PMID: 28053186 PMCID: PMC5335581 DOI: 10.1194/jlr.m071274] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 12/15/2016] [Indexed: 12/27/2022] Open
Abstract
Niemann-Pick type C (NPC) disease is a fatal inherited neurodegenerative disorder caused by loss-of-function mutations in the NPC1 or NPC2 gene. There is no effective way to treat NPC disease. In this study, we used adeno-associated virus (AAV) serotype 9 (AAV9) to deliver a functional NPC1 gene systemically into NPC1-/- mice at postnatal day 4. One single AAV9-NPC1 injection resulted in robust NPC1 expression in various tissues, including brain, heart, and lung. Strikingly, AAV9-mediated NPC1 delivery significantly promoted Purkinje cell survival, restored locomotor activity and coordination, and increased the lifespan of NPC1-/- mice. Our work suggests that AAV-based gene therapy is a promising means to treat NPC disease.
Collapse
Affiliation(s)
- Chang Xie
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Institute for Advanced Studies, Wuhan University, Wuhan 430072, China
- Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Xue-Min Gong
- Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Jie Luo
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Institute for Advanced Studies, Wuhan University, Wuhan 430072, China
| | - Bo-Liang Li
- Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Bao-Liang Song
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Institute for Advanced Studies, Wuhan University, Wuhan 430072, China
| |
Collapse
|
40
|
Nghiem-Rao TH, Patel SB. Investigating Sitosterolemia to Understand Lipid Physiology. ACTA ACUST UNITED AC 2017; 8:649-658. [PMID: 29928317 DOI: 10.2217/clp.13.60] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The cholesterol molecule is at the center of the pathophysiology of many vascular diseases. Whole-body cholesterol pools are maintained by a balance of endogenous synthesis, dietary absorption and elimination from our bodies. While the cellular aspects of cholesterol metabolism received significant impetus from the seminal work of Goldstein and Brown investigating LDL receptor trafficking, how dietary cholesterol was absorbed and eliminated was relatively neglected. The identification of the molecular defect a rare human disorder, Sitosterolemia, led to elucidation of a key mechanism of how we regulate the excretory pathway in the liver and in the intestine. Two proteins, ABCG5 and ABCG8, constitute a heterodimeric transporter that facilitates the extrusion of sterols from the cell into the biliary lumen, with a preference for xenosterols. This mechanism explained how dietary xenosterols are prevented from accumulating in our bodies. In addition, this disease has also highlighted the potential harm of xenosterols; macrothrombocytopenia, liver disease and endocrine disruption are seen when xenosterols accumulate. Mouse models of this disease suggest that there are more dramatic alterations of physiology, suggesting that these highly conserved mechanisms have evolved to prevent these xenosterols from accumulating in our bodies.
Collapse
Affiliation(s)
| | - Shailendra B Patel
- Medical College of Wisconsin, and the Clement J. Zablocki Veterans Affairs Medical Center, Milwaukee, WI
| |
Collapse
|
41
|
Zhu J, Xu K, Zhang X, Cao J, Jia Z, Yang R, Ma C, Chen C, Zhang T, Yan Z. Studies on the regulation of lipid metabolism and its mechanism of the iridoids rich fraction in Valeriana jatamansi Jones. Biomed Pharmacother 2016; 84:1891-1898. [PMID: 27832992 DOI: 10.1016/j.biopha.2016.10.099] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2016] [Revised: 10/16/2016] [Accepted: 10/31/2016] [Indexed: 12/27/2022] Open
Abstract
Valeriana jatamansi Jones, a plant with heart-shaped leaves in the Valeriana genus of Valerianaceae, is widely used in Chinese folk medicine. Iridoid is an important constituent of V. jatamansi that contributes to the pharmacological efficacy of the herb. This study aims to investigate the regulation of lipid metabolism and its mechanism of the iridoids rich fraction in V. jatamansi (IRFV). A high fat diet was used to establish the hyperlipidemia rat model, with 2mg/kg/d of simvastatin as a positive control, fed with 7.5, 15, and 30mg/kg/d of IRFV for 20days to investigate the lipid regulation activity and mechanism of IRFV. Body weight, liver index, total cholesterol (TC), triglyceride (TG), low-density lipoprotein cholesterol (LDL-C), and high-density lipoprotein cholesterol (HDL-C) in both serum and liver, as well as total bile acid (TBA), aspartate aminotransferase (AST), and alanine aminotransferase (ALT) in serum were measured. The lipoprotein lipase (LPL) and hepatic lipase (HL) activities and the apoprotein A5 (ApoA5), peroxisome proliferator-activated receptor α (PPAR-α), sterol regulatory element-binding proteins (SREBP-1c), and liver X receptor α (LXR-α) protein expressions were observed. Liver pathology was described through hematoxylin-eosin (HE) staining. Compared with the model group, three different IRFV dosages can slow down the weight gain of rats, reduce the contents of TG, and increase the contents of HDL-C in serum. Low IRFV dosage can significantly reduce the AST and ALT contents in serum, liver index, and the TG contents in liver, enhance LPL activity. Medium IRFV dosage can significantly decrease the TG and LDL-C contents in liver. High IRFV dosage can significantly reduce LDL-C, TBA, AST, and ALT contents in serum, and enhance HL activity. Three different IRFV dosages can significantly increase the ApoA5 and PPAR-α protein expression and decrease the SREBP-1c protein expression. Furthermore, the LXR-α protein expression decreased in low- and high-dose groups. Liver tissue pathological observation showed that IRFV can improve cell degeneration to a certain extent. These results strongly suggest that IRFV play significant roles in regulating lipid metabolism, the mechanism may be related to the increased ApoA5 protein expression.
Collapse
Affiliation(s)
- Jiali Zhu
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, PR China
| | - Keke Xu
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, PR China
| | - Xuemei Zhang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, PR China
| | - Jiahong Cao
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, PR China
| | - Zhanrong Jia
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, PR China
| | - Ruocong Yang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, PR China
| | - Chaoying Ma
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, PR China
| | - Chang Chen
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, PR China
| | - Tiane Zhang
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine,Chengdu 611137, PR China.
| | - Zhiyong Yan
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, PR China.
| |
Collapse
|
42
|
Soayfane Z, Tercé F, Cantiello M, Robenek H, Nauze M, Bézirard V, Allart S, Payré B, Capilla F, Cartier C, Peres C, Al Saati T, Théodorou V, Nelson DW, Yen CLE, Collet X, Coméra C. Exposure to dietary lipid leads to rapid production of cytosolic lipid droplets near the brush border membrane. Nutr Metab (Lond) 2016; 13:48. [PMID: 27478484 PMCID: PMC4965885 DOI: 10.1186/s12986-016-0107-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 07/21/2016] [Indexed: 12/17/2022] Open
Abstract
Background Intestinal absorption of dietary lipids involves their hydrolysis in the lumen of proximal intestine as well as uptake, intracellular transport and re-assembly of hydrolyzed lipids in enterocytes, leading to the formation and secretion of the lipoproteins chylomicrons and HDL. In this study, we examined the potential involvement of cytosolic lipid droplets (CLD) whose function in the process of lipid absorption is poorly understood. Methods Intestinal lipid absorption was studied in mouse after gavage. Three populations of CLD were purified by density ultracentrifugations, as well as the brush border membranes, which were analyzed by western-blots. Immunofluorescent localization of membranes transporters or metabolic enzymes, as well as kinetics of CLD production, were also studied in intestine or Caco-2 cells. Results We isolated three populations of CLD (ranging from 15 to 1000 nm) which showed differential expression of the major lipid transporters scavenger receptor BI (SR-BI), cluster of differentiation 36 (CD-36), Niemann Pick C-like 1 (NPC1L1), and the ATP-binding cassette transporters ABCG5/G8 but also caveolin 2 and fatty acid binding proteins. The enzyme monoacylglycerol acyltransferase 2 (MGAT2) was identified in the brush border membrane (BBM) in addition to the endoplasmic reticulum, suggesting local synthesis of triglycerides and CLD at both places. Conclusions We show a very fast production of CLD by enterocytes associated with a transfer of apical constituents as lipid transporters. Our findings suggest that following their uptake by enterocytes, lipids can be partially metabolized at the BBM and packaged into CLD for their transportation to the ER. Electronic supplementary material The online version of this article (doi:10.1186/s12986-016-0107-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Zeina Soayfane
