101
|
Hiebl V, Ladurner A, Latkolik S, Dirsch VM. Natural products as modulators of the nuclear receptors and metabolic sensors LXR, FXR and RXR. Biotechnol Adv 2018; 36:1657-1698. [PMID: 29548878 DOI: 10.1016/j.biotechadv.2018.03.003] [Citation(s) in RCA: 97] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 03/02/2018] [Accepted: 03/08/2018] [Indexed: 01/25/2023]
Abstract
Nuclear receptors (NRs) represent attractive targets for the treatment of metabolic syndrome-related diseases. In addition, natural products are an interesting pool of potential ligands since they have been refined under evolutionary pressure to interact with proteins or other biological targets. This review aims to briefly summarize current basic knowledge regarding the liver X (LXR) and farnesoid X receptors (FXR) that form permissive heterodimers with retinoid X receptors (RXR). Natural product-based ligands for these receptors are summarized and the potential of LXR, FXR and RXR as targets in precision medicine is discussed.
Collapse
Affiliation(s)
- Verena Hiebl
- University of Vienna, Department of Pharmacognosy, Althanstrasse 14, 1090 Vienna, Austria
| | - Angela Ladurner
- University of Vienna, Department of Pharmacognosy, Althanstrasse 14, 1090 Vienna, Austria.
| | - Simone Latkolik
- University of Vienna, Department of Pharmacognosy, Althanstrasse 14, 1090 Vienna, Austria
| | - Verena M Dirsch
- University of Vienna, Department of Pharmacognosy, Althanstrasse 14, 1090 Vienna, Austria
| |
Collapse
|
102
|
Ayisi CL, Yamei C, Zhao JL. Genes, transcription factors and enzymes involved in lipid metabolism in fin fish. ACTA ACUST UNITED AC 2018. [DOI: 10.1016/j.aggene.2017.09.006] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
103
|
de Boer JF, Kuipers F, Groen AK. Cholesterol Transport Revisited: A New Turbo Mechanism to Drive Cholesterol Excretion. Trends Endocrinol Metab 2018; 29:123-133. [PMID: 29276134 DOI: 10.1016/j.tem.2017.11.006] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2017] [Revised: 11/26/2017] [Accepted: 11/29/2017] [Indexed: 12/31/2022]
Abstract
A fine-tuned balance between cholesterol uptake and excretion by the body is pivotal to maintain health and to remain free from the deleterious consequences of cholesterol accumulation such as cardiovascular disease. The pathways involved in intracellular and extracellular cholesterol transport are a subject of intense investigation and are being unraveled in increasing detail. In addition, insight into the complex interactions between cholesterol and bile acid metabolism has increased considerably in the last couple of years. This review provides an overview of the mechanisms involved in cholesterol uptake and excretion, with a particular emphasis on the most recent progress in this field. Special attention is given to the transintestinal cholesterol excretion (TICE) pathway, which was recently demonstrated to have a remarkably high transport capacity and to be sensitive to pharmacological modulation.
Collapse
Affiliation(s)
- Jan Freark de Boer
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands; Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
| | - Folkert Kuipers
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands; Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Albert K Groen
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands; Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands; Department of Vascular Medicine, University of Amsterdam Academic Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
104
|
Cuffe H, Liu M, Key CCC, Boudyguina E, Sawyer JK, Weckerle A, Bashore A, Fried SK, Chung S, Parks JS. Targeted Deletion of Adipocyte Abca1 (ATP-Binding Cassette Transporter A1) Impairs Diet-Induced Obesity. Arterioscler Thromb Vasc Biol 2018; 38:733-743. [PMID: 29348118 DOI: 10.1161/atvbaha.117.309880] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2017] [Accepted: 01/01/2018] [Indexed: 01/23/2023]
Abstract
OBJECTIVE Adipose tissue cholesterol increases with adipocyte triglyceride content and size during development of obesity. However, how adipocyte cholesterol affects adipocyte function is poorly understood. The aim of this study was to evaluate the role of the cellular cholesterol exporter, Abca1 (ATP-binding cassette transporter A1), on adipose tissue function during diet-induced obesity. APPROACH AND RESULTS Adiponectin Cre recombinase transgenic mice were crossed with Abca1flox/flox mice to generate ASKO (adipocyte-specific Abca1 knockout) mice. Control and ASKO mice were then fed a high-fat, high-cholesterol (45% calories as fat and 0.2% cholesterol) diet for 16 weeks. Compared with control mice, ASKO mice had a 2-fold increase in adipocyte plasma membrane cholesterol content and significantly lower body weight, epididymal fat pad weight, and adipocyte size. ASKO versus control adipose tissue had decreased PPARγ (peroxisome proliferator-activated receptor γ) and CCAAT/enhancer-binding protein expression, nuclear SREBP1 (sterol regulatory element-binding protein 1) protein, lipogenesis, and triglyceride accretion but similar Akt activation after acute insulin stimulation. Acute siRNA-mediated Abca1 silencing during 3T3L1 adipocyte differentiation reduced adipocyte Abca1 and PPARγ protein expression and triglyceride content. Systemic stimulated triglyceride lipolysis and glucose homeostasis were similar between control and ASKO mice. CONCLUSIONS Adipocyte Abca1 is a key regulator of adipocyte lipogenesis and lipid accretion, likely because of increased adipose tissue membrane cholesterol, resulting in decreased activation of lipogenic transcription factors PPARγ and SREBP1.
Collapse
Affiliation(s)
- Helen Cuffe
- From the Section on Molecular Medicine, Department of Internal Medicine (H.C., M.L., C.-C.C.K., E.B., J.K.S., A.W., A.B., J.S.P.) and Department of Biochemistry (J.S.P.), Wake Forest School of Medicine, Winston-Salem, NC; Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York (S.K.F.); and Department of Nutrition and Health Sciences, University of Nebraska, Lincoln, NE (S.C.)
| | - Mingxia Liu
- From the Section on Molecular Medicine, Department of Internal Medicine (H.C., M.L., C.-C.C.K., E.B., J.K.S., A.W., A.B., J.S.P.) and Department of Biochemistry (J.S.P.), Wake Forest School of Medicine, Winston-Salem, NC; Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York (S.K.F.); and Department of Nutrition and Health Sciences, University of Nebraska, Lincoln, NE (S.C.)
| | - Chia-Chi C Key
- From the Section on Molecular Medicine, Department of Internal Medicine (H.C., M.L., C.-C.C.K., E.B., J.K.S., A.W., A.B., J.S.P.) and Department of Biochemistry (J.S.P.), Wake Forest School of Medicine, Winston-Salem, NC; Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York (S.K.F.); and Department of Nutrition and Health Sciences, University of Nebraska, Lincoln, NE (S.C.)
| | - Elena Boudyguina
- From the Section on Molecular Medicine, Department of Internal Medicine (H.C., M.L., C.-C.C.K., E.B., J.K.S., A.W., A.B., J.S.P.) and Department of Biochemistry (J.S.P.), Wake Forest School of Medicine, Winston-Salem, NC; Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York (S.K.F.); and Department of Nutrition and Health Sciences, University of Nebraska, Lincoln, NE (S.C.)
| | - Janet K Sawyer
- From the Section on Molecular Medicine, Department of Internal Medicine (H.C., M.L., C.-C.C.K., E.B., J.K.S., A.W., A.B., J.S.P.) and Department of Biochemistry (J.S.P.), Wake Forest School of Medicine, Winston-Salem, NC; Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York (S.K.F.); and Department of Nutrition and Health Sciences, University of Nebraska, Lincoln, NE (S.C.)
| | - Allison Weckerle
- From the Section on Molecular Medicine, Department of Internal Medicine (H.C., M.L., C.-C.C.K., E.B., J.K.S., A.W., A.B., J.S.P.) and Department of Biochemistry (J.S.P.), Wake Forest School of Medicine, Winston-Salem, NC; Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York (S.K.F.); and Department of Nutrition and Health Sciences, University of Nebraska, Lincoln, NE (S.C.)
| | - Alexander Bashore
- From the Section on Molecular Medicine, Department of Internal Medicine (H.C., M.L., C.-C.C.K., E.B., J.K.S., A.W., A.B., J.S.P.) and Department of Biochemistry (J.S.P.), Wake Forest School of Medicine, Winston-Salem, NC; Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York (S.K.F.); and Department of Nutrition and Health Sciences, University of Nebraska, Lincoln, NE (S.C.)
| | - Susan K Fried
- From the Section on Molecular Medicine, Department of Internal Medicine (H.C., M.L., C.-C.C.K., E.B., J.K.S., A.W., A.B., J.S.P.) and Department of Biochemistry (J.S.P.), Wake Forest School of Medicine, Winston-Salem, NC; Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York (S.K.F.); and Department of Nutrition and Health Sciences, University of Nebraska, Lincoln, NE (S.C.)
| | - Soonkyu Chung
- From the Section on Molecular Medicine, Department of Internal Medicine (H.C., M.L., C.-C.C.K., E.B., J.K.S., A.W., A.B., J.S.P.) and Department of Biochemistry (J.S.P.), Wake Forest School of Medicine, Winston-Salem, NC; Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York (S.K.F.); and Department of Nutrition and Health Sciences, University of Nebraska, Lincoln, NE (S.C.)
| | - John S Parks
- From the Section on Molecular Medicine, Department of Internal Medicine (H.C., M.L., C.-C.C.K., E.B., J.K.S., A.W., A.B., J.S.P.) and Department of Biochemistry (J.S.P.), Wake Forest School of Medicine, Winston-Salem, NC; Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York (S.K.F.); and Department of Nutrition and Health Sciences, University of Nebraska, Lincoln, NE (S.C.).
| |
Collapse
|
105
|
Phillips MC. Is ABCA1 a lipid transfer protein? J Lipid Res 2018; 59:749-763. [PMID: 29305383 DOI: 10.1194/jlr.r082313] [Citation(s) in RCA: 123] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 01/02/2018] [Indexed: 12/16/2022] Open
Abstract
ABCA1 functions as a lipid transporter because it mediates the transfer of cellular phospholipid (PL) and free (unesterified) cholesterol (FC) to apoA-I and related proteins present in the extracellular medium. ABCA1 is a membrane PL translocase and its enzymatic activity leads to transfer of PL molecules from the cytoplasmic leaflet to the exofacial leaflet of a cell plasma membrane (PM). The presence of active ABCA1 in the PM promotes binding of apoA-I to the cell surface. About 10% of this bound apoA-I interacts directly with ABCA1 and stabilizes the transporter. Most of the pool of cell surface-associated apoA-I is bound to lipid domains in the PM that are created by the activity of ABCA1. The amphipathic α-helices in apoA-I confer detergent-like properties on the protein enabling it to solubilize PL and FC in these membrane domains to create a heterogeneous population of discoidal nascent HDL particles. This review focuses on current understanding of the structure-function relationships of human ABCA1 and the molecular mechanisms underlying HDL particle production.
Collapse
Affiliation(s)
- Michael C Phillips
- Division of Translational Medicine and Human Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-5158
| |
Collapse
|
106
|
Mangat R, Borthwick F, Haase T, Jacome M, Nelson R, Kontush A, Vine DF, Proctor SD. Intestinal lymphatic HDL miR‐223 and ApoA‐I are reduced during insulin resistance and restored with niacin. FASEB J 2018; 32:1602-1612. [DOI: 10.1096/fj.201600298rr] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Affiliation(s)
- Rabban Mangat
- Metabolic and Cardiovascular Diseases Laboratory, Group on the Molecular Cell Biology of Lipids University of Alberta Edmonton Alberta Canada
- Alberta Diabetes Institute University of Alberta Edmonton Alberta Canada
- Mazankowski Alberta Heart Institute University of Alberta Edmonton Alberta Canada
| | - Faye Borthwick
- Metabolic and Cardiovascular Diseases Laboratory, Group on the Molecular Cell Biology of Lipids University of Alberta Edmonton Alberta Canada
- Alberta Diabetes Institute University of Alberta Edmonton Alberta Canada
- Mazankowski Alberta Heart Institute University of Alberta Edmonton Alberta Canada
| | - Tina Haase
- Metabolic and Cardiovascular Diseases Laboratory, Group on the Molecular Cell Biology of Lipids University of Alberta Edmonton Alberta Canada
- Alberta Diabetes Institute University of Alberta Edmonton Alberta Canada
- Mazankowski Alberta Heart Institute University of Alberta Edmonton Alberta Canada
| | - Miriam Jacome
- Metabolic and Cardiovascular Diseases Laboratory, Group on the Molecular Cell Biology of Lipids University of Alberta Edmonton Alberta Canada
- Alberta Diabetes Institute University of Alberta Edmonton Alberta Canada
- Mazankowski Alberta Heart Institute University of Alberta Edmonton Alberta Canada
| | - Randy Nelson
- Metabolic and Cardiovascular Diseases Laboratory, Group on the Molecular Cell Biology of Lipids University of Alberta Edmonton Alberta Canada
- Alberta Diabetes Institute University of Alberta Edmonton Alberta Canada
- Mazankowski Alberta Heart Institute University of Alberta Edmonton Alberta Canada
| | - Anatol Kontush
- National Institute for Health and Medical Research University of Pierre and Marie Curie, Salpétrière University Hospital Paris France
| | - Donna F. Vine
- Metabolic and Cardiovascular Diseases Laboratory, Group on the Molecular Cell Biology of Lipids University of Alberta Edmonton Alberta Canada
- Alberta Diabetes Institute University of Alberta Edmonton Alberta Canada
- Mazankowski Alberta Heart Institute University of Alberta Edmonton Alberta Canada
| | - Spencer D. Proctor
- Metabolic and Cardiovascular Diseases Laboratory, Group on the Molecular Cell Biology of Lipids University of Alberta Edmonton Alberta Canada
- Alberta Diabetes Institute University of Alberta Edmonton Alberta Canada
- Mazankowski Alberta Heart Institute University of Alberta Edmonton Alberta Canada
| |
Collapse
|
107
|
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
|
108
|
Yamauchi Y, Rogers MA. Sterol Metabolism and Transport in Atherosclerosis and Cancer. Front Endocrinol (Lausanne) 2018; 9:509. [PMID: 30283400 PMCID: PMC6157400 DOI: 10.3389/fendo.2018.00509] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 08/14/2018] [Indexed: 01/22/2023] Open
Abstract
Cholesterol is a vital lipid molecule for mammalian cells, regulating fluidity of biological membranes, and serving as an essential constituent of lipid rafts. Mammalian cells acquire cholesterol from extracellular lipoproteins and from de novo synthesis. Cholesterol biosynthesis generates various precursor sterols. Cholesterol undergoes metabolic conversion into oxygenated sterols (oxysterols), bile acids, and steroid hormones. Cholesterol intermediates and metabolites have diverse and important cellular functions. A network of molecular machineries including transcription factors, protein modifiers, sterol transporters/carriers, and sterol sensors regulate sterol homeostasis in mammalian cells and tissues. Dysfunction in metabolism and transport of cholesterol, sterol intermediates, and oxysterols occurs in various pathophysiological settings such as atherosclerosis, cancers, and neurodegenerative diseases. Here we review the cholesterol, intermediate sterol, and oxysterol regulatory mechanisms and intracellular transport machineries, and discuss the roles of sterols and sterol metabolism in human diseases.
