1
|
Valacchi G, Pecorelli A. Role of Scavenger Receptor B1 (SR-B1) in Improving Food Benefits for Human Health. Annu Rev Food Sci Technol 2025; 16:403-432. [PMID: 39899837 DOI: 10.1146/annurev-food-111523-121935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2025]
Abstract
Scavenger receptor class B member 1 (SR-B1) is a multiligand receptor with a broad range of functions spanning from the uptake of cholesteryl esters from high-density lipoproteins (HDLs) and transport of micronutrients such as fat-soluble vitamins and carotenoids across cell membranes to roles in tumor progression, pathogen recognition, and inflammatory responses. As a target of exposome factors such as environmental stressors and unhealthy lifestyle choices, as well as aging, dysregulated expression and activity of SR-B1 can negatively impact human health. Intriguingly, not only is SR-B1 a major determinant of nutrient homeostasis and, hence, metabolic health status, but these same nutrients and some phytochemicals have also demonstrated their ability to modulate SR-B1. Therefore, an integrated approach that, taking into account human health, nutrition, and food technology sciences, aims to produce foods with health-promoting effects should take advantage of the multifaceted properties of SR-B1. Improved functional foods and novel nanoparticle-based delivery systems, rich in nutrients and phytochemicals, with precise targeting to SR-B1 in specific tissues or structures could represent a strategic advance to improve human health and promote well-being.
Collapse
Affiliation(s)
- Giuseppe Valacchi
- Department of Animal Science, North Carolina State University, Kannapolis, North Carolina, USA
- Department of Environmental and Prevention Sciences, University of Ferrara, Ferrara, Italy;
- Department of Food and Nutrition, Kyung Hee University, Seoul, South Korea
| | - Alessandra Pecorelli
- Department of Environmental and Prevention Sciences, University of Ferrara, Ferrara, Italy;
- Department of Food, Bioprocessing and Nutrition Sciences, North Carolina State University, Kannapolis, North Carolina, USA
| |
Collapse
|
2
|
Arrari F, Ortiz-Flores RM, Lhamyani S, Garcia-Fuentes E, Jabri MA, Sebai H, Bermudez-Silva FJ. Protective Effects of Spirulina Against Lipid Micelles and Lipopolysaccharide-Induced Intestinal Epithelium Disruption in Caco-2 Cells: In Silico Molecular Docking Analysis of Phycocyanobilin. Nutrients 2024; 16:4074. [PMID: 39683467 DOI: 10.3390/nu16234074] [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: 10/25/2024] [Revised: 11/15/2024] [Accepted: 11/20/2024] [Indexed: 12/18/2024] Open
Abstract
Damage to intestinal epithelial cells is present in obesity and other diseases because of inflammatory and oxidative processes. This damage compromises the gastrointestinal barrier, killing enterocytes, altering intestinal permeability, and eliciting abnormal immune responses that promote chronic inflammation. Recent evidence shows that spirulina is a potent natural agent with antioxidant and anti-inflammatory properties. OBJECTIVES This study was conducted to evaluate the effect of spirulina aqueous extract (SPAE) on the alterations of the intestinal epithelium induced by lipid micelles (LMs) and/or inflammation induced by lipopolysaccharides (LPSs) in the Caco-2 cell line. METHODS In the current research, we assessed the protective actions of SPAE against cytotoxicity, oxidative stress, inflammation, and epithelial barrier perturbation by using an in vitro model, the intestinal Caco-2 cells, treated with LPSs and/or LMs. We also performed an in silico molecular docking analysis with spirulina's bioactive compound, phycocyanobilin. RESULTS Our results showed that SPAE has no cytotoxic effect on Caco-2 cells. On the contrary, it improved cell viability and exhibited anti-inflammatory and antioxidant actions. SPAE also protected against endoplasmic reticulum stress and tight junction proteins, thus improving the epithelial barrier. The in silico study revealed a strong binding affinity of the phycocyanobilin compound with human SOD and NADPH oxidase and a good binding affinity towards COX-2 and iNOS. CONCLUSIONS Taken together, these findings demonstrate the beneficial actions of SPAE on Caco-2 cells, suggesting it may be useful in preserving the epithelial intestinal barrier in human conditions involving oxidative stress and inflammation such as obesity.
Collapse
Affiliation(s)
- Fatma Arrari
- Laboratory of Functional Physiology and Valorization of Bio-Resources, Higher Institute of Biotechnology of Beja, University of Jendouba, Beja 9000, Tunisia
| | - Rodolfo-Matias Ortiz-Flores
- Centro de Investigacion Biomedica en Red de Diabetes y Enfermedades Metabolicas Asociadas (CIBERDEM), Instituto de Investigacion Biomedica de Malaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Hospital Regional Universitario de Malaga, UGC Endocrinología y Nutricion, 29009 Malaga, Spain
| | - Said Lhamyani
- Centro de Investigacion Biomedica en Red de Diabetes y Enfermedades Metabolicas Asociadas (CIBERDEM), Instituto de Investigacion Biomedica de Malaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Hospital Regional Universitario de Malaga, UGC Endocrinología y Nutricion, 29009 Malaga, Spain
| | - Eduardo Garcia-Fuentes
- Centro de Investigacion Biomedica en Red de Enfermedades Hepaticas y Digestivas (CIBERehd), Instituto de Investigacion Biomedica de Malaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Hospital Universitario Virgen de la Victoria, UGC de Aparato Digestivo, 29010 Malaga, Spain
| | - Mohamed-Amine Jabri
- Laboratory of Functional Physiology and Valorization of Bio-Resources, Higher Institute of Biotechnology of Beja, University of Jendouba, Beja 9000, Tunisia
| | - Hichem Sebai
- Laboratory of Functional Physiology and Valorization of Bio-Resources, Higher Institute of Biotechnology of Beja, University of Jendouba, Beja 9000, Tunisia
| | - Francisco-Javier Bermudez-Silva
- Centro de Investigacion Biomedica en Red de Diabetes y Enfermedades Metabolicas Asociadas (CIBERDEM), Instituto de Investigacion Biomedica de Malaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Hospital Regional Universitario de Malaga, UGC Endocrinología y Nutricion, 29009 Malaga, Spain
| |
Collapse
|
3
|
Reboul E. Proteins involved in fat-soluble vitamin and carotenoid transport across the intestinal cells: New insights from the past decade. Prog Lipid Res 2023; 89:101208. [PMID: 36493998 DOI: 10.1016/j.plipres.2022.101208] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 11/28/2022] [Accepted: 11/29/2022] [Indexed: 12/12/2022]
Abstract
It is now well established that vitamins D, E, and K and carotenoids are not absorbed solely through passive diffusion. Broad-specificity membrane transporters such as SR-BI (scavenger receptor class B type I), CD36 (CD36 molecule), NPC1L1 (Niemann Pick C1-like 1) or ABCA1 (ATP-binding cassette A1) are involved in the uptake of these micronutrients from the lumen to the enterocyte cytosol and in their secretion into the bloodstream. Recently, the existence of efflux pathways from the enterocyte back to the lumen or from the bloodstream to the lumen, involving ABCB1 (P-glycoprotein/MDR1) or the ABCG5/ABCG8 complex, has also been evidenced for vitamins D and K. Surprisingly, no membrane proteins have been involved in dietary vitamin A uptake so far. After an overview of the metabolism of fat-soluble vitamins and carotenoids along the gastrointestinal tract (from the mouth to the colon where interactions with microbiota may occur), a focus is placed on the identified and candidate proteins participating in the apical uptake, intracellular transport, basolateral secretion and efflux back to the lumen of fat-soluble vitamins and carotenoids in enterocytes. This review also highlights the mechanisms that remain to be identified to fully unravel the pathways involved in fat-soluble vitamin and carotenoid intestinal absorption.
Collapse
|
4
|
Axmann M, Plochberger B, Mikula M, Weber F, Strobl WM, Stangl H. Plasma Membrane Lipids: An Important Binding Site for All Lipoprotein Classes. MEMBRANES 2021; 11:membranes11110882. [PMID: 34832111 PMCID: PMC8622984 DOI: 10.3390/membranes11110882] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 11/12/2021] [Accepted: 11/12/2021] [Indexed: 12/23/2022]
Abstract
Cholesterol is one of the main constituents of plasma membranes; thus, its supply is of utmost importance. This review covers the known mechanisms of cholesterol transfer from circulating lipoprotein particles to the plasma membrane, and vice versa. To achieve homeostasis, the human body utilizes cellular de novo synthesis and extracellular transport particles for supply of cholesterol and other lipids via the blood stream. These lipoprotein particles can be classified according to their density: chylomicrons, very low, low, and high-density lipoprotein (VLDL, LDL, and HDL, respectively). They deliver and receive their lipid loads, most importantly cholesterol, to and from cells by several redundant routes. Defects in one of these pathways (e.g., due to mutations in receptors) usually are not immediately fatal. Several redundant pathways, at least temporarily, compensate for the loss of one or more of them, but the defects trigger systemic diseases, such as atherosclerosis later on. Recently, intracellular membrane–membrane contact sites were shown to be involved in intracellular cholesterol transfer and the plasma membrane itself has been proposed to act as a binding site for lipoprotein-mediated cargo unloading.
Collapse
Affiliation(s)
- Markus Axmann
- School of Medical Engineering and Applied Social Sciences, University of Applied Sciences Upper Austria, Garnisonstrasse 21, 4020 Linz, Austria; (M.A.); (B.P.); (F.W.)
| | - Birgit Plochberger
- School of Medical Engineering and Applied Social Sciences, University of Applied Sciences Upper Austria, Garnisonstrasse 21, 4020 Linz, Austria; (M.A.); (B.P.); (F.W.)
| | - Mario Mikula
- Center for Pathobiochemistry and Genetics, Institute for Medical Genetics, Medical University of Vienna, Währingerstrasse 10, 1090 Vienna, Austria;
| | - Florian Weber
- School of Medical Engineering and Applied Social Sciences, University of Applied Sciences Upper Austria, Garnisonstrasse 21, 4020 Linz, Austria; (M.A.); (B.P.); (F.W.)