- Institut des Maladies Métaboliques et Cardiovasculaires - I2MC, UMR 1048, Institut National de la Santé et de la Recherche Médicale, Université Toulouse III Paul Sabatier, Toulouse, F-31000 France
| | - François Tercé
- Institut des Maladies Métaboliques et Cardiovasculaires - I2MC, UMR 1048, Institut National de la Santé et de la Recherche Médicale, Université Toulouse III Paul Sabatier, Toulouse, F-31000 France
| | - Michela Cantiello
- Institut des Maladies Métaboliques et Cardiovasculaires - I2MC, UMR 1048, Institut National de la Santé et de la Recherche Médicale, Université Toulouse III Paul Sabatier, Toulouse, F-31000 France
| | - Horst Robenek
- Leibniz-Institut für Arterioskleroseforschung, Universität Münster, Münster, Germany
| | - Michel Nauze
- Institut des Maladies Métaboliques et Cardiovasculaires - I2MC, UMR 1048, Institut National de la Santé et de la Recherche Médicale, Université Toulouse III Paul Sabatier, Toulouse, F-31000 France
| | - Valérie Bézirard
- UMR 1331 Toxalim, INRA, Université de Toulouse, ENVT, INP-Purpan, 180 chemin de Tournefeuille, BP 93173, 31027 Toulouse, cedex 3, France
| | - Sophie Allart
- INSERM UMR 1043 (INSERM/UPS/CNRS/USC Inra), CHU Purpan, Toulouse, France
| | - Bruno Payré
- CMEAB, Faculté de Médecine Rangueil, Toulouse, France
| | - Florence Capilla
- INSERM/UPS - US006/CREFRE, Service d'Histopathologie, CHU Purpan, Toulouse, France
| | - Christel Cartier
- UMR 1331 Toxalim, INRA, Université de Toulouse, ENVT, INP-Purpan, 180 chemin de Tournefeuille, BP 93173, 31027 Toulouse, cedex 3, France
| | - Christine Peres
- Institut des Maladies Métaboliques et Cardiovasculaires - I2MC, UMR 1048, Institut National de la Santé et de la Recherche Médicale, Université Toulouse III Paul Sabatier, Toulouse, F-31000 France
| | - Talal Al Saati
- INSERM/UPS - US006/CREFRE, Service d'Histopathologie, CHU Purpan, Toulouse, France
| | - Vassilia Théodorou
- UMR 1331 Toxalim, INRA, Université de Toulouse, ENVT, INP-Purpan, 180 chemin de Tournefeuille, BP 93173, 31027 Toulouse, cedex 3, France
| | - David W Nelson
- Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, WI USA
| | - Chi-Liang Eric Yen
- Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, WI USA
| | - Xavier Collet
- Institut des Maladies Métaboliques et Cardiovasculaires - I2MC, UMR 1048, Institut National de la Santé et de la Recherche Médicale, Université Toulouse III Paul Sabatier, Toulouse, F-31000 France
| | - Christine Coméra
- Institut des Maladies Métaboliques et Cardiovasculaires - I2MC, UMR 1048, Institut National de la Santé et de la Recherche Médicale, Université Toulouse III Paul Sabatier, Toulouse, F-31000 France.,UMR 1331 Toxalim, INRA, Université de Toulouse, ENVT, INP-Purpan, 180 chemin de Tournefeuille, BP 93173, 31027 Toulouse, cedex 3, France
| |
Collapse
|
43
|
Nakano T, Inoue I, Takenaka Y, Ono H, Katayama S, Awata T, Murakoshi T. Ezetimibe Promotes Brush Border Membrane-to-Lumen Cholesterol Efflux in the Small Intestine. PLoS One 2016; 11:e0152207. [PMID: 27023132 PMCID: PMC4811413 DOI: 10.1371/journal.pone.0152207] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Accepted: 03/10/2016] [Indexed: 11/19/2022] Open
Abstract
Ezetimibe inhibits Niemann-Pick C1-like 1 (NPC1L1), an apical membrane cholesterol transporter of enterocytes, thereby reduces intestinal cholesterol absorption. This treatment also increases extrahepatic reverse cholesterol transport via an undefined mechanism. To explore this, we employed a trans-intestinal cholesterol efflux (TICE) assay, which directly detects circulation-to-intestinal lumen 3H-cholesterol transit in a cannulated jejunal segment, and found an increase of TICE by 45%. To examine whether such increase in efflux occurs at the intestinal brush border membrane(BBM)-level, we performed luminal perfusion assays, similar to TICE but the jejunal wall was labelled with orally-given 3H-cholesterol, and