1
|
Caleffi‐Marchesini ER, Herling AA, Macente J, Bonan RH, de Freitas Lima P, Moreno R, Alexandre V, Charbe NB, Borghi‐Pangoni FB, Cristofoletti R, Diniz A. Adult and pediatric physiologically-based biopharmaceutics modeling to explain lamotrigine immediate release absorption process. CPT Pharmacometrics Syst Pharmacol 2024; 13:208-221. [PMID: 37916262 PMCID: PMC10864931 DOI: 10.1002/psp4.13071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 09/17/2023] [Accepted: 09/25/2023] [Indexed: 11/03/2023] Open
Abstract
Physiologically-based biopharmaceutics modeling (PBBM) has potential to accelerate the development of new drug and formulations. An important application of PBBM is for special populations such as pediatrics that have pharmacokinetics dependent on the maturation process. Lamotrigine (LTG) is a Biopharmaceutics Classification System (BCS) II drug and is widely prescribed. Therefore, the goal of this study was to assess the biopharmaceutics risk of the low-soluble drug LTG when the ontogeny on gastrointestinal tract (GIT) physiological parameters are considered. An oral physiologically-based pharmacokinetic model and a PBBM were developed and verified using GastroPlus™ software for both adults and children (2-12 years old, 12-52 kg). The biopharmaceutics properties and GIT physiological parameters were evaluated by sensitivity analysis. High doses were simulated assuming a worst case scenario, that is, the dose of 200 mg for adults and 5 mg/kg (up to the maximum of 200 mg) for 2-year-old children. Although several authors have suggested that ontogeny may have an effect on gastrointestinal fluid volume, our study found no evidence of interference between fluid and dose volumes with in vivo dissolution of LTG. The most impactful parameter was found to be the gastric transit time. Therefore, the hypothesis is developed to examine whether LTG exhibits characteristics of a BCS II classification in vitro while showing BCS I-like behavior in vivo. This hypothesis could act as a base for conducting novel studies on model-informed precision dosing, tailored to specific populations and clinical conditions. In addition, it could be instrumental in assessing the influence of various release profiles on in vivo performance for both adult and pediatric populations.
Collapse
Affiliation(s)
| | - Amanda Antunes Herling
- Pharmacokinetics and Biopharmaceutics LaboratoryState University of MaringáMaringáPRBrazil
| | - Julia Macente
- Pharmacokinetics and Biopharmaceutics LaboratoryState University of MaringáMaringáPRBrazil
| | | | | | - Rafaela Moreno
- Centro Universitário Barão de MauáRibeirão PretoSPBrazil
| | - Veriano Alexandre
- Hospital das Clínicas, Faculdade de MedicinaUniversidade de São PauloRibeirão PretoSPBrazil
| | - Nitin Bharat Charbe
- Center for Pharmacometrics & Systems PharmacologyUniversity of FloridaOrlandoFloridaUSA
| | | | - Rodrigo Cristofoletti
- Center for Pharmacometrics & Systems PharmacologyUniversity of FloridaOrlandoFloridaUSA
| | - Andréa Diniz
- Pharmacokinetics and Biopharmaceutics LaboratoryState University of MaringáMaringáPRBrazil
| |
Collapse
|
2
|
Eroglu A, Al'Abri IS, Kopec RE, Crook N, Bohn T. Carotenoids and Their Health Benefits as Derived via Their Interactions with Gut Microbiota. Adv Nutr 2023; 14:238-255. [PMID: 36775788 DOI: 10.1016/j.advnut.2022.10.007] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 09/21/2022] [Accepted: 10/28/2022] [Indexed: 12/23/2022] Open
Abstract
Carotenoids have been related to a number of health benefits. Their dietary intake and circulating levels have been associated with a reduced incidence of obesity, diabetes, certain types of cancer, and even lower total mortality. Their potential interaction with the gut microbiota (GM) has been generally overlooked but may be of relevance, as carotenoids largely bypass absorption in the small intestine and are passed on to the colon, where they appear to be in part degraded into unknown metabolites. These may include apo-carotenoids that may have biological effects because of higher aqueous solubility and higher electrophilicity that could better target transcription factors, i.e., NF-κB, PPARγ, and RAR/RXRs. If absorbed in the colon, they could have both local and systemic effects. Certain microbes that may be supplemented were also reported to produce carotenoids in the colon. Although some bactericidal aspects of carotenoids have been shown in vitro, a few studies have also demonstrated a prebiotic-like effect, resulting in bacterial shifts with health-associated properties. Also, stimulation of IgA could play a role in this respect. Carotenoids may further contribute to mucosal and gut barrier health, such as stabilizing tight junctions. This review highlights potential gut-related health-beneficial effects of carotenoids and emphasizes the current research gaps regarding carotenoid-GM interactions.
Collapse
Affiliation(s)
- Abdulkerim Eroglu
- Department of Molecular and Structural Biochemistry, College of Agriculture and Life Sciences, North Carolina State University, Raleigh, NC, USA; Plants for Human Health Institute, North Carolina Research Campus, North Carolina State University, Kannapolis, NC, USA.
| | - Ibrahim S Al'Abri
- Department of Chemical and Biomolecular Engineering, College of Engineering, North Carolina State University, Raleigh, NC, USA
| | - Rachel E Kopec
- Human Nutrition Program, Department of Human Sciences, The Ohio State University, Columbus, OH, USA; Foods for Health Discovery Theme, The Ohio State University, Columbus, OH, USA
| | - Nathan Crook
- Department of Chemical and Biomolecular Engineering, College of Engineering, North Carolina State University, Raleigh, NC, USA
| | - Torsten Bohn
- Nutrition and Health Research Group, Department of Precision Health, Luxembourg Institute of Health, rue 1 A-B, Thomas Edison, L-1445 Strassen, Luxembourg.
| |
Collapse
|
3
|
Borel P, Dangles O, Kopec RE. Fat-soluble vitamin and phytochemical metabolites: Production, gastrointestinal absorption, and health effects. Prog Lipid Res 2023; 90:101220. [PMID: 36657621 DOI: 10.1016/j.plipres.2023.101220] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 12/12/2022] [Accepted: 01/12/2023] [Indexed: 01/18/2023]
Abstract
Consumption of diets rich in fruits and vegetables, which provide some fat-soluble vitamins and many phytochemicals, is associated with a lower risk of developing certain degenerative diseases. It is well accepted that not only the parent compounds, but also their derivatives formed upon enzymatic or nonenzymatic transformations, can produce protective biological effects. These derivatives can be formed during food storage, processing, or cooking. They can also be formed in the lumen of the upper digestive tract during digestion, or via metabolism by microbiota in the colon. This review compiles the known metabolites of fat-soluble vitamins and fat-soluble phytochemicals (FSV and FSP) that have been identified in food and in the human digestive tract, or could potentially be present based on the known reactivity of the parent compounds in normal or pathological conditions, or following surgical interventions of the digestive tract or consumption of xenobiotics known to impair lipid absorption. It also covers the very limited data available on the bioavailability (absorption, intestinal mucosa metabolism) and summarizes their effects on health. Notably, despite great interest in identifying bioactive derivatives of FSV and FSP, studying their absorption, and probing their putative health effects, much research remains to be conducted to understand and capitalize on the potential of these molecules to preserve health.
Collapse
Affiliation(s)
- Patrick Borel
- C2VN, INRAE, INSERM, Aix-Marseille Univ, Marseille, France.
| | | | - Rachel E Kopec
- Human Nutrition Program, Department of Human Sciences, Foods for Health Discovery Theme, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
4
|
Efficacy of Polyphenols in the Management of Dyslipidemia: A Focus on Clinical Studies. Nutrients 2021; 13:nu13020672. [PMID: 33669729 PMCID: PMC7922034 DOI: 10.3390/nu13020672] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/15/2021] [Accepted: 02/16/2021] [Indexed: 02/06/2023] Open
Abstract
Polyphenols (PLPs), phytochemicals found in a wide range of plant-based foods, have gained extensive attention in view of their antioxidant, anti-inflammatory, immunomodulatory and several additional beneficial activities. The health-promoting effects noted in animal models of various non-communicable diseases explain the growing interest in these molecules. In particular, in vitro and animal studies reported an attenuation of lipid disorders in response to PLPs. However, despite promising preclinical investigations, the effectiveness of PLPs in human dyslipidemia (DLP) is less clear and necessitates revision of available literature. Therefore, the present review analyzes the role of PLPs in managing clinical DLP, notably by dissecting their potential in ameliorating lipid/lipoprotein metabolism and alleviating hyperlipidemia, both postprandially and in long-term interventions. To this end, PubMed was used for article search. The search terms included polyphenols, lipids, triglycerides, cholesterol, LDL-cholesterol and /or HDL-cholesterol. The critical examination of the trials published to date illustrates certain benefits on blood lipids along with co-morbidities in participant’s health status. However, inconsistent results document significant research gaps, potentially owing to study heterogeneity and lack of rigor in establishing PLP bioavailability during supplementation. This underlines the need for further efforts in order to elucidate and support a potential role of PLPs in fighting DLP.
Collapse
|
5
|
Levy E, Beaulieu JF, Spahis S. From Congenital Disorders of Fat Malabsorption to Understanding Intra-Enterocyte Mechanisms Behind Chylomicron Assembly and Secretion. Front Physiol 2021; 12:629222. [PMID: 33584351 PMCID: PMC7873531 DOI: 10.3389/fphys.2021.629222] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 01/06/2021] [Indexed: 11/13/2022] Open
Abstract
During the last two decades, a large body of information on the events responsible for intestinal fat digestion and absorption has been accumulated. In particular, many groups have extensively focused on the absorptive phase in order to highlight the critical "players" and the main mechanisms orchestrating the assembly and secretion of chylomicrons (CM) as essential vehicles of alimentary lipids. The major aim of this article is to review understanding derived from basic science and clinical conditions associated with impaired packaging and export of CM. We have particularly insisted on inborn metabolic pathways in humans as well as on genetically modified animal models (recapitulating pathological features). The ultimate goal of this approach is that "experiments of nature" and in vivo model strategy collectively allow gaining novel mechanistic insight and filling the gap between the underlying genetic defect and the apparent clinical phenotype. Thus, uncovering the cause of disease contributes not only to understanding normal physiologic pathway, but also to capturing disorder onset, progression, treatment and prognosis.
Collapse
Affiliation(s)
- Emile Levy
- Research Centre, CHU Ste-Justine, Université de Montréal, Montreal, QC, Canada
- Department of Nutrition, Université de Montréal, Montreal, QC, Canada
- Department of Pediatrics, Université de Montréal, Montreal, QC, Canada
| | - Jean François Beaulieu
- Laboratory of Intestinal Physiopathology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Schohraya Spahis
- Research Centre, CHU Ste-Justine, Université de Montréal, Montreal, QC, Canada
- Department of Nutrition, Université de Montréal, Montreal, QC, Canada
| |
Collapse
|
6
|
Schachter J, Martel J, Lin CS, Chang CJ, Wu TR, Lu CC, Ko YF, Lai HC, Ojcius DM, Young JD. Effects of obesity on depression: A role for inflammation and the gut microbiota. Brain Behav Immun 2018; 69:1-8. [PMID: 28888668 DOI: 10.1016/j.bbi.2017.08.026] [Citation(s) in RCA: 148] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2017] [Revised: 08/29/2017] [Accepted: 08/31/2017] [Indexed: 12/14/2022] Open
Abstract
Depression is a mental disorder associated with environmental, genetic and psychological factors. Recent studies indicate that chronic neuro-inflammation may affect brain physiology and alter mood and behavior. Consumption of a high-fat diet leads to obesity and chronic systemic inflammation. The gut microbiota mediates many effects of a high-fat diet on human physiology and may also influence the mood and behavior of the host. We review here recent studies suggesting the existence of a link between obesity, the gut microbiota and depression, focusing on the mechanisms underlying the effects of a high-fat diet on chronic inflammation and brain physiology. This body of research suggests that modulating the composition of the gut microbiota using prebiotics and probiotics may produce beneficial effects on anxiety and depression.