| | - Witta Monika Strobl
- Center for Pathobiochemistry and Genetics, Institute for Medical Chemistry, Medical University of Vienna, Währingerstrasse 10, 1090 Vienna, Austria;
| | - Herbert Stangl
- Center for Pathobiochemistry and Genetics, Institute for Medical Chemistry, Medical University of Vienna, Währingerstrasse 10, 1090 Vienna, Austria;
- Correspondence:
| |
Collapse
|
5
|
Zembroski AS, Xiao C, Buhman KK. The Roles of Cytoplasmic Lipid Droplets in Modulating Intestinal Uptake of Dietary Fat. Annu Rev Nutr 2021; 41:79-104. [PMID: 34283920 DOI: 10.1146/annurev-nutr-110320-013657] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Dietary fat absorption is required for health but also contributes to hyperlipidemia and metabolic disease when dysregulated. One step in the process of dietary fat absorption is the formation of cytoplasmic lipid droplets (CLDs) in small intestinal enterocytes; these CLDs serve as dynamic triacylglycerol storage organelles that influence the rate at which dietary fat is absorbed. Recent studies have uncovered novel factors regulating enterocyte CLD metabolism that in turn influence the absorption of dietary fat. These include peroxisome proliferator-activated receptor α activation, compartmentalization of different lipid pools, the gut microbiome, liver X receptor and farnesoid X receptor activation, obesity, and physiological factors stimulating CLD mobilization. Understanding how enterocyte CLD metabolism is regulated is key in modulating the absorption of dietary fat in the prevention of hyperlipidemia and its associated metabolic disorders. Expected final online publication date for the Annual Review of Nutrition, Volume 41 is September 2021. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Alyssa S Zembroski
- Department of Nutrition Science, Purdue University, West Lafayette, Indiana 47907, USA;
| | - Changting Xiao
- Department of Anatomy, Physiology and Pharmacology, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5E5, Canada
| | - Kimberly K Buhman
- Department of Nutrition Science, Purdue University, West Lafayette, Indiana 47907, USA;
| |
Collapse
|
6
|
Postal BG, Aguanno D, Thenet S, Carrière V. Rapid Evaluation of Intestinal Paracellular Permeability Using the Human Enterocytic-Like Caco-2/TC7 Cell Line. Methods Mol Biol 2021; 2367:13-26. [PMID: 33730353 DOI: 10.1007/7651_2021_366] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Paracellular permeability of the intestinal epithelium is a feature of the intestinal barrier, which plays an important role in the physiology of gut and the whole organism. Intestinal paracellular permeability is controlled by complex processes and is involved in the passage of ions and fluids (called pore pathway) and macromolecules (called leak pathway) through tight junctions, which seal the intercellular space. Impairment of intestinal paracellular permeability is associated with several diseases. The identification of a defect in intestinal paracellular permeability may help to understand the implication of gut barrier as a cause or a consequence in human pathology. Here we describe two complementary methods to evaluate alteration of paracellular permeability in cell culture, using the human intestinal cell line Caco-2 and its clone Caco-2/TC7.
Collapse
Affiliation(s)
- Bárbara Graziela Postal
- Centre de Recherche sur l'Inflammation, INSERM UMR1149, Université de Paris, Paris, France
- Biology and Genetics of Bacterial Cell Wall Unit, Pasteur Institute, Paris, France
| | - Doriane Aguanno
- Centre de Recherche Saint-Antoine, INSERM UMRS 938, Sorbonne Université, INSERM, Paris, France
| | - Sophie Thenet
- Centre de Recherche Saint-Antoine, INSERM UMRS 938, Sorbonne Université, INSERM, Paris, France
- EPHE, PSL University, Paris, France
- Center for Microbiome Medicine (PaCeMM) FHU, AP-HP, Paris, Ile-de-France, France
| | - Véronique Carrière
- Centre de Recherche Saint-Antoine, INSERM UMRS 938, Sorbonne Université, INSERM, Paris, France.
- Center for Microbiome Medicine (PaCeMM) FHU, AP-HP, Paris, Ile-de-France, France.
| |
Collapse
|
7
|
Postal BG, Ghezzal S, Aguanno D, André S, Garbin K, Genser L, Brot-Laroche E, Poitou C, Soula H, Leturque A, Clément K, Carrière V. AhR activation defends gut barrier integrity against damage occurring in obesity. Mol Metab 2020; 39:101007. [PMID: 32360426 PMCID: PMC7267728 DOI: 10.1016/j.molmet.2020.101007] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 04/03/2020] [Accepted: 04/22/2020] [Indexed: 12/24/2022] Open
Abstract
Objective Obesity is characterized by systemic and low-grade tissue inflammation. In the intestine, alteration of the intestinal barrier and accumulation of inflammatory cells in the epithelium are important contributors of gut inflammation. Recent studies demonstrated the role of the aryl hydrocarbon receptor (AhR) in the maintenance of immune cells at mucosal barrier sites. A wide range of ligands of external and local origin can activate this receptor. We studied the causal relationship between AhR activation and gut inflammation in obesity. Methods Jejunum samples from subjects with normal weight and severe obesity were phenotyped according to T lymphocyte infiltration in the epithelium from lamina propria and assayed for the mRNA level of AhR target genes. The effect of an AhR agonist was studied in mice and Caco-2/TC7 cells. AhR target gene expression, permeability to small molecules and ions, and location of cell-cell junction proteins were recorded under conditions of altered intestinal permeability. Results We showed that a low AhR tone correlated with a high inflammatory score in the intestinal epithelium in severe human obesity. Moreover, AhR activation protected junctional complexes in the intestinal epithelium in mice challenged by an oral lipid load. AhR ligands prevented chemically induced damage to barrier integrity and cytokine expression in Caco-2/TC7 cells. The PKC and p38MAPK signaling pathways were involved in this AhR action. Conclusions The results of these series of human, mouse, and cell culture experiments demonstrate the protective effect of AhR activation in the intestine targeting particularly tight junctions and cytokine expression. We propose that AhR constitutes a valuable target to protect intestinal functions in metabolic diseases, which can be achieved in the future via food or drug ligands. Intestinal inflammation in obese subjects correlates with low AhR activity. AhR activation protects intestinal epithelium from damages. AhR activation prevents cytokine induction in response to damages. Protective role of AhR activation targets junctional complexes. Protein kinases-dependent signaling pathways are involved in AhR action.
Collapse
Affiliation(s)
- Bárbara G Postal
- Cordeliers Research Center, Sorbonne University, Paris Dauphine University 05, INSERM, CNRS, F-75006, Paris, France; Saint-Antoine Research Center, Sorbonne University, INSERM, F-75012, Paris, France
| | - Sara Ghezzal
- Cordeliers Research Center, Sorbonne University, Paris Dauphine University 05, INSERM, CNRS, F-75006, Paris, France
| | - Doriane Aguanno
- Cordeliers Research Center, Sorbonne University, Paris Dauphine University 05, INSERM, CNRS, F-75006, Paris, France; Saint-Antoine Research Center, Sorbonne University, INSERM, F-75012, Paris, France; EPHE, PSL Research University, F-75006, Paris, France
| | - Sébastien André
- Sorbonne University, INSERM, NutriOmics Research Unit Paris, F-75013, France
| | - Kevin Garbin
- CHIC Platform of Cordeliers Research Center, Sorbonne University, UPD Univ Paris 05, INSERM, F-75006, Paris, France
| | - Laurent Genser
- Sorbonne University, INSERM, NutriOmics Research Unit Paris, F-75013, France
| | - Edith Brot-Laroche
- Cordeliers Research Center, Sorbonne University, Paris Dauphine University 05, INSERM, CNRS, F-75006, Paris, France
| | - Christine Poitou
- Sorbonne University, INSERM, NutriOmics Research Unit Paris, F-75013, France; Assistance Publique-Hôpitaux de Paris, Pitié-Salpêtrière Hospital, Nutrition Department, CRNH Ile de France, F-75013, Paris, France
| | - Hédi Soula
- Sorbonne University, INSERM, NutriOmics Research Unit Paris, F-75013, France
| | - Armelle Leturque
- Cordeliers Research Center, Sorbonne University, Paris Dauphine University 05, INSERM, CNRS, F-75006, Paris, France; Sorbonne University, INSERM, NutriOmics Research Unit Paris, F-75013, France
| | - Karine Clément
- Sorbonne University, INSERM, NutriOmics Research Unit Paris, F-75013, France; Assistance Publique-Hôpitaux de Paris, Pitié-Salpêtrière Hospital, Nutrition Department, CRNH Ile de France, F-75013, Paris, France
| | - Véronique Carrière
- Cordeliers Research Center, Sorbonne University, Paris Dauphine University 05, INSERM, CNRS, F-75006, Paris, France; Saint-Antoine Research Center, Sorbonne University, INSERM, F-75012, Paris, France.
| |
Collapse
|
8
|
Hiebl V, Schachner D, Ladurner A, Heiss EH, Stangl H, Dirsch VM. Caco-2 Cells for Measuring Intestinal Cholesterol Transport - Possibilities and Limitations. Biol Proced Online 2020; 22:7. [PMID: 32308567 PMCID: PMC7149936 DOI: 10.1186/s12575-020-00120-w] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 04/01/2020] [Indexed: 12/19/2022] Open
Abstract
Background The human Caco-2 cell line is a common in vitro model of the intestinal epithelial barrier. As the intestine is a major interface in cholesterol turnover and represents a non-biliary pathway for cholesterol excretion, Caco-2 cells are also a valuable model for studying cholesterol homeostasis, including cholesterol uptake and efflux. Currently available protocols are, however, either sketchy or not consistent among different laboratories. Our aim was therefore to generate a collection of optimized protocols, considering the different approaches of the different laboratories and to highlight possibilities and limitations of measuring cholesterol transport with this cell line. Results We developed comprehensive and quality-controlled protocols for the cultivation of Caco-2 cells on filter inserts in a single tight monolayer. A cholesterol uptake as well as a cholesterol efflux assay is described in detail, including suitable positive controls. We further show that Caco-2 cells can be efficiently transfected for luciferase reporter gene assays in order to determine nuclear receptor activation, main transcriptional regulators of cholesterol transporters (ABCA1, ABCB1, ABCG5/8, NPC1L1). Detection of protein and mRNA levels of cholesterol transporters in cells grown on filter inserts can pose challenges for which we highlight essential steps and alternative approaches for consideration. A protocol for viability assays with cells differentiated on filter inserts is provided for the first time. Conclusions The Caco-2 cell line is widely used in the scientific community as model for the intestinal epithelium, although with highly divergent protocols. The herein provided information and protocols can be a common basis for researchers intending to use Caco-2 cells in the context of cellular cholesterol homeostasis.