determined elevated BBM-to-lumen cholesterol efflux by 3.5-fold with ezetimibe. Such increased efflux probably promotes circulation-to-lumen cholesterol transit eventually; thus increases TICE. Next, we wondered how inhibition of NPC1L1, an influx transporter, resulted in increased efflux. When we traced orally-given 3H-cholesterol in mice, we found that lumen-to-BBM 3H-cholesterol transit was rapid and less sensitive to ezetimibe treatment. Comparison of the efflux and fractional cholesterol absorption revealed an inverse correlation, indicating the efflux as an opposite-regulatory factor for cholesterol absorption efficiency and counteracting to the naturally-occurring rapid cholesterol influx to the BBM. These suggest that the ezetimibe-stimulated increased efflux is crucial in reducing cholesterol absorption. Ezetimibe-induced increase in cholesterol efflux was approximately 2.5-fold greater in mice having endogenous ATP-binding cassette G5/G8 heterodimer, the major sterol efflux transporter of enterocytes, than the knockout counterparts, suggesting that the heterodimer confers additional rapid BBM-to-lumen cholesterol efflux in response to NPC1L1 inhibition. The observed framework for intestinal cholesterol fluxes may provide ways to modulate the flux to dispose of endogenous cholesterol efficiently for therapeutic purposes.
Collapse
Affiliation(s)
- Takanari Nakano
- Department of Biochemistry, Faculty of Medicine, Saitama Medical University, Iruma-gun, Saitama, Japan
- * E-mail:
| | - Ikuo Inoue
- Department of Diabetes and Endocrinology, Faculty of Medicine, Saitama Medical University, Iruma-gun, Saitama, Japan
| | - Yasuhiro Takenaka
- Department of Diabetes and Endocrinology, Faculty of Medicine, Saitama Medical University, Iruma-gun, Saitama, Japan
| | - Hiraku Ono
- Department of Diabetes and Endocrinology, Faculty of Medicine, Saitama Medical University, Iruma-gun, Saitama, Japan
| | - Shigehiro Katayama
- Department of Diabetes and Endocrinology, Faculty of Medicine, Saitama Medical University, Iruma-gun, Saitama, Japan
| | - Takuya Awata
- Department of Diabetes, Endocrinology and Metabolism, International University of Health and Welfare Hospital, Nasushiobara-shi, Tochigi, Japan
| | - Takayuki Murakoshi
- Department of Biochemistry, Faculty of Medicine, Saitama Medical University, Iruma-gun, Saitama, Japan
| |
Collapse
|
44
|
Characterization of the NPC1L1 gene and proteome from an exceptional responder to ezetimibe. Atherosclerosis 2015; 246:78-86. [PMID: 26761771 DOI: 10.1016/j.atherosclerosis.2015.12.032] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Revised: 12/17/2015] [Accepted: 12/22/2015] [Indexed: 12/27/2022]
Abstract
BACKGROUND Strategies to reduce LDL-cholesterol involve reductions in cholesterol synthesis or absorption. We identified a familial hypercholesterolemia patient with an exceptional response to the cholesterol absorption inhibitor, ezetimibe. Niemann-Pick C 1-like 1 (NPC1L1) is the molecular target of ezetimibe. METHODS AND RESULTS Sequencing identified nucleotide changes predicted to change amino acids 52 (L52P), 300 (I300T) and 489 (S489G) in exceptional NPC1L1. In silico analyses identified increased stability and cholesterol binding affinity in L52P-NPC1L1 versus WT-NPC1L1. HEK293 cells overexpressing WT-NPC1L1 or NPC1L1 harboring amino acid changes singly or in combination (Comb-NPC1L1) had reduced cholesterol uptake in Comb-NPC1L1 when ezetimibe was present. Cholesterol uptake was reduced by ezetimibe in L52P-NPC1L1, I300T-NPC1L1, but increased in S489G-NPC1L1 overexpressing cells. Immunolocalization studies found preferential plasma membrane localization of mutant NPC1L1 independent of ezetimibe. Flotillin 1 and 2 expression was reduced and binding to Comb-NPC1L1 was reduced independent of ezetimibe exposure. Proteomic analyses identified increased association with proteins that modulate intermediate filament proteins in Comb-NPC1L1 versus WT-NPC1L1 treated with ezetimibe. CONCLUSION This is the first detailed analysis of the role of NPC1L1 mutations in an exceptional responder to ezetimibe. The results point to a complex set of events in which the combined mutations were shown to affect cholesterol uptake in the presence of ezetimibe. Proteomic analysis suggests that the exceptional response may also lie in the nature of interactions with cytosolic proteins.