Collapse
Affiliation(s)
- Julieta Schachter
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal de Rio de Janeiro, 21941-902, 373 Avenida Carlos Chagas Filho, Cidade Universitária - Ilha do Fundão, Rio de Janeiro, Brazil; Center for Molecular and Clinical Immunology, Chang Gung University, 259 Wen-Hua First Road, Taoyuan 33302, Taiwan
| | - Jan Martel
- Center for Molecular and Clinical Immunology, Chang Gung University, 259 Wen-Hua First Road, Taoyuan 33302, Taiwan; Chang Gung Immunology Consortium, Linkou Chang Gung Memorial Hospital, 5 Fu-Hsing Street, Taoyuan 33305, Taiwan
| | - Chuan-Sheng Lin
- Center for Molecular and Clinical Immunology, Chang Gung University, 259 Wen-Hua First Road, Taoyuan 33302, Taiwan; Chang Gung Immunology Consortium, Linkou Chang Gung Memorial Hospital, 5 Fu-Hsing Street, Taoyuan 33305, Taiwan; Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, 259 Wen-Hua First Road, Taoyuan 33302, Taiwan; Research Center of Bacterial Pathogenesis, Chang Gung University, 259 Wen-Hua First Road, Taoyuan 33302, Taiwan; Department of Microbiology and Immunology, College of Medicine, Chang Gung University, 259 Wen-Hua First Road, Taoyuan 33302, Taiwan
| | - Chih-Jung Chang
- Center for Molecular and Clinical Immunology, Chang Gung University, 259 Wen-Hua First Road, Taoyuan 33302, Taiwan; Chang Gung Immunology Consortium, Linkou Chang Gung Memorial Hospital, 5 Fu-Hsing Street, Taoyuan 33305, Taiwan; Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, 259 Wen-Hua First Road, Taoyuan 33302, Taiwan; Research Center of Bacterial Pathogenesis, Chang Gung University, 259 Wen-Hua First Road, Taoyuan 33302, Taiwan; Department of Microbiology and Immunology, College of Medicine, Chang Gung University, 259 Wen-Hua First Road, Taoyuan 33302, Taiwan
| | - Tsung-Ru Wu
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, 259 Wen-Hua First Road, Taoyuan 33302, Taiwan
| | - Chia-Chen Lu
- Department of Respiratory Therapy, Fu Jen Catholic University, 510 Zhong-Zheng Street, New Taipei City 24205, Taiwan
| | - Yun-Fei Ko
- Chang Gung Immunology Consortium, Linkou Chang Gung Memorial Hospital, 5 Fu-Hsing Street, Taoyuan 33305, Taiwan; Chang Gung Biotechnology Corporation, 201 Tung-Hua North Road, Taipei 10508, Taiwan; Biochemical Engineering Research Center, Ming Chi University of Technology, 84 Gungjuan Road, New Taipei City 24301, Taiwan
| | - Hsin-Chih Lai
- Center for Molecular and Clinical Immunology, Chang Gung University, 259 Wen-Hua First Road, Taoyuan 33302, Taiwan; Chang Gung Immunology Consortium, Linkou Chang Gung Memorial Hospital, 5 Fu-Hsing Street, Taoyuan 33305, Taiwan; Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, 259 Wen-Hua First Road, Taoyuan 33302, Taiwan; Research Center of Bacterial Pathogenesis, Chang Gung University, 259 Wen-Hua First Road, Taoyuan 33302, Taiwan; Department of Microbiology and Immunology, College of Medicine, Chang Gung University, 259 Wen-Hua First Road, Taoyuan 33302, Taiwan; Department of Laboratory Medicine, Linkou Chang Gung Memorial Hospital, 5 Fu-Hsing Street, Taoyuan 33305, Taiwan; Research Center for Industry of Human Ecology, College of Human Ecology, Chang Gung University of Science and Technology, 261 Wen-Hua First Road, Taoyuan 33303, Taiwan; Graduate Institute of Health Industry and Technology, College of Human Ecology, Chang Gung University of Science and Technology, 261 Wen-Hua First Road, Taoyuan 33303, Taiwan
| | - David M Ojcius
- Center for Molecular and Clinical Immunology, Chang Gung University, 259 Wen-Hua First Road, Taoyuan 33302, Taiwan; Chang Gung Immunology Consortium, Linkou Chang Gung Memorial Hospital, 5 Fu-Hsing Street, Taoyuan 33305, Taiwan; Department of Biomedical Sciences, University of the Pacific, Arthur Dugoni School of Dentistry, 155 Fifth Street, San Francisco, CA 94103, USA
| | - John D Young
- Center for Molecular and Clinical Immunology, Chang Gung University, 259 Wen-Hua First Road, Taoyuan 33302, Taiwan; Chang Gung Immunology Consortium, Linkou Chang Gung Memorial Hospital, 5 Fu-Hsing Street, Taoyuan 33305, Taiwan; Chang Gung Biotechnology Corporation, 201 Tung-Hua North Road, Taipei 10508, Taiwan; Biochemical Engineering Research Center, Ming Chi University of Technology, 84 Gungjuan Road, New Taipei City 24301, Taiwan; Laboratory of Cellular Physiology and Immunology, Rockefeller University, 1230 York Avenue, New York, NY 10021, USA.
| |
Collapse
|
7
|
Auclair N, Melbouci L, St-Pierre D, Levy E. Gastrointestinal factors regulating lipid droplet formation in the intestine. Exp Cell Res 2018; 363:1-14. [PMID: 29305172 DOI: 10.1016/j.yexcr.2017.12.031] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Revised: 12/28/2017] [Accepted: 12/29/2017] [Indexed: 12/22/2022]
Abstract
Cytoplasmic lipid droplets (CLD) are considered as neutral lipid reservoirs, which protect cells from lipotoxicity. It became clear that these fascinating dynamic organelles play a role not only in energy storage and metabolism, but also in cellular lipid and protein handling, inter-organelle communication, and signaling among diverse functions. Their dysregulation is associated with multiple disorders, including obesity, liver steatosis and cardiovascular diseases. The central aim of this review is to highlight the link between intra-enterocyte CLD dynamics and the formation of chylomicrons, the main intestinal dietary lipid vehicle, after overviewing the morphology, molecular composition, biogenesis and functions of CLD.
Collapse
Affiliation(s)
- N Auclair
- Research Centre, CHU Sainte-Justine and Department of Montreal, Quebec, Canada H3T 1C5; Nutrition, Université de Montréal, Montreal, Quebec, Canada H3T 1C5
| | - L Melbouci
- Research Centre, CHU Sainte-Justine and Department of Montreal, Quebec, Canada H3T 1C5; Department of Sciences and Physical Activities, UQAM, Quebec, Canada H2X 1Y4
| | - D St-Pierre
- Research Centre, CHU Sainte-Justine and Department of Montreal, Quebec, Canada H3T 1C5; Department of Sciences and Physical Activities, UQAM, Quebec, Canada H2X 1Y4
| | - E Levy
- Research Centre, CHU Sainte-Justine and Department of Montreal, Quebec, Canada H3T 1C5; Nutrition, Université de Montréal, Montreal, Quebec, Canada H3T 1C5; Institute of Nutrition and Functional Foods (INAF), Université Laval, Quebec, Quebec, Canada G1V 0A6.
| |
Collapse
|
8
|
Poquet L, Wooster TJ. Infant digestion physiology and the relevance of in vitro biochemical models to test infant formula lipid digestion. Mol Nutr Food Res 2017; 60:1876-95. [PMID: 27279140 DOI: 10.1002/mnfr.201500883] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Revised: 03/05/2016] [Accepted: 05/31/2016] [Indexed: 01/30/2023]
Abstract
Lipids play an important role in the diet of preterm and term infants providing a key energy source and essential lipid components for development. While a lot is known about adult lipid digestion, our understanding of infant digestion physiology is still incomplete, the greatest gap being on the biochemistry of the small intestine, particularly the activity and relative importance of the various lipases active in the intestine. The literature has been reviewed to identify the characteristics of lipid digestion of preterm and term infants, but also to better understand the physiology of the infant gastrointestinal tract compared to adults that impacts the absorption of lipids. The main differences are a higher gastric pH, submicellar bile salt concentration, a far more important role of gastric lipases as well as differences at the level of the intestinal barrier. Importantly, the consequences of improper in vitro replication of gastric digestions conditions (pH and lipase specificity) are demonstrated using examples from the most recent of studies. It is true that some animal models could be adapted to study infant lipid digestion physiology, however the ethical relevance of such models is questionable, hence the development of accurate in vitro models is a must. In vitro models that combine up to date knowledge of digestion biochemistry with intestinal cells in culture are the best choice to replicate digestion and absorption in infant population, this would allow the adaptation of infant formula for a better digestion and absorption of dietary lipids by preterm and term infants.
Collapse
Affiliation(s)
- Laure Poquet
- Nestlé Research Center, Vers-Chez-Les-Blanc, Lausanne 26, Switzerland
| | - Tim J Wooster
- Nestlé Research Center, Vers-Chez-Les-Blanc, Lausanne 26, Switzerland
| |
Collapse
|
9
|
Guerville M, Boudry G. Gastrointestinal and hepatic mechanisms limiting entry and dissemination of lipopolysaccharide into the systemic circulation. Am J Physiol Gastrointest Liver Physiol 2016; 311:G1-G15. [PMID: 27151941 DOI: 10.1152/ajpgi.00098.2016] [Citation(s) in RCA: 116] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Accepted: 04/30/2016] [Indexed: 01/31/2023]
Abstract
The human microbiota consists of 100 trillion microorganisms that provide important metabolic and biological functions benefiting the host. However, the presence in host plasma of a gut-derived bacteria component, the lipopolysaccharide (LPS), has been identified as a causal or complicating factor in multiple serious diseases such as sepsis and septic shock and, more recently, obesity-associated metabolic disorders. Understanding the precise mechanisms by which gut-derived LPS is transported from the gut lumen to the systemic circulation is crucial to advance our knowledge of LPS-associated diseases and elaborate targeted strategies for their prevention. The aim of this review is to synthetize current knowledge on the host mechanisms limiting the entry and dissemination of LPS into the systemic circulation. To prevent bacterial colonization and penetration, the intestinal epithelium harbors multiple defense mechanisms including the secretion of antimicrobial peptides and mucins as well as detoxification enzymes. Despite this first line of defense, LPS can reach the apical site of intestinal epithelial cells (IECs) and, because of its large size, likely crosses IECs via transcellular transport, either lipid raft- or clathrin-mediated endocytosis or goblet cell-associated passage. However, the precise pathway remains poorly described. Finally, if LPS crosses the gut mucosa, it is directed via the portal vein to the liver, where major detoxification processes occur by deacetylation and excretion through the bile. If this disposal process is not sufficient, LPS enters the systemic circulation, where it is handled by numerous transport proteins that clear it back to the liver for further excretion.
Collapse
Affiliation(s)
| | - Gaëlle Boudry
- INRA UR1341 ADNC, Domaine de la Prise, Saint-Gilles, France
| |
Collapse
|
10
|
Prevention of oxidative stress, inflammation and mitochondrial dysfunction in the intestine by different cranberry phenolic fractions. Clin Sci (Lond) 2014; 128:197-212. [PMID: 25069567 DOI: 10.1042/cs20140210] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Cranberry fruit has been reported to have high antioxidant effectiveness that is potentially linked to its richness in diversified polyphenolic content. The aim of the present study was to determine the role of cranberry polyphenolic fractions in oxidative stress (OxS), inflammation and mitochondrial functions using intestinal Caco-2/15 cells. The combination of HPLC and UltraPerformance LC®-tandem quadrupole (UPLC-TQD) techniques allowed us to characterize the profile of low, medium and high molecular mass polyphenolic compounds in cranberry extracts. The medium molecular mass fraction was enriched with flavonoids and procyanidin dimers whereas procyanidin oligomers (DP > 4) were the dominant class of polyphenols in the high molecular mass fraction. Pre-incubation of Caco-2/15 cells with these cranberry extracts prevented iron/ascorbate-mediated lipid peroxidation and counteracted lipopolysaccharide-mediated inflammation as evidenced by the decrease in pro-inflammatory cytokines (TNF-α and interleukin-6), cyclo-oxygenase-2 and prostaglandin E2. Cranberry polyphenols (CP) fractions limited both nuclear factor κB activation and Nrf2 down-regulation. Consistently, cranberry procyanidins alleviated OxS-dependent mitochondrial dysfunctions as shown by the rise in ATP production and the up-regulation of Bcl-2, as well as the decline of protein expression of cytochrome c and apoptotic-inducing factor. These mitochondrial effects were associated with a significant stimulation of peroxisome-proliferator-activated receptor γ co-activator-1-α, a central inducing factor of mitochondrial biogenesis and transcriptional co-activator of numerous downstream mediators. Finally, cranberry procyanidins forestalled the effect of iron/ascorbate on the protein expression of mitochondrial transcription factors (mtTFA, mtTFB1, mtTFB2). Our findings provide evidence for the capacity of CP to reduce intestinal OxS and inflammation while improving mitochondrial dysfunction.
