1
|
Do TTM, Vu CD, Dien TM, Can TBN, Nguyen TTN, Nguyen HH, Tran VK, Nguyen NL, Tran HT, Mai TTC, Nguyen KN. Phenotypes, Genotypes, Treatment, and Outcomes of 14 Children with Sitosterolemia at Vietnam National Children's Hospital. J Clin Med 2025; 14:325. [PMID: 39860331 PMCID: PMC11765834 DOI: 10.3390/jcm14020325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 01/03/2025] [Accepted: 01/03/2025] [Indexed: 01/27/2025] Open
Abstract
Background: Sitosterolemia is a rare autosomal recessive disorder characterized by diverse clinical manifestations ranging from asymptomatic cases to the development of xanthomas, hypercholesterolemia, premature atherosclerosis, or even sudden death during childhood. It results from homozygous or compound heterozygous pathogenic variants in the ABCG5 or ABCG8 genes. Prompt detection and intervention are essential to managing this condition and preventing severe outcomes. Methods: This study aims to retrospectively analyze the phenotype, genotype, treatment, and outcomes of 14 children-seven boys and seven girls-all of Vietnamese origin, diagnosed with sitosterolemia at the Vietnam National Children's Hospital between March 2015 and July 2024. Results: The median ages at disease onset and diagnosis were 5.7 years (range: 1.5-17.9) and 7.2 years (range: 1.7-17.9), respectively. Xanthomas were observed in 85.7% of patients (12/14), arthralgia in 14.3% (2/14), and anemia in 7.1% (1/14), with no cases of thrombocytopenia. At diagnosis, all patients exhibited elevated total cholesterol (TC) and low-density lipoprotein cholesterol (LDL-C), with considerably higher levels in patients with xanthomas compared to those without. Mutations in the ABCG5 gene were identified in 71.4% (10/14) of the patients, while 28.6% (4/14) had mutations in the ABCG8 gene. Fourteen variants were detected, nine in ABCG5 and five in ABCG8, with five variants reported for the first time in sitosterolemia patients. Initial management for all patients involved dietary modifications. After three months, 10 patients with persistently elevated TC and LDL-C received ezetimibe or cholestyramine treatment. Among the eight patients who continued treatment for over three months, the median TC and LDL-C concentrations decreased by 54.9% and 67.3%, respectively. Conclusions: Among Vietnamese patients with sitosterolemia, variants in the ABCG5 gene were more prevalent than those in the ABCG8 gene. Patients showed a positive response to ezetimibe or cholestyramine treatment. Genetic testing is essential for establishing a diagnosis of sitosterolemia and guiding accurate management strategies.
Collapse
Affiliation(s)
- Thi Thanh Mai Do
- Hanoi Medical University, 1st Ton That Tung Street, Hanoi 11521, Vietnam; (T.T.M.D.); (C.D.V.); (T.T.C.M.)
- Center of Endocrinology, Metabolism, Genetic/Genomics and Molecular Therapy, Vietnam National Children’s Hospital, 18/879 La Thanh, Dong Da, Hanoi 11512, Vietnam;
| | - Chi Dung Vu
- Hanoi Medical University, 1st Ton That Tung Street, Hanoi 11521, Vietnam; (T.T.M.D.); (C.D.V.); (T.T.C.M.)
- Center of Endocrinology, Metabolism, Genetic/Genomics and Molecular Therapy, Vietnam National Children’s Hospital, 18/879 La Thanh, Dong Da, Hanoi 11512, Vietnam;
| | - Tran Minh Dien
- Vietnam National Children’s Hospital, 18/879 La Thanh, Dong Da, Hanoi 11512, Vietnam;
| | - Thi Bich Ngoc Can
- Center of Endocrinology, Metabolism, Genetic/Genomics and Molecular Therapy, Vietnam National Children’s Hospital, 18/879 La Thanh, Dong Da, Hanoi 11512, Vietnam;
| | - Thi Thanh Ngan Nguyen
- Institute of Genome Research, Vietnam Academy of Science and Technology, 18 Hoang Quoc Viet Street, Cau Giay, Hanoi 100000, Vietnam; (T.T.N.N.); (H.H.N.)