Collapse
Affiliation(s)
- Yoshio Yamauchi
- Nutri-Life Science Laboratory, Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, University of Tokyo, Tokyo, Japan
- AMED-CREST, Japan Agency for Medical Research and Development, Tokyo, Japan
- *Correspondence: Yoshio Yamauchi
| | - Maximillian A. Rogers
- Division of Cardiovascular Medicine, Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
109
|
Taghipoor Asramy A, Ghanbari-Niaki A, Hakemi S, Naghizadeh Qomi M, Moghanny Bashi MM. Effect of 12 Weeks of Intense Endurance Training and Bee Pollen Consumption on ABCA1 Gene Expression in Small Intestine, Liver and Gastrocnemius Muscle of Male Rats. MEDICAL LABORATORY JOURNAL 2018. [DOI: 10.29252/mlj.12.1.11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
|
110
|
Herbert KE, Erridge C. Regulation of low-density lipoprotein cholesterol by intestinal inflammation and the acute phase response. Cardiovasc Res 2017; 114:226-232. [DOI: 10.1093/cvr/cvx237] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Accepted: 11/29/2017] [Indexed: 02/07/2023] Open
Abstract
AbstractSystemic inflammation, induced by disease or experimental intervention, is well established to result in elevated levels of circulating triglycerides, and reduced levels of high-density lipoprotein-cholesterol (HDL-C), in most mammalian species. However, the relationship between inflammation and low-density lipoprotein-cholesterol (LDL-C) concentrations is less clear. Most reports indicate that systemic inflammation, as observed during sepsis or following high dose experimental endotoxaemia, lowers total, and LDL-C in man. However, isolated reports have suggested that certain inflammatory conditions are associated with increased LDL-C. In this review, we summarize the emerging evidence that low-grade inflammation specifically of intestinal origin may be associated with increased serum LDL-C levels. Preliminary insights into potential mechanisms that may mediate these effects, including those connecting inflammation to trans-intestinal cholesterol efflux (TICE), are considered. We conclude that this evidence supports the potential downregulation of major mediators of TICE by inflammatory mediators in vitro and during intestinal inflammation in vivo. The TICE-inflammation axis therefore merits further study in terms of its potential to regulate serum LDL-C, and as a readily druggable target for hypercholesterolaemia.
Collapse
Affiliation(s)
- Karl E Herbert
- Department of Cardiovascular Sciences, Glenfield Hospital, University of Leicester, Groby Road, Leicester, Leicestershire, LE3 9QP, UK
| | - Clett Erridge
- Department of Cardiovascular Sciences, Glenfield Hospital, University of Leicester, Groby Road, Leicester, Leicestershire, LE3 9QP, UK
- Department of Biomedical and Forensic Sciences, Anglia Ruskin University, East Road, Cambridge, Cambridgeshire, CB1 1PT, UK
| |
Collapse
|
111
|
Zannis VI, Su S, Fotakis P. Role of apolipoproteins, ABCA1 and LCAT in the biogenesis of normal and aberrant high density lipoproteins. J Biomed Res 2017; 31:471. [PMID: 29109329 PMCID: PMC6307667 DOI: 10.7555/jbr.31.20160082] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 08/30/2016] [Indexed: 12/28/2022] Open
Abstract
In this review, we focus on the pathway of biogenesis of HDL, the essential role of apoA-I, ATP binding cassette transporter A1 (ABCA1), and lecithin: cholesterol acyltransferase (LCAT) in the formation of plasma HDL; the generation of aberrant forms of HDL containing mutant apoA-I forms and the role of apoA-IV and apoE in the formation of distinct HDL subpopulations. The biogenesis of HDL requires functional interactions of the ABCA1 with apoA-I (and to a lesser extent with apoE and apoA-IV) and subsequent interactions of the nascent HDL species thus formed with LCAT. Mutations in apoA-I, ABCA1 and LCAT either prevent or impair the formation of HDL and may also affect the functionality of the HDL species formed. Emphasis is placed on three categories of apoA-I mutations. The first category describes a unique bio-engineered apoA-I mutation that disrupts interactions between apoA-I and ABCA1 and generates aberrant preβ HDL subpopulations that cannot be converted efficiently to α subpopulations by LCAT. The second category describes natural and bio-engineered apoA-I mutations that generate preβ and small size α4 HDL subpopulations, and are associated with low plasma HDL levels. These phenotypes can be corrected by excess LCAT. The third category describes bio-engineered apoA-I mutations that induce hypertriglyceridemia that can be corrected by excess lipoprotein lipase and also have defective maturation of HDL. The HDL phenotypes described here may serve in the future for diagnosis, prognoses and potential treatment of abnormalities that affect the biogenesis and functionality of HDL.
Collapse
Affiliation(s)
- Vassilis I. Zannis
- . Molecular Genetics, Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA 02118, USA
- . Department University of Crete, School of Medicine, Heraklion, Crete, Greece
| | - Shi Su
- . Molecular Genetics, Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA 02118, USA
| | - Panagiotis Fotakis
- . Molecular Genetics, Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA 02118, USA
- . Department University of Crete, School of Medicine, Heraklion, Crete, Greece
| |
Collapse
|
112
|
Nikolova V, Papacleovoulou G, Bellafante E, Borges Manna L, Jansen E, Baron S, Abu-Hayyeh S, Parker M, Williamson C. Changes in LXR signaling influence early-pregnancy lipogenesis and protect against dysregulated fetoplacental lipid homeostasis. Am J Physiol Endocrinol Metab 2017; 313:E463-E472. [PMID: 28420650 PMCID: PMC5689017 DOI: 10.1152/ajpendo.00449.2016] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Revised: 04/13/2017] [Accepted: 04/13/2017] [Indexed: 12/13/2022]
Abstract
Human pregnancy is associated with enhanced de novo lipogenesis in the early stages followed by hyperlipidemia during advanced gestation. Liver X receptors (LXRs) are oxysterol-activated nuclear receptors that stimulate de novo lipogenesis and also promote the efflux of cholesterol from extrahepatic tissues followed by its transport back to the liver for biliary excretion. Although LXR is recognized as a master regulator of triglyceride and cholesterol homeostasis, it is unknown whether it facilitates the gestational adaptations in lipid metabolism. To address this question, biochemical profiling, protein quantification, and gene expression studies were used, and gestational metabolic changes in T0901317-treated wild-type mice and Lxrab-/- mutants were investigated. Here, we show that altered LXR signaling contributes to the enhanced lipogenesis in early pregnancy by increasing the expression of hepatic Fas and stearoyl-CoA desaturase 1 (Scd1). Both the pharmacological activation of LXR with T0901317 and the genetic ablation of its two isoforms disrupted the increase in hepatic fatty acid biosynthesis and the development of hypertriglyceridemia during early gestation. We also demonstrate that absence of LXR enhances maternal white adipose tissue lipolysis, causing abnormal accumulation of triglycerides, cholesterol, and free fatty acids in the fetal liver. Together, these data identify LXR as an important factor in early-pregnancy lipogenesis that is also necessary to protect against abnormalities in fetoplacental lipid homeostasis.
Collapse
Affiliation(s)
- Vanya Nikolova
- Women's Health Academic Centre, King's College London, London, United Kingdom
| | | | - Elena Bellafante
- Women's Health Academic Centre, King's College London, London, United Kingdom
| | - Luiza Borges Manna
- Institute of Reproductive and Developmental Biology, Imperial College London, London, United Kingdom
| | - Eugene Jansen
- Centre for Health Protection, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - Silvère Baron
- Laboratoire Génétique Reproduction et Développement, Université Clermont Auvergne, Clermont-Ferrand, France; and
| | - Shadi Abu-Hayyeh
- Women's Health Academic Centre, King's College London, London, United Kingdom
| | - Malcolm Parker
- Institute of Reproductive and Developmental Biology, Imperial College London, London, United Kingdom
| | | |
Collapse
|
113
|
Khalil A, Kamtchueng Simo O, Ikhlef S, Berrougui H. The role of paraoxonase 1 in regulating high-density lipoprotein functionality during aging. Can J Physiol Pharmacol 2017; 95:1254-1262. [DOI: 10.1139/cjpp-2017-0117] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Pharmacological interventions to increase the concentration of high-density lipoprotein (HDL) have led to disappointing results and have contributed to the emergence of the concept of HDL functionality. The anti-atherogenic activity of HDLs can be explained by their functionality or quality. The capacity of HDLs to maintain cellular cholesterol homeostasis and to transport cholesterol from peripheral cells to the liver for elimination is one of their principal anti-atherogenic activities. However, HDLs possess several other attributes that contribute to their protective effect against cardiovascular diseases. HDL functionality is regulated by various proteins and lipids making up HDL particles. However, several studies investigated the role of paraoxonase 1 (PON1) and suggest a significant role of this protein in the regulation of the functionality of HDLs. Moreover, research on PON1 attracted much interest following several studies indicating that it is involved in cardiovascular protection. However, the mechanisms by which PON1 exerts these effects remain to be elucidated.
Collapse
Affiliation(s)
- Abdelouahed Khalil
- Research Centre on Aging, Sherbrooke, QC J1H 4C4, Canada
- Department of Medicine, Geriatrics Service, Faculty of Medicine and Biological Sciences, University of Sherbrooke, Sherbrooke, QC J1H 4N4, Canada
| | | | - Souade Ikhlef
- Research Centre on Aging, Sherbrooke, QC J1H 4C4, Canada
| | - Hicham Berrougui
- Department of Biology, Polydisciplinary Faculty, University Sultan Moulay Slimane, BP 592, 23000 Beni Mellal, Morocco
| |
Collapse
|
114
|
Dunbar RL, Movva R, Bloedon LT, Duffy D, Norris RB, Navab M, Fogelman AM, Rader DJ. Oral Apolipoprotein A-I Mimetic D-4F Lowers HDL-Inflammatory Index in High-Risk Patients: A First-in-Human Multiple-Dose, Randomized Controlled Trial. Clin Transl Sci 2017; 10:455-469. [PMID: 28795506 PMCID: PMC5673907 DOI: 10.1111/cts.12487] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Accepted: 06/13/2017] [Indexed: 12/26/2022] Open
Abstract
A single dose of the apolipoprotein (apo)A-I mimetic peptide D-4F rendered high-density lipoprotein (HDL) less inflammatory, motivating the first multiple-dose study. We aimed to assess safety/tolerability, pharmacokinetics, and pharmacodynamics of daily, orally administered D-4F. High-risk coronary heart disease (CHD) subjects added double-blinded placebo or D-4F to statin for 13 days, randomly assigned 1:3 to ascending cohorts of 100, 300, then 500 mg (n = 62; 46 men/16 women). D-4F was safe and well-tolerated. Mean ± SD plasma D-4F area under the curve (AUC, 0-8h) was 6.9 ± 5.7 ng/mL*h (100 mg), 22.7 ± 19.6 ng/mL*h (300 mg), and 104.0 ± 60.9 ng/mL*h (500 mg) among men, higher among women. Whereas placebo dropped HDL inflammatory index (HII) 28% 8 h postdose (range, 1.25-0.86), 300-500 mg D-4F effectively halved HII: 1.35-0.57 and 1.22-0.63, respectively (P < 0.03 vs. placebo). Oral D-4F peptide dose predicted HII suppression, whereas plasma D-4F exposure was dissociated, suggesting plasma penetration is unnecessary. In conclusion, oral D-4F dosing rendered HDL less inflammatory, affirming oral D-4F as a potential therapy to improve HDL function.
Collapse
Affiliation(s)
- Richard L Dunbar
- Department of Medicine, Division of Translational Medicine and Human Genetics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Department of Medicine, Division of Cardiovascular Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Institute for Translational Medicine and Therapeutics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA.,The Cardiovascular Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Rajesh Movva
- Eastern Maine Medical Center, Bangor, Maine, USA
| | | | - Danielle Duffy
- Department of Medicine, Division of Cardiology, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Robert B Norris
- Department of Medicine, Division of Cardiovascular Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Mohamad Navab
- University of California, Los Angeles, California, USA
| | | | - Daniel J Rader
- Department of Medicine, Division of Translational Medicine and Human Genetics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Department of Medicine, Division of Cardiovascular Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Institute for Translational Medicine and Therapeutics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA.,The Cardiovascular Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
115
|
Lim FT, Lim SM, Ramasamy K. Cholesterol lowering by Pediococcus acidilactici LAB4 and Lactobacillus plantarum LAB12 in adult zebrafish is associated with improved memory and involves an interplay between npc1l1 and abca1. Food Funct 2017; 8:2817-2828. [PMID: 28725889 DOI: 10.1039/c7fo00764g] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
This study assessed the cholesterol lowering effect of Pediococcus acidilactici LAB4 and Lactobacillus plantarum LAB12 using adult zebrafish. Animals were fed with a high cholesterol diet (HCD) with/without LAB for seven weeks. Serum and liver cholesterol was quantified using colorimetric and dye staining methods. Expressions of npc1l1 and abca1 in the liver and intestine and appa in the brain were quantified using RT-PCR. Serum and liver cholesterol was significantly lowered in LAB4- and LAB12-fed zebrafish (≤64% and ≤71%, respectively), with reduced liver cholesterol deposition. The cholesterol lowering effect was accompanied by down-regulation of npc1l1 in intestines (≤28.7%), up-regulation of abca1 in the liver (≥30.5%) and down-regulation of appa in the brain (≤24.5%). A moderately strong positive Pearson correlation (r = 0.617, p < 0.01) was found between appa and serum cholesterol. LAB-fed zebrafish exhibited improved spatial learning and memory. LAB4 and LAB12 can be potentially used in preventing hypercholesterolaemia and Alzheimer's diseases.