Collapse
|
11
|
Grenier E, Mailhot G, Dion D, Ravid Z, Spahis S, Bendayan M, Levy E. Role of the apical and basolateral domains of the enterocyte in the regulation of cholesterol transport by a high glucose concentration. Biochem Cell Biol 2013; 91:476-86. [PMID: 24219290 DOI: 10.1139/bcb-2013-0053] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
We have recently shown that a high glucose (HG) concentration raised intestinal cholesterol (CHOL) transport and metabolism in intestinal epithelial cells. The objective of the present work is to determine whether the stimulus for increased CHOL absorption by glucose originates from the apical site (corresponding to the intestinal lumen) or from the basolateral site (related to blood circulation). We tackled this issue by using differentiated Caco-2/15 cells. Only basolateral medium, supplemented with 25 mmol/L glucose, stimulated [(14)C]-CHOL uptake via the up-regulation of the critical CHOL transporter NPC1L1 protein, as confirmed by its specific ezetimibe inhibitor that abolished the rise in glucose-mediated CHOL capture. No significant changes were noted in SR-BI and CD36. Elevated CHOL uptake was associated with an increase in the transcription factors SREBP-2, LXR-β, and ChREBP, along with a fall in RXR-α. Interestingly, although the HG concentration in the apical medium caused modest changes in CHOL processing, its impact was synergetic with that of the basolateral medium. Our results suggest that HG concentration influences positively intestinal CHOL uptake when present in the basolateral medium. In addition, excessive consumption of diets containing high levels of carbohydrates may strengthen intestinal CHOL uptake in metabolic syndrome, thereby contributing to elevated levels of circulating CHOL and, consequently, the risk of developing type 2 diabetes and cardiovascular disease.
Collapse
Affiliation(s)
- Emilie Grenier
- a Research Centre, CHU Ste-Justine, 3175 Ste-Catherine Road, Montreal, QC H3T 1C5, Canada
| | | | | | | | | | | | | |
Collapse
|
12
|
Levy E, Ben Djoudi Ouadda A, Spahis S, Sane AT, Garofalo C, Grenier É, Emonnot L, Yara S, Couture P, Beaulieu JF, Ménard D, Seidah NG, Elchebly M. PCSK9 plays a significant role in cholesterol homeostasis and lipid transport in intestinal epithelial cells. Atherosclerosis 2013; 227:297-306. [PMID: 23422832 DOI: 10.1016/j.atherosclerosis.2013.01.023] [Citation(s) in RCA: 106] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2012] [Revised: 01/14/2013] [Accepted: 01/16/2013] [Indexed: 12/30/2022]
Abstract
OBJECTIVES The proprotein convertase subtillisin/kexin type 9 (PCSK9) regulates cholesterol metabolism via degradation of low-density lipoprotein receptor (LDLr). Although PCSK9 is abundantly expressed in the intestine, limited data are available on its functions. The present study aims at determining whether PCSK9 plays important roles in cholesterol homeostasis and lipid transport in the gut. METHODS AND RESULTS Caco-2/15 cells were used allowing the exploration of the PCSK9 secretory route through the apical and basolateral compartments corresponding to intestinal lumen and serosal circulation, respectively. The output of PCSK9 occurred through the basolateral membrane, a site characterized by the location of LDLr. Co-immunoprecipitation studies indicated an association between PCSK9 and LDLr. Addition of purified recombinant wild type and D374Y gain-of function PCSK9 proteins to the basolateral medium was followed by a decrease in LDLr concomitantly with the accumulation of both forms of PCSK9. Furthermore, the latter caused a significant enhancement in cholesterol uptake also evidenced by a raised protein expression of cholesterol transporters NPC1L1 and CD36 without changes in SR-BI, ABCA1, and ABCG5/G8. Moreover, exogenous PCSK9 altered the activity of HMG-CoA reductase and acylcoenzyme A: cholesterol acyltransferase, and was able to enhance chylomicron secretion by positively modulating lipids and apolipoprotein B-48 biogenesis. Importantly, PCSK9 silencing led to opposite findings, which validate our data on the role of PCSK9 in lipid transport and metabolism. Moreover, PCSK9-mediated changes persisted despite LDLr knockdown. CONCLUSIONS These findings indicate that, in addition to its effect on LDLr, PCSK9 modulates cholesterol transport and metabolism, as well as production of apo B-containing lipoproteins in intestinal cells.
Collapse
Affiliation(s)
- Emile Levy
- Research Centre, CHU Ste-Justine, Université de Montréal, Québec, Canada.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Abstract
PURPOSE OF REVIEW This review highlights the diverse roles of the high-affinity HDL receptor scavenger receptor class B, type I (SR-BI) in the modulation of global cholesterol homeostasis and vascular cell function, and the potential implications of these processes in atherosclerosis. RECENT FINDINGS SR-BI in the liver plays a critical role in reverse cholesterol transport and it dramatically impacts the characteristics of the HDL particle, and through reverse cholesterol transport it promotes an antiatherogenic lipid environment in the vascular wall. SR-BI in macrophages may influence their inflammatory phenotype. In endothelial cells, SR-BI mediates HDL-induced endothelial nitric oxide synthase activation and proliferation and migration, and in platelets SR-BI may be prothrombotic in the setting of dyslipidemia. Several polymorphisms of SR-BI have been reported in humans that influence receptor expression or function. SUMMARY In addition to regulating global lipid metabolism, SR-BI influences the functions of a variety of vascular cells relevant to atherosclerosis. Studies of SR-BI genetics in humans partially support the conclusions drawn from experimental models. However, because of the multiple functions of SR-BI, the diversity of cell types in which it is expressed, and the influence of the receptor on the characteristics of its own ligand, our understanding of the biology of the receptor is just emerging.
Collapse
Affiliation(s)
- Chieko Mineo
- Division of Pulmonary and Vascular Biology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9063, USA.
| | | |
Collapse
|
14
|
Grenier E, Garofalo C, Delvin E, Levy E. Modulatory role of PYY in transport and metabolism of cholesterol in intestinal epithelial cells. PLoS One 2012; 7:e40992. [PMID: 22844422 PMCID: PMC3402548 DOI: 10.1371/journal.pone.0040992] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2011] [Accepted: 06/19/2012] [Indexed: 12/17/2022] Open
Abstract
Background Gastrointestinal peptides are involved in modulating appetite. Other biological functions attributed to them include the regulation of lipid homeostasis. However, data concerning PYY remain fragmentary. The objectives of the study were: (i) To determine the effect of PYY on intestinal transport and synthesis of cholesterol, the biogenesis of apolipoproteins (apos) and assembly of lipoproteins and (ii) To analyze whether the effects of PYY are similar according to whether cells are exposed to PYY on apical or basolateral surface. Methodology/Principal Findings Caco-2/15 cells were incubated with PYY (1–36) administered either to the apical or basolateral medium, at concentrations of 50 or 200 nM for 24 hours. De novo synthesis of cholesterol, cholesterol uptake, and assembly of lipoproteins were evaluated through the incorporation of [14C]-acetate, [14C]-cholesterol, and [14C]-oleate, respectively. Biogenesis of apos (A-I, A-IV, E, B-48 and B-100) was examined by the incorporation of [35S]-methionine. The influence of PYY on protein and mRNA levels of many key mediators of lipid metabolism was analyzed by Western blot and PCR, respectively. Our results show that PYY influenced cholesterol metabolism in Caco-2/15 cells depending on the site of PYY delivery. Apical addition of PYY significantly lowered the incorporation of [14C]-cholesterol likely via the reduction of NPC1L1, stimulated intracellular cholesterol synthesis probably through an increase in SREBP-2 expression, whereas it concomitantly increased apo A-I synthesis and decreased LDL secretion. In contrast, basolateral PYY reduced the production of chylomicrons (CM) as well as the biogenesis of apos B-48 and B-100, while lowering the expression of the transcription factors RXRα and PPAR(α,β). Conclusions/Significance PYY is capable of influencing cholesterol homeostasis in intestinal Caco-2/15 cells depending on the site delivery. Apical PYY was able to decrease cholesterol uptake via NPC1L1 downregulation, whereas basolateral PYY diminished CM output through the biogenesis decline of apos B-48 and B-100.
Collapse
Affiliation(s)
- Emilie Grenier
- Research Centre, Centre Hospitalier Universitaire (CHU) Ste-Justine, Montreal, Quebec, Canada
- Department of Nutrition, Université de Montréal, Montreal, Quebec, Canada
| | - Carole Garofalo
- Research Centre, Centre Hospitalier Universitaire (CHU) Ste-Justine, Montreal, Quebec, Canada
| | - Edgard Delvin
- Research Centre, Centre Hospitalier Universitaire (CHU) Ste-Justine, Montreal, Quebec, Canada
- Department of Biochemistry, Université de Montréal, Montreal, Quebec, Canada
| | - Emile Levy
- Research Centre, Centre Hospitalier Universitaire (CHU) Ste-Justine, Montreal, Quebec, Canada
- Department of Nutrition, Université de Montréal, Montreal, Quebec, Canada
- * E-mail:
| |
Collapse
|
15
|
Eckhardt ERM, Witta J, Zhong J, Arsenescu R, Arsenescu V, Wang Y, Ghoshal S, de Beer MC, de Beer FC, de Villiers WJS. Intestinal epithelial serum amyloid A modulates bacterial growth in vitro and pro-inflammatory responses in mouse experimental colitis. BMC Gastroenterol 2010; 10:133. [PMID: 21067563 PMCID: PMC2992040 DOI: 10.1186/1471-230x-10-133] [Citation(s) in RCA: 87] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2010] [Accepted: 11/10/2010] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Serum Amyloid A (SAA) is a major acute phase protein of unknown function. SAA is mostly expressed in the liver, but also in other tissues including the intestinal epithelium. SAA reportedly has anti-bacterial effects, and because inflammatory bowel diseases (IBD) result from a breakdown in homeostatic interactions between intestinal epithelia and bacteria, we hypothesized that SAA is protective during experimental colitis. METHODS Intestinal SAA expression was measured in mouse and human samples. Dextran sodium sulfate (DSS) colitis was induced in SAA 1/2 double knockout (DKO) mice and in wildtype controls. Anti-bacterial effects of SAA1/2 were tested in intestinal epithelial cell lines transduced with adenoviral vectors encoding the CE/J SAA isoform or control vectors prior to exposure to live Escherichia coli. RESULTS Significant levels of SAA1/SAA2 RNA and SAA protein were detected by in situ hybridization and immunohistochemistry in mouse colonic epithelium. SAA3 expression was weaker, but similarly distributed. SAA1/2 RNA was present in the ileum and colon of conventional mice and in the colon of germfree mice. Expression of SAA3 was strongly regulated by bacterial lipopolysaccharides in cultured epithelial cell lines, whereas SAA1/2 expression was constitutive and not LPS inducible. Overexpression of SAA1/2 in cultured epithelial cell lines reduced the viability of co-cultured E. coli. This might partially explain the observed increase in susceptibility of DKO mice to DSS colitis. SAA1/2 expression was increased in colon samples obtained from Crohn's Disease patients compared to controls. CONCLUSIONS Intestinal epithelial SAA displays bactericidal properties in vitro and could play a protective role in experimental mouse colitis. Altered expression of SAA in intestinal biopsies from Crohn's Disease patients suggests that SAA is involved in the disease process..