Collapse
|
45
|
Giménez-Bastida JA, Zieliński H. Buckwheat as a Functional Food and Its Effects on Health. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2015; 63:7896-913. [PMID: 26270637 DOI: 10.1021/acs.jafc.5b02498] [Citation(s) in RCA: 167] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
Buckwheat (BW) is a gluten-free pseudocereal that belongs to the Polygonaceae family. BW grain is a highly nutritional food component that has been shown to provide a wide range of beneficial effects. Health benefits attributed to BW include plasma cholesterol level reduction, neuroprotection, anticancer, anti-inflammatory, antidiabetic effects, and improvement of hypertension conditions. In addition, BW has been reported to possess prebiotic and antioxidant activities. In vitro and animal studies suggest that BW's bioactive compounds, such as D-chiro-inositol (DCI), BW proteins (BWP), and BW flavonoids (mainly rutin and quercetin) may be partially responsible for the observed effects. The purpose of this paper is to review the recent research regarding the health benefits of BW, in vitro and in vivo, focusing on the specific role of its bioactive compounds and on the mechanisms by which these effects are exerted.
Collapse
Affiliation(s)
- Juan Antonio Giménez-Bastida
- Division of Food Science, Institute of Animal Reproduction and Food Research of the Polish Academy of Sciences , Tuwima 10, P.O. Box 55, 10-748 Olsztyn, Poland
| | - Henryk Zieliński
- Division of Food Science, Institute of Animal Reproduction and Food Research of the Polish Academy of Sciences , Tuwima 10, P.O. Box 55, 10-748 Olsztyn, Poland
| |
Collapse
|
46
|
Xie P, Zhu H, Jia L, Ma Y, Tang W, Wang Y, Xue B, Shi H, Yu L. Genetic demonstration of intestinal NPC1L1 as a major determinant of hepatic cholesterol and blood atherogenic lipoprotein levels. Atherosclerosis 2014; 237:609-17. [PMID: 25463095 DOI: 10.1016/j.atherosclerosis.2014.09.036] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2013] [Revised: 09/08/2014] [Accepted: 09/17/2014] [Indexed: 12/21/2022]
Abstract
OBJECTIVE The correlation between intestinal cholesterol absorption values and plasma low-density lipoprotein-cholesterol (LDL-C) levels remains controversial. Niemann-Pick-C1-Like 1 (NPC1L1) is essential for intestinal cholesterol absorption, and is the target of ezetimibe, a cholesterol absorption inhibitor. However, studies with NPC1L1 knockout mice or ezetimibe cannot definitively clarify this correlation because NPC1L1 expression is not restricted to intestine in humans and mice. In this study we sought to genetically address this issue. METHODS AND RESULTS We developed a mouse model that lacks endogenous (NPC1L1) and LDL receptor (LDLR) (DKO), but transgenically expresses human NPC1L1 in gastrointestinal tract only (DKO/L1(IntOnly) mice). Our novel model eliminated potential effects of non-intestinal NPC1L1 on cholesterol homeostasis. We found that human NPC1L1 was localized at the intestinal brush border membrane of DKO/L1(IntOnly) mice. Cholesterol feeding induced formation of NPC1L1-positive vesicles beneath this membrane in an ezetimibe-sensitive manner. Compared to DKO mice, DKO/L1(IntOnly) mice showed significant increases in cholesterol absorption and blood/hepatic/biliary cholesterol. Increased blood cholesterol was restricted to very low-density lipoprotein (VLDL) and LDL fractions, which was associated with increased secretion and plasma levels of apolipoproteins B100 and B48. Additionally, DKO/L1(IntOnly) mice displayed decreased fecal cholesterol excretion and hepatic/intestinal expression of cholesterologenic genes. Ezetimibe treatment virtually reversed all of the transgene-related phenotypes in DKO/L1(IntOnly) mice. CONCLUSION Our findings from DKO/L1(IntOnly) mice clearly demonstrate that NPC1L1-mediated cholesterol absorption is a major determinant of blood levels of apolipoprotein B-containing atherogenic lipoproteins, at least in mice.