Collapse
Affiliation(s)
- Fei Tieng Lim
- Collaborative Drug Discovery Research (CDDR) Group, Faculty of Pharmacy, University Teknologi MARA (UiTM), 42300 Bandar Puncak Alam, Selangor Darul Ehsan, Malaysia.
| | - Siong Meng Lim
- Collaborative Drug Discovery Research (CDDR) Group, Faculty of Pharmacy, University Teknologi MARA (UiTM), 42300 Bandar Puncak Alam, Selangor Darul Ehsan, Malaysia.
| | - Kalavathy Ramasamy
- Collaborative Drug Discovery Research (CDDR) Group, Faculty of Pharmacy, University Teknologi MARA (UiTM), 42300 Bandar Puncak Alam, Selangor Darul Ehsan, Malaysia.
| |
Collapse
|
116
|
Liu M, Chung S, Shelness GS, Parks JS. Hepatic ABCA1 deficiency is associated with delayed apolipoprotein B secretory trafficking and augmented VLDL triglyceride secretion. Biochim Biophys Acta Mol Cell Biol Lipids 2017; 1862:1035-1043. [PMID: 28694219 DOI: 10.1016/j.bbalip.2017.07.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 06/30/2017] [Accepted: 07/05/2017] [Indexed: 11/30/2022]
Abstract
ATP binding cassette transporter A1 (ABCA1) is a membrane transporter that facilitates nascent HDL formation. Tangier disease subjects with complete ABCA1 deficiency have <5% of normal levels of plasma HDL, elevated triglycerides (TGs), and defective vesicular trafficking in fibroblasts and macrophages. Hepatocyte-specific ABCA1 knockout mice (HSKO) have a similar lipid phenotype with 20% of normal plasma HDL levels and a two-fold elevation of plasma TGs due to hepatic overproduction of large, triglyceride-enriched VLDL. We hypothesized that enhanced VLDL TG secretion in the absence of hepatocyte ABCA1 is due to altered intracellular trafficking of apolipoprotein B (apoB), resulting in augmented TG addition to nascent VLDL. We found that trafficking of newly synthesized apoB through the secretory pathway was delayed in ABCA1-silenced rat hepatoma cells and HSKO primary hepatocytes, relative to controls. Endoglycosidase H treatment of cellular apoB revealed a likely delay in apoB trafficking in post-ER compartments. The reduced rate of protein trafficking was also observed for an adenoviral-expressed GPI-linked fluorescent fusion protein, but not albumin, suggesting a selective delay of secretory cargoes in the absence of hepatocyte ABCA1. Our results suggest an important role for hepatic ABCA1 in regulating secretory trafficking and modulating VLDL expansion during the TG accretion phase of hepatic lipoprotein particle assembly.
Collapse
Affiliation(s)
- Mingxia Liu
- Department of Internal Medicine-Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA.
| | - Soonkyu Chung
- Department of Internal Medicine-Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Gregory S Shelness
- Department of Internal Medicine-Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - John S Parks
- Department of Internal Medicine-Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA; Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC, USA
| |
Collapse
|
117
|
Chuang JC, Lopez AM, Turley SD. Quantitation of the rates of hepatic and intestinal cholesterol synthesis in lysosomal acid lipase-deficient mice before and during treatment with ezetimibe. Biochem Pharmacol 2017; 135:116-125. [PMID: 28322747 PMCID: PMC5489310 DOI: 10.1016/j.bcp.2017.03.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Accepted: 03/14/2017] [Indexed: 01/28/2023]
Abstract
Esterified cholesterol (EC) and triglycerides, contained within lipoproteins taken up by cells, are hydrolysed by lysosomal acid lipase (LAL) in the late endosomal/lysosomal (E/L) compartment. The resulting unesterified cholesterol (UC) is transported via Niemann-Pick type C2 and C1 into the cytosolic compartment where it enters a putative pool of metabolically active cholesterol that is utilized in accordance with cellular needs. Loss-of-function mutations in LIPA, the gene encoding LAL, result in dramatic increases in tissue concentrations of EC, a hallmark feature of Wolman disease and cholesteryl ester storage disease (CESD). The lysosomal sequestration of EC causes cells to respond to a perceived deficit of sterol by increasing their rate of cholesterol synthesis, particularly in the liver. A similar compensatory response occurs with treatments that disrupt the enterohepatic movement of cholesterol or bile acids. Here we measured rates of cholesterol synthesis in vivo in the liver and small intestine of a mouse model for CESD given the cholesterol absorption inhibitor ezetimibe from weaning until early adulthood. Consistent with previous findings, this treatment significantly reduced the amount of EC sequestered in the liver (from 132.43±7.35 to 70.07±6.04mg/organ) and small intestine (from 2.78±0.21 to 1.34±0.09mg/organ) in the LAL-deficient mice even though their rates of hepatic and intestinal cholesterol synthesis were either comparable to, or exceeded those in matching untreated Lal-/- mice. These data reveal the role of intestinal cholesterol absorption in driving the expansion of tissue EC content and disease progression in LAL deficiency.
Collapse
Affiliation(s)
- Jen-Chieh Chuang
- Department of Internal Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX, United States.
| | - Adam M Lopez
- Department of Internal Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX, United States.
| | - Stephen D Turley
- Department of Internal Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX, United States.
| |
Collapse
|
118
|
Papachristou NI, Blair HC, Kypreos KE, Papachristou DJ. High-density lipoprotein (HDL) metabolism and bone mass. J Endocrinol 2017; 233:R95-R107. [PMID: 28314771 PMCID: PMC5598779 DOI: 10.1530/joe-16-0657] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 03/17/2017] [Indexed: 02/06/2023]
Abstract
It is well appreciated that high-density lipoprotein (HDL) and bone physiology and pathology are tightly linked. Studies, primarily in mouse models, have shown that dysfunctional and/or disturbed HDL can affect bone mass through many different ways. Specifically, reduced HDL levels have been associated with the development of an inflammatory microenvironment that affects the differentiation and function of osteoblasts. In addition, perturbation in metabolic pathways of HDL favors adipoblastic differentiation and restrains osteoblastic differentiation through, among others, the modification of specific bone-related chemokines and signaling cascades. Increased bone marrow adiposity also deteriorates bone osteoblastic function and thus bone synthesis, leading to reduced bone mass. In this review, we present the current knowledge and the future directions with regard to the HDL-bone mass connection. Unraveling the molecular phenomena that underline this connection will promote the deeper understanding of the pathophysiology of bone-related pathologies, such as osteoporosis or bone metastasis, and pave the way toward the development of novel and more effective therapies against these conditions.
Collapse
Affiliation(s)
- Nicholaos I Papachristou
- Department of Anatomy-Histology-EmbryologyUnit of Bone and Soft Tissue Studies, University of Patras Medical School, Patras, Greece
| | - Harry C Blair
- Department of PathologyUniversity of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Pittsburgh VA Medical CenterPittsburgh, Pennsylvania, USA
| | - Kyriakos E Kypreos
- Department of PharmacologyUniversity of Patras Medical School, Patras, Greece
| | - Dionysios J Papachristou
- Department of Anatomy-Histology-EmbryologyUnit of Bone and Soft Tissue Studies, University of Patras Medical School, Patras, Greece
- Department of PathologyUniversity of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
119
|
Doktorova M, Zwarts I, Zutphen TV, Dijk THV, Bloks VW, Harkema L, Bruin AD, Downes M, Evans RM, Verkade HJ, Jonker JW. Intestinal PPARδ protects against diet-induced obesity, insulin resistance and dyslipidemia. Sci Rep 2017; 7:846. [PMID: 28404991 PMCID: PMC5429805 DOI: 10.1038/s41598-017-00889-z] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 03/21/2017] [Indexed: 01/03/2023] Open
Abstract
Peroxisome proliferator-activated receptor δ (PPARδ) is a ligand-activated transcription factor that has an important role in lipid metabolism. Activation of PPARδ stimulates fatty acid oxidation in adipose tissue and skeletal muscle and improves dyslipidemia in mice and humans. PPARδ is highly expressed in the intestinal tract but its physiological function in this organ is not known. Using mice with an intestinal epithelial cell-specific deletion of PPARδ, we show that intestinal PPARδ protects against diet-induced obesity, insulin resistance and dyslipidemia. Furthermore, absence of intestinal PPARδ abolished the ability of PPARδ agonist GW501516 to increase plasma levels of HDL-cholesterol. Together, our findings show that intestinal PPARδ is important in maintaining metabolic homeostasis and suggest that intestinal-specific activation of PPARδ could be a therapeutic approach for treatment of the metabolic syndrome and dyslipidemia, while avoiding systemic toxicity.
Collapse
Affiliation(s)
- Marcela Doktorova
- Section of Molecular Metabolism and Nutrition, Department of Pediatrics, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
| | - Irene Zwarts
- Section of Molecular Metabolism and Nutrition, Department of Pediatrics, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
| | - Tim van Zutphen
- Section of Molecular Metabolism and Nutrition, Department of Pediatrics, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
| | - Theo H van Dijk
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
| | - Vincent W Bloks
- Section of Molecular Metabolism and Nutrition, Department of Pediatrics, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
| | - Liesbeth Harkema
- Dutch Molecular Pathology Center, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 1, 3584, CL, Utrecht, The Netherlands
| | - Alain de Bruin
- Section of Molecular Metabolism and Nutrition, Department of Pediatrics, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
- Dutch Molecular Pathology Center, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 1, 3584, CL, Utrecht, The Netherlands
| | - Michael Downes
- Howard Hughes Medical Institute and Gene Expression Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, California, 92037, USA
| | - Ronald M Evans
- Howard Hughes Medical Institute and Gene Expression Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, California, 92037, USA
| | - Henkjan J Verkade
- Section of Molecular Metabolism and Nutrition, Department of Pediatrics, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
| | - Johan W Jonker
- Section of Molecular Metabolism and Nutrition, Department of Pediatrics, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands.
| |
Collapse
|
120
|
Li R, Yang J, Saffari A, Jacobs J, Baek KI, Hough G, Larauche MH, Ma J, Jen N, Moussaoui N, Zhou B, Kang H, Reddy S, Henning SM, Campen MJ, Pisegna J, Li Z, Fogelman AM, Sioutas C, Navab M, Hsiai TK. Ambient Ultrafine Particle Ingestion Alters Gut Microbiota in Association with Increased Atherogenic Lipid Metabolites. Sci Rep 2017; 7:42906. [PMID: 28211537 PMCID: PMC5314329 DOI: 10.1038/srep42906] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Accepted: 01/17/2017] [Indexed: 12/22/2022] Open
Abstract
Ambient particulate matter (PM) exposure is associated with atherosclerosis and inflammatory bowel disease. Ultrafine particles (UFP, dp < 0.1–0.2 μm) are redox active components of PM. We hypothesized that orally ingested UFP promoted atherogenic lipid metabolites in both the intestine and plasma via altered gut microbiota composition. Low density lipoprotein receptor-null (Ldlr−/−) mice on a high-fat diet were orally administered with vehicle control or UFP (40 μg/mouse/day) for 3 days a week. After 10 weeks, UFP ingested mice developed macrophage and neutrophil infiltration in the intestinal villi, accompanied by elevated cholesterol but reduced coprostanol levels in the cecum, as well as elevated atherogenic lysophosphatidylcholine (LPC 18:1) and lysophosphatidic acids (LPAs) in the intestine and plasma. At the phylum level, Principle Component Analysis revealed significant segregation of microbiota compositions which was validated by Beta diversity analysis. UFP-exposed mice developed increased abundance in Verrocomicrobia but decreased Actinobacteria, Cyanobacteria, and Firmicutes as well as a reduced diversity in microbiome. Spearman’s analysis negatively correlated Actinobacteria with cecal cholesterol, intestinal and plasma LPC18:1, and Firmicutes and Cyanobacteria with plasma LPC 18:1. Thus, ultrafine particles ingestion alters gut microbiota composition, accompanied by increased atherogenic lipid metabolites. These findings implicate the gut-vascular axis in a atherosclerosis model.