Collapse
Affiliation(s)
- Verena Hiebl
- 1Department of Pharmacognosy, University of Vienna, Althanstrasse 14, 1090 Vienna, Austria
| | - Daniel Schachner
- 1Department of Pharmacognosy, University of Vienna, Althanstrasse 14, 1090 Vienna, Austria
| | - Angela Ladurner
- 1Department of Pharmacognosy, University of Vienna, Althanstrasse 14, 1090 Vienna, Austria
| | - Elke H Heiss
- 1Department of Pharmacognosy, University of Vienna, Althanstrasse 14, 1090 Vienna, Austria
| | - Herbert Stangl
- 2Institute of Medical Chemistry, Center for Pathobiochemistry and Genetics, Medical University of Vienna, Vienna, Austria
| | - Verena M Dirsch
- 1Department of Pharmacognosy, University of Vienna, Althanstrasse 14, 1090 Vienna, Austria
| |
Collapse
|
9
|
Palmitic acid damages gut epithelium integrity and initiates inflammatory cytokine production. Biochim Biophys Acta Mol Cell Biol Lipids 2020; 1865:158530. [DOI: 10.1016/j.bbalip.2019.158530] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 07/16/2019] [Accepted: 09/13/2019] [Indexed: 02/06/2023]
|
10
|
Cedó L, Farràs M, Lee-Rueckert M, Escolà-Gil JC. Molecular Insights into the Mechanisms Underlying the Cholesterol- Lowering Effects of Phytosterols. Curr Med Chem 2019; 26:6704-6723. [DOI: 10.2174/0929867326666190822154701] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 01/18/2019] [Accepted: 02/22/2019] [Indexed: 12/11/2022]
Abstract
Dietary phytosterols, which comprise plant sterols and stanols, reduce plasma Low-Density Lipoprotein-Cholesterol (LDL-C) levels when given 2 g/day. Since this dose has not been reported to cause health-related side effects in long-term human studies, food products containing these plant compounds are used as potential therapeutic dietary options to reduce LDL-C and cardiovascular disease risk. Several mechanisms have been proposed to explain the cholesterol-lowering action of phytosterols. They may compete with dietary and biliary cholesterol for micellar solubilization in the intestinal lumen, impairing intestinal cholesterol absorption. Recent evidence indicates that phytosterols may also regulate other pathways. Impaired intestinal cholesterol absorption is usually associated with reduced cholesterol transport to the liver, which may reduce the incorporation of cholesterol into Very-Low- Density Lipoprotein (VLDL) particles, thereby lowering the rate of VLDL assembly and secretion. Impaired liver VLDL production may reduce the rate of LDL production. On the other hand, significant evidence supports a role for plant sterols in the Transintestinal Cholesterol Excretion (TICE) pathway, although the exact mechanisms by which they promote the flow of cholesterol from the blood to enterocytes and the intestinal lumen remains unknown. Dietary phytosterols may also alter the conversion of bile acids into secondary bile acids, and may lower the bile acid hydrophobic/hydrophilic ratio, thereby reducing intestinal cholesterol absorption. This article reviews the progress to date in research on the molecular mechanisms underlying the cholesterol-lowering effects of phytosterols.
Collapse
Affiliation(s)
- Lídia Cedó
- Institut d'Investigacions Biomediques (IIB) Sant Pau, Barcelona, Spain
| | - Marta Farràs
- Integrative Systems Medicine and Digestive Disease Division, Department of Surgery and Cancer, Imperial College London, London, United Kingdom
| | | | | |
Collapse
|
11
|
Bakker GJ, Schnitzler JG, Bekkering S, de Clercq NC, Koopen AM, Hartstra AV, Meessen ECE, Scheithauer TP, Winkelmeijer M, Dallinga‐Thie GM, Cani PD, Kemper EM, Soeters MR, Kroon J, Groen AK, van Raalte DH, Herrema H, Nieuwdorp M. Oral vancomycin treatment does not alter markers of postprandial inflammation in lean and obese subjects. Physiol Rep 2019; 7:e14199. [PMID: 31423751 PMCID: PMC6698488 DOI: 10.14814/phy2.14199] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 07/14/2019] [Indexed: 12/30/2022] Open
Abstract
Intake of a high-fat meal induces a systemic inflammatory response in the postprandial which is augmented in obese subjects. However, the underlying mechanisms of this response have not been fully elucidated. We aimed to assess the effect of gut microbiota modulation on postprandial inflammatory response in lean and obese subjects. Ten lean and ten obese subjects with metabolic syndrome received oral vancomycin 500 mg four times per day for 7 days. Oral high-fat meal tests (50 g fat/m2 body surface area) were performed before and after vancomycin intervention. Gut microbiota composition, leukocyte counts, plasma lipopolysaccharides (LPS), LPS-binding protein (LBP), IL-6 and MCP-1 concentrations and monocyte CCR2 and cytokine expression were determined before and after the high-fat meal. Oral vancomycin treatment resulted in profound changes in gut microbiota composition and significantly decreased bacterial diversity in both groups (phylogenetic diversity pre- versus post-intervention: lean, 56.9 ± 7.8 vs. 21.4 ± 6.6, P < 0.001; obese, 53.9 ± 7.8 vs. 21.0 ± 5.9, P < 0.001). After intervention, fasting plasma LPS significantly increased (lean, median [IQR] 0.81 [0.63-1.45] EU/mL vs. 2.23 [1.33-3.83] EU/mL, P = 0.017; obese, median [IQR] 0.76 [0.45-1.03] EU/mL vs. 1.44 [1.11-4.24], P = 0.014). However, postprandial increases in leukocytes and plasma LPS were unaffected by vancomycin in both groups. Moreover, we found no changes in plasma LBP, IL-6 and MCP-1 or in monocyte CCR2 expression. Despite major vancomycin-induced disruption of the gut microbiota and increased fasting plasma LPS, the postprandial inflammatory phenotype in lean and obese subjects was unaffected in this study.
Collapse
Affiliation(s)
- Guido J. Bakker
- Department of Vascular MedicineAmsterdam UMC, Location AMC at University of AmsterdamAmsterdamThe Netherlands
| | - Johan G. Schnitzler
- Department of Experimental Vascular MedicineAmsterdam UMC, Location AMC at University of AmsterdamAmsterdamThe Netherlands
| | - Siroon Bekkering
- Department of Experimental Internal MedicineRadboud University Medical CentreNijmegenThe Netherlands
| | - Nicolien C. de Clercq
- Department of Vascular MedicineAmsterdam UMC, Location AMC at University of AmsterdamAmsterdamThe Netherlands
| | - Annefleur M. Koopen
- Department of Vascular MedicineAmsterdam UMC, Location AMC at University of AmsterdamAmsterdamThe Netherlands
| | - Annick V. Hartstra
- Department of Vascular MedicineAmsterdam UMC, Location AMC at University of AmsterdamAmsterdamThe Netherlands
| | - Emma C. E. Meessen
- Department of Endocrinology and MetabolismAmsterdam UMC, Location AMC at University of AmsterdamAmsterdamThe Netherlands
| | - Torsten P. Scheithauer
- Department of Internal Medicine, Diabetes CenterAmsterdam UMC, Location VUMC at Vrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Maaike Winkelmeijer
- Department of Experimental Vascular MedicineAmsterdam UMC, Location AMC at University of AmsterdamAmsterdamThe Netherlands
| | - Geesje M. Dallinga‐Thie
- Department of Experimental Vascular MedicineAmsterdam UMC, Location AMC at University of AmsterdamAmsterdamThe Netherlands
| | - Patrice D. Cani
- WELBIO – Walloon Excellence in Life Sciences and Biotechnology, Metabolism and NutritionLouvain Drug Research Institute, Université Catholique de LouvainBrusselsBelgium
| | - Elles Marleen Kemper
- Department of Clinical PharmacyAmsterdam UMC, Location AMC at University of AmsterdamAmsterdamThe Netherlands
| | - Maarten R. Soeters
- Department of Endocrinology and MetabolismAmsterdam UMC, Location AMC at University of AmsterdamAmsterdamThe Netherlands
| | - Jeffrey Kroon
- Department of Vascular MedicineAmsterdam UMC, Location AMC at University of AmsterdamAmsterdamThe Netherlands
- Department of Experimental Vascular MedicineAmsterdam UMC, Location AMC at University of AmsterdamAmsterdamThe Netherlands
| | - Albert K. Groen
- Department of Experimental Vascular MedicineAmsterdam UMC, Location AMC at University of AmsterdamAmsterdamThe Netherlands
| | - Daniël H. van Raalte
- Department of Internal Medicine, Diabetes CenterAmsterdam UMC, Location VUMC at Vrije Universiteit AmsterdamAmsterdamThe Netherlands
- Amsterdam UMC, ICar at Vrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Hilde Herrema
- Department of Experimental Vascular MedicineAmsterdam UMC, Location AMC at University of AmsterdamAmsterdamThe Netherlands
| | - Max Nieuwdorp
- Department of Vascular MedicineAmsterdam UMC, Location AMC at University of AmsterdamAmsterdamThe Netherlands
- Department of Experimental Vascular MedicineAmsterdam UMC, Location AMC at University of AmsterdamAmsterdamThe Netherlands
- Department of Internal Medicine, Diabetes CenterAmsterdam UMC, Location VUMC at Vrije Universiteit AmsterdamAmsterdamThe Netherlands
- Amsterdam UMC, ICar at Vrije Universiteit AmsterdamAmsterdamThe Netherlands
- Department of Molecular and Clinical Medicine, Wallenberg LaboratorySahlgrenska Academy, University of GothenburgGothenburgSweden
| |
Collapse
|
12
|
Fuentes M, Santander N, Cortés V. Insulin increases cholesterol uptake, lipid droplet content, and apolipoprotein B secretion in CaCo-2 cells by upregulating SR-BI via a PI3K, AKT, and mTOR-dependent pathway. J Cell Biochem 2019; 120:1550-1559. [PMID: 30278109 DOI: 10.1002/jcb.27410] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 07/10/2018] [Indexed: 01/24/2023]
Abstract
The actions of insulin on intestinal cholesterol absorption and lipoprotein secretion are not well understood. Herein, we determined the effects of insulin on the levels of cholesterol transporter scavenger receptor, class B, type I (SR-BI), cellular cholesterol uptake, intracellular lipid accumulation, and lipoprotein secretion in a cellular model of human intestinal epithelium. METHODS CaCo-2 cells were cultured to postconfluency in Transwell filters and stimulated with glucose (25 mM) in the presence or absence of insulin (100 nM) at their basolateral surface. SR-BI mRNA and protein levels were quantified by quantitative reverse transcription-PCR and immunoblot, respectively. Polarized localization of SR-BI was determined by cell surface proteins biotinylation and streptavidin precipitation. Activities of PI3K, AKT, mTOR, and SR-BI were pharmacologically antagonized. Cholesterol uptake was assessed by NBD-cholesterol incorporation. Apolipoprotein (apo) B concentration was quantified by ELISA. Subcellular localization of neutral lipids (BODIPY) and SR-BI (immunofluorescence) was determined by confocal microscopy. RESULTS In polarized CaCo-2 cells, insulin increased SR-BI at the mRNA and protein levels. SR-BI was exclusively present at apical cell surface, as indicated by biotinylation and confocal microscopy analysis. Glucose did not modify SR-BI abundance or subcellular localization. Effects of insulin on SR-BI levels were abrogated by PI3K, AKT, or mTOR pharmacological antagonism. Cholesterol uptake, neutral lipid abundance, and apo B secretion were increased by insulin in CaCo-2 cells, and these effects were prevented by SR-BI pharmacological antagonism with block lipid transport-1. CONCLUSIONS insulin promotes cholesterol uptake, intracellular lipid store, and apo B-containing lipoproteins secretion by SR-BI-dependent mechanisms in a model of human intestinal epithelium.