| | - Huy Hoang Nguyen
- Institute of Genome Research, Vietnam Academy of Science and Technology, 18 Hoang Quoc Viet Street, Cau Giay, Hanoi 100000, Vietnam; (T.T.N.N.); (H.H.N.)
| | - Van Khanh Tran
- Center for Gene and Protein Research, Hanoi Medical University, 1st Ton That Tung Street, Hanoi 11521, Vietnam; (V.K.T.); (N.L.N.)
| | - Ngoc Lan Nguyen
- Center for Gene and Protein Research, Hanoi Medical University, 1st Ton That Tung Street, Hanoi 11521, Vietnam; (V.K.T.); (N.L.N.)
| | - Huy Thinh Tran
- Biochemistry Department, Hanoi Medical University, 1st Ton That Tung Street, Hanoi 11521, Vietnam;
| | - Tran Thi Chi Mai
- Hanoi Medical University, 1st Ton That Tung Street, Hanoi 11521, Vietnam; (T.T.M.D.); (C.D.V.); (T.T.C.M.)
| | - Khanh Ngoc Nguyen
- Hanoi Medical University, 1st Ton That Tung Street, Hanoi 11521, Vietnam; (T.T.M.D.); (C.D.V.); (T.T.C.M.)
- Center of Endocrinology, Metabolism, Genetic/Genomics and Molecular Therapy, Vietnam National Children’s Hospital, 18/879 La Thanh, Dong Da, Hanoi 11512, Vietnam;
| |
Collapse
|
2
|
Chen Z, Ding H, Zhu H, Huang S, Yan C, Chen ZY. Additional mechanism for selective absorption of cholesterol and phytosterols. Food Chem 2024; 458:140300. [PMID: 38964108 DOI: 10.1016/j.foodchem.2024.140300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 06/16/2024] [Accepted: 06/29/2024] [Indexed: 07/06/2024]
Abstract
Phytosterols are structurally similar to cholesterol but they are much less absorbed (<2%) than cholesterol (>50%) in the intestine. We hypothesize that phytosterols are poor substrates of intestinal acyl-CoA: cholesterol acyltransferase 2 (ACAT2), and thus minimal phytosterol esters are formed and packed into chylomicrons, leading to their low absorption. Two isotope tracing models, including a radioactive hamster microsomal ACAT2 reaction model and a differentiated Caco-2 cell model, were established to examine the specificity of ACAT2 to various sterols, including cholesterol, sitosterol, stigmasterol, and campesterol. Both models consistently demonstrated that only cholesterol but not phytosterols could be efficiently esterified by ACAT2 in a time- and dose-dependent manner. Molecular docking further suggested that unfavorable interactions existed between ACAT2 and phytosterols. In conclusion, phytosterols are poor substrates of ACAT2 and thus minimally absorbed. This work provides a theoretical basis for the use of phytosterol-based supplements in treating dyslipidemia and preventing heart diseases.
Collapse
Affiliation(s)
- Zixing Chen
- School of Life Sciences, The Chinese University of Hong Kong, Hong Kong 999077, China..
| | - Huafang Ding
- School of Life Sciences, The Chinese University of Hong Kong, Hong Kong 999077, China..
| | - Hanyue Zhu
- School of Food Science and Engineering / Guangdong Provincial Key Laboratory of Intelligent Food Manufacturing, Foshan University, Foshan, Guangdong, China
| | - Shouhe Huang
- School of Life Sciences, The Chinese University of Hong Kong, Hong Kong 999077, China..
| | - Chi Yan
- School of Life Sciences, The Chinese University of Hong Kong, Hong Kong 999077, China..
| | - Zhen-Yu Chen
- School of Life Sciences, The Chinese University of Hong Kong, Hong Kong 999077, China..