Collapse
Affiliation(s)
- Erik RM Eckhardt
- Graduate Center for Nutritional Sciences, University of Kentucky, Lexington, USA
- Internal Medicine Department, University of Kentucky, Lexington, USA
| | - Jassir Witta
- Internal Medicine Department, University of Kentucky, Lexington, USA
| | - Jian Zhong
- Internal Medicine Department, University of Kentucky, Lexington, USA
| | - Razvan Arsenescu
- Internal Medicine Department, University of Kentucky, Lexington, USA
| | - Violeta Arsenescu
- Graduate Center for Nutritional Sciences, University of Kentucky, Lexington, USA
- Internal Medicine Department, University of Kentucky, Lexington, USA
| | - Yu Wang
- Graduate Center for Nutritional Sciences, University of Kentucky, Lexington, USA
| | - Sarbani Ghoshal
- Internal Medicine Department, University of Kentucky, Lexington, USA
| | | | | | | |
Collapse
|
16
|
Boudry G, David ES, Douard V, Monteiro IM, Le Huërou-Luron I, Ferraris RP. Role of intestinal transporters in neonatal nutrition: carbohydrates, proteins, lipids, minerals, and vitamins. J Pediatr Gastroenterol Nutr 2010; 51:380-401. [PMID: 20808244 DOI: 10.1097/mpg.0b013e3181eb5ad6] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
To support rapid growth and a high metabolic rate, infants require enormous amounts of nutrients. The small intestine must have the complete array of transporters that absorb the nutrients released from digested food. Failure of intestinal transporters to function properly often presents symptoms as "failure to thrive" because nutrients are not absorbed and as diarrhea because unabsorbed nutrients upset luminal osmolality or become substrates of intestinal bacteria. We enumerate the nutrients that constitute human milk and various infant milk formulas, explain their importance in neonatal nutrition, then describe for each nutrient the transporter(s) that absorbs it from the intestinal lumen into the enterocyte cytosol and from the cytosol to the portal blood. More than 100 membrane and cytosolic transporters are now thought to facilitate absorption of minerals and vitamins as well as products of digestion of the macronutrients carbohydrates, proteins, and lipids. We highlight research areas that should yield information needed to better understand the important role of these transporters during normal development.
Collapse
Affiliation(s)
- Gaëlle Boudry
- Institut National de Recherche Agronomique, UMR1079 Système d'Elevage, Nutrition, Animale et Humaine, St-Gilles, France
| | | | | | | | | | | |
Collapse
|
17
|
Mohanty S, Anderson CL, Robinson JM. The expression of caveolin-1 and the distribution of caveolae in the murine placenta and yolk sac: parallels to the human placenta. Placenta 2009; 31:144-50. [PMID: 20004972 DOI: 10.1016/j.placenta.2009.11.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2009] [Revised: 11/16/2009] [Accepted: 11/17/2009] [Indexed: 01/15/2023]
Abstract
The expression pattern of caveolin-1 and the distribution of caveolae in the murine placental labyrinth and visceral yolk sac have been determined. Immunoblot analysis demonstrates that both placenta and yolk sac express the protein caveolin-1. Immunofluorescence microscopy was used to determine which cell types in the placental labyrinth and yolk sac express caveolin-1. In yolk sac, detectable caveolin-1 was restricted to endothelial cells and smooth muscle cells of the vitelline vasculature and to mesothelial cells. Endoderm, the major cell type in the yolk sac, does not express caveolin-1 as assessed by this assay. In the labyrinth region of the placenta, endothelial cells express caveolin-1 but this protein was not detectable in any of the three trophoblast layers. These tissues were also examined by electron microscopy to determine which cell types contain the specialized plasma membrane microdomains known as caveolae. Morphologically detectable caveolae were present in endothelial and smooth muscle cells, as well as mesothelial cells of the yolk sac and in endothelial cells of the placental labyrinth. Neither endodermal cells of the yolk sac nor trophoblastic cells in the placental labyrinth contained caveolae-like structures. We conclude that caveolin-1 and caveolae have restricted distribution in the murine placenta and yolk sac and that this parallels the situation in human placenta.
Collapse
Affiliation(s)
- S Mohanty
- Department of Internal Medicine, Ohio State University, Columbus, OH 43210, USA
| | | | | |
Collapse
|
18
|
Levy E, Ménard D, Delvin E, Montoudis A, Beaulieu JF, Mailhot G, Dubé N, Sinnett D, Seidman E, Bendayan M. Localization, function and regulation of the two intestinal fatty acid-binding protein types. Histochem Cell Biol 2009; 132:351-67. [PMID: 19499240 DOI: 10.1007/s00418-009-0608-y] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/13/2009] [Indexed: 01/20/2023]
Abstract
Although intestinal (I) and liver (L) fatty acid binding proteins (FABP) have been widely studied, the physiological significance of the presence of the two FABP forms (I- and L-FABP) in absorptive cells remains unknown as do the differences related to their distribution along the crypt-villus axis, regional expression, ontogeny and regulation in the human intestine. Our morphological experiments supported the expression of I- and L-FABP as early as 13 weeks of gestation. Whereas cytoplasmic immunofluorescence staining of L-FABP was barely detectable in the lower half of the villus and in the crypt epithelial cells, I-FABP was visualized in epithelial cells of the crypt-villus axis in all intestinal segments until the adult period in which the staining was maximized in the upper part of the villus. Immunoelectron microscopy revealed more intense labeling of L-FABP compared with I-FABP, accompanied with a heterogeneous distribution in the cytoplasm, microvilli and basolateral membranes. By western blot analysis, I- and L-FABP at 15 weeks of gestation appeared predominant in jejunum compared with duodenum, ileum, proximal and distal colon. Exploration of the maturation aspect documented a rise in L-FABP in adult tissues. Permanent transfections of Caco-2 cells with I-FABP cDNA resulted in decreased lipid export, apolipoprotein (apo) biogenesis and chylomicron secretion. Additionally, supplementation of Caco-2 with insulin, hydrocortisone and epidermal growth factor differentially modulated the expression of I- and L-FABP, apo B-48 and microsomal triglyceride transfer protein (MTP), emphasizing that these key proteins do not exhibit a parallel modulation. Overall, our findings indicate that the two FABPs display differences in localization, regulation and developmental pattern.
Collapse
Affiliation(s)
- Emile Levy
- Department of Nutrition, CHU-Sainte-Justine, University of Montreal, 3175 Côte Ste-Catherine Road, Montreal, QC, H3T 1C5, Canada.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Leblond F, Seidah NG, Précourt LP, Delvin E, Dominguez M, Levy E. Regulation of the proprotein convertase subtilisin/kexin type 9 in intestinal epithelial cells. Am J Physiol Gastrointest Liver Physiol 2009; 296:G805-15. [PMID: 19179626 DOI: 10.1152/ajpgi.90424.2008] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Proprotein convertase subtilisin/kexin type 9 (PCSK9) posttranslationally promotes the degradation of the low-density lipoprotein receptor (LDLr) in hepatocytes and increases plasma LDL cholesterol. It is not clear, however, whether PCSK9 plays a role in the small intestine. Here, we characterized the patterns of variations of PCSK9 and LDLr in fully differentiated Caco-2/15 cells as a function of various potential effectors. Cholesterol (100 microM) solubilized in albumin or micelles significantly downregulated PCSK9 gene (30%, P<0.05) and protein expression (50%, P<0.05), surprisingly in concert with a decrease in LDLr protein levels (45%, P<0.05). Cells treated with 25-hydroxycholesterol (50 microM) also displayed significant reduction in PCSK9 gene (37%, P<0.01) and protein (75% P<0.001) expression, whereas LDLr showed a decrease at the gene (30%, P<0.05) and protein (57%, P<0.01) levels, respectively. The amounts of PCSK9 mRNA and protein in Caco-2/15 cells were associated to the regulation of 3-hydroxy-3-methylglutaryl-CoA reductase and sterol regulatory element binding protein-2 (SREBP-2) that can transcriptionally activate PCSK9 via sterol-regulatory elements located in its proximal promoter region. On the other hand, depletion of cholesterol content by hydroxypropyl-beta-cyclodextrin upregulated PCSK9 transcripts (20%, P<0.05) and protein mass (540%, P<0.001), in parallel with SREBP-2 protein levels. The addition of bile acids (BA) taurocholate and deoxycholate to the apical culture medium lowered PCSK9 gene expression (25%, P<0.01) and raised PCSK9 protein expression (30%, P<0.01), respectively, probably via the modulation of farnesoid X receptor. Furthermore, unconjugated and conjugated BA exhibited different effects on PCSK9 and LDLr. Altogether, these data indicate that intestinal PCSK9 is highly modulated by sterols and emphasize the distinct effects of BA species.
Collapse
Affiliation(s)
- François Leblond
- Department of Nutrition, Clinical Research Institute of Montréal, Montreal, Quebec, Canada, H3T 1C5
| | | | | | | | | | | |
Collapse
|
20
|
Comparative expression analysis reveals differences in the regulation of intestinal paraoxonase family members. Int J Biochem Cell Biol 2009; 41:1628-37. [PMID: 19401157 DOI: 10.1016/j.biocel.2009.02.013] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2008] [Revised: 11/29/2008] [Accepted: 02/09/2009] [Indexed: 01/31/2023]
Abstract
The paraoxonase (PON) gene cluster contains three members (PON1, PON2, and PON3), located on chromosome 7q21.3-22.1. Until now there has been little insight into their regulation in human intestine. This study was designed to determine the regulation of PONs by oxidative stress and inflammatory factors. Differentiated Caco-2/15 cells, cultured on polycarbonate Transwell filter inserts, exhibited transcripts of the 3 PONs whereas Western blot revealed the protein expression of PON2 and PON3 only. Iron-ascorbate-mediated lipid peroxidation, lipopolysaccharides (LPS), tumor necrosis factor-alpha and interferon-gamma induced differential effects on the gene expression and protein mass of PONs. In particular, LPS down-regulated PON2 protein expression, which was accompanied with decreased levels of IkappaBalpha, the inhibitor of the proinflammatory transcription factor nuclear factor-kappa B (NF-kappaB). Selective inactivation of NF-kappaB by the action of caffeic acid phenethyl ester (CAPE) partially attenuated but did not abolish LPS-triggered decline of PON2. However, the combination of CAPE and antioxidants completely abrogated the negative impact of LPS on PON2. Therefore, our data indicate that oxidative stress and proinflammatory agents selectively affect the expression of PONs. Our findings also suggest that both NF-kappaB pathway and lipid peroxidation are implicated in LPS-dependent diminution of PON2.
Collapse
|
21
|
Béaslas O, Cueille C, Delers F, Chateau D, Chambaz J, Rousset M, Carrière V. Sensing of dietary lipids by enterocytes: a new role for SR-BI/CLA-1. PLoS One 2009; 4:e4278. [PMID: 19169357 PMCID: PMC2627924 DOI: 10.1371/journal.pone.0004278] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2008] [Accepted: 12/17/2008] [Indexed: 11/23/2022] Open
Abstract
Background The intestine is responsible for absorbing dietary lipids and delivering them to the organism as triglyceride-rich lipoproteins (TRL). It is important to determine how this process is regulated in enterocytes, the absorptive cells of the intestine, as prolonged postprandial hypertriglyceridemia is a known risk factor for atherosclerosis. During the postprandial period, dietary lipids, mostly triglycerides (TG) hydrolyzed by pancreatic enzymes, are combined with bile products and reach the apical membrane of enterocytes as postprandial micelles (PPM). Our aim was to determine whether these micelles induce, in enterocytes, specific early cell signaling events that could control the processes leading to TRL secretion. Methodology/Principal Findings The effects of supplying PPM to the apex of Caco-2/TC7 enterocytes were analyzed. Micelles devoid of TG hydrolysis products, like those present in the intestinal lumen in the interprandial period, were used as controls. The apical delivery of PPM specifically induced a number of cellular events that are not induced by interprandial micelles. These early events included the trafficking of apolipoprotein B, a structural component of TRL, from apical towards secretory domains, and the rapid, dose-dependent activation of ERK and p38MAPK. PPM supply induced the scavenger receptor SR-BI/CLA-1 to cluster at the apical brush border membrane and to move from non-raft to raft domains. Competition, inhibition or knockdown of SR-BI/CLA-1 impaired the PPM-dependent apoB trafficking and ERK activation. Conclusions/Significance These results are the first evidence that enterocytes specifically sense postprandial dietary lipid-containing micelles. SR-BI/CLA-1 is involved in this process and could be a target for further study with a view to modifying intestinal TRL secretion early in the control pathway.