Collapse
Affiliation(s)
- Rongsong Li
- Division of Cardiology, Department of Medicine, School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Jieping Yang
- Division of Clinical Nutrition, Department of Medicine, School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Arian Saffari
- Civil and Environmental Engineering, University of Southern California, Los Angeles, CA 90089, USA
| | - Jonathan Jacobs
- Division of Gastroenterology and Hepatology, Department of Medicine, School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Kyung In Baek
- Department of Bioengineering, School of Engineering &Applied Science, University of California, Los Angeles, CA 90095, USA
| | - Greg Hough
- Division of Cardiology, Department of Medicine, School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Muriel H Larauche
- Division of Gastroenterology and Hepatology, Department of Medicine, School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Jianguo Ma
- Division of Cardiology, Department of Medicine, School of Medicine, University of California, Los Angeles, CA 90095, USA.,Department of Bioengineering, School of Engineering &Applied Science, University of California, Los Angeles, CA 90095, USA
| | - Nelson Jen
- Division of Cardiology, Department of Medicine, School of Medicine, University of California, Los Angeles, CA 90095, USA.,Department of Bioengineering, School of Engineering &Applied Science, University of California, Los Angeles, CA 90095, USA
| | - Nabila Moussaoui
- Division of Gastroenterology and Hepatology, Department of Medicine, School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Bill Zhou
- Division of Cardiology, Department of Medicine, School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Hanul Kang
- Division of Cardiology, Department of Medicine, School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Srinivasa Reddy
- Division of Cardiology, Department of Medicine, School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Susanne M Henning
- Division of Clinical Nutrition, Department of Medicine, School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Matthew J Campen
- Department of Pharmaceutical Sciences, School of Pharmacy, University of New Mexico, Albuquerque, NM 87131, USA
| | - Joseph Pisegna
- Division of Gastroenterology and Hepatology, Department of Medicine, School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Zhaoping Li
- Division of Clinical Nutrition, Department of Medicine, School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Alan M Fogelman
- Division of Cardiology, Department of Medicine, School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Constantinos Sioutas
- Civil and Environmental Engineering, University of Southern California, Los Angeles, CA 90089, USA
| | - Mohamad Navab
- Division of Cardiology, Department of Medicine, School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Tzung K Hsiai
- Division of Cardiology, Department of Medicine, School of Medicine, University of California, Los Angeles, CA 90095, USA.,Department of Bioengineering, School of Engineering &Applied Science, University of California, Los Angeles, CA 90095, USA
| |
Collapse
|
121
|
Takei K, Nakagawa Y, Wang Y, Han SI, Satoh A, Sekiya M, Matsuzaka T, Shimano H. Effects of K-877, a novel selective PPARα modulator, on small intestine contribute to the amelioration of hyperlipidemia in low-density lipoprotein receptor knockout mice. J Pharmacol Sci 2017; 133:214-222. [PMID: 28366492 DOI: 10.1016/j.jphs.2017.02.003] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 01/26/2017] [Accepted: 02/06/2017] [Indexed: 11/18/2022] Open
Abstract
Peroxisome proliferator-activated receptor α (PPARα) is a well-known therapeutic target for treating hyperlipidemia. K-877 is a novel selective PPARα modulator (SPPARMα) that enhances PPARα transcriptional activity with high selectivity and potency, resulting in reduced plasma lipid levels. This study aimed to evaluate the effects of K-877 on hyperlipidemia in low-density lipoprotein receptor knockout (Ldlr-/-) mice, a mouse model of atherosclerosis. We revealed that K-877 administration significantly decreased plasma triglyceride (TG) and total cholesterol (TC) levels and increased plasma high-density lipoprotein cholesterol (HDL-C) levels in Ldlr-/- mice. K-877 administration to Ldlr-/- mice efficiently increased the gene expression of PPARα and its target genes related to fatty acid oxidation in the liver and small intestine. The same treatment significantly increased ATP-binding cassette a1 gene expression in the liver and small intestine and reduced Niemann Pick C1-like 1 gene expression in the small intestine, suggesting that K-877 administration induced HDL-C production in the liver and small intestine and reduced cholesterol absorption in the small intestine. In conclusion, K-877 administration had pronounced effects on the liver and small intestine in Ldlr-/- mice. K-877 is an attractive PPARα-modulating drug for treating hyperlipidemia that works equally well in both the liver and small intestine.
Collapse
Affiliation(s)
- Kenta Takei
- Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
| | - Yoshimi Nakagawa
- Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575, Japan; International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575, Japan.
| | - Yunong Wang
- Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
| | - Song-Iee Han
- Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
| | - Aoi Satoh
- Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
| | - Motohiro Sekiya
- Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
| | - Takashi Matsuzaka
- Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
| | - Hitoshi Shimano
- Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575, Japan; International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575, Japan; Life Science Center, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8577, Japan.
| |
Collapse
|
122
|
Yamanashi Y, Takada T, Kurauchi R, Tanaka Y, Komine T, Suzuki H. Transporters for the Intestinal Absorption of Cholesterol, Vitamin E, and Vitamin K. J Atheroscler Thromb 2017; 24:347-359. [PMID: 28100881 PMCID: PMC5392472 DOI: 10.5551/jat.rv16007] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Humans cannot synthesize fat-soluble vitamins such as vitamin E and vitamin K. For this reason, they must be obtained from the diet via intestinal absorption. As the deficiency or excess of these vitamins has been reported to cause several types of diseases and disorders in humans, the intestinal absorption of these nutrients must be properly regulated to ensure good health. However, the mechanism of their intestinal absorption remains poorly understood. Recent studies on cholesterol using genome-edited mice, genome-wide association approaches, gene mutation analyses, and the development of cholesterol absorption inhibitors have revealed that several membrane proteins play crucial roles in the intestinal absorption of cholesterol. Surprisingly, detailed analyses of these cholesterol transporters have revealed that they can also transport vitamin E and vitamin K, providing clues to uncover the molecular mechanisms underlying the intestinal absorption of these fat-soluble vitamins. In this review, we focus on the membrane proteins (Niemann-Pick C1 like 1, scavenger receptor class B type I, cluster of differentiation 36, and ATP-binding cassette transporter A1) that are (potentially) involved in the intestinal absorption of cholesterol, vitamin E, and vitamin K and discuss their physiological and pharmacological importance. We also discuss the related uncertainties that need to be explored in future studies.
Collapse
Affiliation(s)
- Yoshihide Yamanashi
- Department of Pharmacy, the University of Tokyo Hospital, Faculty of Medicine, the University of Tokyo
| | | | | | | | | | | |
Collapse
|
123
|
Hu WY, Ma XH, Zhou WY, Li XX, Sun TT, Sun H. Preventive effect of Silibinin in combination with Pu-erh tea extract on non-alcoholic fatty liver disease in ob/ob mice. Food Funct 2017; 8:1105-1115. [DOI: 10.1039/c6fo01591c] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
This study investigates the synergistic effect of Silibinin combined with Pu-erh tea extract against NAFLD.
Collapse
Affiliation(s)
- Wen-Yi Hu
- School of Pharmaceutical Science and Technology
- Tianjin University
- Tianjin 300072
- China
- State Key Laboratory of Core Technology in Innovative Chinese medicine
| | - Xiao-Hui Ma
- State Key Laboratory of Core Technology in Innovative Chinese medicine
- Division of Pharmacology and Toxicology
- Tasly Holding Group Co
- Ltd
- Tianjin 300410
| | - Wang-Yi Zhou
- State Key Laboratory of Core Technology in Innovative Chinese medicine
- Division of Pharmacology and Toxicology
- Tasly Holding Group Co
- Ltd
- Tianjin 300410
| | - Xin-Xin Li
- State Key Laboratory of Core Technology in Innovative Chinese medicine
- Division of Pharmacology and Toxicology
- Tasly Holding Group Co
- Ltd
- Tianjin 300410
| | - Ting-Ting Sun
- State Key Laboratory of Core Technology in Innovative Chinese medicine
- Division of Pharmacology and Toxicology
- Tasly Holding Group Co
- Ltd
- Tianjin 300410
| | - He Sun
- School of Pharmaceutical Science and Technology
- Tianjin University
- Tianjin 300072
- China
- State Key Laboratory of Core Technology in Innovative Chinese medicine
| |
Collapse
|
124
|
Drouin-Chartier JP, Tremblay AJ, Lemelin V, Lépine MC, Lamarche B, Couture P. Ezetimibe increases intestinal expression of the LDL receptor gene in dyslipidaemic men with insulin resistance. Diabetes Obes Metab 2016; 18:1226-1235. [PMID: 27460541 DOI: 10.1111/dom.12749] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Revised: 07/11/2016] [Accepted: 07/21/2016] [Indexed: 01/25/2023]
Abstract
AIM To gain further insight into intestinal cholesterol homeostasis in dyslipidaemic men with insulin resistance (IR) by examining the impact of treatment with ezetimibe on the expression of key genes involved in cholesterol synthesis and LDL receptor (R)-mediated uptake of lipoproteins. METHODS A total of 25 men with dyslipidaemia and IR were recruited to participate in this double-blind, randomized, crossover, placebo-controlled trial. Participants received 10 mg/day ezetimibe or placebo for periods of 12 weeks each. Intestinal gene expression was measured by quantitative PCR in duodenal biopsy samples collected by gastroduodenoscopy at the end of each treatment. RESULTS A total of 20 participants completed the protocol. Treatment with ezetimibe significantly increased intestinal LDLR (+16.2%; P = .01), 3-hydroxy-3-methyl-glutaryl-CoA reductase (HMG-CoAR; +14.0%; P = .04) and acetyl-Coenzyme A acetyltransferase 2 (ACAT-2) mRNA expression (+12.5%; P = .03). Changes in sterol regulatory element-binding transcription factor 2 (SREBP-2) expression were significantly correlated with changes in HMG-CoAR (r = 0.55; P < .05), ACAT-2 (r = 0.69; P < .001) and proprotein convertase substilisin/kexin type 9 (PCSK9) expression (r = 0.45; P < .05). CONCLUSIONS These results show that inhibition of intestinal cholesterol absorption by ezetimibe increases expression of the LDLR gene, supporting the concept that increased LDL clearance with ezetimibe treatment occurs not only in the liver but also in the small intestine.
Collapse
Affiliation(s)
| | - André J Tremblay
- Department of Medicine, Institute of Nutrition and Functional Foods, Laval University, Quebec City, Canada
| | - Valéry Lemelin
- Department of Gastroenterology, CHU de Québec-Université Laval, Quebec City, Canada
| | - Marie-Claude Lépine
- Department of Medicine, Institute of Nutrition and Functional Foods, Laval University, Quebec City, Canada
| | - Benoît Lamarche
- Department of Medicine, Institute of Nutrition and Functional Foods, Laval University, Quebec City, Canada
| | - Patrick Couture
- Department of Medicine, Institute of Nutrition and Functional Foods, Laval University, Quebec City, Canada
- Department of Medicine, Lipid Research Center, CHU de Québec-Université Laval, Quebec City, Canada
| |
Collapse
|
125
|
Katsube A, Hayashi H, Kusuhara H. Pim-1L Protects Cell Surface–Resident ABCA1 From Lysosomal Degradation in Hepatocytes and Thereby Regulates Plasma High-Density Lipoprotein Level. Arterioscler Thromb Vasc Biol 2016; 36:2304-2314. [DOI: 10.1161/atvbaha.116.308472] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Accepted: 10/05/2016] [Indexed: 01/13/2023]
Abstract
Objective—
ATP-binding cassette transporter A1 (ABCA1) exerts an atheroprotective action through the biogenesis of high-density lipoprotein in hepatocytes and prevents the formation of foam cells from macrophages. Controlling ABCA1 is a rational approach to improving atherosclerotic cardiovascular disease. Although much is known about the regulatory mechanism of ABCA1 synthesis, the molecular mechanism underpinning its degradation remains to be clearly described.
Approach and Results—
ABCA1 possesses potential sites of phosphorylation by serine/threonine-protein kinase Pim-1 (Pim-1). Pim-1 depletion decreased the expression of cell surface–resident ABCA1 (csABCA1) and apolipoprotein A-I–mediated [
3
H]cholesterol efflux in the human hepatoma cell line HepG2, but not in peritoneal macrophages from mice. In vitro kinase assay, immunoprecipitation, and immunocytochemistry suggested phosphorylation of csABCA1 by the long form of Pim-1 (Pim-1L). Cell surface biotinylation indicated that Pim-1L inhibited lysosomal degradation of csABCA1 involving the liver X receptor β, which interacts with csABCA1 and thereby protects it from ubiquitination and subsequent lysosomal degradation. Cell surface coimmunoprecipitation with COS-1 cells expressing extracellularly hemagglutinin-tagged ABCA1 showed that Pim-1L–mediated phosphorylation of csABCA1 facilitated the interaction between csABCA1 and liver X receptor β and thereby stabilized the csABCA1–Pim-1L complex. Mice deficient in Pim-1 kinase activity showed lower expression of ABCA1 in liver plasma membranes and lower plasma high-density lipoprotein levels than control mice.
Conclusions—
Pim-1L protects hepatic csABCA1 from lysosomal degradation by facilitating the physical interaction between csABCA1 and liver X receptor β and subsequent stabilization of the csABCA1–Pim-1L complex and thereby regulates the circulating level of high-density lipoprotein. Our findings may aid the development of high-density lipoprotein–targeted therapy.
Collapse
Affiliation(s)
- Akira Katsube
- From the Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Japan
| | - Hisamitsu Hayashi
- From the Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Japan
| | - Hiroyuki Kusuhara
- From the Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Japan
| |
Collapse
|
126
|
Lim FT, Lim SM, Ramasamy K. Pediococcus acidilactici LAB4 and Lactobacillus plantarum LAB12 assimilate cholesterol and modulate ABCA1, CD36, NPC1L1 and SCARB1 in vitro. Benef Microbes 2016; 8:97-109. [PMID: 27903090 DOI: 10.3920/bm2016.0048] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
There is growing interest in the use of probiotic lactic acid bacteria (LAB) for prevention of hypercholesterolaemia. This study assessed the cholesterol lowering ability of Pediococcus acidilactici LAB4 and Lactobacillus plantarum LAB12 in growth media. Both LAB yielded >98% (39.2 μg/ml) cholesterol lowering in growth media. Nile Red staining indicated direct assimilation of cholesterol by the LAB. The LAB were then explored for their prophylactic (pre-treatment of HT29 cells with LAB prior to cholesterol exposure) and biotherapeutic (treatment of HT29 cells with LAB after exposure to cholesterol) use against short and prolonged exposure of HT29 cells to cholesterol, respectively. For HT29 cells pre-treated with LAB, cholesterol lowering was accompanied by down-regulation of ATP-binding cassette family transporter-type A1 (ABCA1), cluster of differentiation 36 (CD36) and scavenger receptor class B member 1 (SCARB1). HT29 cells treated with LAB after prolonged exposure to cholesterol source, on the other hand, was associated with up-regulation of ABCA1, restoration of CD36 to basal level and down-regulation of Neimann-Pick C1-Like 1 (NPC1L1). The present findings implied the potential use of LAB4 and LAB12 as part of the strategies in prevention and management of hypercholesterolaemia.