Collapse
Affiliation(s)
- Marcela Fuentes
- Department of Nutrition, Diabetes and Metabolism, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Nicolás Santander
- Department of Nutrition, Diabetes and Metabolism, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Víctor Cortés
- Department of Nutrition, Diabetes and Metabolism, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
13
|
Reboul E. Vitamin E intestinal absorption: Regulation of membrane transport across the enterocyte. IUBMB Life 2018; 71:416-423. [PMID: 30308094 DOI: 10.1002/iub.1955] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 08/28/2018] [Accepted: 08/31/2018] [Indexed: 12/29/2022]
Abstract
Vitamin E is an essential molecule for our development and health. It has long been thought that it was absorbed and transported through cellular membranes by a passive diffusion process. However, data obtained during the past 15 years showed that its absorption is actually mediated, at least in part, by cholesterol membrane transporters including the scavenger receptor class B type I (SR-BI), CD36 molecule (CD36), NPC1-like transporter 1 (NPC1L1), and ATP-binding cassettes A1 and G1 (ABCA1 and ABCG1). This review focuses on the absorption process of vitamin E across the enterocyte. A special attention is given to the regulation of this process, including the possible competition with other fat-soluble micronutrients, and the modulation of transporter expressions. Overall, recent results noticeably increased the comprehension of vitamin E intestinal transport, but additional investigations are still required to fully appreciate the mechanisms governing vitamin E bioavailability. © 2018 IUBMB Life, 71(4):416-423, 2019.
Collapse
|
14
|
Genser L, Aguanno D, Soula HA, Dong L, Trystram L, Assmann K, Salem JE, Vaillant JC, Oppert JM, Laugerette F, Michalski MC, Wind P, Rousset M, Brot-Laroche E, Leturque A, Clément K, Thenet S, Poitou C. Increased jejunal permeability in human obesity is revealed by a lipid challenge and is linked to inflammation and type 2 diabetes. J Pathol 2018; 246:217-230. [DOI: 10.1002/path.5134] [Citation(s) in RCA: 133] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 06/19/2018] [Accepted: 06/28/2018] [Indexed: 12/27/2022]
Affiliation(s)
- Laurent Genser
- Sorbonne Université, INSERM, Nutriomics Team; Paris France
- Assistance Publique-Hôpitaux de Paris; Pitié-Salpêtrière Hospital, Department of Hepato-Biliary and Pancreatic Surgery; Paris France
| | - Doriane Aguanno
- Centre de Recherche des Cordeliers, Sorbonne Université, EPHE, PSL University, Sorbonne Cités, UPD Univ Paris 05; INSERM, CNRS; Paris France
| | - Hédi A Soula
- Centre de Recherche des Cordeliers, Sorbonne Université, EPHE, PSL University, Sorbonne Cités, UPD Univ Paris 05; INSERM, CNRS; Paris France
| | - Liping Dong
- Centre de Recherche des Cordeliers, Sorbonne Université, EPHE, PSL University, Sorbonne Cités, UPD Univ Paris 05; INSERM, CNRS; Paris France
| | - Laurence Trystram
- Assistance Publique-Hôpitaux de Paris, Pitié-Salpêtrière Hospital, Department of Functional Coprology; Paris France
| | - Karen Assmann
- Sorbonne Université, INSERM, Nutriomics Team; Paris France
| | - Joe-Elie Salem
- Assistance Publique-Hôpitaux de Paris, Pitié-Salpêtrière Hospital, Department of Pharmacology and CIC-1421; Paris France
| | - Jean-Christophe Vaillant
- Assistance Publique-Hôpitaux de Paris; Pitié-Salpêtrière Hospital, Department of Hepato-Biliary and Pancreatic Surgery; Paris France
| | - Jean-Michel Oppert
- Assistance Publique-Hôpitaux de Paris, Pitié-Salpêtrière Hospital, Nutrition Department; CRNH Ile de France; Paris France
| | - Fabienne Laugerette
- Lyon University, CarMeN laboratory, INRA U1397, Univ Lyon-1, INSERM U1060; INSA Lyon, Villeurbanne France
| | - Marie-Caroline Michalski
- Lyon University, CarMeN laboratory, INRA U1397, Univ Lyon-1, INSERM U1060; INSA Lyon, Villeurbanne France
| | - Philippe Wind
- Assistance Publique-Hôpitaux de Paris, Avicenne Hospital, Department of Digestive and Metabolic Surgery; Université Paris XIII-UFR SMBH ‘Léonard de Vinci’; Bobigny France
| | - Monique Rousset
- Centre de Recherche des Cordeliers, Sorbonne Université, EPHE, PSL University, Sorbonne Cités, UPD Univ Paris 05; INSERM, CNRS; Paris France
| | - Edith Brot-Laroche
- Centre de Recherche des Cordeliers, Sorbonne Université, EPHE, PSL University, Sorbonne Cités, UPD Univ Paris 05; INSERM, CNRS; Paris France
| | - Armelle Leturque
- Centre de Recherche des Cordeliers, Sorbonne Université, EPHE, PSL University, Sorbonne Cités, UPD Univ Paris 05; INSERM, CNRS; Paris France
| | - Karine Clément
- Sorbonne Université, INSERM, Nutriomics Team; Paris France
- Assistance Publique-Hôpitaux de Paris, Pitié-Salpêtrière Hospital, Nutrition Department; CRNH Ile de France; Paris France
| | - Sophie Thenet
- Centre de Recherche des Cordeliers, Sorbonne Université, EPHE, PSL University, Sorbonne Cités, UPD Univ Paris 05; INSERM, CNRS; Paris France
| | - Christine Poitou
- Sorbonne Université, INSERM, Nutriomics Team; Paris France
- Assistance Publique-Hôpitaux de Paris, Pitié-Salpêtrière Hospital, Nutrition Department; CRNH Ile de France; Paris France
| |
Collapse
|
15
|
Morel E, Ghezzal S, Lucchi G, Truntzer C, Pais de Barros JP, Simon-Plas F, Demignot S, Mineo C, Shaul PW, Leturque A, Rousset M, Carrière V. Cholesterol trafficking and raft-like membrane domain composition mediate scavenger receptor class B type 1-dependent lipid sensing in intestinal epithelial cells. Biochim Biophys Acta Mol Cell Biol Lipids 2017; 1863:199-211. [PMID: 29196159 DOI: 10.1016/j.bbalip.2017.11.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 11/13/2017] [Accepted: 11/27/2017] [Indexed: 02/02/2023]
Abstract
Scavenger receptor Class B type 1 (SR-B1) is a lipid transporter and sensor. In intestinal epithelial cells, SR-B1-dependent lipid sensing is associated with SR-B1 recruitment in raft-like/ detergent-resistant membrane domains and interaction of its C-terminal transmembrane domain with plasma membrane cholesterol. To clarify the initiating events occurring during lipid sensing by SR-B1, we analyzed cholesterol trafficking and raft-like domain composition in intestinal epithelial cells expressing wild-type SR-B1 or the mutated form SR-B1-Q445A, defective in membrane cholesterol binding and signal initiation. These features of SR-B1 were found to influence both apical cholesterol efflux and intracellular cholesterol trafficking from plasma membrane to lipid droplets, and the lipid composition of raft-like domains. Lipidomic analysis revealed likely participation of d18:0/16:0 sphingomyelin and 16:0/0:0 lysophosphatidylethanolamine in lipid sensing by SR-B1. Proteomic analysis identified proteins, whose abundance changed in raft-like domains during lipid sensing, and these included molecules linked to lipid raft dynamics and signal transduction. These findings provide new insights into the role of SR-B1 in cellular cholesterol homeostasis and suggest molecular links between SR-B1-dependent lipid sensing and cell cholesterol and lipid droplet dynamics.
Collapse
Affiliation(s)
- Etienne Morel
- Centre de Recherche des Cordeliers, INSERM, UMPC Université Paris 6, Université Paris Descartes Paris 5, CNRS, F-75006 Paris, France
| | - Sara Ghezzal
- Centre de Recherche des Cordeliers, INSERM, UMPC Université Paris 6, Université Paris Descartes Paris 5, CNRS, F-75006 Paris, France
| | - Géraldine Lucchi
- Clinical Innovation Proteomic Platform CLIPP, Université Bourgogne Franche-Comté, F-21000 Dijon, France
| | - Caroline Truntzer
- Clinical Innovation Proteomic Platform CLIPP, Université Bourgogne Franche-Comté, F-21000 Dijon, France
| | - Jean-Paul Pais de Barros
- Plateforme de Lipidomique, INSERM UMR1231, Université de Bourgogne Franche Comté, F-21000 Dijon, France
| | - Françoise Simon-Plas
- Agroécologie, AgroSup Dijon, CNRS, INRA, Université Bourgogne Franche-Comté, F-21000 Dijon, France
| | - Sylvie Demignot
- Centre de Recherche des Cordeliers, INSERM, UMPC Université Paris 6, Université Paris Descartes Paris 5, CNRS, F-75006 Paris, France; EPHE, PSL Research University, F-75006 Paris, France
| | - Chieko Mineo
- Center for Pulmonary and Vascular Biology, Department of Pediatrics, University of Texas, Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Philip W Shaul
- Center for Pulmonary and Vascular Biology, Department of Pediatrics, University of Texas, Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Armelle Leturque
- Centre de Recherche des Cordeliers, INSERM, UMPC Université Paris 6, Université Paris Descartes Paris 5, CNRS, F-75006 Paris, France
| | - Monique Rousset
- Centre de Recherche des Cordeliers, INSERM, UMPC Université Paris 6, Université Paris Descartes Paris 5, CNRS, F-75006 Paris, France
| | - Véronique Carrière
- Centre de Recherche des Cordeliers, INSERM, UMPC Université Paris 6, Université Paris Descartes Paris 5, CNRS, F-75006 Paris, France.
| |
Collapse
|
16
|
Vitamin E Bioavailability: Mechanisms of Intestinal Absorption in the Spotlight. Antioxidants (Basel) 2017; 6:antiox6040095. [PMID: 29165370 PMCID: PMC5745505 DOI: 10.3390/antiox6040095] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 11/14/2017] [Accepted: 11/16/2017] [Indexed: 01/06/2023] Open
Abstract
Vitamin E is an essential fat-soluble micronutrient whose effects on human health can be attributed to both antioxidant and non-antioxidant properties. A growing number of studies aim to promote vitamin E bioavailability in foods. It is thus of major interest to gain deeper insight into the mechanisms of vitamin E absorption, which remain only partly understood. It was long assumed that vitamin E was absorbed by passive diffusion, but recent data has shown that this process is actually far more complex than previously thought. This review describes the fate of vitamin E in the human gastrointestinal lumen during digestion and focuses on the proteins involved in the intestinal membrane and cellular transport of vitamin E across the enterocyte. Special attention is also given to the factors modulating both vitamin E micellarization and absorption. Although these latest results significantly improve our understanding of vitamin E intestinal absorption, further studies are still needed to decipher the molecular mechanisms driving this multifaceted process.