Collapse
Affiliation(s)
- Olivier Béaslas
- Université Pierre et Marie Curie - Paris 6, UMR S 872, Les Cordeliers, Paris, France
- INSERM, U 872, Paris, France
- Université Paris Descartes, UMR S 872, Paris, France
| | - Carine Cueille
- Université Pierre et Marie Curie - Paris 6, UMR S 872, Les Cordeliers, Paris, France
- INSERM, U 872, Paris, France
- Université Paris Descartes, UMR S 872, Paris, France
| | - François Delers
- Université Pierre et Marie Curie - Paris 6, UMR S 872, Les Cordeliers, Paris, France
- INSERM, U 872, Paris, France
- Université Paris Descartes, UMR S 872, Paris, France
| | - Danielle Chateau
- Université Pierre et Marie Curie - Paris 6, UMR S 872, Les Cordeliers, Paris, France
- INSERM, U 872, Paris, France
- Université Paris Descartes, UMR S 872, Paris, France
| | - Jean Chambaz
- Université Pierre et Marie Curie - Paris 6, UMR S 872, Les Cordeliers, Paris, France
- INSERM, U 872, Paris, France
- Université Paris Descartes, UMR S 872, Paris, France
| | - Monique Rousset
- Université Pierre et Marie Curie - Paris 6, UMR S 872, Les Cordeliers, Paris, France
- INSERM, U 872, Paris, France
- Université Paris Descartes, UMR S 872, Paris, France
| | - Véronique Carrière
- Université Pierre et Marie Curie - Paris 6, UMR S 872, Les Cordeliers, Paris, France
- INSERM, U 872, Paris, France
- Université Paris Descartes, UMR S 872, Paris, France
- * E-mail:
| |
Collapse
|
22
|
Ravid Z, Bendayan M, Delvin E, Sane AT, Elchebly M, Lafond J, Lambert M, Mailhot G, Levy E. Modulation of intestinal cholesterol absorption by high glucose levels: impact on cholesterol transporters, regulatory enzymes, and transcription factors. Am J Physiol Gastrointest Liver Physiol 2008; 295:G873-85. [PMID: 18772361 DOI: 10.1152/ajpgi.90376.2008] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Growing evidence suggests that the small intestine may contribute to excessive postprandial lipemia, which is highly prevalent in insulin-resistant/Type 2 diabetic individuals and substantially increases the risk of cardiovascular disease. The aim of the present study was to determine the role of high glucose levels on intestinal cholesterol absorption, cholesterol transporter expression, enzymes controlling cholesterol homeostasis, and the status of transcription factors. To this end, we employed highly differentiated and polarized cells (20 days of culture), plated on permeable polycarbonate filters. In the presence of [(14)C]cholesterol, glucose at 25 mM stimulated cholesterol uptake compared with Caco-2/15 cells supplemented with 5 mM glucose (P < 0.04). Because combination of 5 mM glucose with 20 mM of the structurally related mannitol or sorbitol did not change cholesterol uptake, we conclude that extracellular glucose concentration is uniquely involved in the regulation of intestinal cholesterol transport. The high concentration of glucose enhanced the protein expression of the critical cholesterol transporter NPC1L1 and that of CD36 (P < 0.02) and concomitantly decreased SR-BI protein mass (P < 0.02). No significant changes were observed in the protein expression of ABCA1 and ABCG8, which act as efflux pumps favoring cholesterol export out of absorptive cells. At the same time, 3-hydroxy-3-methylglutaryl-coenzyme A reductase activity was decreased (P < 0.007), whereas ACAT activity remained unchanged. Finally, increases were noted in the transcription factors LXR-alpha, LXR-beta, PPAR-beta, and PPAR-gamma along with a drop in the protein expression of SREBP-2. Collectively, our data indicate that glucose at high concentrations may regulate intestinal cholesterol transport and metabolism in Caco-2/15 cells, thus suggesting a potential influence on the cholesterol absorption process in Type 2 diabetes.
Collapse
Affiliation(s)
- Z Ravid
- Research Centre, CHU-Sainte-Justine, 3175 Côte Ste-Catherine, Montréal, Québec, Canada H3T 1C5
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Ghoshal S, Witta J, Zhong J, de Villiers W, Eckhardt E. Chylomicrons promote intestinal absorption of lipopolysaccharides. J Lipid Res 2008; 50:90-7. [PMID: 18815435 DOI: 10.1194/jlr.m800156-jlr200] [Citation(s) in RCA: 474] [Impact Index Per Article: 27.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Recent data suggest that dietary fat promotes intestinal absorption of lipopolysaccharides (LPS) from the gut microflora, which might contribute to various inflammatory disorders. The mechanism of fat-induced LPS absorption is unclear, however. Intestinal-epithelial cells can internalize LPS from the apical surface and transport LPS to the Golgi. The Golgi complex also contains newly formed chylomicrons, the lipoproteins that transport dietary long-chain fat through mesenteric lymph and blood. Because LPS has affinity for chylomicrons, we hypothesized that chylomicron formation promotes LPS absorption. In agreement with our hypothesis, we found that CaCo-2 cells released more cell-associated LPS after incubation with oleic-acid (OA), a long-chain fatty acid that induces chylomicron formation, than with butyric acid (BA), a short-chain fatty acid that does not induce chylomicron formation. Moreover, the effect of OA was blocked by the inhibitor of chylomicron formation, Pluronic L-81. We also observed that intragastric triolein (TO) gavage was followed by increased plasma LPS, whereas gavage with tributyrin (TB), or TO plus Pluronic L-81, was not. Most intestinally absorbed LPS was present on chylomicron remnants (CM-R) in the blood. Chylomicron formation also promoted transport of LPS through mesenteric lymph nodes (MLN) and the production of TNFalpha mRNA in the MLN. Together, our data suggest that intestinal epithelial cells may release LPS on chylomicrons from cell-associated pools. Chylomicron-associated LPS may contribute to postprandial inflammatory responses or chronic diet-induced inflammation in chylomicron target tissues.
Collapse
Affiliation(s)
- Sarbani Ghoshal
- Department of Internal Medicine, Division of Digestive Diseases and Nutrition, University of Kentucky, Lexington, KY, USA
| | | | | | | | | |
Collapse
|
24
|
Montoudis A, Seidman E, Boudreau F, Beaulieu JF, Menard D, Elchebly M, Mailhot G, Sane AT, Lambert M, Delvin E, Levy E. Intestinal fatty acid binding protein regulates mitochondrion beta-oxidation and cholesterol uptake. J Lipid Res 2008; 49:961-72. [PMID: 18235139 DOI: 10.1194/jlr.m700363-jlr200] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
The role of intestinal fatty acid binding protein (I-FABP) in lipid metabolism remains elusive. To address this issue, normal human intestinal epithelial cells (HIEC-6) were transfected with cDNA to overexpress I-FABP and compared with cells treated with empty pQCXIP vector. I-FABP overexpression stimulated mitochondrial [U-14C]oleate oxidation to CO2 and acid-soluble metabolites via mechanisms including the upregulation of protein expression and the activity of carnitine palmitoyltransferase 1, a critical enzyme controlling the entry of fatty acid (FA) into mitochondria, and increased activity of 3-hydroxyacyl-CoA dehydrogenase, a mitochondrial beta-oxidation enzyme. On the other hand, the gene and protein expression of the key enzymes FA synthase and acetyl-coenzyme A carboxylase 2 was decreased, suggesting diminished lipogenesis. Furthermore, I-FABP overexpression caused a decline in [14C]free cholesterol (CHOL) incorporation. Accordingly, a significant lessening was observed in the gene expression of Niemann Pick C1-Like 1, a mediator of CHOL uptake, along with an increase in the transcripts and protein content of ABCA1 and ABCG5/ABCG8, acting as CHOL efflux pumps. Furthermore, I-FABP overexpression resulted in increased levels of mRNA, protein mass, and activity of HMG-CoA reductase, the rate-limiting step in CHOL synthesis. Scrutiny of the nuclear receptors revealed augmented peroxisome proliferator-activated receptor alpha,gamma and reduced liver X receptor-alpha in HIEC-6 overexpressing I-FABP. Finally, I-FABP overexpression did not influence acyl-coenzyme A oxidase 1, which catalyzes the first rate-limiting step in peroxisomal FA beta-oxidation. Overall, our data suggest that I-FABP may influence mitochondrial FA oxidation and CHOL transport by regulating gene expression and interaction with nuclear receptors.
Collapse
Affiliation(s)
- Alain Montoudis
- Department of Nutrition, Centre Hospitalier Universitaire Sainte-Justine, Université de Montréal, Montreal, Quebec, Canada
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Levy E, Trudel K, Bendayan M, Seidman E, Delvin E, Elchebly M, Lavoie JC, Precourt LP, Amre D, Sinnett D. Biological role, protein expression, subcellular localization, and oxidative stress response of paraoxonase 2 in the intestine of humans and rats. Am J Physiol Gastrointest Liver Physiol 2007; 293:G1252-61. [PMID: 17916643 DOI: 10.1152/ajpgi.00369.2007] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Oxidative stress is a cardinal manifestation of various intestinal disorders. However, very little knowledge is available on the intestine's inherent defense mechanisms against free radicals. This study was designed to determine the protein expression, subcellular localization and oxidative stress response of paraoxonase 2 (PON2), a member of a powerful antioxidant family in human and rat intestine. Biochemical and ultrastructural experiments all showed a substantial expression of PON2 in human and rat intestine. Western blot analysis disclosed higher levels of PON2 in the jejunum than in the duodenum, ileum, and colon. Cell fractionation revealed a predominant PON2 association with microsomes and lysosomes in the human jejunum, which differed from that in rats. PON2 was detected in the intestine as early as week 15 of gestation and was significantly increased by week 20. Iron ascorbate-mediated lipid peroxidation induced a marked decrease in PON2 expression in intestinal specimens coincidental to an abundant rise in malondialdehyde (MDA). On the other hand, preincubation with potent antioxidants, such as butylated hydroxytoluene, Trolox, and N-acetylcysteine, prevented iron-ascorbate-generating PON2 reduction in parallel with MDA suppression. Finally, the preincubation of permeabilized Caco-2 cells with purified PON2 led to a protection against iron-ascorbate-induced lipid peroxidation. These observations demonstrate that the human intestine is preferentially endowed with a marked PON2 expression compared with the rat intestine and this expression shows a developmental and intracellular pattern of distribution. Furthermore, our observations suggest PON2 protective effects against prooxidant stimuli in the small intestine.
Collapse
Affiliation(s)
- Emile Levy
- Department of Nutrition, Université de Montréal, Research Centre, CHU Sainte Justine, Montréal, Québec, Canada.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Zhang Y, Ahmed AM, Tran TL, Lin J, McFarlane N, Boreham DR, Igdoura SA, Truant R, Trigatti BL. The inhibition of endocytosis affects HDL-lipid uptake mediated by the human scavenger receptor class B type I. Mol Membr Biol 2007; 24:442-54. [PMID: 17710648 DOI: 10.1080/09687680701300410] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The scavenger receptor SR-BI plays an important role in the hepatic clearance of HDL cholesterol and other lipids, driving reverse cholesterol transport and contributing to protection against atherosclerosis in mouse models. We characterized the role of endocytosis in lipid uptake from HDL, mediated by the human SR-BI, using a variety of approaches to inhibit endocytosis, including hypertonic shock, potassium or energy depletion and disassembly of the actin cytoskeleton. Our studies revealed that unlike mouse SR-BI, human SR-BI-mediated HDL-lipid uptake was reduced by inhibition of endocytosis. This was not dependent on the cytoplasmic C-terminus of SR-BI. Monitoring the uptake of both the protein and lipid components of HDL revealed that although overall lipid uptake was decreased, the degree of selective lipid uptake was increased. These data suggest that that endocytosis is a dynamic regulator of SR-BI's selective lipid uptake activity.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Biochemistry, McMaster University, Hamilton, Ontario, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Levy E, Lavoie MA, Delvin E, Seidman E, Lambert M, Sinnett D, Sané AT, Leblond F, Spahis S, Roy CC. Avancées dans la dissection fonctionnelle du transport intestinal des lipides. Med Sci (Paris) 2007; 23:1014-9. [DOI: 10.1051/medsci/200723111014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
28
|
Pshezhetsky AV, Fedjaev M, Ashmarina L, Mazur A, Budman L, Sinnett D, Labuda D, Beaulieu JF, Ménard D, Nifant'ev I, Levy E. Subcellular proteomics of cell differentiation: quantitative analysis of the plasma membrane proteome of Caco-2 cells. Proteomics 2007; 7:2201-15. [PMID: 17549793 DOI: 10.1002/pmic.200600956] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Human colorectal carcinoma (Caco-2) cells undergo in culture spontaneous enterocytic differentiation, characterized by polarization and appearance of the functional apical brush border membrane. To provide insights into the biology of differentiation, we have performed a comparative proteomic analysis of the plasma membranes from proliferating cells (PCs) and the apical membranes from differentiated cells (DCs). Proteins were resolved by SDS-PAGE, in-gel digested and analyzed by RP-LC and MS/MS. Alternatively, proteins were digested in solution, and tryptic peptides were labeled with isotopic tags and analyzed by 2-D LC followed by MS/MS. Among the 1125 proteins identified in both proteomes, 76 were found to be significantly increased in the membranes of DCs and 61 were increased in PCs. Majority of the proteins increased in the apical membranes were metabolic enzymes, proteins involved in the maintenance of cellular structure, transmembrane transporters, and proteins regulating vesicular transport. In contrast, majority of the proteins increased in the membranes of PCs were involved in gene expression, protein synthesis, and folding. Both groups contained many novel proteins with yet to be identified functions, which could provide potential new markers of the intestinal cells or of colorectal cancer.