Collapse
Affiliation(s)
- F T Lim
- 1 Faculty of Pharmacy, Universiti Teknologi MARA (UiTM), 42300 Bandar Puncak Alam, Selangor Darul Ehsan, Malaysia.,2 Collaborative Drug Discovery Research (CDDR) Group, Pharmaceutical and Life Sciences Community of Research, Universiti Teknologi MARA (UiTM), 40450 Shah Alam, Selangor Darul Ehsan, Malaysia
| | - S M Lim
- 1 Faculty of Pharmacy, Universiti Teknologi MARA (UiTM), 42300 Bandar Puncak Alam, Selangor Darul Ehsan, Malaysia.,2 Collaborative Drug Discovery Research (CDDR) Group, Pharmaceutical and Life Sciences Community of Research, Universiti Teknologi MARA (UiTM), 40450 Shah Alam, Selangor Darul Ehsan, Malaysia
| | - K Ramasamy
- 1 Faculty of Pharmacy, Universiti Teknologi MARA (UiTM), 42300 Bandar Puncak Alam, Selangor Darul Ehsan, Malaysia.,2 Collaborative Drug Discovery Research (CDDR) Group, Pharmaceutical and Life Sciences Community of Research, Universiti Teknologi MARA (UiTM), 40450 Shah Alam, Selangor Darul Ehsan, Malaysia
| |
Collapse
|
127
|
Oda MN. Lipid-free apoA-I structure - Origins of model diversity. Biochim Biophys Acta Mol Cell Biol Lipids 2016; 1862:221-233. [PMID: 27890580 DOI: 10.1016/j.bbalip.2016.11.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Revised: 10/20/2016] [Accepted: 11/20/2016] [Indexed: 01/22/2023]
Abstract
Apolipoprotein A-I (apoA-I) is a prominent member of the exchangeable apolipoprotein class of proteins, capable of transitioning between lipid-bound and lipid-free states. It is the primary structural and functional protein of high density lipoprotein (HDL). Lipid-free apoA-I is critical to de novo HDL formation as it is the preferred substrate of the lipid transporter, ATP Binding Cassette Transporter A1 (ABCA1) Remaley et al. (2001) [1]. Lipid-free apoA-I is an important element in reverse cholesterol transport and comprehension of its structure is a core issue in our understanding of cholesterol metabolism. However, lipid-free apoA-I is highly conformationally dynamic making it a challenging subject for structural analysis. Over the past 20years there have been significant advances in overcoming the dynamic nature of lipid-free apoA-I, which have resulted in a multitude of proposed conformational models.
Collapse
Affiliation(s)
- Michael N Oda
- Children's Hospital Oakland Research Institute, Oakland, CA 94609, United States.
| |
Collapse
|
128
|
Kikuchi T, Orihara K, Oikawa F, Han SI, Kuba M, Okuda K, Satoh A, Osaki Y, Takeuchi Y, Aita Y, Matsuzaka T, Iwasaki H, Yatoh S, Sekiya M, Yahagi N, Suzuki H, Sone H, Nakagawa Y, Yamada N, Shimano H. Intestinal CREBH overexpression prevents high-cholesterol diet-induced hypercholesterolemia by reducing Npc1l1 expression. Mol Metab 2016; 5:1092-1102. [PMID: 27818935 PMCID: PMC5081412 DOI: 10.1016/j.molmet.2016.09.004] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Revised: 09/06/2016] [Accepted: 09/10/2016] [Indexed: 12/12/2022] Open
Abstract
Objective The transcription factor cyclic AMP-responsive element-binding protein H (CREBH, encoded by Creb3l3) is highly expressed in the liver and small intestine. Hepatic CREBH contributes to glucose and triglyceride metabolism by regulating fibroblast growth factor 21 (Fgf21) expression. However, the intestinal CREBH function remains unknown. Methods To investigate the influence of intestinal CREBH on cholesterol metabolism, we compared plasma, bile, fecal, and tissue cholesterol levels between wild-type (WT) mice and mice overexpressing active human CREBH mainly in the small intestine (CREBH Tg mice) under different dietary conditions. Results Plasma cholesterol, hepatic lipid, and cholesterol crystal formation in the gallbladder were lower in CREBH Tg mice fed a lithogenic diet (LD) than in LD-fed WTs, while fecal cholesterol output was higher in the former. These results suggest that intestinal CREBH overexpression suppresses cholesterol absorption, leading to reduced plasma cholesterol, limited hepatic supply, and greater excretion. The expression of Niemann–Pick C1-like 1 (Npc1l1), a rate-limiting transporter mediating intestinal cholesterol absorption, was reduced in the small intestine of CREBH Tg mice. Adenosine triphosphate-binding cassette transporter A1 (Abca1), Abcg5/8, and scavenger receptor class B, member 1 (Srb1) expression levels were also reduced in CREBH Tg mice. Promoter assays revealed that CREBH directly regulates Npc1l1 expression. Conversely, CREBH null mice exhibited higher intestinal Npc1l1 expression, elevated plasma and hepatic cholesterol, and lower fecal output. Conclusion Intestinal CREBH regulates dietary cholesterol flow from the small intestine by controlling the expression of multiple intestinal transporters. We propose that intestinal CREBH could be a therapeutic target for hypercholesterolemia. Plasma cholesterol, hepatic lipid, and gallstones were lower in CREBH Tg mice. Expression of intestinal Npc1l1 was reduced in CREBH Tg mice. CREBH directly down-regulates mouse Npc1l1 promoter activity. Intestinal CREBH regulates dietary cholesterol flow from the small intestine.
Collapse
Key Words
- ABCG5/8, adenosine triphosphate-binding cassette transporter G5/G8
- ALT, alanine aminotransferase
- AST, aspartate aminotransferase
- Abca1, ATP-binding cassette, sub-family A1
- Apoa4, apolipoprotein A-IV
- CREBH
- CREBH, cyclic AMP-responsive element-binding protein H
- Cholesterol
- Cpt1a, carnitine palmitoyltransferase 1a, liver
- Cyp7a1, cytochrome P450, family 7, subfamily a, polypeptide 1
- ER, endoplasmic reticulum
- FGF21, fibroblast growth factor 21
- FXR, Farnesoid X receptor
- Intestine
- LD, lithogenic diet
- LPL, lipoprotein lipase
- LXR, liver X receptor
- NEFA, non-esterified fatty acids
- NPC1L1, Nieman Pick C1-like 1
- Npc1l1
- PPARα, proliferator activated receptor alpha
- RCT, reverse cholesterol transport
- SREBP, sterol regulatory element-binding protein
- Shp, small heterodimer partner
- Srb1, scavenger receptor class B, member 1
- Srebf, sterol regulatory element-binding factor
- TG, triglyceride
- WT, wild type
Collapse
Affiliation(s)
- Takuya Kikuchi
- Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Kana Orihara
- Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Fusaka Oikawa
- Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Song-Iee Han
- Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Motoko Kuba
- Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Kanako Okuda
- Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Aoi Satoh
- Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Yoshinori Osaki
- Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Yoshinori Takeuchi
- Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Yuichi Aita
- Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Takashi Matsuzaka
- Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Hitoshi Iwasaki
- Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Shigeru Yatoh
- Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Motohiro Sekiya
- Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Naoya Yahagi
- Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Hiroaki Suzuki
- Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Hirohito Sone
- Department of Hematology, Endocrinology and Metabolism, Niigata University Faculty of Medicine, Niigata, Niigata 951-8510, Japan
| | - Yoshimi Nakagawa
- Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan; International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan.
| | - Nobuhiro Yamada
- Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Hitoshi Shimano
- Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan; International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan; Life Science Center, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba 305-8577, Japan.
| |
Collapse
|
129
|
Soayfane Z, Tercé F, Cantiello M, Robenek H, Nauze M, Bézirard V, Allart S, Payré B, Capilla F, Cartier C, Peres C, Al Saati T, Théodorou V, Nelson DW, Yen CLE, Collet X, Coméra C. Exposure to dietary lipid leads to rapid production of cytosolic lipid droplets near the brush border membrane. Nutr Metab (Lond) 2016; 13:48. [PMID: 27478484 PMCID: PMC4965885 DOI: 10.1186/s12986-016-0107-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 07/21/2016] [Indexed: 12/17/2022] Open
Abstract
Background Intestinal absorption of dietary lipids involves their hydrolysis in the lumen of proximal intestine as well as uptake, intracellular transport and re-assembly of hydrolyzed lipids in enterocytes, leading to the formation and secretion of the lipoproteins chylomicrons and HDL. In this study, we examined the potential involvement of cytosolic lipid droplets (CLD) whose function in the process of lipid absorption is poorly understood. Methods Intestinal lipid absorption was studied in mouse after gavage. Three populations of CLD were purified by density ultracentrifugations, as well as the brush border membranes, which were analyzed by western-blots. Immunofluorescent localization of membranes transporters or metabolic enzymes, as well as kinetics of CLD production, were also studied in intestine or Caco-2 cells. Results We isolated three populations of CLD (ranging from 15 to 1000 nm) which showed differential expression of the major lipid transporters scavenger receptor BI (SR-BI), cluster of differentiation 36 (CD-36), Niemann Pick C-like 1 (NPC1L1), and the ATP-binding cassette transporters ABCG5/G8 but also caveolin 2 and fatty acid binding proteins. The enzyme monoacylglycerol acyltransferase 2 (MGAT2) was identified in the brush border membrane (BBM) in addition to the endoplasmic reticulum, suggesting local synthesis of triglycerides and CLD at both places. Conclusions We show a very fast production of CLD by enterocytes associated with a transfer of apical constituents as lipid transporters. Our findings suggest that following their uptake by enterocytes, lipids can be partially metabolized at the BBM and packaged into CLD for their transportation to the ER. Electronic supplementary material The online version of this article (doi:10.1186/s12986-016-0107-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Zeina Soayfane
- Institut des Maladies Métaboliques et Cardiovasculaires - I2MC, UMR 1048, Institut National de la Santé et de la Recherche Médicale, Université Toulouse III Paul Sabatier, Toulouse, F-31000 France
| | - François Tercé
- Institut des Maladies Métaboliques et Cardiovasculaires - I2MC, UMR 1048, Institut National de la Santé et de la Recherche Médicale, Université Toulouse III Paul Sabatier, Toulouse, F-31000 France
| | - Michela Cantiello
- Institut des Maladies Métaboliques et Cardiovasculaires - I2MC, UMR 1048, Institut National de la Santé et de la Recherche Médicale, Université Toulouse III Paul Sabatier, Toulouse, F-31000 France
| | - Horst Robenek
- Leibniz-Institut für Arterioskleroseforschung, Universität Münster, Münster, Germany
| | - Michel Nauze
- Institut des Maladies Métaboliques et Cardiovasculaires - I2MC, UMR 1048, Institut National de la Santé et de la Recherche Médicale, Université Toulouse III Paul Sabatier, Toulouse, F-31000 France
| | - Valérie Bézirard
- UMR 1331 Toxalim, INRA, Université de Toulouse, ENVT, INP-Purpan, 180 chemin de Tournefeuille, BP 93173, 31027 Toulouse, cedex 3, France
| | - Sophie Allart
- INSERM UMR 1043 (INSERM/UPS/CNRS/USC Inra), CHU Purpan, Toulouse, France
| | - Bruno Payré
- CMEAB, Faculté de Médecine Rangueil, Toulouse, France
| | - Florence Capilla
- INSERM/UPS - US006/CREFRE, Service d'Histopathologie, CHU Purpan, Toulouse, France
| | - Christel Cartier
- UMR 1331 Toxalim, INRA, Université de Toulouse, ENVT, INP-Purpan, 180 chemin de Tournefeuille, BP 93173, 31027 Toulouse, cedex 3, France
| | - Christine Peres
- Institut des Maladies Métaboliques et Cardiovasculaires - I2MC, UMR 1048, Institut National de la Santé et de la Recherche Médicale, Université Toulouse III Paul Sabatier, Toulouse, F-31000 France
| | - Talal Al Saati
- INSERM/UPS - US006/CREFRE, Service d'Histopathologie, CHU Purpan, Toulouse, France
| | - Vassilia Théodorou
- UMR 1331 Toxalim, INRA, Université de Toulouse, ENVT, INP-Purpan, 180 chemin de Tournefeuille, BP 93173, 31027 Toulouse, cedex 3, France
| | - David W Nelson
- Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, WI USA
| | - Chi-Liang Eric Yen
- Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, WI USA
| | - Xavier Collet
- Institut des Maladies Métaboliques et Cardiovasculaires - I2MC, UMR 1048, Institut National de la Santé et de la Recherche Médicale, Université Toulouse III Paul Sabatier, Toulouse, F-31000 France
| | - Christine Coméra
- Institut des Maladies Métaboliques et Cardiovasculaires - I2MC, UMR 1048, Institut National de la Santé et de la Recherche Médicale, Université Toulouse III Paul Sabatier, Toulouse, F-31000 France.,UMR 1331 Toxalim, INRA, Université de Toulouse, ENVT, INP-Purpan, 180 chemin de Tournefeuille, BP 93173, 31027 Toulouse, cedex 3, France
| |
Collapse
|
130
|
Zhang T, Zhou L, Li CC, Shi H, Zhou X. TSH increases synthesis of hepatic ATP-binding cassette subfamily A member 1 in hypercholesterolemia. Biochem Biophys Res Commun 2016; 476:75-81. [PMID: 27179782 DOI: 10.1016/j.bbrc.2016.05.054] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Accepted: 05/10/2016] [Indexed: 10/21/2022]
Abstract
Epidemiological evidence suggests that thyrotropin (TSH) levels are closely correlated with the severity of hypercholesterolemia. Reverse cholesterol transfer (RCT) plays an important role in regulating bloodcholesterol. However, the molecular mechanism of hypercholesterolemia in subclinical hypothyroidism (SCH) has not been fully clarified. The SCH mouse model, which is characterized by elevated serum TSH but not thyroid hormone levels, demonstrated a significant increase in plasma cholesterol compared with controls. Interestingly, Tshr KO mice, with normal thyroid hormone levels after thyroid hormone supplementation, showed lower plasma cholesterol levels compared with their wild-type littermates. ATP binding cassette subfamily A member 1(ABCA1) is a member of the ABC superfamily, which induces transfer of intracellular cholesterol to extracellular apolipoprotein. TSH upregulated hepatic ABCA1 to promote the efflux of intercellular cumulative cholesterol, resulting in increased plasma cholesterol. These data might partially explain the pathogenesis of hypercholesterolemia in SCH.