Collapse
|
17
|
Hersoug LG, Møller P, Loft S. Gut microbiota-derived lipopolysaccharide uptake and trafficking to adipose tissue: implications for inflammation and obesity. Obes Rev 2016; 17:297-312. [PMID: 26712364 DOI: 10.1111/obr.12370] [Citation(s) in RCA: 206] [Impact Index Per Article: 22.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2015] [Revised: 11/18/2015] [Accepted: 11/19/2015] [Indexed: 12/12/2022]
Abstract
The composition of the gut microbiota and excessive ingestion of high-fat diets (HFD) are considered to be important factors for development of obesity. In this review we describe a coherent mechanism of action for the development of obesity, which involves the composition of gut microbiota, HFD, low-grade inflammation, expression of fat translocase and scavenger receptor CD36, and the scavenger receptor class B type 1 (SR-BI). SR-BI binds to both lipids and lipopolysaccharide (LPS) from Gram-negative bacteria, which may promote incorporation of LPS in chylomicrons (CMs). These CMs are transported via lymph to the circulation, where LPS is transferred to other lipoproteins by translocases, preferentially to HDL. LPS increases the SR-BI binding, transcytosis of lipoproteins over the endothelial barrier,and endocytosis in adipocytes. Especially large size adipocytes with high metabolic activity absorb LPS-rich lipoproteins. In addition, macrophages in adipose tissue internalize LPS-lipoproteins. This may contribute to the polarization from M2 to M1 phenotype, which is a consequence of increased LPS delivery into the tissue during hypertrophy. In conclusion, evidence suggests that LPS is involved in the development of obesity as a direct targeting molecule for lipid delivery and storage in adipose tissue.
Collapse
Affiliation(s)
- L-G Hersoug
- Section of Environmental Health, Department of Public Health, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - P Møller
- Section of Environmental Health, Department of Public Health, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - S Loft
- Section of Environmental Health, Department of Public Health, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
18
|
Sparks SM, Zhou H, Generaux C, Harston L, Moncol D, Jayawickreme C, Parham J, Condreay P, Rimele T. Identification of nonabsorbable inhibitors of the scavenger receptor-BI (SR-BI) for tissue-specific administration. Bioorg Med Chem Lett 2016; 26:1901-4. [PMID: 26988301 DOI: 10.1016/j.bmcl.2016.03.025] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Revised: 03/07/2016] [Accepted: 03/08/2016] [Indexed: 01/01/2023]
Abstract
The identification of a low-permeability scavenger receptor BI (SR-BI) inhibitor starting from the ITX-5061 template is described. Structure-activity and structure-permeability relationships were assessed for analogs leading to the identification of compound 8 as a potent and nonabsorbable SR-BI inhibitor.
Collapse
Affiliation(s)
- Steven M Sparks
- GlaxoSmithKline, Enteroendocrine Discovery Performance Unit and Platform Technology and Science, 5 Moore Drive, Research Triangle Park, NC 27709, United States.
| | - Huiqiang Zhou
- GlaxoSmithKline, Enteroendocrine Discovery Performance Unit and Platform Technology and Science, 5 Moore Drive, Research Triangle Park, NC 27709, United States
| | - Claudia Generaux
- GlaxoSmithKline, Enteroendocrine Discovery Performance Unit and Platform Technology and Science, 5 Moore Drive, Research Triangle Park, NC 27709, United States
| | - Lindsey Harston
- GlaxoSmithKline, Enteroendocrine Discovery Performance Unit and Platform Technology and Science, 5 Moore Drive, Research Triangle Park, NC 27709, United States
| | - David Moncol
- GlaxoSmithKline, Enteroendocrine Discovery Performance Unit and Platform Technology and Science, 5 Moore Drive, Research Triangle Park, NC 27709, United States
| | - Channa Jayawickreme
- GlaxoSmithKline, Enteroendocrine Discovery Performance Unit and Platform Technology and Science, 5 Moore Drive, Research Triangle Park, NC 27709, United States
| | - Janet Parham
- GlaxoSmithKline, Enteroendocrine Discovery Performance Unit and Platform Technology and Science, 5 Moore Drive, Research Triangle Park, NC 27709, United States
| | - Patrick Condreay
- GlaxoSmithKline, Enteroendocrine Discovery Performance Unit and Platform Technology and Science, 5 Moore Drive, Research Triangle Park, NC 27709, United States
| | - Thomas Rimele
- GlaxoSmithKline, Enteroendocrine Discovery Performance Unit and Platform Technology and Science, 5 Moore Drive, Research Triangle Park, NC 27709, United States
| |
Collapse
|
19
|
Briand O, Touche V, Colin S, Brufau G, Davalos A, Schonewille M, Bovenga F, Carrière V, de Boer JF, Dugardin C, Riveau B, Clavey V, Tailleux A, Moschetta A, Lasunción MA, Groen AK, Staels B, Lestavel S. Liver X Receptor Regulates Triglyceride Absorption Through Intestinal Down-regulation of Scavenger Receptor Class B, Type 1. Gastroenterology 2016; 150:650-8. [PMID: 26602218 DOI: 10.1053/j.gastro.2015.11.015] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Revised: 10/30/2015] [Accepted: 11/10/2015] [Indexed: 12/02/2022]
Abstract
BACKGROUND & AIMS Reducing postprandial triglyceridemia may be a promising strategy to lower the risk of cardiovascular disorders associated with obesity and type 2 diabetes. In enterocytes, scavenger receptor class B, type 1 (SR-B1, encoded by SCARB1) mediates lipid-micelle sensing to promote assembly and secretion of chylomicrons. The nuclear receptor subfamily 1, group H, members 2 and 3 (also known as liver X receptors [LXRs]) regulate genes involved in cholesterol and fatty acid metabolism. We aimed to determine whether intestinal LXRs regulate triglyceride absorption. METHODS C57BL/6J mice were either fed a cholesterol-enriched diet or given synthetic LXR agonists (GW3965 or T0901317). We measured the production of chylomicrons and localized SR-B1 by immunohistochemistry. Mechanisms of postprandial triglyceridemia and SR-B1 regulation were studied in Caco-2/TC7 cells incubated with LXR agonists. RESULTS In mice and in the Caco-2/TC7 cell line, LXR agonists caused localization of intestinal SR-B1 from apical membranes to intracellular organelles and reduced chylomicron secretion. In Caco-2/TC7 cells, LXR agonists reduced SR-B1-dependent lipidic-micelle-induced Erk phosphorylation. LXR agonists also reduced intracellular trafficking of the apical apolipoprotein B pool toward secretory compartments. LXR reduced levels of SR-B1 in Caco-2/TC7 cells via a post-transcriptional mechanism that involves microRNAs. CONCLUSION In Caco-2/TC7 cells and mice, intestinal activation of LXR reduces the production of chylomicrons by a mechanism dependent on the apical localization of SR-B1.
Collapse
Affiliation(s)
- Olivier Briand
- University Lille, Inserm, Centre Hospitalier Universitaire (CHU) de Lille, Institut Pasteur de Lille, U1011-European Genomic Institute for Diabetes, Lille, France
| | - Véronique Touche
- University Lille, Inserm, Centre Hospitalier Universitaire (CHU) de Lille, Institut Pasteur de Lille, U1011-European Genomic Institute for Diabetes, Lille, France
| | - Sophie Colin
- University Lille, Inserm, Centre Hospitalier Universitaire (CHU) de Lille, Institut Pasteur de Lille, U1011-European Genomic Institute for Diabetes, Lille, France
| | - Gemma Brufau
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Alberto Davalos
- Madrid Institute for Advanced Studies (IMDEA) Food Institute, Laboratory of Disorders of Lipid Metabolism and Molecular Nutrition, Campus de Excelencia Internacional (CEI), Universidad Autónoma de Madrid (UAM), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Marleen Schonewille
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Fabiola Bovenga
- National Research Cancer Center, Giovanni Paolo II, and University of Bari, Bari, Italy
| | - Véronique Carrière
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France; Université Paris Descartes, Sorbonne Paris Cit, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France; Servicio de Bioquímica-Investigación, Hospital Ramón y Cajal, Instituto Ramón y Cajal de Investigatión Sanitaria (IRYCIS), Madrid, Spain
| | - Jan Freark de Boer
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Camille Dugardin
- University Lille, Inserm, Centre Hospitalier Universitaire (CHU) de Lille, Institut Pasteur de Lille, U1011-European Genomic Institute for Diabetes, Lille, France
| | - Béatrice Riveau
- Sorbonne Universités, Université Pierre et Marie Curie (UPMC), UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France; Institut National de la Santé et de la Recherche Médicale (INSERM), UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France; Université Paris Descartes, Sorbonne Paris Cit, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France
| | - Véronique Clavey
- University Lille, Inserm, Centre Hospitalier Universitaire (CHU) de Lille, Institut Pasteur de Lille, U1011-European Genomic Institute for Diabetes, Lille, France
| | - Anne Tailleux
- University Lille, Inserm, Centre Hospitalier Universitaire (CHU) de Lille, Institut Pasteur de Lille, U1011-European Genomic Institute for Diabetes, Lille, France
| | - Antonio Moschetta
- National Research Cancer Center, Giovanni Paolo II, and University of Bari, Bari, Italy
| | - Miguel A Lasunción
- Servicio de Bioquímica-Investigación, Hospital Ramón y Cajal, Instituto Ramón y Cajal de Investigatión Sanitaria (IRYCIS), Madrid, Spain; Centro de Investigatión Biomedica en Red (CIBER) de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Spain
| | - Albert K Groen
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Bart Staels
- University Lille, Inserm, Centre Hospitalier Universitaire (CHU) de Lille, Institut Pasteur de Lille, U1011-European Genomic Institute for Diabetes, Lille, France.
| | - Sophie Lestavel
- University Lille, Inserm, Centre Hospitalier Universitaire (CHU) de Lille, Institut Pasteur de Lille, U1011-European Genomic Institute for Diabetes, Lille, France
| |
Collapse
|
20
|
Lino M, Farr S, Baker C, Fuller M, Trigatti B, Adeli K. Intestinal scavenger receptor class B type I as a novel regulator of chylomicron production in healthy and diet-induced obese states. Am J Physiol Gastrointest Liver Physiol 2015; 309:G350-9. [PMID: 26138463 DOI: 10.1152/ajpgi.00086.2015] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Accepted: 06/25/2015] [Indexed: 01/31/2023]
Abstract
The small intestine contributes to diabetic dyslipidemia through the overproduction of apolipoprotein B48 (apoB48)-containing chylomicron particles. An important regulator of chylomicron generation is dietary lipid absorption, underlining the potential involvement of intestinal lipid transporters for developing dyslipidemia. Intestinal expression of scavenger receptor class B type I (SR-BI) has been found to be upregulated in animal models of insulin resistance. Here we characterized the potential importance of SR-BI in contributing to chylomicron production and postprandial hypertriglyceridemia in vivo. Postprandial triglyceride (TG)-rich lipoprotein (TRL) production was characterized in hamsters treated with the SR-BI inhibitor to block lipid transport-1 (BLT-1) under healthy conditions or conditions of diet-induced obesity and dyslipidemia. BLT-1 (1 mg/kg) or vehicle was administered acutely in chow-fed hamsters or gavaged twice daily over 10 days during high-fructose, high-fat, high-cholesterol (FFC) feeding. Effects of acute SR-BI inhibition by BLT-1 were confirmed in healthy fat-loaded rats. Finally, plasma lipid levels were compared between SR-BI(-/-) mice and their wild-type counterparts fed either chow or a 12-wk high-fat diet. Acute BLT-1 treatment reduced postprandial plasma and TRL TG levels in healthy hamsters and rats. Chronic BLT-1 treatment of FFC-fed hamsters blunted diet-induced weight gain and fasting hypertriglyceridemia, and lowered postprandial TRL-TG, -cholesterol, and -apoB48 levels. Finally, SR-BI(-/-) mice displayed lower plasma and TRL TG levels relative to wild type, and diet-induced weight gain and postprandial hypertriglyceridemia were hindered in SR-BI(-/-) mice. We conclude that intestinal SR-BI is a critical regulator of postprandial lipoprotein production, emphasizing its potential as a target for preventing diabetic dyslipidemia.