Collapse
Affiliation(s)
- Ping Xie
- Department of Pathology Section on Lipid Sciences, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157, USA; Department of Biochemistry, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157, USA; Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, China
| | - Hongling Zhu
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD 20742, USA
| | - Lin Jia
- Department of Pathology Section on Lipid Sciences, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157, USA; Department of Biochemistry, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157, USA
| | - Yinyan Ma
- Department of Pathology Section on Lipid Sciences, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157, USA; Department of Biochemistry, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157, USA; Department of Animal and Avian Sciences, University of Maryland, College Park, MD 20742, USA
| | - Weiqing Tang
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD 20742, USA
| | - Youlin Wang
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD 20742, USA
| | - Bingzhong Xue
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA
| | - Hang Shi
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA
| | - Liqing Yu
- Department of Pathology Section on Lipid Sciences, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157, USA; Department of Biochemistry, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157, USA; Department of Animal and Avian Sciences, University of Maryland, College Park, MD 20742, USA.
| |
Collapse
|
47
|
Dražić T, Molčanov K, Sachdev V, Malnar M, Hećimović S, Patankar JV, Obrowsky S, Levak-Frank S, Habuš I, Kratky D. Novel amino-β-lactam derivatives as potent cholesterol absorption inhibitors. Eur J Med Chem 2014; 87:722-34. [PMID: 25305716 PMCID: PMC4237514 DOI: 10.1016/j.ejmech.2014.10.014] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Revised: 10/02/2014] [Accepted: 10/06/2014] [Indexed: 11/17/2022]
Abstract
Two new trans-(3R,4R)-amino-β-lactam derivatives and their diastereoisomeric mixtures were synthesized as ezetimibe bioisosteres and tested in in vitro and in vivo experiments as novel β-lactam cholesterol absorption inhibitors. Both compounds exhibited low cytotoxicity in MDCKII, hNPC1L1/MDCKII, and HepG2 cell lines and potent inhibitory effect in hNPC1L1/MDCKII cells. In addition, these compounds markedly reduced cholesterol absorption in mice, resulting in reduced cholesterol concentrations in plasma, liver, and intestine. We determined the crystal structure of one amino-β-lactam derivative to establish unambiguously both the absolute and relative configuration at the new stereogenic centre C17, which was assigned to be S. The pKa values for both compounds are 9.35, implying that the amino-β-lactam derivatives and their diastereoisomeric mixtures are in form of ammonium salt in blood and the intestine. The IC50 value for the diastereoisomeric mixture is 60 μM. In vivo, it efficiently inhibited cholesterol absorption comparable to ezetimibe.
Collapse
Affiliation(s)
- Tonko Dražić
- Ruđer Bošković Institute, Bijenička c. 54, HR-10002 Zagreb, Croatia.
| | | | - Vinay Sachdev
- Institute of Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Martina Malnar
- Ruđer Bošković Institute, Bijenička c. 54, HR-10002 Zagreb, Croatia
| | - Silva Hećimović
- Ruđer Bošković Institute, Bijenička c. 54, HR-10002 Zagreb, Croatia
| | - Jay V Patankar
- Institute of Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Sascha Obrowsky
- Institute of Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Sanja Levak-Frank
- Institute of Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Ivan Habuš
- Ruđer Bošković Institute, Bijenička c. 54, HR-10002 Zagreb, Croatia
| | - Dagmar Kratky
- Institute of Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria.