Collapse
Affiliation(s)
- Alexey V Pshezhetsky
- Sainte-Justine Hospital Research Center, University of Montreal, Montreal, Quebec, Canada.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Knöpfel M, Davies JP, Duong PT, Kvaernø L, Carreira EM, Phillips MC, Ioannou YA, Hauser H. Multiple plasma membrane receptors but not NPC1L1 mediate high-affinity, ezetimibe-sensitive cholesterol uptake into the intestinal brush border membrane. Biochim Biophys Acta Mol Cell Biol Lipids 2007; 1771:1140-7. [PMID: 17689140 DOI: 10.1016/j.bbalip.2007.05.011] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2007] [Revised: 05/01/2007] [Accepted: 05/02/2007] [Indexed: 11/30/2022]
Abstract
We compared cholesterol uptake into brush border membrane vesicles (BBMV) made from the small intestines of either wild-type or Niemann-Pick C1-like 1 (NPC1L1) knockout mice to elucidate the contribution of NPC1L1 to facilitated uptake; this uptake involves cholesterol transport from lipid donor particles into the BBM of enterocytes. The lack of NPC1L1 in the BBM of the knockout mice had no effect on the rate of cholesterol uptake. It follows that NPC1L1 cannot be the putative high-affinity, ezetimibe-sensitive cholesterol transporter in the brush border membrane (BBM) as has been proposed by others. The following findings substantiate this conclusion: (I) NPC1L1 is not a brush border membrane protein but very likely localized to intracellular membranes; (II) the cholesterol absorption inhibitor ezetimibe and its analogues reduce cholesterol uptake to the same extent in wild-type and NPC1L1 knockout mouse BBMV. These findings indicate that the prevailing belief that NPC1L1 facilitates intestinal cholesterol uptake into the BBM and its interaction with ezetimibe is responsible for the inhibition of this process can no longer be sustained.
Collapse
Affiliation(s)
- Martin Knöpfel
- Laboratorium für Organische Chemie, ETH Zürich, CH-8093 Zürich, Switzerland
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Orlowski S, Coméra C, Tercé F, Collet X. Lipid rafts: dream or reality for cholesterol transporters? EUROPEAN BIOPHYSICS JOURNAL: EBJ 2007; 36:869-85. [PMID: 17576551 DOI: 10.1007/s00249-007-0193-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2007] [Revised: 05/11/2007] [Accepted: 05/15/2007] [Indexed: 01/12/2023]
Abstract
As a key constituent of the cell membranes, cholesterol is an endogenous component of mammalian cells of primary importance, and is thus subjected to highly regulated homeostasis at the cellular level as well as at the level of the whole body. This regulation requires adapted mechanisms favoring the handling of cholesterol in aqueous compartments, as well as its transfer into or out of membranes, involving membrane proteins. A membrane exhibits functional properties largely depending on its lipid composition and on its structural organization, which very often involves cholesterol-rich microdomains. Then there is the appealing possibility that cholesterol may regulate its own transmembrane transport at a purely functional level, independently of any transcriptional regulation based on cholesterol-sensitive nuclear factors controling the expression level of lipid transport proteins. Indeed, the main cholesterol "transporters" presently believed to mediate for instance the intestinal absorption of cholesterol, that are SR-BI, NPC1L1, ABCA1, ABCG1, ABCG5/G8 and even P-glycoprotein, all present privileged functional relationships with membrane cholesterol-containing microdomains. In particular, they all more or less clearly induce membrane disorganization, supposed to facilitate cholesterol exchanges with the close aqueous medium. The actual lipid substrates handled by these transporters are not yet unambiguously determined, but they likely concern the components of membrane microdomains. Conversely, raft alterations may provide specific modulations of the transporter activities, as well as they can induce indirect effects via local perturbations of the membrane. Finally, these cholesterol transporters undergo regulated intracellular trafficking, with presumably some relationships to rafts which remain to be clarified.
Collapse
Affiliation(s)
- Stéphane Orlowski
- SB2SM/IBTS and URA 2096 CNRS, CEA, Centre de Saclay, 91191, Gif-sur-Yvette cedex, France.
| | | | | | | |
Collapse
|
31
|
Levy E, Spahis S, Sinnett D, Peretti N, Maupas-Schwalm F, Delvin E, Lambert M, Lavoie MA. Intestinal cholesterol transport proteins: an update and beyond. Curr Opin Lipidol 2007; 18:310-8. [PMID: 17495606 DOI: 10.1097/mol.0b013e32813fa2e2] [Citation(s) in RCA: 96] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW Various studies have delineated the causal role of dietary cholesterol in atherogenesis. Strategies have thus been developed to minimize cholesterol absorption, and cholesterol transport proteins found at the apical membrane of enterocytes have been extensively investigated. This review focuses on recent progress related to various brush-border proteins that are potentially involved in alimentary cholesterol transport. RECENT FINDINGS Molecular mechanisms responsible for dietary cholesterol and plant sterol uptake have not been completely defined. Growing evidence, however, supports the concept that several proteins are involved in mediating intestinal cholesterol transport, including SR-BI, NPC1L1, CD36, aminopeptidase N, P-glycoprotein, and the caveolin-1/annexin-2 heterocomplex. Other ABC family members (ABCA1 and ABCG5/ABCG8) act as efflux pumps favoring cholesterol export out of absorptive cells into the lumen or basolateral compartment. Several of these cholesterol carriers influence intracellular cholesterol homeostasis and are controlled by transcription factors, including RXR, LXR, SREBP-2 and PPARalpha. The lack of responsiveness of NPC1L1-deficient mice to ezetimibe suggests that NPC1L1 is likely to be the principal target of this cholesterol-lowering drug. SUMMARY The understanding of the role, genetic regulation and coordinated function of proteins mediating intestinal cholesterol transport may lead to novel ways of treating cardiovascular disease.
Collapse
Affiliation(s)
- Emile Levy
- Research Centre, CHU-Sainte-Justine, Québec, Canada.
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Wang T, Jiao Y, Montell C. Dissection of the pathway required for generation of vitamin A and for Drosophila phototransduction. ACTA ACUST UNITED AC 2007; 177:305-16. [PMID: 17452532 PMCID: PMC2064138 DOI: 10.1083/jcb.200610081] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Dietary carotenoids are precursors for the production of retinoids, which participate in many essential processes, including the formation of the photopigment rhodopsin. Despite the importance of conversion of carotenoids to vitamin A (all-trans-retinol), many questions remain concerning the mechanisms that promote this process, including the uptake of carotenoids. We use the Drosophila visual system as a genetic model to study retinoid formation from β-carotene. In a screen for mutations that affect the biosynthesis of rhodopsin, we identified a class B scavenger receptor, SANTA MARIA. We demonstrate that SANTA MARIA functions upstream of vitamin A formation in neurons and glia, which are outside of the retina. The protein is coexpressed and functionally coupled with the β, β-carotene-15, 15′-monooxygenase, NINAB, which converts β-carotene to all-trans-retinal. Another class B scavenger receptor, NINAD, functions upstream of SANTA MARIA in the uptake of carotenoids, enabling us to propose a pathway involving multiple extraretinal cell types and proteins essential for the formation of rhodopsin.
Collapse
Affiliation(s)
- Tao Wang
- Department of Biological Chemistry, Department of Neuroscience, and Center for Sensory Biology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | | | | |
Collapse
|
33
|
Peretti N, Delvin E, Sinnett D, Marcil V, Garofalo C, Levy E. Asymmetrical regulation of scavenger receptor class B type I by apical and basolateral stimuli using Caco-2 cells. J Cell Biochem 2007; 100:421-33. [PMID: 16927335 DOI: 10.1002/jcb.20882] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Cholesterol uptake and the mechanisms that regulate cholesterol translocation from the intestinal lumen into enterocytes remain for the most part unclear. Since scavenger receptor class B type I (SR-BI) has been suggested to play a role in cholesterol absorption, we investigated cellular SR-BI modulation by various potential effectors administered in both apical and basolateral sides of Caco-2 cells. With differentiation, Caco-2 cells increased SR-BI protein expression. Western blot analysis showed the ability of cholesterol and oxysterols in both cell compartments to reduce SR-BI protein expression. Among the n-3, n-6, and n-9 fatty acid families, only eicosapentaenoic acid was able to lower SR-BI protein expression on both sides, whereas apical alpha-linolenic acid decreased SR-BI abundance and basolateral arachidonic acid (AA) raised it. Epidermal growth factor and growth hormone, either in the apical or basolateral medium, diminished SR-BI cellular content, while insulin displayed the same effect only on the basolateral side. In the presence of proinflammatory agents (LPS, TNF-alpha, IFN-gamma), Caco-2 cells exhibited differential behavior. SR-BI was downregulated by lipopolysaccharide on both sides. Finally, WY-14643 fibrate diminished SR-BI protein expression when it was added to the apical medium. Biotinylation studies in response to selected stimuli revealed that regulatory modifications in SR-BI protein expression occurred for the most part at the apical cell surface irrespective of the effector location. Our data indicate that various effectors supplied to the apical and basolateral compartments may impact on SR-BI at the apical membrane, thus suggesting potential regulation of intestinal cholesterol absorption and distribution in various intracellular pools.
Collapse
Affiliation(s)
- N Peretti
- Centre de Recherche, CHU-Sainte-Justine, Université de Montréal, Montréal (Québec), Canada, H3T 1C5
| | | | | | | | | | | |
Collapse
|
34
|
Abstract
alpha-Tocopherol is the only form of vitamin E in vitamin supplements, whereas gamma-tocopherol is the predominant form of vitamin E in the US diet. gamma-Tocopherol has beneficial properties as an anti-inflammatory and possibly anti-atherogenic and anticancer agent. Excess a-tocopherol taken in supplements causes a reduction of gamma-tocopherol concentration in plasma. The biochemical mechanism of this effect, which is important to human nutrition, has recently been elucidated.