Collapse
Affiliation(s)
- Tiantian Zhang
- Department of Endocrinology, Shandong Provincial Hospital affiliated to Shandong University, Shandong Clinical Medical Center of Endocrinology and Metabolism, Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, China
| | - Lingyan Zhou
- Department of Endocrinology, Shandong Provincial Hospital affiliated to Shandong University, Shandong Clinical Medical Center of Endocrinology and Metabolism, Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, China
| | - Cong Cong Li
- Jinan Central Hospital affiliated to Shandong University, Jinan, Shandong, 250021, China
| | - Hong Shi
- Department of Pediatrics, The Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, China
| | - Xinli Zhou
- Department of Endocrinology, Shandong Provincial Hospital affiliated to Shandong University, Shandong Clinical Medical Center of Endocrinology and Metabolism, Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, China.
| |
Collapse
|
131
|
TTC39B deficiency stabilizes LXR reducing both atherosclerosis and steatohepatitis. Nature 2016; 535:303-7. [PMID: 27383786 PMCID: PMC4947007 DOI: 10.1038/nature18628] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Accepted: 06/02/2016] [Indexed: 01/07/2023]
Abstract
Cellular mechanisms that mediate steato-hepatitis, an increasingly prevalent condition in the Western world for which no therapies are available1, are poorly understood. Despite the fact its synthetic agonists induce fatty liver, the Liver X receptor (LXR) transcription factor remains a target of interest because of its anti-atherogenic, cholesterol removal and anti-inflammatory activities. We discovered that tetratricopeptide repeat (TPR) domain protein 39B (Ttc39b, C9orf52) (T39), a high density lipoprotein (HDL) gene discovered in human genome wide association studies (GWAS)2, promotes the ubiquitination and degradation of LXR. Chow-fed T39-/- mice displayed increased HDL cholesterol levels associated with increased enterocyte ATP binding cassette transporter A1 (Abca1) expression and increased LXR protein without change in LXR mRNA. When challenged with a high fat/high cholesterol/bile salt (HF/HC/BS) diet, T39-/- mice or mice with hepatocyte-specific T39 deficiency showed increased hepatic LXR protein and target gene expression, and unexpectedly protection from steato-hepatitis and death. Western Type Diet (WTD)-fed Low density lipoprotein receptor (Ldlr)-/-T39-/- mice showed decreased fatty liver, increased HDL, decreased LDL and reduced atherosclerosis. In addition to increasing hepatic Abcg5/8 expression and limiting dietary cholesterol absorption, T39 deficiency inhibited hepatic sterol regulatory element binding protein 1 (SREBP-1, ADD1) processing. This was explained by an increase in microsomal phospholipids containing polyunsaturated fatty acids (PUFA), linked to an LXRα-dependent increase in expression of enzymes mediating PC biosynthesis and incorporation of PUFA into phospholipids. The preservation of endogenous LXR protein activates a beneficial profile of gene expression that promotes cholesterol removal and inhibits lipogenesis. T39 inhibition could be an effective strategy for reducing both steato-hepatitis and atherosclerosis.
Collapse
|
132
|
Molecular characterization and developmental expression patterns of apolipoprotein A-I in Senegalese sole (Solea senegalensis Kaup). Gene Expr Patterns 2016; 21:7-18. [PMID: 27261260 DOI: 10.1016/j.gep.2016.05.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Revised: 05/24/2016] [Accepted: 05/26/2016] [Indexed: 12/24/2022]
Abstract
The apolipoprotein A-I (ApoA-I) is an essential component of the high density lipoproteins (HDL). In this study, the cDNA and genomic sequences of this apolipoprotein were characterized for first time in Solea senegalensis. The predicted polypeptide revealed conserved structural features including ten repeats in the lipid-binding domain and some residues involved in cholesterol interaction and binding. The gene structure analysis identified four exons and three introns. Moreover, the synteny analysis revealed that apoA-I did not localize with other apolipoproteins indicating a divergent evolution with respect to the apoA-IV and apoE cluster. The phylogenetic analyses identified two distinct apoA-I paralogs in Ostariophysi (referred to as Ia and Ib) and only one (Ib) in Acanthopterygii. Whole-mount in situ hybridization located the apoA-I signal mainly in the yolk syncytial layer in lecitotrophic larval stages. Later at mouth opening, the mRNA signals were detected mainly in liver and intestine compatible with its role in the HDL formation. Moreover, a clear signal was detected in some regions of the brain, retina and neural cord suggesting a role in local regulation of cholesterol homeostasis. After metamorphosis, apoA-I was also detected in other tissues such as gills, head kidney and spleen suggesting a putative role in immunity. Expression analyses in larvae fed two diets with different triacylglycerol levels indicated that apoA-I mRNA levels were more associated to larval size and development than dietary lipid levels. Finally, qPCR analyses of immature and mature transcripts revealed distinct expression profiles suggesting a posttranscriptional regulatory mechanism.
Collapse
|
133
|
Meriwether D, Sulaiman D, Wagner A, Grijalva V, Kaji I, Williams KJ, Yu L, Fogelman S, Volpe C, Bensinger SJ, Anantharamaiah GM, Shechter I, Fogelman AM, Reddy ST. Transintestinal transport of the anti-inflammatory drug 4F and the modulation of transintestinal cholesterol efflux. J Lipid Res 2016; 57:1175-93. [PMID: 27199144 DOI: 10.1194/jlr.m067025] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Indexed: 01/28/2023] Open
Abstract
The site and mechanism of action of the apoA-I mimetic peptide 4F are incompletely understood. Transintestinal cholesterol efflux (TICE) is a process involved in the clearance of excess cholesterol from the body. While TICE is responsible for at least 30% of the clearance of neutral sterols from the circulation into the intestinal lumen, few pharmacological agents have been identified that modulate this pathway. We show first that circulating 4F selectively targets the small intestine (SI) and that it is predominantly transported into the intestinal lumen. This transport of 4F into the SI lumen is transintestinal in nature, and it is modulated by TICE. We also show that circulating 4F increases reverse cholesterol transport from macrophages and cholesterol efflux from lipoproteins via the TICE pathway. We identify the cause of this modulation of TICE either as 4F being a cholesterol acceptor with respect to enterocytes, from which 4F enhances cholesterol efflux, or as 4F being an intestinal chaperone with respect to TICE. Our results assign a novel role for 4F as a modulator of the TICE pathway and suggest that the anti-inflammatory functions of 4F may be a partial consequence of the codependent intestinal transport of both 4F and cholesterol.
Collapse
Affiliation(s)
- David Meriwether
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA Department of Medical and Molecular Pharmacology, University of California Los Angeles, Los Angeles, CA
| | - Dawoud Sulaiman
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA Molecular Toxicology Interdepartmental Degree Program, University of California Los Angeles, Los Angeles, CA
| | - Alan Wagner
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA
| | - Victor Grijalva
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA
| | - Izumi Kaji
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA
| | - Kevin J Williams
- Department of Medical and Molecular Pharmacology, University of California Los Angeles, Los Angeles, CA
| | - Liqing Yu
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD
| | - Spencer Fogelman
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA
| | - Carmen Volpe
- Division of Laboratory Animal Medicine, University of California Los Angeles, Los Angeles, CA
| | - Steven J Bensinger
- Department of Medical and Molecular Pharmacology, University of California Los Angeles, Los Angeles, CA Department of Microbiology, Immunology and Molecular Genetics, University of California Los Angeles, Los Angeles, CA
| | - G M Anantharamaiah
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | - Ishaiahu Shechter
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA
| | - Alan M Fogelman
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA
| | - Srinivasa T Reddy
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA Department of Medical and Molecular Pharmacology, University of California Los Angeles, Los Angeles, CA Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA
| |
Collapse
|
134
|
Subbaiah PV, Dammanahalli KJ, Yang P, Bi J, O'Donnell JM. Enhanced incorporation of dietary DHA into lymph phospholipids by altering its molecular carrier. Biochim Biophys Acta Mol Cell Biol Lipids 2016; 1861:723-9. [PMID: 27178174 DOI: 10.1016/j.bbalip.2016.05.002] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Revised: 04/27/2016] [Accepted: 05/07/2016] [Indexed: 12/11/2022]
Abstract
Several previous studies indicated that for optimal uptake by the brain, docosahexaenoic acid (DHA) should be present as phospholipid in the plasma. However most of dietary DHA is absorbed as triacylglycerol (TAG) because it is released as free fatty acid during digestion of either TAG-DHA (fish oil) or sn-2-DHA phospholipid (krill oil), and subsequently incorporated into TAG of chylomicrons. We tested the hypothesis that the absorption of DHA as phospholipid can be increased if it is present in the sn-1 position of dietary phospholipid or in lysophosphatidylcholine (LPC), because it would escape the hydrolysis by pancreatic phospholipase A2. We infused micelle containing the DHA either as LPC or as free acid, into the duodenum of lymph cannulated rats, and analyzed the chylomicrons and HDL of the lymph for the DHA-containing lipids. The results show that while the total amount of DHA absorbed was comparable from the two types of micelle, the percentage of DHA recovered in lymph phospholipids was 5 times greater with LPC-DHA, compared to free DHA. Furthermore, the amount of DHA recovered in lymph HDL was increased by 2-fold when LPC-DHA micelle was infused. These results could potentially lead to a novel strategy to increase brain DHA levels through the diet.
Collapse
Affiliation(s)
- Papasani V Subbaiah
- Department of Medicine, University of Illinois at Chicago, Chicago, IL 60612, United States; Department of Biochemistry & Molecular Genetics, University of Illinois at Chicago, Chicago, IL 60612, United States; Jesse Brown VA Medical Center, Chicago, IL 60612, United States.
| | | | - Peng Yang
- Department of Medicine, University of Illinois at Chicago, Chicago, IL 60612, United States
| | - Jian Bi
- Department of Physiology & Biophysics, University of Illinois at Chicago, Chicago, IL 60612, United States
| | - J Michael O'Donnell
- Department of Physiology & Biophysics, University of Illinois at Chicago, Chicago, IL 60612, United States
| |
Collapse
|
135
|
Zhao D. Challenges associated with elucidating the mechanisms of the hypocholesterolaemic activity of saponins. J Funct Foods 2016. [DOI: 10.1016/j.jff.2016.02.023] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
|
136
|
Heidker RM, Caiozzi GC, Ricketts ML. Grape Seed Procyanidins and Cholestyramine Differentially Alter Bile Acid and Cholesterol Homeostatic Gene Expression in Mouse Intestine and Liver. PLoS One 2016; 11:e0154305. [PMID: 27111442 PMCID: PMC4844140 DOI: 10.1371/journal.pone.0154305] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2016] [Accepted: 04/12/2016] [Indexed: 01/05/2023] Open
Abstract
Bile acid (BA) sequestrants, lipid-lowering agents, may be prescribed as a monotherapy or combination therapy to reduce the risk of coronary artery disease. Over 33% of adults in the United States use complementary and alternative medicine strategies, and we recently reported that grape seed procyanidin extract (GSPE) reduces enterohepatic BA recirculation as a means to reduce serum triglyceride (TG) levels. The current study was therefore designed to assess the effects on BA, cholesterol and TG homeostatic gene expression following co-administration with GSPE and the BA sequestrant, cholestyramine (CHY). Eight-week old male C57BL/6 mice were treated for 4 weeks with either a control or 2% CHY-supplemented diet, after which, they were administered vehicle or GSPE for 14 hours. Liver and intestines were harvested and gene expression was analyzed. BA, cholesterol, non-esterified fatty acid and TG levels were also analyzed in serum and feces. Results reveal that GSPE treatment alone, and co-administration with CHY, regulates BA, cholesterol and TG metabolism differently than CHY administration alone. Notably, GSPE decreased intestinal apical sodium-dependent bile acid transporter (Asbt) gene expression, while CHY significantly induced expression. Administration with GSPE or CHY robustly induced hepatic BA biosynthetic gene expression, especially cholesterol 7α-hydroxylase (Cyp7a1), compared to control, while co-administration further enhanced expression. Treatment with CHY induced both intestinal and hepatic cholesterologenic gene expression, while co-administration with GSPE attenuated the CHY-induced increase in the liver but not intestine. CHY also induced hepatic lipogenic gene expression, which was attenuated by co-administration with GSPE. Consequently, a 25% decrease in serum TG levels was observed in the CHY+GSPE group, compared to the CHY group. Collectively, this study presents novel evidence demonstrating that GSPE provides additive and complementary efficacy as a lipid-lowering combination therapy in conjunction with CHY by attenuating hepatic cholesterol synthesis, enhancing BA biosynthesis and decreasing lipogenesis, which warrants further investigation.
Collapse
Affiliation(s)
- Rebecca M. Heidker
- Department of Agriculture, Nutrition and Veterinary Sciences, University of Nevada Reno, Reno, Nevada, United States of America
| | - Gianella C. Caiozzi
- Department of Agriculture, Nutrition and Veterinary Sciences, University of Nevada Reno, Reno, Nevada, United States of America
| | - Marie-Louise Ricketts
- Department of Agriculture, Nutrition and Veterinary Sciences, University of Nevada Reno, Reno, Nevada, United States of America
- * E-mail:
| |
Collapse
|
137
|
Lee-Rueckert M, Escola-Gil JC, Kovanen PT. HDL functionality in reverse cholesterol transport--Challenges in translating data emerging from mouse models to human disease. Biochim Biophys Acta Mol Cell Biol Lipids 2016; 1861:566-83. [PMID: 26968096 DOI: 10.1016/j.bbalip.2016.03.004] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2015] [Revised: 02/26/2016] [Accepted: 03/04/2016] [Indexed: 12/18/2022]
Abstract
Whereas LDL-derived cholesterol accumulates in atherosclerotic lesions, HDL particles are thought to facilitate removal of cholesterol from the lesions back to the liver thereby promoting its fecal excretion from the body. Because generation of cholesterol-loaded macrophages is inherent to atherogenesis, studies on the mechanisms stimulating the release of cholesterol from these cells and its ultimate excretion into feces are crucial to learn how to prevent lesion development or even induce lesion regression. Modulation of this key anti-atherogenic pathway, known as the macrophage-specific reverse cholesterol transport, has been extensively studied in several mouse models with the ultimate aim of applying the emerging knowledge to humans. The present review provides a detailed comparison and critical analysis of the various steps of reverse cholesterol transport in mouse and man. We attempt to translate this in vivo complex scenario into practical concepts, which could serve as valuable tools when developing novel HDL-targeted therapies.