| |
Collapse
|
3
|
Brañes MC, Gillet R, Valenzuela R. Nuclear receptors behind the therapeutic effects of plant sterols on metabolism: A review. Lipids 2024; 59:169-180. [PMID: 39077818 DOI: 10.1002/lipd.12409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 07/14/2024] [Accepted: 07/15/2024] [Indexed: 07/31/2024]
Abstract
Plant sterols are known for their hypocholesterolemic action, and the molecular mechanisms behind this within the gut have been extensively discussed and demonstrated to the point that there is a degree of consensus. However, recent studies show that these molecules exert an additional umbrella of therapeutic effects in other tissues, which are related to immune function, lipid metabolism, and glucose metabolism. A strong hypothesis to explain these effects is the structural relationship between plant sterols and the ligands of a group of nuclear receptors. This review delves into the molecular aspects of therapeutic effects related with lipid and energy metabolism that have been observed and demonstrated for plant sterols, and turns the perspective to explore the involvement of nuclear receptors as part of these mechanisms.
Collapse
Affiliation(s)
| | | | - Rodrigo Valenzuela
- Department of Nutrition, Faculty of Medicine, University of Chile, Santiago, Chile
| |
Collapse
|
4
|
Evtyugin DD, Evtuguin DV, Casal S, Domingues MR. Advances and Challenges in Plant Sterol Research: Fundamentals, Analysis, Applications and Production. Molecules 2023; 28:6526. [PMID: 37764302 PMCID: PMC10535520 DOI: 10.3390/molecules28186526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 09/04/2023] [Accepted: 09/05/2023] [Indexed: 09/29/2023] Open
Abstract
Plant sterols (PS) are cholesterol-like terpenoids widely spread in the kingdom Plantae. Being the target of extensive research for more than a century, PS have topped with evidence of having beneficial effects in healthy subjects and applications in food, cosmetic and pharmaceutical industries. However, many gaps in several fields of PS's research still hinder their widespread practical applications. In fact, many of the mechanisms associated with PS supplementation and their health benefits are still not fully elucidated. Furthermore, compared to cholesterol data, many complex PS chemical structures still need to be fully characterized, especially in oxidized PS. On the other hand, PS molecules have also been the focus of structural modifications for applications in diverse areas, including not only the above-mentioned but also in e.g., drug delivery systems or alternative matrixes for functional foods and fats. All the identified drawbacks are also superimposed by the need of new PS sources and technologies for their isolation and purification, taking into account increased environmental and sustainability concerns. Accordingly, current and future trends in PS research warrant discussion.
Collapse
Affiliation(s)
- Dmitry D. Evtyugin
- CICECO, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal; (D.D.E.); (D.V.E.)
- LAQV-REQUIMTE, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
- Mass Spectrometry Centre, LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal
| | - Dmitry V. Evtuguin
- CICECO, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal; (D.D.E.); (D.V.E.)