Collapse
Affiliation(s)
- Marsel Lino
- Department of Molecular Structure and Function, Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada; and
| | - Sarah Farr
- Department of Molecular Structure and Function, Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada; and
| | - Chris Baker
- Department of Molecular Structure and Function, Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Mark Fuller
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Bernardo Trigatti
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Khosrow Adeli
- Department of Molecular Structure and Function, Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada; and
| |
Collapse
|
21
|
Goncalves A, Gontero B, Nowicki M, Margier M, Masset G, Amiot MJ, Reboul E. Micellar lipid composition affects micelle interaction with class B scavenger receptor extracellular loops. J Lipid Res 2015; 56:1123-33. [PMID: 25833688 DOI: 10.1194/jlr.m057612] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Indexed: 11/20/2022] Open
Abstract
Scavenger receptors (SRs) like cluster determinant 36 (CD36) and SR class B type I (SR-BI) play a debated role in lipid transport across the intestinal brush border membrane. We used surface plasmon resonance to analyze real-time interactions between the extracellular protein loops and various ligands ranging from single lipid molecules to mixed micelles. Micelles mimicking physiological structures were necessary for optimal binding to both the extracellular loop of CD36 (lCD36) and the extracellular loop of SR-BI (lSR-BI). Cholesterol, phospholipid, and fatty acid micellar content significantly modulated micelle binding to and dissociation from the transporters. In particular, high phospholipid micellar concentrations inhibited micelle binding to both receptors (-53.8 and -74.4% binding at 0.32 mM compared with 0.04 mM for lCD36 and lSR-BI, respectively, P < 0.05). The presence of fatty acids was crucial for micelle interactions with both proteins (94.4 and 81.3% binding with oleic acid for lCD36 and lSR-BI, respectively, P < 0.05) and fatty acid type substitution within the micelles was the component that most impacted micelle binding to the transporters. These effects were partly due to subsequent modifications in micellar size and surface electric charge, and could be correlated to micellar vitamin D uptake by Caco-2 cells. Our findings show for the first time that micellar lipid composition and micellar properties are key factors governing micelle interactions with SRs.
Collapse
Affiliation(s)
- Aurélie Goncalves
- INRA, UMR 1260 "Nutrition, Obesity and Risk of Thrombosis," F-13385 Marseille, France INSERM, UMR 1062, F-13385 Marseille, France Aix-Marseille Université, F-13385 Marseille, France
| | - Brigitte Gontero
- Aix-Marseille Université CNRS, BIP, UMR 7281, F-13402 Marseille, France
| | - Marion Nowicki
- INRA, UMR 1260 "Nutrition, Obesity and Risk of Thrombosis," F-13385 Marseille, France INSERM, UMR 1062, F-13385 Marseille, France Aix-Marseille Université, F-13385 Marseille, France
| | - Marielle Margier
- INRA, UMR 1260 "Nutrition, Obesity and Risk of Thrombosis," F-13385 Marseille, France INSERM, UMR 1062, F-13385 Marseille, France Aix-Marseille Université, F-13385 Marseille, France
| | - Gabriel Masset
- INRA, UMR 1260 "Nutrition, Obesity and Risk of Thrombosis," F-13385 Marseille, France INSERM, UMR 1062, F-13385 Marseille, France Aix-Marseille Université, F-13385 Marseille, France
| | - Marie-Josèphe Amiot
- INRA, UMR 1260 "Nutrition, Obesity and Risk of Thrombosis," F-13385 Marseille, France INSERM, UMR 1062, F-13385 Marseille, France Aix-Marseille Université, F-13385 Marseille, France
| | - Emmanuelle Reboul
- INRA, UMR 1260 "Nutrition, Obesity and Risk of Thrombosis," F-13385 Marseille, France INSERM, UMR 1062, F-13385 Marseille, France Aix-Marseille Université, F-13385 Marseille, France
| |
Collapse
|
22
|
Zhao Y, Hoekstra M, Korporaal SJA, Van Berkel TJC, Van Eck M. HDL Receptor Scavenger Receptor BI. Atherosclerosis 2015. [DOI: 10.1002/9781118828533.ch25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
23
|
Andres SF, Santoro MA, Mah AT, Keku JA, Bortvedt AE, Blue RE, Lund PK. Deletion of intestinal epithelial insulin receptor attenuates high-fat diet-induced elevations in cholesterol and stem, enteroendocrine, and Paneth cell mRNAs. Am J Physiol Gastrointest Liver Physiol 2015; 308:G100-11. [PMID: 25394660 PMCID: PMC4297856 DOI: 10.1152/ajpgi.00287.2014] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The insulin receptor (IR) regulates nutrient uptake and utilization in multiple organs, but its role in the intestinal epithelium is not defined. This study developed a mouse model with villin-Cre (VC) recombinase-mediated intestinal epithelial cell (IEC)-specific IR deletion (VC-IR(Δ/Δ)) and littermate controls with floxed, but intact, IR (IR(fl/fl)) to define in vivo roles of IEC-IR in mice fed chow or high-fat diet (HFD). We hypothesized that loss of IEC-IR would alter intestinal growth, biomarkers of intestinal epithelial stem cells (IESC) or other lineages, body weight, adiposity, and glucose or lipid handling. In lean, chow-fed mice, IEC-IR deletion did not affect body or fat mass, plasma glucose, or IEC proliferation. In chow-fed VC-IR(Δ/Δ) mice, mRNA levels of the Paneth cell marker lysozyme (Lyz) were decreased, but markers of other differentiated lineages were unchanged. During HFD-induced obesity, IR(fl/fl) and VC-IR(Δ/Δ) mice exhibited similar increases in body and fat mass, plasma insulin, mRNAs encoding several lipid-handling proteins, a decrease in Paneth cell number, and impaired glucose tolerance. In IR(fl/fl) mice, HFD-induced obesity increased circulating cholesterol; numbers of chromogranin A (CHGA)-positive enteroendocrine cells (EEC); and mRNAs encoding Chga, glucose-dependent insulinotrophic peptide (Gip), glucagon (Gcg), Lyz, IESC biomarkers, and the enterocyte cholesterol transporter Scarb1. All these effects were attenuated or lost in VC-IR(Δ/Δ) mice. These results demonstrate that IEC-IR is not required for normal growth of the intestinal epithelium in lean adult mice. However, our findings provide novel evidence that, during HFD-induced obesity, IEC-IR contributes to increases in EEC, plasma cholesterol, and increased expression of Scarb1 or IESC-, EEC-, and Paneth cell-derived mRNAs.
Collapse
Affiliation(s)
- Sarah F. Andres
- 1Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; and
| | - M. Agostina Santoro
- 1Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; and
| | - Amanda T. Mah
- 2Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - J. Adeola Keku
- 1Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; and
| | - Amy E. Bortvedt
- 1Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; and
| | - R. Eric Blue
- 1Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; and
| | - P. Kay Lund
- 1Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; and
| |
Collapse
|
24
|
Khaldoun SA, Emond-Boisjoly MA, Chateau D, Carrière V, Lacasa M, Rousset M, Demignot S, Morel E. Autophagosomes contribute to intracellular lipid distribution in enterocytes. Mol Biol Cell 2013; 25:118-32. [PMID: 24173715 PMCID: PMC3873883 DOI: 10.1091/mbc.e13-06-0324] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Delivery of alimentary lipids induces immediate autophagic response in enterocytes. Forming autophagosomes are recruited to the ER membrane, where they capture nascent lipid droplets and later fuse with lysosomes, illustrating for the first time the role of autophagy in neutral-lipid distribution in enterocytes. Enterocytes, the intestinal absorptive cells, have to deal with massive alimentary lipids upon food consumption. They orchestrate complex lipid-trafficking events that lead to the secretion of triglyceride-rich lipoproteins and/or the intracellular transient storage of lipids as lipid droplets (LDs). LDs originate from the endoplasmic reticulum (ER) membrane and are mainly composed of a triglyceride (TG) and cholesterol-ester core surrounded by a phospholipid and cholesterol monolayer and specific coat proteins. The pivotal role of LDs in cellular lipid homeostasis is clearly established, but processes regulating LD dynamics in enterocytes are poorly understood. Here we show that delivery of alimentary lipid micelles to polarized human enterocytes induces an immediate autophagic response, accompanied by phosphatidylinositol-3-phosphate appearance at the ER membrane. We observe a specific and rapid capture of newly synthesized LD at the ER membrane by nascent autophagosomal structures. By combining pharmacological and genetic approaches, we demonstrate that autophagy is a key player in TG targeting to lysosomes. Our results highlight the yet-unraveled role of autophagy in the regulation of TG distribution, trafficking, and turnover in human enterocytes.
Collapse
Affiliation(s)
- Salem Ait Khaldoun
- Centre de Recherche des Cordeliers, UMR S 872, Université Pierre et Marie Curie-Paris 6, Institut National de la Santé et de la Recherche Médicale, U 872 and UMR S 872, Université Paris Descartes-Paris 5, F-75006 Paris, France Laboratoire de Pharmacologie Cellulaire et Moléculaire, Ecole Pratique des Hautes Etudes, F-75006 Paris, France
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Buttet M, Traynard V, Tran TTT, Besnard P, Poirier H, Niot I. From fatty-acid sensing to chylomicron synthesis: role of intestinal lipid-binding proteins. Biochimie 2013; 96:37-47. [PMID: 23958439 DOI: 10.1016/j.biochi.2013.08.011] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2013] [Accepted: 08/05/2013] [Indexed: 12/31/2022]
Abstract
Today, it is well established that the development of obesity and associated diseases results, in part, from excessive lipid intake associated with a qualitative imbalance. Among the organs involved in lipid homeostasis, the small intestine is the least studied even though it determines lipid bioavailability and largely contributes to the regulation of postprandial hyperlipemia (triacylglycerols (TG) and free fatty acids (FFA)). Several Lipid-Binding Proteins (LBP) are expressed in the small intestine. Their supposed intestinal functions were initially based on what was reported in other tissues, and took no account of the physiological specificity of the small intestine. Progressively, the identification of regulating factors of intestinal LBP and the description of the phenotype of their deletion have provided new insights into cellular and molecular mechanisms involved in fat absorption. This review will discuss the physiological contribution of each LBP in the main steps of intestinal absorption of long-chain fatty acids (LCFA): uptake, trafficking and reassembly into chylomicrons (CM). Moreover, current data indicate that the small intestine is able to adapt its lipid absorption capacity to the fat content of the diet, especially through the coordinated induction of LBP. This adaptation requires the existence of a mechanism of intestinal lipid sensing. Emerging data suggest that the membrane LBP CD36 may operate as a lipid receptor that triggers an intracellular signal leading to the modulation of the expression of LBP involved in CM formation. This event could be the starting point for the optimized synthesis of large CM, which are efficiently degraded in blood. Better understanding of this intestinal lipid sensing might provide new approaches to decrease the prevalence of postprandial hypertriglyceridemia, which is associated with cardiovascular diseases, insulin resistance and obesity.