Collapse
|
138
|
Uwiera RR, Mangat R, Kelly S, Uwiera TC, Proctor SD. Long-Term Catheterization of the Intestinal Lymph Trunk and Collection of Lymph in Neonatal Pigs. J Vis Exp 2016. [PMID: 27023826 DOI: 10.3791/53457] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Catheterization of the intestinal lymph trunk in neonatal pigs is a technique allowing for the long-term collection of large quantities of intestinal (central) efferent lymph. Importantly, the collection of central lymph from the intestine enables researchers to study both the mechanisms and lipid constitutes associated with lipid metabolism, intestinal inflammation and cancer metastasis, as well as cells involved in immune function and immunosurveillance. A ventral mid-line surgical approach permits excellent surgical exposure to the cranial abdomen and relatively easy access to the intestinal lymph trunk vessel that lies near the pancreas and the right ventral segment of the portal vein underneath the visceral aspect of the right liver lobe. The vessel is meticulously dissected and released from the surrounding fascia and then dilated with sutures allowing for insertion and subsequent securing of the catheter into the vessel. The catheter is exteriorized and approximately 1 L/24 hr of lymph is collected over a 7 day period. While this technique enables the collection of large quantities of central lymph over an extended period of time, the success depends on careful surgical dissection, tissue handling and close attention to proper surgical technique. This is particularly important with surgeries in young animals as the lymph vessels can easily tear, potentially leading to surgical and experimental failure. The video demonstrates an excellent surgical technique for the collection of intestinal lymph.
Collapse
Affiliation(s)
- Richard R Uwiera
- Department of Agricultural, Food and Nutritional Science, University of Alberta;
| | - Rabban Mangat
- Department of Agricultural, Food and Nutritional Science, University of Alberta
| | - Sandra Kelly
- Department of Agricultural, Food and Nutritional Science, University of Alberta
| | | | - Spencer D Proctor
- Department of Agricultural, Food and Nutritional Science, University of Alberta
| |
Collapse
|
139
|
Papageorgiou N, Zacharia E, Androulakis E, Briasoulis A, Charakida M, Tousoulis D. HDL as a prognostic biomarker for coronary atherosclerosis: the role of inflammation. Expert Opin Ther Targets 2016; 20:907-21. [PMID: 26854521 DOI: 10.1517/14728222.2016.1152264] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
INTRODUCTION Emerging evidence suggests that the role of high density lipoprotein (HDL) in the atherosclerotic process is not as clear as previously thought, since atheroprotective HDL becomes atherogenic in states of increased inflammatory processes. AREAS COVERED In this review we aim to elucidate the role of HDL as a prognostic biomarker and we discuss therapeutic approaches that aim to increase HDL and their possible clinical benefit. EXPERT OPINION Given the structural variability and biological complexity of the HDL particle, its role in the atherosclerotic process is far from clear. According to current evidence, the atheroprotective role of HDL turns atherogenic in states of increased inflammatory processes, while even minor alterations in systemic inflammation are likely to hinder the endothelial protective effects of HDL. In accordance, significant data have revealed that HDL-related drugs may be effective in reducing cardiovascular mortality; however they are not as encouraging or unanimous as expected. Possible future goals could be to quantify either HDL subclasses or functions in an attempt to reach safer conclusions as to the prognostic importance of HDL in coronary atherosclerosis. Having achieved that, a more targeted therapy that would aim to raise either HDL functionality or to remodel HDL structure would be more easily designed.
Collapse
Affiliation(s)
| | - Effimia Zacharia
- b 1st Department of Cardiology , Hippokration Hospital, University of Athens , Athens , Greece
| | | | - Alexandros Briasoulis
- d Division of Cardiology , Wayne State University/Detroit Medical Center , Detroit , MI , USA
| | - Marietta Charakida
- e Vascular Physiology Unit, Institute of Cardiovascular Science , University College London , London , UK
| | - Dimitris Tousoulis
- b 1st Department of Cardiology , Hippokration Hospital, University of Athens , Athens , Greece
| |
Collapse
|
140
|
Lhermusier T, Severin S, Van Rothem J, Garcia C, Bertrand-Michel J, Le Faouder P, Hechler B, Broccardo C, Couvert P, Chimini G, Sié P, Payrastre B. ATP-binding cassette transporter 1 (ABCA1) deficiency decreases platelet reactivity and reduces thromboxane A2 production independently of hematopoietic ABCA1. J Thromb Haemost 2016; 14:585-95. [PMID: 26749169 DOI: 10.1111/jth.13247] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Indexed: 02/03/2023]
Abstract
UNLABELLED ESSENTIALS: The role of ATP-binding cassette transporter 1 (ABCA1) in platelet functions is poorly characterized. We studied the impact of ABCA1 deficiency on platelet responses in a mouse model and two Tangier patients. ABCA1-deficient platelets exhibit reduced positive feedback loop mechanisms. This reduced reactivity is dependent on external environment and independent of hematopoietic ABCA1. SUMMARY BACKGROUND The ATP-binding cassette transporter ABCA1 is required for the conversion of apolipoprotein A-1 to high-density lipoprotein (HDL), and its defect causes Tangier disease, a rare disorder characterized by an absence of HDL and accumulation of cholesterol in peripheral tissues. The role of ABCA1 in platelet functions remains poorly characterized. OBJECTIVE To determine the role of ABCA1 in platelet functions and to clarify controversies concerning its implication in processes as fundamental as platelet phosphatidylserine exposure and control of platelet membrane lipid composition. METHODS AND RESULTS We studied the impact of ABCA1 deficiency on platelet responses in a mouse model and in two Tangier patients. We show that platelets in ABCA1-deficient mice are slightly larger in size and exhibit aggregation and secretion defects in response to low concentrations of thrombin and collagen. These platelets have normal cholesterol and major phospholipid composition, granule morphology, or calcium-induced phosphatidylserine exposure. Interestingly, ABCA1-deficient platelets display a reduction in positive feedback loop mechanisms, particularly in thromboxane A2 (TXA2) production. Hematopoietic chimera mice demonstrated that defective eicosanoids production, particularly TXA2, was primarily dependent on external environment and not on the hematopoietic ABCA1. Decreased aggregation and production of TXA2 and eicosanoids were also observed in platelets from Tangier patients. CONCLUSIONS Absence of ABCA1 and low HDL level induce reduction of platelet reactivity by decreasing positive feedback loops, particularly TXA2 production through a hematopoietic ABCA1-independent mechanism.
Collapse
Affiliation(s)
- T Lhermusier
- Inserm, U1048 et Université Toulouse 3, I2MC, Toulouse, France
- Département de Cardiologie, CHU de Toulouse, Toulouse, France
| | - S Severin
- Inserm, U1048 et Université Toulouse 3, I2MC, Toulouse, France
| | - J Van Rothem
- Inserm, U1048 et Université Toulouse 3, I2MC, Toulouse, France
- Département de Cardiologie, CHU de Toulouse, Toulouse, France
| | - C Garcia
- Laboratoire d'Hématologie, CHU de Toulouse, Toulouse, France
| | | | - P Le Faouder
- I2MC, Lipidomic Core Facility-MetaToul, Toulouse, France
| | - B Hechler
- Inserm UMR-S949, Etablissement Français du Sang-Alsace, Strasbourg, France
| | | | - P Couvert
- Service de Biochimie Endocrinienne et Oncologique, Hôpital Pitié Salpêtrière, Paris, France
| | - G Chimini
- CIML, Parc Scientifique de Luminy, Case 906, Marseille Luminy, France
| | - P Sié
- Inserm, U1048 et Université Toulouse 3, I2MC, Toulouse, France
- Laboratoire d'Hématologie, CHU de Toulouse, Toulouse, France
| | - B Payrastre
- Inserm, U1048 et Université Toulouse 3, I2MC, Toulouse, France
- Laboratoire d'Hématologie, CHU de Toulouse, Toulouse, France
| |
Collapse
|
141
|
Baldán Á, de Aguiar Vallim TQ. miRNAs and High-Density Lipoprotein metabolism. Biochim Biophys Acta Mol Cell Biol Lipids 2016; 1861:2053-2061. [PMID: 26869447 DOI: 10.1016/j.bbalip.2016.01.021] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Revised: 01/28/2016] [Accepted: 01/29/2016] [Indexed: 12/16/2022]
Abstract
Altered lipoprotein metabolism plays a key role during atherogenesis. For over 50years, epidemiological data have fueled the proposal that HDL-cholesterol (HDL-c) in circulation is inversely correlated to cardiovascular risk. However, the atheroprotective role of HDL is currently the focus of much debate and remains an active field of research. The emerging picture from research in the past decade suggests that HDL function, rather than HDL-c content, is important in disease. Recent developments demonstrate that miRNAs play an important role in fine-tuning the expression of key genes involved in HDL biogenesis, lipidation, and clearance, as well as in determining the amounts of HDL-c in circulation. Thus, it has been proposed that miRNAs that affect HDL metabolism might be exploited therapeutically in patients. Whether HDL-based therapies, alone or in combination with LDL-based treatments (e.g. statins), provide superior outcomes in patients has been recently questioned by human genetics studies and clinical trials. The switch in focus from "HDL-cholesterol" to "HDL function" opens a new paradigm to understand the physiology and therapeutic potential of HDL, and to find novel modulators of cardiovascular risk. In this review we summarize the current knowledge on the regulation of HDL metabolism and function by miRNAs. This article is part of a Special Issue entitled: MicroRNAs and lipid/energy metabolism and related diseases edited by Carlos Fernández-Hernando and Yajaira Suárez.
Collapse
Affiliation(s)
- Ángel Baldán
- Edward A. Doisy Department of Biochemistry & Molecular Biology, Center for Cardiovascular Research, and Liver Center, 1100 S. Grand Blvd., Saint Louis University, Saint Louis, MO 63104, United States.
| | - Thomas Q de Aguiar Vallim
- Department of Medicine, Division of Cardiology, 650 Charles E. Young Drive S, A2-237 CHS, UCLA Los Angeles, Los Angeles, CA 90095, United States.
| |
Collapse
|
142
|
Singh SA, Andraski AB, Pieper B, Goh W, Mendivil CO, Sacks FM, Aikawa M. Multiple apolipoprotein kinetics measured in human HDL by high-resolution/accurate mass parallel reaction monitoring. J Lipid Res 2016; 57:714-28. [PMID: 26862155 DOI: 10.1194/jlr.d061432] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Indexed: 01/10/2023] Open
Abstract
Endogenous labeling with stable isotopes is used to study the metabolism of proteins in vivo. However, traditional detection methods such as GC/MS cannot measure tracer enrichment in multiple proteins simultaneously, and multiple reaction monitoring MS cannot measure precisely the low tracer enrichment in slowly turning-over proteins as in HDL. We exploited the versatility of the high-resolution/accurate mass (HR/AM) quadrupole Orbitrap for proteomic analysis of five HDL sizes. We identified 58 proteins in HDL that were shared among three humans and that were organized into five subproteomes according to HDL size. For seven of these proteins, apoA-I, apoA-II, apoA-IV, apoC-III, apoD, apoE, and apoM, we performed parallel reaction monitoring (PRM) to measure trideuterated leucine tracer enrichment between 0.03 to 1.0% in vivo, as required to study their metabolism. The results were suitable for multicompartmental modeling in all except apoD. These apolipoproteins in each HDL size mainly originated directly from the source compartment, presumably the liver and intestine. Flux of apolipoproteins from smaller to larger HDL or the reverse contributed only slightly to apolipoprotein metabolism. These novel findings on HDL apolipoprotein metabolism demonstrate the analytical breadth and scope of the HR/AM-PRM technology to perform metabolic research.
Collapse
Affiliation(s)
- Sasha A Singh
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Allison B Andraski
- Department of Nutrition, Harvard T. H. Chan School of Public Health, Boston, MA
| | - Brett Pieper
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Wilson Goh
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | | | - Frank M Sacks
- Department of Nutrition, Harvard T. H. Chan School of Public Health, Boston, MA Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Masanori Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
143
|
Extended-Release Niacin/Laropiprant Improves Overall Efficacy of Postprandial Reverse Cholesterol Transport. Arterioscler Thromb Vasc Biol 2016; 36:285-94. [DOI: 10.1161/atvbaha.115.306834] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Accepted: 11/30/2015] [Indexed: 11/16/2022]
|
144
|
Alphonse PAS, Jones PJH. Revisiting Human Cholesterol Synthesis and Absorption: The Reciprocity Paradigm and its Key Regulators. Lipids 2015. [PMID: 26620375 DOI: 10.1007/s11745‐015‐4096‐7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Hypercholesterolemia is a major risk factor for cardiovascular disease. Cholesterol homeostasis in the body is governed by the interplay between absorption, synthesis, and excretion or conversion of cholesterol into bile acids. A reciprocal relationship between cholesterol synthesis and absorption is known to regulate circulating cholesterol in response to dietary or therapeutic interventions. However, the degree to which these factors affect synthesis and absorption and the extent to which one vector shifts in response to the other are not thoroughly understood. Also, huge inter-individual variability exists in the manner in which the two systems act in response to any cholesterol-lowering treatment. Various factors are known to account for this variability and in light of recent experimental advances new players such as gene-gene interactions, gene-environmental effects, and gut microbiome hold immense potential in offering an explanation to the complex traits of inter-individual variability in human cholesterol metabolism. In this context, the objective of the present review is to provide an overview on cholesterol metabolism and discuss the role of potential factors such as genetics, epigenetics, epistasis, and gut microbiome, as well as other regulators in modulating cholesterol metabolism, especially emphasizing the reciprocal relationship between cholesterol synthesis and absorption. Furthermore, an evaluation of the implications of this push-pull mechanism on cholesterol-lowering strategies is presented.
Collapse
Affiliation(s)
- Peter A S Alphonse
- Human Nutritional Sciences, University of Manitoba, Winnipeg, MB, Canada.