| | - Susana Casal
- LAQV-REQUIMTE, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Maria Rosário Domingues
- Mass Spectrometry Centre, LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal
- CESAM, Centre for Environmental and Marine Studies, Department of Chemistry, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal
| |
Collapse
|
5
|
Windler E, Beil FU, Berthold HK, Gouni-Berthold I, Kassner U, Klose G, Lorkowski S, März W, Parhofer KG, Plat J, Silbernagel G, Steinhagen-Thiessen E, Weingärtner O, Zyriax BC, Lütjohann D. Phytosterols and Cardiovascular Risk Evaluated against the Background of Phytosterolemia Cases-A German Expert Panel Statement. Nutrients 2023; 15:nu15040828. [PMID: 36839186 PMCID: PMC9963617 DOI: 10.3390/nu15040828] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 01/23/2023] [Accepted: 01/25/2023] [Indexed: 02/08/2023] Open
Abstract
Phytosterols (PSs) have been proposed as dietary means to lower plasma LDL-C. However, concerns are raised that PSs may exert atherogenic effects, which would offset this benefit. Phytosterolemia was thought to mimic increased plasma PSs observed after the consumption of PS-enriched foods. This expert statement examines the possibility of specific atherogenicity of PSs based on sterol metabolism, experimental, animal, and human data. Observational studies show no evidence that plasma PS concentrations would be associated with an increased risk of atherosclerosis or cardiovascular (CV) events. Since variants of the ABCG5/8 transporter affect the absorption of cholesterol and non-cholesterol sterols, Mendelian randomization studies examining the effects of ABCG5/8 polymorphisms cannot support or refute the potential atherogenic effects of PSs due to pleiotropy. In homozygous patients with phytosterolemia, total PS concentrations are ~4000% higher than under physiological conditions. The prevalence of atherosclerosis in these individuals is variable and may mainly relate to concomitant elevated LDL-C. Consuming PS-enriched foods increases PS concentrations by ~35%. Hence, PSs, on a molar basis, would need to have 20-40 times higher atherogenicity than cholesterol to offset their cholesterol reduction benefit. Based on their LDL-C lowering and absence of adverse safety signals, PSs offer a dietary approach to cholesterol management. However, their clinical benefits have not been established in long-term CV endpoint studies.
Collapse
Affiliation(s)
- Eberhard Windler
- Preventive Medicine, University Heart Center, University Hospital Hamburg-Eppendorf, Hamburg-Eppendorf, Martinistr. 52-Bldg. N26, 20246 Hamburg, Germany
| | - Frank-Ulrich Beil
- Ambulanzzentrum, Universitätsklinikum Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany
| | - Heiner K. Berthold
- Department of Internal Medicine and Geriatrics, Bethel Clinic, 33611 Bielefeld, Germany
| | - Ioanna Gouni-Berthold
- Center for Endocrinology, Diabetes and Preventive Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Kerpener Str. 62, 50937 Cologne, Germany
| | - Ursula Kassner
- Lipid Clinic at the Interdisciplinary Metabolism Center, Charite-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Gerald Klose
- Praxen Dres. T. Beckenbauer & S. Maierhof, Am Markt 11, 28195 Bremen und Dres. I. van de Loo & K. Spieker, Gerold Janssen Straße 2 A, 28359 Bremen, Germany
| | - Stefan Lorkowski
- Institute of Nutritional Science and Competence Cluster for Nutrition and Cardiovascular Health (nutriCARD), Halle-Jena-Leipzig, Friedrich Schiller University Jena, Dornburger Str. 25, 07743 Jena, Germany
| | - Winfried März
- SYNLAB Akademie für Ärztliche Fortbildung, SYNLAB Holding Deutschland GmbH, P5,7, 68161 Mannheim, Germany
- Medical Clinic V, Medical Faculty Mannheim, University of Heidelberg, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, 8010 Graz, Austria
- Correspondence:
| | - Klaus G. Parhofer
- Medizinische Klinik IV, Klinikum der Universität München, Grosshadern, Marchioninistr. 15, 81377 München, Germany
| | - Jogchum Plat
- Department of Nutrition and Movement Sciences, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, 6211 LK Maastricht, The Netherlands
| | - Günter Silbernagel
- Division of Vascular Medicine, Department of Internal Medicine, Medical University of Graz, 8010 Graz, Austria
| | - Elisabeth Steinhagen-Thiessen
- Arbeitsbereich Lipidstoffwechsel der Medizinischen Klinik für Endokrinologie und Stoffwechselmedizin, Charité—Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Oliver Weingärtner
- Klinik für Innere Medizin I, Universitätskliniken Jena, Friedrich-Schiller-Universität Jena, 07743 Jena, Germany