| |
Collapse
|
48
|
Malhotra P, Soni V, Kumar A, Anbazhagan AN, Dudeja A, Saksena S, Gill RK, Dudeja PK, Alrefai WA. Epigenetic modulation of intestinal cholesterol transporter Niemann-Pick C1-like 1 (NPC1L1) gene expression by DNA methylation. J Biol Chem 2014; 289:23132-23140. [PMID: 24904062 PMCID: PMC4132811 DOI: 10.1074/jbc.m113.546283] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2013] [Revised: 06/02/2014] [Indexed: 01/05/2023] Open
Abstract
Intestinal NPC1L1 transporter is essential for cholesterol absorption and the maintenance of cholesterol homeostasis in the body. NPC1L1 is differentially expressed along the gastrointestinal tract with very low levels in the colon as compared with the small intestine. This study was undertaken to examine whether DNA methylation was responsible for segment-specific expression of NPC1L1. Treatment of mice with 5-azacytidine (i.p.) resulted in a significant dose-dependent increase in NPC1L1 mRNA expression in the colon. The lack of expression of NPC1L1 in the normal colon was associated with high levels of methylation in the area flanking the 3-kb fragment upstream of the initiation site of the mouse NPC1L1 gene in mouse colon as analyzed by EpiTYPER® MassARRAY®. The high level of methylation in the colon was observed in specific CpG dinucleotides and was significantly decreased in response to 5-azacytidine. Similar to mouse NPC1L1, 5-azacytidine treatment also increased the level of human NPC1L1 mRNA expression in the intestinal HuTu-80 cell line in a dose- and time-dependent manner. Silencing the expression of DNA methyltransferase DNMT1, -2, -3A, and -3B alone by siRNA did not affect NPC1L1 expression in HuTu-80 cells. However, the simultaneous attenuation of DNMT1 and -3B expression caused a significant increase in NPC1L1 mRNA expression as compared with control. Also, in vitro methylation of the human NPC1L1 promoter significantly decreased NPC1L1 promoter activity in human intestinal Caco2 cells. In conclusion, our data demonstrated for the first time that DNA methylation in the promoter region of the NPC1L1 gene appears to be a major mechanism underlying differential expression of NPC1L1 along the length of the gastrointestinal tract.
Collapse
Affiliation(s)
- Pooja Malhotra
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois 60612
| | - Vinay Soni
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois 60612
| | - Anoop Kumar
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois 60612
| | - Arivarasu N Anbazhagan
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois 60612
| | - Amish Dudeja
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois 60612
| | - Seema Saksena
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois 60612
| | - Ravinder K Gill
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois 60612
| | - Pradeep K Dudeja
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois 60612; Jesse Brown Veterans Affairs Medical Center, University of Illinois at Chicago, Chicago, Illinois 60612
| | - Waddah A Alrefai
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois 60612; Jesse Brown Veterans Affairs Medical Center, University of Illinois at Chicago, Chicago, Illinois 60612.
| |
Collapse
|
49
|
The clathrin adaptor Numb regulates intestinal cholesterol absorption through dynamic interaction with NPC1L1. Nat Med 2013; 20:80-6. [PMID: 24336247 DOI: 10.1038/nm.3417] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2013] [Accepted: 10/30/2013] [Indexed: 11/08/2022]
Abstract
Hypercholesterolemia, typically due to excessive cholesterol uptake, is a major risk factor for cardiovascular disease, which is responsible for ∼50% of all deaths in developed societies. Although it has been shown that intestinal cholesterol absorption is mediated by vesicular endocytosis of the Niemann-Pick C1-like 1 (NPC1L1) protein, the mechanism of sterol-stimulated NPC1L1 internalization is still mysterious. Here, we identified an endocytic peptide signal, YVNXXF (where X stands for any amino acid), in the cytoplasmic C-terminal tail of NPC1L1. Cholesterol binding on the N-terminal domain of NPC1L1 released the YVNXXF-containing region of NPC1L1 from association with the plasma membrane and enabled Numb binding. We also found that Numb, a clathrin adaptor, specifically recognized this motif and recruited clathrin for internalization. Disrupting the NPC1L1-Numb interaction decreased cholesterol uptake. Ablation of Numb in mouse intestine significantly reduced dietary cholesterol absorption and plasma cholesterol level. Together, these data show that Numb is a pivotal protein for intestinal cholesterol absorption and may provide a therapeutic target for hypercholesterolemia.
Collapse
|
50
|
Hayashi H, Kawamura M. Lowering LDL cholesterol, but not raising LDL receptor activity, by ezetimibe. J Clin Lipidol 2013; 7:632-6. [DOI: 10.1016/j.jacl.2013.06.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2013] [Revised: 06/06/2013] [Accepted: 06/27/2013] [Indexed: 10/26/2022]
|