- Richardson Centre for Functional Foods and Nutraceuticals (RCFFN), University of Manitoba, 196, Innovation Drive, SmartPark, Winnipeg, MB, R3T 2N2, Canada.
| | - Peter J H Jones
- Human Nutritional Sciences, University of Manitoba, Winnipeg, MB, Canada
- Richardson Centre for Functional Foods and Nutraceuticals (RCFFN), University of Manitoba, 196, Innovation Drive, SmartPark, Winnipeg, MB, R3T 2N2, Canada
- Food Science, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
145
|
Alphonse PAS, Jones PJH. Revisiting Human Cholesterol Synthesis and Absorption: The Reciprocity Paradigm and its Key Regulators. Lipids 2015; 51:519-36. [PMID: 26620375 DOI: 10.1007/s11745-015-4096-7] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 11/09/2015] [Indexed: 12/22/2022]
Abstract
Hypercholesterolemia is a major risk factor for cardiovascular disease. Cholesterol homeostasis in the body is governed by the interplay between absorption, synthesis, and excretion or conversion of cholesterol into bile acids. A reciprocal relationship between cholesterol synthesis and absorption is known to regulate circulating cholesterol in response to dietary or therapeutic interventions. However, the degree to which these factors affect synthesis and absorption and the extent to which one vector shifts in response to the other are not thoroughly understood. Also, huge inter-individual variability exists in the manner in which the two systems act in response to any cholesterol-lowering treatment. Various factors are known to account for this variability and in light of recent experimental advances new players such as gene-gene interactions, gene-environmental effects, and gut microbiome hold immense potential in offering an explanation to the complex traits of inter-individual variability in human cholesterol metabolism. In this context, the objective of the present review is to provide an overview on cholesterol metabolism and discuss the role of potential factors such as genetics, epigenetics, epistasis, and gut microbiome, as well as other regulators in modulating cholesterol metabolism, especially emphasizing the reciprocal relationship between cholesterol synthesis and absorption. Furthermore, an evaluation of the implications of this push-pull mechanism on cholesterol-lowering strategies is presented.
Collapse
Affiliation(s)
- Peter A S Alphonse
- Human Nutritional Sciences, University of Manitoba, Winnipeg, MB, Canada. .,Richardson Centre for Functional Foods and Nutraceuticals (RCFFN), University of Manitoba, 196, Innovation Drive, SmartPark, Winnipeg, MB, R3T 2N2, Canada.
| | - Peter J H Jones
- Human Nutritional Sciences, University of Manitoba, Winnipeg, MB, Canada.,Richardson Centre for Functional Foods and Nutraceuticals (RCFFN), University of Manitoba, 196, Innovation Drive, SmartPark, Winnipeg, MB, R3T 2N2, Canada.,Food Science, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
146
|
Meadows DN, Bahous RH, Best AF, Rozen R. High Dietary Folate in Mice Alters Immune Response and Reduces Survival after Malarial Infection. PLoS One 2015; 10:e0143738. [PMID: 26599510 PMCID: PMC4658061 DOI: 10.1371/journal.pone.0143738] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Accepted: 11/09/2015] [Indexed: 12/20/2022] Open
Abstract
Malaria is a significant global health issue, with nearly 200 million cases in 2013 alone. Parasites obtain folate from the host or synthesize it de novo. Folate consumption has increased in many populations, prompting concerns regarding potential deleterious consequences of higher intake. The impact of high dietary folate on the host’s immune function and response to malaria has not been examined. Our goal was to determine whether high dietary folate would affect response to malarial infection in a murine model of cerebral malaria. Mice were fed control diets (CD, recommended folate level for rodents) or folic acid-supplemented diets (FASD, 10x recommended level) for 5 weeks before infection with Plasmodium berghei ANKA. Survival, parasitemia, numbers of immune cells and other infection parameters were assessed. FASD mice had reduced survival (p<0.01, Cox proportional hazards) and higher parasitemia (p< 0.01, joint model of parasitemia and survival) compared with CD mice. FASD mice had lower numbers of splenocytes, total T cells, and lower numbers of specific T and NK cell sub-populations, compared with CD mice (p<0.05, linear mixed effects). Increased brain TNFα immunoreactive protein (p<0.01, t-test) and increased liver Abca1 mRNA (p<0.01, t-test), a modulator of TNFα, were observed in FASD mice; these variables correlated positively (rs = 0.63, p = 0.01). Bcl-xl/Bak mRNA was increased in liver of FASD mice (p<0.01, t-test), suggesting reduced apoptotic potential. We conclude that high dietary folate increases parasite replication, disturbs the immune response and reduces resistance to malaria in mice. These findings have relevance for malaria-endemic regions, when considering anti-folate anti-malarials, food fortification or vitamin supplementation programs.
Collapse
Affiliation(s)
- Danielle N. Meadows
- Department of Human Genetics, McGill University, McGill University Health Center, Montreal, Quebec, Canada
| | - Renata H. Bahous
- Department of Human Genetics, McGill University, McGill University Health Center, Montreal, Quebec, Canada
| | - Ana F. Best
- Department of Mathematics and Statistics, McGill University, Montreal, Quebec, Canada
| | - Rima Rozen
- Department of Human Genetics, McGill University, McGill University Health Center, Montreal, Quebec, Canada
- Department of Pediatrics, McGill University, McGill University Health Center, Montreal, Quebec, Canada
- * E-mail:
| |
Collapse
|
147
|
Jacobo-Albavera L, Posadas-Romero C, Vargas-Alarcón G, Romero-Hidalgo S, Posadas-Sánchez R, González-Salazar MDC, Carnevale A, Canizales-Quinteros S, Medina-Urrutia A, Antúnez-Argüelles E, Villarreal-Molina T. Dietary fat and carbohydrate modulate the effect of the ATP-binding cassette A1 (ABCA1) R230C variant on metabolic risk parameters in premenopausal women from the Genetics of Atherosclerotic Disease (GEA) Study. Nutr Metab (Lond) 2015; 12:45. [PMID: 26579206 PMCID: PMC4647664 DOI: 10.1186/s12986-015-0040-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 11/09/2015] [Indexed: 12/16/2022] Open
Abstract
Background Although the R230C-ATP-binding cassette A1 (ABCA1) variant has been consistently associated with HDL-C levels, its association with diabetes and other metabolic parameters is unclear. Estrogen and dietary factors are known to regulate ABCA1 expression in different tissues. Thus, we aimed to explore whether gender, menopausal status and macronutrient proportions of diet modulate the effect of this variant on various metabolic parameters. Methods One thousand five hundred ninety-eight controls from the GEA study were included (787 men, 363 premenopausal women and 448 menopausal women), previously assessed for anthropometric and biochemical measurements and visceral to subcutaneous abdominal fat (VAT/SAT) ratio on computed tomography. Taqman assays were performed for genotyping. Diet macronutrient proportions were assessed using a food frequency questionnaire validated for the Mexican population. Multivariate regression models were constructed to assess the interaction between the proportion of dietary macronutrients and the R230C polymorphism on metabolic parameters. Results All significant interactions were observed in premenopausal women. Those carrying the risk allele and consuming higher carbohydrate/lower fat diets showed an unfavorable metabolic pattern [lower HDL-C and adiponectin levels, higher VAT/SAT ratio, homeostasis model assessment for insulin resistance (HOMA-IR) and higher gamma-glutamyl transpeptidase (GGT) and alkaline phosphatase (ALP) levels]. Conversely, premenopausal women carrying the risk allele and consuming lower carbohydrate/higher fat diets showed a more favorable metabolic pattern (higher HDL-C and adiponectin levels, and lower VAT/SAT ratio, HOMA-IR, GGT and ALP levels). Conclusion This is the first study reporting a gender-specific interaction between ABCA1/R230C variant and dietary carbohydrate and fat percentages affecting VAT/SAT ratio, GGT, ALP, adiponectin levels and HOMA index. Our study confirmed the previously reported gender-specific ABCA1-diet interaction affecting HDL-C levels observed in an independent study. Our results show how gene-environment interactions may help further understand how certain gene variants confer metabolic risk, and may provide information useful to design diet intervention studies. Electronic supplementary material The online version of this article (doi:10.1186/s12986-015-0040-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Leonor Jacobo-Albavera
- Laboratorio de Genómica de Enfermedades Cardiovasculares, Instituto Nacional de Medicina Genómica, Mexico City, Mexico
| | - Carlos Posadas-Romero
- Departamento de Endocrinología, Instituto Nacional de Cardiología "Ignacio Chávez", Mexico City, Mexico
| | - Gilberto Vargas-Alarcón
- Departamento de Biología Molecular, Instituto Nacional de Cardiología "Ignacio Chávez", Mexico City, Mexico
| | - Sandra Romero-Hidalgo
- Departamento de Genómica Computacional, Instituto Nacional de Medicina Genómica, Mexico City, Mexico
| | - Rosalinda Posadas-Sánchez
- Departamento de Endocrinología, Instituto Nacional de Cardiología "Ignacio Chávez", Mexico City, Mexico
| | | | - Alessandra Carnevale
- Laboratorio de Enfermedades Mendelianas, Instituto Nacional de Medicina Genómica, Mexico City, Mexico
| | - Samuel Canizales-Quinteros
- Unidad de Genómica de Poblaciones Aplicada a la Salud, Facultad de Química UNAM-INMEGEN, Mexico City, Mexico
| | - Aida Medina-Urrutia
- Departamento de Endocrinología, Instituto Nacional de Cardiología "Ignacio Chávez", Mexico City, Mexico
| | - Erika Antúnez-Argüelles
- Laboratorio de Genómica de Enfermedades Cardiovasculares, Instituto Nacional de Medicina Genómica, Periférico Sur 4809 Colonia Arenal Tepepan, CP 14610 México, D.F. Mexico
| | - Teresa Villarreal-Molina
- Laboratorio de Genómica de Enfermedades Cardiovasculares, Instituto Nacional de Medicina Genómica, Periférico Sur 4809 Colonia Arenal Tepepan, CP 14610 México, D.F. Mexico
| |
Collapse
|
148
|
Dysfunctional High-Density Lipoprotein: An Innovative Target for Proteomics and Lipidomics. CHOLESTEROL 2015; 2015:296417. [PMID: 26634153 PMCID: PMC4655037 DOI: 10.1155/2015/296417] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/23/2015] [Revised: 10/12/2015] [Accepted: 10/12/2015] [Indexed: 02/02/2023]
Abstract
High-Density Lipoprotein-Cholesterol (HDL-C) is regarded as an important protective factor against cardiovascular disease, with abundant evidence of an inverse relationship between its serum levels and risk of cardiovascular disease, as well as various antiatherogenic, antioxidant, and anti-inflammatory properties. Nevertheless, observations of hereditary syndromes featuring scant HDL-C concentration in absence of premature atherosclerotic disease suggest HDL-C levels may not be the best predictor of cardiovascular disease. Indeed, the beneficial effects of HDL may not depend solely on their concentration, but also on their quality. Distinct subfractions of this lipoprotein appear to be constituted by specific protein-lipid conglomerates necessary for different physiologic and pathophysiologic functions. However, in a chronic inflammatory microenvironment, diverse components of the HDL proteome and lipid core suffer alterations, which propel a shift towards a dysfunctional state, where HDL-C becomes proatherogenic, prooxidant, and proinflammatory. This heterogeneity highlights the need for further specialized molecular studies in this aspect, in order to achieve a better understanding of this dysfunctional state; with an emphasis on the potential role for proteomics and lipidomics as valuable methods in the search of novel therapeutic approaches for cardiovascular disease.
Collapse
|
149
|
Jeon BH, Lee YH, Yun MR, Kim SH, Lee BW, Kang ES, Lee HC, Cha BS. Increased expression of ATP-binding cassette transporter A1 (ABCA1) as a possible mechanism for the protective effect of cilostazol against hepatic steatosis. Metabolism 2015; 64:1444-53. [PMID: 26362727 DOI: 10.1016/j.metabol.2015.07.014] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Revised: 06/24/2015] [Accepted: 07/15/2015] [Indexed: 01/01/2023]
Abstract
OBJECTIVE Cilostazol, a phosphodiesterase 3, has been widely used in patients with arterial disease and is known to have additional beneficial effects on dyslipidemia. However, the effect of cilostazol on hepatic steatosis has not been fully elucidated. We investigated the effect of cilostazol on hepatic ABCA1 expression and hepatic steatosis in diet-induced obesity mice model. METHODS Hepatic ABCA1 expression and lipid accumulation were analyzed in HepG2 cell lines treated with cilostazol. Male C57BL/6 mice were randomly divided into three groups: (1) fed normal chow diet with vehicle; (2) fed high-fat diet (HFD) with vehicle; (3) fed HFD with cilostazol. Cilostazol (30 mg/kg) was orally administered once daily for 9 weeks. RESULTS Cilostazol significantly enhanced ABCA1 expression and restored ABCA1 expression reduced by palmitate in HepG2 cells. Cilostazol treatment ameliorated lipid accumulation induced by palmitate, and this effect was diminished when ABCA1 or LRP1 was silenced by small interference RNA. After silencing of LRP1, ABCA1 expression was decreased in HepG2 cells. Cilostazol significantly enhanced hepatic ABCA1 expression and decreased hepatic fat in HFD-fed mice. Hepatic expression of cleaved caspase-3 and PARP1 was also decreased in HFD-fed mice treated with cilostazol. CONCLUSIONS Cilostazol ameliorated hepatic steatosis and increased ABCA1 expression in the hepatocytes. Enhancing ABCA1 expression with cilostazol represents a potential therapeutic avenue for treatment of hepatic steatosis.
Collapse
Affiliation(s)
- Byung Hun Jeon
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Yong-ho Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Mi Ra Yun
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Soo Hyun Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Byung Wan Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Eun Seok Kang
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Hyun Chul Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Bong-Soo Cha
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea; Division of Endocrinology and Metabolism, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea.
| |
Collapse
|
150
|
Rødgaard T, Heegaard PM, Callesen H. Non-invasive assessment of in-vitro embryo quality to improve transfer success. Reprod Biomed Online 2015; 31:585-92. [DOI: 10.1016/j.rbmo.2015.08.003] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Revised: 08/05/2015] [Accepted: 08/06/2015] [Indexed: 12/12/2022]
|