Collapse
Affiliation(s)
- Marjorie Buttet
- Physiologie de la Nutrition et Toxicologie Team (NUTox), UMR U866 INSERM, Université de Bourgogne, AgroSup Dijon, 1 Esplanade Erasme, 21000 Dijon, France
| | | | | | | | | | | |
Collapse
|
26
|
Bura KS, Lord C, Marshall S, McDaniel A, Thomas G, Warrier M, Zhang J, Davis MA, Sawyer JK, Shah R, Wilson MD, Dikkers A, Tietge UJF, Collet X, Rudel LL, Temel RE, Brown JM. Intestinal SR-BI does not impact cholesterol absorption or transintestinal cholesterol efflux in mice. J Lipid Res 2013; 54:1567-1577. [PMID: 23564696 DOI: 10.1194/jlr.m034454] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Reverse cholesterol transport (RCT) can proceed through the classic hepatobiliary route or through the nonbiliary transintestinal cholesterol efflux (TICE) pathway. Scavenger receptor class B type I (SR-BI) plays a critical role in the classic hepatobiliary route of RCT. However, the role of SR-BI in TICE has not been studied. To examine the role of intestinal SR-BI in TICE, sterol balance was measured in control mice and mice transgenically overexpressing SR-BI in the proximal small intestine (SR-BI(hApoCIII-ApoAIV-Tg)). SR-BI(hApoCIII-ApoAIV-Tg) mice had significantly lower plasma cholesterol levels compared with wild-type controls, yet SR-BI(hApoCIII-ApoAIV-Tg) mice had normal fractional cholesterol absorption and fecal neutral sterol excretion. Both in the absence or presence of ezetimibe, intestinal SR-BI overexpression had no impact on the amount of cholesterol excreted in the feces. To specifically study effects of intestinal SR-BI on TICE we crossed SR-BI(hApoCIII-ApoAIV-Tg) mice into a mouse model that preferentially utilized the TICE pathway for RCT (Niemann-Pick C1-like 1 liver transgenic), and likewise found no alterations in cholesterol absorption or fecal sterol excretion. Finally, mice lacking SR-BI in all tissues also exhibited normal cholesterol absorption and fecal cholesterol disposal. Collectively, these results suggest that SR-BI is not rate limiting for intestinal cholesterol absorption or for fecal neutral sterol loss through the TICE pathway.
Collapse
Affiliation(s)
- Kanwardeep S Bura
- Department of Pathology, Section on Lipid Sciences, Wake Forest School of Medicine, Winston-Salem, NC
| | - Caleb Lord
- Department of Pathology, Section on Lipid Sciences, Wake Forest School of Medicine, Winston-Salem, NC
| | - Stephanie Marshall
- Department of Pathology, Section on Lipid Sciences, Wake Forest School of Medicine, Winston-Salem, NC
| | - Allison McDaniel
- Department of Pathology, Section on Lipid Sciences, Wake Forest School of Medicine, Winston-Salem, NC
| | - Gwyn Thomas
- Department of Pathology, Section on Lipid Sciences, Wake Forest School of Medicine, Winston-Salem, NC
| | - Manya Warrier
- Department of Pathology, Section on Lipid Sciences, Wake Forest School of Medicine, Winston-Salem, NC
| | - Jun Zhang
- Department of Pathology, Section on Lipid Sciences, Wake Forest School of Medicine, Winston-Salem, NC
| | - Matthew A Davis
- Department of Pathology, Section on Lipid Sciences, Wake Forest School of Medicine, Winston-Salem, NC
| | - Janet K Sawyer
- Department of Pathology, Section on Lipid Sciences, Wake Forest School of Medicine, Winston-Salem, NC
| | - Ramesh Shah
- Department of Pathology, Section on Lipid Sciences, Wake Forest School of Medicine, Winston-Salem, NC
| | - Martha D Wilson
- Department of Pathology, Section on Lipid Sciences, Wake Forest School of Medicine, Winston-Salem, NC
| | - Arne Dikkers
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Uwe J F Tietge
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Xavier Collet
- INSERM U1048, Institute of Metabolic and Cardiovascular Diseases of Rangueil Hospital, BP 84225, Toulouse, France
| | - Lawrence L Rudel
- Department of Pathology, Section on Lipid Sciences, Wake Forest School of Medicine, Winston-Salem, NC
| | - Ryan E Temel
- Department of Pathology, Section on Lipid Sciences, Wake Forest School of Medicine, Winston-Salem, NC.
| | - J Mark Brown
- Department of Pathology, Section on Lipid Sciences, Wake Forest School of Medicine, Winston-Salem, NC.
| |
Collapse
|
27
|
Affiliation(s)
- Mary Johnson
- Synatom Research, Princeton, New Jersey, United States
| |
Collapse
|
28
|
Ferruzza S, Rossi C, Scarino ML, Sambuy Y. A protocol for differentiation of human intestinal Caco-2 cells in asymmetric serum-containing medium. Toxicol In Vitro 2012; 26:1252-5. [DOI: 10.1016/j.tiv.2012.01.008] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2011] [Revised: 12/13/2011] [Accepted: 01/06/2012] [Indexed: 11/30/2022]
|
29
|
Saddar S, Carriere V, Lee WR, Tanigaki K, Yuhanna IS, Parathath S, Morel E, Warrier M, Sawyer JK, Gerard RD, Temel RE, Brown JM, Connelly M, Mineo C, Shaul PW. Scavenger receptor class B type I is a plasma membrane cholesterol sensor. Circ Res 2012; 112:140-51. [PMID: 23023567 DOI: 10.1161/circresaha.112.280081] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
RATIONALE Signal initiation by the high-density lipoprotein (HDL) receptor scavenger receptor class B, type I (SR-BI), which is important to actions of HDL on endothelium and other processes, requires cholesterol efflux and the C-terminal transmembrane domain. The C-terminal transmembrane domain uniquely interacts with plasma membrane (PM) cholesterol. OBJECTIVE The molecular basis and functional significance of SR-BI interaction with PM cholesterol are unknown. We tested the hypotheses that the interaction is required for SR-BI signaling, and that it enables SR-BI to serve as a PM cholesterol sensor. METHODS AND RESULTS In studies performed in COS-M6 cells, mutation of a highly conserved C-terminal transmembrane domain glutamine to alanine (SR-BI-Q445A) decreased PM cholesterol interaction with the receptor by 71% without altering HDL binding or cholesterol uptake or efflux, and it yielded a receptor incapable of HDL-induced signaling. Signaling prompted by cholesterol efflux to methyl-β-cyclodextrin also was prevented, indicating that PM cholesterol interaction with the receptor enables it to serve as a PM cholesterol sensor. Using SR-BI-Q445A, we further demonstrated that PM cholesterol sensing by SR-BI does not influence SR-BI-mediated reverse cholesterol transport to the liver in mice. However, the PM cholesterol sensing does underlie apolipoprotein B intracellular trafficking in response to postprandial micelles or methyl-β-cyclodextrin in cultured enterocytes, and it is required for HDL activation of endothelial NO synthase and migration in cultured endothelial cells and HDL-induced angiogenesis in vivo. CONCLUSIONS Through interaction with PM cholesterol, SR-BI serves as a PM cholesterol sensor, and the resulting intracellular signaling governs processes in both enterocytes and endothelial cells.
Collapse
Affiliation(s)
- Sonika Saddar
- Department of Pediatrics, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Shimotoyodome A, Osaki N, Onizawa K, Mizuno T, Suzukamo C, Okahara F, Fukuoka D, Hase T. Dietary 1-monoolein decreases postprandial GIP release by reducing jejunal transport of glucose and fatty acid in rodents. Am J Physiol Gastrointest Liver Physiol 2012; 303:G298-310. [PMID: 22651926 DOI: 10.1152/ajpgi.00457.2011] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Postprandial secretion of insulin and glucose-dependent insulinotropic polypeptide (GIP) is differentially regulated by not only dietary carbohydrate but also fat. Recent studies have shown that the ingestion of diacylglycerol (DAG) results in lower postprandial insulin and GIP release than that of triacylglycerol (TAG), suggesting a possible mechanism for the antiobesity effect of DAG. The structural and metabolic characteristics of DAG are believed to be responsible for its beneficial effects. This study was designed to clarify the effect of 1-monoacylglycerol [oleic acid-rich (1-MO)], the characteristic metabolite of DAG, on postprandial insulin and GIP secretion, and the underlying mechanism. Dietary 1-MO dose dependently stimulated whole body fat utilization, and reduced high-fat diet-induced body weight gain and visceral fat accumulation in mice, both of which are consistent with the physiological effect of dietary DAG. Although glucose-stimulated insulin and GIP release was augmented by the addition of fat, coingestion of 1-MO reduced the postprandial hormone release in a dose-dependent manner. Either glucose or fatty acid transport into the everted intestinal sacs and enteroendocrine HuTu-80 cells was also reduced by the addition of 1-MO. Reduction of either glucose or fatty acid transport or the nutrient-stimulated GIP release by 1-MO was nullified when the intestine was pretreated with sodium-glucose cotransporter-1 (SGLT-1) or fatty acid translocase (FAT)/CD36 inhibitor. We conclude that dietary 1-MO attenuates postprandial GIP and insulin secretion by reducing the intestinal transport of the GIP secretagogues, which may be mediated via SGLT-1 and FAT/CD36. Reduced secretion of these anabolic hormones by 1-MO may be related to the antiobesity effect of DAG.
Collapse
|
31
|
Colin S, Briand O, Touche V, Wouters K, Baron M, Pattou F, Hanf R, Tailleux A, Chinetti G, Staels B, Lestavel S. Activation of intestinal peroxisome proliferator-activated receptor-α increases high-density lipoprotein production. Eur Heart J 2012; 34:2566-74. [PMID: 22843443 DOI: 10.1093/eurheartj/ehs227] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
AIMS Peroxisome proliferator-activated receptor (PPAR)-α is a transcription factor controlling lipid metabolism in liver, heart, muscle, and macrophages. Peroxisome proliferator-activated receptor-α activation increases plasma HDL cholesterol and exerts hypotriglyceridaemic actions via the liver. However, the intestine expresses PPAR-α, produces HDL and chylomicrons, and is exposed to diet-derived PPAR-α ligands. Therefore, we examined the effects of PPAR-α activation on intestinal lipid and lipoprotein metabolism. METHODS AND RESULTS The impact of PPAR-α activation was evaluated in term of HDL-related gene expression in mice, ex vivo in human jejunal biopsies and in Caco-2/TC7 cells. Apolipoprotein-AI/HDL secretion, cholesterol esterification, and trafficking were also studied in vitro. In parallel to improving plasma lipid profiles and increasing liver and intestinal expression of fatty acid oxidation genes, treatment with the dual PPAR-α/δ ligand GFT505 resulted in a more pronounced increase in plasma HDL compared with fenofibrate in mice. GFT505, but not fenofibrate, increased the expression of HDL production genes such as apolipoprotein-AI and ATP-binding cassette A1 transporter in murine intestines. A similar increase was observed upon PPAR-α activation of human biopsies and Caco-2/TC7 cells. Additionally, HDL secretion by Caco-2/TC7 cells increased. Moreover, PPAR-α activation decreased the cholesterol esterification capacity of Caco-2/TC7 cells, modified cholesterol trafficking, and reduced apolipoprotein-B secretion. CONCLUSION Peroxisome proliferator-activated receptor-α activation reduces cholesterol esterification, suppresses chylomicron, and increases HDL secretion by enterocytes. These results identify the intestine as a target organ of PPAR-α ligands with entero-hepatic tropism to reduce atherogenic dyslipidaemia.