Collapse
Affiliation(s)
- George Wolf
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, USA.
| |
Collapse
|
35
|
Sané AT, Sinnett D, Delvin E, Bendayan M, Marcil V, Ménard D, Beaulieu JF, Levy E. Localization and role of NPC1L1 in cholesterol absorption in human intestine. J Lipid Res 2006; 47:2112-20. [PMID: 16829661 DOI: 10.1194/jlr.m600174-jlr200] [Citation(s) in RCA: 122] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Recent studies have documented the presence of Niemann-Pick C1-Like 1 (NPC1L1) in the small intestine and its capacity to transport cholesterol in mice and rats. The current investigation was undertaken to explore the localization and function of NPC1L1 in human enterocytes. Cell fractionation experiments revealed an NPC1L1 association with apical membrane of the enterocyte in human jejunum. Signal was also detected in lysosomes, endosomes, and mitochondria. Confirmation of cellular NPC1L1 distribution was obtained by immunocytochemistry. Knockdown of NPC1L1 caused a decline in the ability of Caco-2 cells to capture micellar [(14)C]free cholesterol. Furthermore, this NPC1L1 suppression resulted in increased and decreased mRNA levels and activity of HMG-CoA reductase, the rate-limiting step in cholesterol synthesis, and of ACAT, the key enzyme in cholesterol esterification, respectively. An increase was also noted in the transcriptional factor sterol-regulatory element binding protein that modulates cholesterol homeostasis. Efforts were devoted to define the impact of NPC1L1 knockdown on other mediators of cholesterol uptake. RT-PCR evidence is presented to show the significant decrease in the levels of scavenger receptor class B type I (SR-BI) with no changes in ABCA1, ABCG5, and cluster determinant 36 in NPC1L1-deficient Caco-2 cells. Together, our data suggest that NPC1L1 contributes to intestinal cholesterol homeostasis and possibly cooperates with SR-BI to mediate cholesterol absorption in humans.
Collapse
Affiliation(s)
- Alain Théophile Sané
- Department of Nutrition, CHU-Sainte-Justine, University of Montreal, Montreal, Quebec H3T 1C5, Canada
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Jiang Y, Oliver P, Davies KE, Platt N. Identification and characterization of murine SCARA5, a novel class A scavenger receptor that is expressed by populations of epithelial cells. J Biol Chem 2006; 281:11834-45. [PMID: 16407294 DOI: 10.1074/jbc.m507599200] [Citation(s) in RCA: 121] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Epithelia are positioned at a critical interface to prevent invasion by microorganisms from the environment. Pattern recognition receptors are important components of innate immunity because of their ability to interact with specific microbe-associated structures and initiate immune responses. Several distinct groups of receptors have been recognized. One of these, the scavenger receptors, has been classified into at least eight separate classes. The class A scavenger receptors are characterized by the presence of a collagen-like domain and include macrophage scavenger receptor type A (SR-A1 I/II, SCARA1) and MARCO (SCARA2). These receptors are known to make important contributions to host defense. Here, we identify a novel murine scavenger receptor, SCARA5, which has a structure typical of this class. The cDNA encodes 491 amino acids, which predict a type II protein that contains C-terminal intracellular, transmembrane, extracellular spacer, collagenous, and N-terminal scavenger receptor cysteine rich domains. Expression in Chinese hamster ovary cells confirmed that the receptor assembles as a homotrimer and is expressed at the plasma membrane. SCARA5-transfected cells bound Escherichia coli and Staphylococcus aureus, but not zymosan, in a polyanionic-inhibitable manner. Unlike other class A scavenger receptors, the receptor was unable to endocytose acetylated or oxidized low density lipoprotein. Quantitative RT-PCR and in situ hybridization demonstrate SCARA5 has a tissue and cellular distribution unique among class A scavenger receptors. Because of the restriction of SCARA5 transcripts to populations of epithelial cells, we propose that this receptor may play important roles in the innate immune activities of these cells.
Collapse
Affiliation(s)
- Yanyan Jiang
- Department of Human Anatomy and Genetics, University of Oxford, Oxford OX1 3QX
| | | | | | | |
Collapse
|
37
|
Reboul E, Klein A, Bietrix F, Gleize B, Malezet-Desmoulins C, Schneider M, Margotat A, Lagrost L, Collet X, Borel P. Scavenger receptor class B type I (SR-BI) is involved in vitamin E transport across the enterocyte. J Biol Chem 2005; 281:4739-45. [PMID: 16380385 PMCID: PMC2045115 DOI: 10.1074/jbc.m509042200] [Citation(s) in RCA: 186] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Although cellular uptake of vitamin E was initially described as a passive process, recent studies in the liver and brain have shown that SR-BI (scavenger receptor class B type I) is involved in this phenomenon. As SR-BI is expressed at high levels in the intestine, the present study addressed the involvement of SR-BI in vitamin E trafficking across enterocytes. Apical uptake and efflux of the main dietary forms of vitamin E were examined using Caco-2 TC-7 cell monolayers as a model of human intestinal epithelium. (R,R,R)-gamma-tocopherol bioavailability was compared between wild-type mice and mice overexpressing SR-BI in the intestine. The effect of vitamin E on enterocyte SR-BI mRNA levels was measured by real-time quantitative reverse transcription-PCR. Concentration-dependent curves for vitamin E uptake were similar for (R,R,R)-alpha-, (R,R,R)-gamma-, and dl-alpha-tocopherol. (R,R,R)-alpha-tocopherol transport was dependent on incubation temperature, with a 60% reduction in absorption at 4 degrees C compared with 37 degrees C (p < 0.05). Vitamin E flux in enterocytes was directed from the apical to the basal side, with a relative 10-fold reduction in the transfer process when measured in the opposite direction (p < 0.05). Co-incubation with cholesterol, gamma-tocopherol, or lutein significantly impaired alpha-tocopherol absorption. Anti-human SR-BI antibodies and BLT1 (a chemical inhibitor of lipid transport via SR-BI) blocked up to 80% of vitamin E uptake and up to 30% of apical vitamin E efflux (p < 0.05), and similar results were obtained for (R,R,R)-gamma-tocopherol. SR-BI mRNA levels were not significantly modified after a 24-h incubation of Caco-2 cells with vitamin E. Finally, (R,R,R)-gamma-tocopherol bioavailability was 2.7-fold higher in mice overexpressing SR-BI than in wild-type mice (p < 0.05). The present data show for the first time that vitamin E intestinal absorption is, at least in part, mediated by SR-BI.
Collapse
Affiliation(s)
- Emmanuelle Reboul
- Nutrition humaine et lipides : Biodisponibilité, métabolisme et régulation
INSERM : U476 INRA : UMR1260Université de Provence - Aix-Marseille I Université de la Méditerranée - Aix-Marseille IIFaculté de Médecine Secteur Timone
27, Boulevard Jean Moulin
13385 MARSEILLE CEDEX 05,FR
| | - Alexis Klein
- Métabolisme des Lipoprotéines Humaines et Interactions Vasculaires
INSERM : U498Université de BourgogneHôpital du Bocage
Av du Maréchal de Lattre de Tassigny
21079 DIJON CEDEX,FR
| | - Florence Bietrix
- Centre de Physiopathologie Toulouse Purpan
INSERM : U563 IFR30Université Paul Sabatier - Toulouse IIIHôpital de Purpan TOULOUSE
Place du Docteur Baylac
31024 TOULOUSE CEDEX 3,FR
| | - Béatrice Gleize
- Nutrition humaine et lipides : Biodisponibilité, métabolisme et régulation
INSERM : U476 INRA : UMR1260Université de Provence - Aix-Marseille I Université de la Méditerranée - Aix-Marseille IIFaculté de Médecine Secteur Timone
27, Boulevard Jean Moulin
13385 MARSEILLE CEDEX 05,FR
| | - Christiane Malezet-Desmoulins
- Nutrition humaine et lipides : Biodisponibilité, métabolisme et régulation
INSERM : U476 INRA : UMR1260Université de Provence - Aix-Marseille I Université de la Méditerranée - Aix-Marseille IIFaculté de Médecine Secteur Timone
27, Boulevard Jean Moulin
13385 MARSEILLE CEDEX 05,FR
| | - Martina Schneider
- Métabolisme des Lipoprotéines Humaines et Interactions Vasculaires
INSERM : U498Université de BourgogneHôpital du Bocage
Av du Maréchal de Lattre de Tassigny
21079 DIJON CEDEX,FR
| | - Alain Margotat
- Nutrition humaine et lipides : Biodisponibilité, métabolisme et régulation
INSERM : U476 INRA : UMR1260Université de Provence - Aix-Marseille I Université de la Méditerranée - Aix-Marseille IIFaculté de Médecine Secteur Timone
27, Boulevard Jean Moulin
13385 MARSEILLE CEDEX 05,FR
| | - Laurent Lagrost
- Métabolisme des Lipoprotéines Humaines et Interactions Vasculaires
INSERM : U498Université de BourgogneHôpital du Bocage
Av du Maréchal de Lattre de Tassigny
21079 DIJON CEDEX,FR
| | - Xavier Collet
- Centre de Physiopathologie Toulouse Purpan
INSERM : U563 IFR30Université Paul Sabatier - Toulouse IIIHôpital de Purpan TOULOUSE
Place du Docteur Baylac
31024 TOULOUSE CEDEX 3,FR
| | - Patrick Borel
- Nutrition humaine et lipides : Biodisponibilité, métabolisme et régulation
INSERM : U476 INRA : UMR1260Université de Provence - Aix-Marseille I Université de la Méditerranée - Aix-Marseille IIFaculté de Médecine Secteur Timone
27, Boulevard Jean Moulin
13385 MARSEILLE CEDEX 05,FR
- * Correspondence should be adressed to: Patrick Borel
| |
Collapse
|
38
|
Harder CJ, Vassiliou G, McBride HM, McPherson R. Hepatic SR-BI-mediated cholesteryl ester selective uptake occurs with unaltered efficiency in the absence of cellular energy. J Lipid Res 2005; 47:492-503. [PMID: 16339112 DOI: 10.1194/jlr.m500444-jlr200] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Scavenger receptor class B type I (SR-BI) plays a critical role in the delivery of HDL cholesterol and cholesteryl esters (CEs) to liver and steroidogenic tissues by a selective process that does not result in significant degradation of HDL protein. Recently, SR-BI-mediated endocytosis and recycling of HDL have been demonstrated. However, it remains unclear whether efficient SR-BI-mediated selective uptake occurs strictly at the plasma membrane or at additional sites along its endocytic itinerary. To examine the requirement for SR-BI endocytosis in HDL selective uptake, we determined the effects of energy depletion on the levels of cell-associated HDL protein and CE in primary mouse hepatocytes. Compared with CHO cells, we observed a much larger energy-dependent effect on CE uptake in primary mouse hepatocytes. Although varying the levels of caveolin-1 and carboxyl ester lipase altered the efficiency of selective uptake, neither was able to account for the energy-dependent component of HDL-CE uptake. Finally, we demonstrate that the hepatocyte-specific, energy-dependent effects on HDL-apolipoprotein A-I and -CE uptake are independent of SR-BI and are not required to achieve efficient SR-BI-mediated selective uptake of CE. Together, these data support the conclusion that neither the intracellular trafficking of HDL nor any energy-dependent cellular process affects the ability of the cell to maximally acquire CE through SR-BI-mediated selective uptake from HDL.
Collapse
Affiliation(s)
- Chris J Harder
- Lipoprotein and Atherosclerosis Group, University of Ottawa Heart Institute, Ottawa, Ontario, Canada K1Y 4W7
| | | | | | | |
Collapse
|
39
|
Montoudis A, Delvin E, Menard D, Beaulieu JF, Jean D, Tremblay E, Bendayan M, Levy E. Intestinal-fatty acid binding protein and lipid transport in human intestinal epithelial cells. Biochem Biophys Res Commun 2005; 339:248-54. [PMID: 16297872 DOI: 10.1016/j.bbrc.2005.10.202] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2005] [Accepted: 10/30/2005] [Indexed: 11/15/2022]
Abstract
Intestinal-fatty acid binding protein (I-FABP) is a 14-15 kDa cytoplasmic molecule highly expressed in the enterocyte. Although different functions have been proposed for various FABP family members, the specific function of I-FABP in human intestine remains unclear. Here, we studied the role of I-FABP in molecularly modified normal human intestinal epithelial cells (HIEC-6). cDNA transfection resulted in 90-fold I-FABP overexpression compared to cells treated with empty pQCXIP vector. The high-resolution immunogold technique revealed labeling mainly in the cytosol and confirmed the marked phenotype abundance of I-FABP in cDNA transfected cells. I-FABP overexpression was not associated with alterations in cell proliferation and viability. Studies using these transfected cells cultured with [14C]oleic acid did not reveal higher efficiency in de novo synthesis or secretion of triglycerides, phospholipids, and cholesteryl esters compared to cells treated with empty pQCXIP vector only. Similarly, the incubation with [35S]methionine did not disclose a superiority in the biogenesis of apolipoproteins (apo) A-I, A-IV, B-48, and B-100. Finally, cells transfected with I-FABP did not exhibit an increased production of chylomicrons, VLDL, LDL, and HDL. Our observations establish that I-FABP overexpression in normal HIEC-6 is not related to cell proliferation, lipid esterification, apo synthesis, and lipoprotein assembly, and, therefore, exclude its role in intestinal fat transport.