Collapse
Affiliation(s)
- Sophie Colin
- Université Lille Nord de France, Lille F-59000, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Respective contributions of intestinal Niemann-Pick C1-like 1 and scavenger receptor class B type I to cholesterol and tocopherol uptake: in vivo v. in vitro studies. Br J Nutr 2011; 107:1296-304. [PMID: 21929836 DOI: 10.1017/s0007114511004405] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
The intestinal absorption of cholesterol and lipid micronutrients such as vitamin E has been shown to share some common pathways. The present study aims to further compare the uptake of cholesterol ([3H]cholesterol v. 22-(N-(7-nitrobenz-2-oxa-1,3-diazol-4-yl)amino)-23,24-bisnor-5-cholen-3-ol (NBD-cholesterol)) and tocopherol in Caco-2 TC-7 cells and in mouse intestine, with special focus on the respective roles of scavenger receptor class B type I (SR-BI) and Niemann-Pick C1-like 1 (NPC1L1). Conversely to NBD-cholesterol, the uptakes of [3H]cholesterol and tocopherol by Caco-2 cells were impaired by both block lipid transport-1 and ezetimibe, which inhibit SR-BI and NPC1L1, respectively. These inhibitions occurred only when cholesterol or tocopherol was delivered to cells included in micelles that contained biliary acid and at least oleic acid as a lipid. In vivo, after 2 h of digestion in mice, the uptake of the two cholesterol analogues and of tocopherol all showed distinct patterns along the duodenum-jejunum axis. [3H]Cholesterol uptake, which correlated closely to NPC1L1 mRNA expression in wild-type (wt) mice, was strongly inhibited by ezetimibe. Intestinal SR-BI overexpression did not change NPC1L1 expression and led to a significant increase in [3H]cholesterol uptake in the distal jejunum. Conversely, neither ezetimibe treatment nor SR-BI overexpression had an effect on NBD-cholesterol uptake. However, in contrast with SR-BI mRNA expression, tocopherol absorption increased strongly up to the distal jejunum in wt mice where it was specifically inhibited by ezetimibe, and was increased in the proximal intestine of intestinal SR-BI-overexpressing mice. Thus, cholesterol and tocopherol uptakes share common pathways in cell culture models, but display different in vivo absorption patterns associated with distinct contributions of SR-BI and NPC1L1.
Collapse
|
33
|
Xiao C, Hsieh J, Adeli K, Lewis GF. Gut-liver interaction in triglyceride-rich lipoprotein metabolism. Am J Physiol Endocrinol Metab 2011; 301:E429-46. [PMID: 21693689 DOI: 10.1152/ajpendo.00178.2011] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The liver and intestine have complementary and coordinated roles in lipoprotein metabolism. Despite their highly specialized functions, assembly and secretion of triglyceride-rich lipoproteins (TRL; apoB-100-containing VLDL in the liver and apoB-48-containing chylomicrons in the intestine) are regulated by many of the same hormonal, inflammatory, nutritional, and metabolic factors. Furthermore, lipoprotein metabolism in these two organs may be affected in a similar fashion by certain disorders. In insulin resistance, for example, overproduction of TRL by both liver and intestine is a prominent component of and underlies other features of a complex dyslipidemia and increased risk of atherosclerosis. The intestine is gaining increasing recognition for its importance in affecting whole body lipid homeostasis, in part through its interaction with the liver. This review aims to integrate recent advances in our understanding of these processes and attempts to provide insight into the factors that coordinate lipid homeostasis in these two organs in health and disease.
Collapse
|
34
|
Hayashi AA, Webb J, Choi J, Baker C, Lino M, Trigatti B, Trajcevski KE, Hawke TJ, Adeli K. Intestinal SR-BI is upregulated in insulin-resistant states and is associated with overproduction of intestinal apoB48-containing lipoproteins. Am J Physiol Gastrointest Liver Physiol 2011; 301:G326-37. [PMID: 21546579 DOI: 10.1152/ajpgi.00425.2010] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Intestinal lipid dysregulation is a common feature of insulin-resistant states. The present study investigated alterations in gene expression of key proteins involved in the active absorption of dietary fat and cholesterol in response to development of insulin resistance. Studies were conducted in two diet-induced animal models of insulin resistance: fructose-fed hamster and high-fat-fed mouse. Changes in the mRNA abundance of lipid transporters, adenosine triphosphate cassette (ABC) G5, ABCG8, FA-CoA ligase fatty acid translocase P4, Niemann-Pick C1-Like1 (NPC1L1), fatty acid transport protein 4 (FATP4), and Scavenger Receptor Class B Type I (SR-BI), were assessed in intestinal fragments (duodenum, jejunum, and ileum) using quantitative real-time PCR. Of all the transporters evaluated, SR-B1 showed the most significant changes in both animal models examined. A marked stimulation of SR-B1 expression was observed in all intestinal segments examined in both insulin-resistant animal models. The link between SR-BI expression and intestinal lipoprotein production was then examined in the Caco-2 cell model. SR-B1 overexpression in Caco-2 cells increased apolipoprotein B (apoB) 100 and apoB48 secretion, whereas RNAi knock down of SR-B1 decreased secretion of both apoB100 and apoB48. We also observed changes in subcellular distribution of SR-B1 in response to exogenous lipid and insulin. Confocal microscopy revealed marked changes in SR-BI subcellular distribution in response to both exogenous lipids (oleate) and insulin. In summary, marked stimulation of intestinal SR-BI occurs in vivo in animal models of diet-induced insulin resistance, and modulation of SR-BI in vitro regulates production of apoB-containing lipoprotein particles. We postulate that apical and/or basolateral SR-BI may play an important role in intestinal chylomicron production and may contribute to chylomicron overproduction normally observed in insulin-resistant states.
Collapse
Affiliation(s)
- Amanda A Hayashi
- Molecular Structure & Function, Research Institute, The Hospital for Sick Children, University of Toronto, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Aspects of Future R&D Regarding Targeted Lipid Nanoemulsions. ACTA ACUST UNITED AC 2011. [DOI: 10.1016/b978-0-444-53798-0.00029-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
|
36
|
Julia Z, Duchene E, Fournier N, Bellanger N, Chapman MJ, Le Goff W, Guerin M. Postprandial lipemia enhances the capacity of large HDL2 particles to mediate free cholesterol efflux via SR-BI and ABCG1 pathways in type IIB hyperlipidemia. J Lipid Res 2010; 51:3350-8. [DOI: 10.1194/jlr.p009746] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
37
|
Clifton JD, Lucumi E, Myers MC, Napper A, Hama K, Farber SA, Smith AB, Huryn DM, Diamond SL, Pack M. Identification of novel inhibitors of dietary lipid absorption using zebrafish. PLoS One 2010; 5:e12386. [PMID: 20811635 PMCID: PMC2928291 DOI: 10.1371/journal.pone.0012386] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2010] [Accepted: 06/11/2010] [Indexed: 11/19/2022] Open
Abstract
Pharmacological inhibition of dietary lipid absorption induces favorable changes in serum lipoprotein levels in patients that are at risk for cardiovascular disease and is considered an adjuvant or alternative treatment with HMG-CoA reductase inhibitors (statins). Here we demonstrate the feasibility of identifying novel inhibitors of intestinal lipid absorption using the zebrafish system. A pilot screen of an unbiased chemical library identified novel compounds that inhibited processing of fluorescent lipid analogues in live zebrafish larvae. Secondary assays identified those compounds suitable for testing in mammals and provided insight into mechanism of action, which for several compounds could be distinguished from ezetimibe, a drug used to inhibit cholesterol absorption in humans that broadly inhibited lipid absorption in zebrafish larvae. These findings support the utility of zebrafish screening assays to identify novel compounds that target complex physiological processes.
Collapse
Affiliation(s)
- Justin D. Clifton
- Department Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Edinson Lucumi
- Penn Center for Molecular Discovery, Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Michael C. Myers
- Department of Chemistry, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Andrew Napper
- Department of Chemistry, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Kotaro Hama
- Department of Embryology, Carnegie Institution, Baltimore, Maryland, United States of America
| | - Steven A. Farber
- Department of Embryology, Carnegie Institution, Baltimore, Maryland, United States of America
| | - Amos B. Smith
- Penn Center for Molecular Discovery, Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- Department of Chemistry, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Donna M. Huryn
- Penn Center for Molecular Discovery, Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- Department of Chemistry, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Scott L. Diamond
- Penn Center for Molecular Discovery, Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- Department of Chemistry, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Michael Pack
- Department Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
- Department of Cell and Developmental Biology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
| |
Collapse
|
38
|
Abstract
PURPOSE OF REVIEW Remnant lipoproteins that persist in the bloodstream after each meal have become increasingly important contributors to atherosclerotic vascular disease, owing to the spread of overnutrition, underexertion, obesity, insulin resistance, and type 2 diabetes. Here, we review recent work that clarified long-standing controversies over the molecular mediators of remnant clearance by the liver, as well as their dysregulation - but possible correction - during alterations in caloric balance. RECENT FINDINGS Two endocytic receptors, the syndecan-1 heparan sulfate proteoglycan (HSPG) and the LDL receptor, plus one docking receptor, SR-BI, significantly contribute to normal hepatic remnant catabolism. Compelling evidence exists for dysfunction of the syndecan-1 HSPG in diabetic states. The major molecular defect identified so far in poorly controlled type 1 diabetes is impaired hepatic HSPG assembly. In contrast, the primary defect in hepatic HSPGs in type 2 diabetes appears to arise from accelerated de-sulfation, owing to the induction of a sulfatase. Moreover, short-term caloric restriction restores hepatic expression of this sulfatase towards normal. SUMMARY Correct identification of hepatic remnant receptors has finally allowed investigations of their molecular dysregulation in diabetes and related conditions. New work points to novel therapeutic targets to correct postprandial dyslipoproteinemia and its consequent arterial damage.
Collapse
Affiliation(s)
- Kevin Jon Williams
- Temple University School of Medicine, Philadelphia, Pennsylvania 19140, USA.
| | | |
Collapse
|
39
|
|