Collapse
Affiliation(s)
- Alain Montoudis
- Department of Nutrition, Université de Montréal and Research Center, CHU Sainte Justine, 3175 Côte Ste-Catherine, Montréal, Que., Canada H3T 1C5
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Joly E, Bendayan M, Roduit R, Saha AK, Ruderman NB, Prentki M. Malonyl-CoA decarboxylase is present in the cytosolic, mitochondrial and peroxisomal compartments of rat hepatocytes. FEBS Lett 2005; 579:6581-6. [PMID: 16298369 DOI: 10.1016/j.febslet.2005.10.050] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2005] [Accepted: 10/25/2005] [Indexed: 10/25/2022]
Abstract
A role for cytosolic malonyl-CoA decarboxylase (MCD) as a regulator of fatty acid oxidation has been postulated. However, there is no direct evidence that MCD is present in the cytosol. To address this issue, we performed cell fractionation and electron microscopic colloidal gold studies of rat liver to determine the location and activity of MCD. By both methods, substantial amounts of MCD protein and activity were found in the cytosol, mitochondria and peroxisomes, the latter with the highest specific activity. MCD species with different electrophoretic mobility were observed in the three fractions. The data demonstrate that active MCD is present in the cytosol, mitochondria and peroxisomes of rat liver, consistent with the view that MCD participates in the regulation of cytosolic malonyl-CoA levels and of hepatic fatty acid oxidation.
Collapse
Affiliation(s)
- Erik Joly
- Molecular Nutrition Unit and the Montreal Diabetes Research Center, Centre de recherche du CHUM, Pavillon de Sève, Y-4603, 1560 Sherbrooke Est, and the Department of Nutrition and Biochemistry, Université de Montréal, Montréal PQ, Canada, H3T 1C5
| | | | | | | | | | | |
Collapse
|
41
|
Kvaernø L, Werder M, Hauser H, Carreira EM. Synthesis and in vitro evaluation of inhibitors of intestinal cholesterol absorption. J Med Chem 2005; 48:6035-53. [PMID: 16162006 DOI: 10.1021/jm050422p] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
We have utilized our recently developed in vitro assay to address two key questions in the design of small-molecule cholesterol absorption inhibitors using ezetimibe, the only drug yet approved for the inhibition of cholesterol absorption in the small intestine, as a starting point: (1) the role of glycosylation and (2) the importance of the beta-lactam scaffold of ezetimibe for inhibitory activity. A wide range of nonhydrolyzable phenolic glycosides of ezetimibe were synthesized and demonstrated to be active inhibitors of cholesterol absorption using the brush border membrane vesicle assay. The analogous azetidines provided access to a variety of inhibitors in vitro, suggesting that the beta-lactam of ezetimibe merely serves as a ring scaffold to appropriately position the required substituents. Our findings highlight several promising strategies for the design of alternative small-molecule cholesterol absorption inhibitors that could ultimately be useful in preventing cardiovascular disease by lowering blood cholesterol levels.
Collapse
Affiliation(s)
- Lisbet Kvaernø
- Laboratorium für Organische Chemie der ETH-Zürich, HCI H 335, Wolfgang Pauli Strasse 10, CH-8093, Zurich, Switzerland
| | | | | | | |
Collapse
|
42
|
Temel RE, Lee RG, Kelley KL, Davis MA, Shah R, Sawyer JK, Wilson MD, Rudel LL. Intestinal cholesterol absorption is substantially reduced in mice deficient in both ABCA1 and ACAT2. J Lipid Res 2005; 46:2423-31. [PMID: 16150828 DOI: 10.1194/jlr.m500232-jlr200] [Citation(s) in RCA: 92] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The process of cholesterol absorption has yet to be completely defined at the molecular level. Because of its ability to esterify cholesterol for packaging into nascent chylomicrons, ACAT2 plays an important role in cholesterol absorption. However, it has been found that cholesterol absorption is not completely inhibited in ACAT2-deficient (ACAT2 KO) mice. Because ABCA1 mRNA expression was increased 3-fold in the small intestine of ACAT2 KO mice, we hypothesized that ABCA1-dependent cholesterol efflux sustains cholesterol absorption in the absence of ACAT2. To test this hypothesis, cholesterol absorption was measured in mice deficient in both ABCA1 and ACAT2 (DKO). Compared with wild-type, ABCA1 KO, or ACAT2 KO mice, DKO mice displayed the lowest level of cholesterol absorption. The concentrations of hepatic free and esterified cholesterol and gallbladder bile cholesterol were significantly reduced in DKO compared with wild-type and ABCA1 KO mice, although these measures of hepatic cholesterol metabolism were very similar in DKO and ACAT2 KO mice. We conclude that ABCA1, especially in the absence of ACAT2, can have a significant effect on cholesterol absorption, although ACAT2 has a more substantial role in this process than ABCA1.
Collapse
Affiliation(s)
- Ryan E Temel
- Department of Pathology, Section on Lipid Sciences, Wake Forest University Health Sciences, Winston-Salem, NC, USA
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Peretti N, Marcil V, Drouin E, Levy E. Mechanisms of lipid malabsorption in Cystic Fibrosis: the impact of essential fatty acids deficiency. Nutr Metab (Lond) 2005; 2:11. [PMID: 15869703 PMCID: PMC1134666 DOI: 10.1186/1743-7075-2-11] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2005] [Accepted: 05/03/2005] [Indexed: 12/26/2022] Open
Abstract
Transport mechanisms, whereby alimentary lipids are digested and packaged into small emulsion particles that enter intestinal cells to be translocated to the plasma in the form of chylomicrons, are impaired in cystic fibrosis. The purpose of this paper is to focus on defects that are related to intraluminal and intracellular events in this life-limiting genetic disorder. Specific evidence is presented to highlight the relationship between fat malabsorption and essential fatty acid deficiency commonly found in patients with cystic fibrosis that are often related to the genotype. Given the interdependency of pulmonary disease, pancreatic insufficiency and nutritional status, greater attention should be paid to the optimal correction of fat malabsorption and essential fatty acid deficiency in order to improve the quality of life and extend the life span of patients with cystic fibrosis.
Collapse
Affiliation(s)
- N Peretti
- Department of Nutrition, CHU-Sainte-Justine, Université de Montréal, Montréal, Québec, Canada
| | - V Marcil
- Department of Nutrition, CHU-Sainte-Justine, Université de Montréal, Montréal, Québec, Canada
| | - E Drouin
- Department of Pediatrics, CHU-Sainte-Justine, Université de Montréal, Montréal, Québec, Canada
| | - E Levy
- Department of Nutrition, CHU-Sainte-Justine, Université de Montréal, Montréal, Québec, Canada
| |
Collapse
|
44
|
van Bennekum A, Werder M, Thuahnai ST, Han CH, Duong P, Williams DL, Wettstein P, Schulthess G, Phillips MC, Hauser H. Class B scavenger receptor-mediated intestinal absorption of dietary beta-carotene and cholesterol. Biochemistry 2005; 44:4517-25. [PMID: 15766282 DOI: 10.1021/bi0484320] [Citation(s) in RCA: 202] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
There is now a general consensus that the intestinal absorption of water-insoluble, dietary lipids is protein-mediated, but the assignment of protein(s) to this function is still a matter of debate. To address this issue, we measured beta-carotene and cholesterol absorption in wild-type and SR-BI knockout mice and the uptake of these lipids in vitro using brush border membrane (BBM) vesicles. From the comparison of the in vivo and in vitro results we conclude that both BBM-resident class B scavenger receptors, SR-BI and CD36, can facilitate the absorption of beta-carotene and cholesterol. SR-BI is essential for beta-carotene absorption, at least in mice on a high fat diet. This is due to the fact that the absorption of beta-carotene is restricted to the duodenum and SR-BI is the predominant receptor in the mouse duodenum. In contrast, SR-BI may be involved but is not essential for cholesterol absorption in the small intestine. The question of whether SR-BI contributes to cholesterol absorption in vivo is still unresolved. Transfection of COS-7 cells with SR-BI or CD36 confers on these cells lipid uptake properties closely resembling those of enterocytes and BBM vesicles. Both scavenger receptors facilitate the uptake of dietary lipids such as beta-carotene, free and esterified cholesterol, phospholipids, and fatty acids into COS-7 cells. This lipid uptake is effected from three different lipid donor particles: mixed bile salt micelles, phospholipid small unilamellar vesicles, and trioleoylglycerol emulsions which are all likely to be present in the small intestine. Ezetimibe, a representative of a new class of drugs that inhibit intestinal cholesterol absorption, blocks SR-BI- and CD36-facilitated uptake of cholesterol into COS-7 cells.
Collapse
MESH Headings
- Animal Feed
- Animals
- Anticholesteremic Agents/administration & dosage
- Azetidines/administration & dosage
- CD36 Antigens/genetics
- CD36 Antigens/metabolism
- CD36 Antigens/physiology
- COS Cells
- Chlorocebus aethiops
- Cholesterol, Dietary/administration & dosage
- Cholesterol, Dietary/antagonists & inhibitors
- Cholesterol, Dietary/metabolism
- Ezetimibe
- Female
- Intestinal Absorption/drug effects
- Intestinal Absorption/genetics
- Intestine, Small/chemistry
- Intestine, Small/metabolism
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Microvilli/metabolism
- Rabbits
- Receptors, Immunologic/deficiency
- Receptors, Immunologic/genetics
- Receptors, Immunologic/metabolism
- Receptors, Immunologic/physiology
- Receptors, Scavenger
- Scavenger Receptors, Class B
- Transfection
- beta Carotene/administration & dosage
- beta Carotene/antagonists & inhibitors
- beta Carotene/metabolism
Collapse
Affiliation(s)
- Ariëtte van Bennekum
- Children's Hospital of Philadelphia and University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104-4318, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Marcil V, Peretti N, Delvin E, Levy E. Les processus digestifs et absorptifs des lipides alimentaires. ACTA ACUST UNITED AC 2004; 28:1257-66. [PMID: 15671937 DOI: 10.1016/s0399-8320(04)95219-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Affiliation(s)
- Valérie Marcil
- Centre de Recherche Hôpital Sainte-Justine, Département de Nutrition, Université de Montréal, Canada
| | | | | | | |
Collapse
|
46
|
Connelly MA, Williams DL. Scavenger receptor BI: a scavenger receptor with a mission to transport high density lipoprotein lipids. Curr Opin Lipidol 2004; 15:287-95. [PMID: 15166784 DOI: 10.1097/00041433-200406000-00008] [Citation(s) in RCA: 127] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW This review will survey recent findings on the cholesterol transport and scavenger functions of scavenger receptor BI. Although scavenger receptor BI and CD36 bind many of the same ligands, these two receptors have very specific lipid transport functions: CD36 facilitates the uptake of long chain fatty acids and SR-BI mediates the transport of cholesterol and cholesteryl ester from HDL particles. Scavenger receptor BI is a physiologically relevant HDL receptor that, along with HDL, is protective against cardiovascular disease. Its atheroprotective role has been hypothesized to be due to its function in the reverse cholesterol transport pathway. RECENT FINDINGS Recent studies suggest that scavenger receptor BI function is not only crucial for cholesterol delivery to the liver but is also important for cholesterol efflux at the vessel wall. Therefore, the receptor acts at both ends of the reverse cholesterol transport pathway. In addition, it stimulates nitric oxide production in endothelial cells, which may also contribute to its positive influence on the vasculature. Lastly, the glycoprotein was cloned as a scavenger receptor and in some cases is still thought to operate in this fashion. SUMMARY It will be interesting to follow future research on scavenger receptor BI that will delineate its functions in cholesterol transport as well as its scavenger functions. Additionally, we are only beginning to learn of the glycoprotein's effects on disease states besides atherosclerosis and cardiovascular disease.
Collapse
Affiliation(s)
- Margery A Connelly
- Department of Pharmacological Sciences, University Medical Center, State University of New York at Stony Brook, Stony Brook, NY 11794-8651, USA
| | | |
Collapse
|