| | - Birgit-Christiane Zyriax
- Midwifery Science—Health Care Research and Prevention, Research Group, Preventive Medicine and Nutrition, Institute for Health Services Research in Dermatology and Nursing (IVDP), University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany
| | - Dieter Lütjohann
- Institute of Clinical Chemistry and Clinical Pharmacology, University Clinics Bonn, 53127 Bonn, Germany
| |
Collapse
|
6
|
Effect of Coffee on the Bioavailability of Sterols. Foods 2022; 11:foods11192935. [PMID: 36230011 PMCID: PMC9563500 DOI: 10.3390/foods11192935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 09/12/2022] [Accepted: 09/16/2022] [Indexed: 11/24/2022] Open
Abstract
Absorption at the intestinal epithelium is a major determinant of cholesterol levels in the organism, influencing the entry of dietary cholesterol and the excretion of endogenous cholesterol. Several strategies are currently being followed to reduce cholesterol absorption, using both pharmacological agents or food ingredients with hypocholesterolemic properties. Coffee has recently been shown to affect cholesterol bioaccessibility, although it has not been shown if this translates into a decrease on cholesterol bioavailability. In this work, coffee obtained with different commercial roasting (light and dark) and grinding (finer and coarser) was evaluated regarding their effect on cholesterol absorption through Caco-2 monolayers, mimicking the intestinal epithelium. The fluorescent dehydroergosterol was used as a sterol model, which was shown to permeate Caco-2 monolayers with a low-to-moderate permeability coefficient depending on its concentration. In the presence of coffee extracts, a 50% decrease of the sterol permeability coefficient was observed, showing their potential to affect sterol bioavailability. This was attributed to an increased sterol precipitation and its deposition on the apical epithelial surface. A higher hypocholesterolemic effect was observed for the dark roasting and finer grinding, showing that the modulation of these technological processing parameters may produce coffees with optimized hypocholesterolemic activity.
Collapse
|
7
|
A teenager boy with a novel variant of Sitosterolemia presented with pancytopenia. Clin Chim Acta 2022; 529:61-66. [DOI: 10.1016/j.cca.2022.02.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 02/02/2022] [Accepted: 02/02/2022] [Indexed: 11/20/2022]
|
8
|
Li X, Xin Y, Mo Y, Marozik P, He T, Guo H. The Bioavailability and Biological Activities of Phytosterols as Modulators of Cholesterol Metabolism. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27020523. [PMID: 35056839 PMCID: PMC8781140 DOI: 10.3390/molecules27020523] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 01/05/2022] [Accepted: 01/13/2022] [Indexed: 12/12/2022]
Abstract
Phytosterols are natural sterols widely found in plants that have a variety of physiological functions, and their role in reducing cholesterol absorption has garnered much attention. Although the bioavailability of phytosterols is only 0.5–2%, they can still promote cholesterol balance in the body. A mechanism of phytosterols for lowering cholesterol has now been proposed. They not only reduce the uptake of cholesterol in the intestinal lumen and affect its transport, but also regulate the metabolism of cholesterol in the liver. In addition, phytosterols can significantly reduce the plasma concentration of total cholesterol, triglycerides, and low-density lipoprotein cholesterol (LDL-C), with a dose-response relationship. Ingestion of 3 g of phytosterols per day can reach the platform period, and this dose can reduce LDL-C by about 10.7%. On the other hand, phytosterols can also activate the liver X receptor α-CPY7A1 mediated bile acids excretion pathway and accelerate the transformation and metabolism of cholesterol. This article reviews the research progress of phytosterols as a molecular regulator of cholesterol and the mechanism of action for this pharmacological effect.
Collapse
Affiliation(s)
- Xiang Li
- Department of Nutrition, School of Public Health, Guangdong Medical University, Zhanjiang 524023, China;
| | - Yan Xin
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan 523808, China; (Y.X.); (Y.M.)
| | - Yuqian Mo
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan 523808, China; (Y.X.); (Y.M.)
| | - Pavel Marozik
- Laboratory of Human Genetics, Institute of Genetics and Cytology of the National Academy of Sciences of Belarus, 220072 Minsk, Belarus;
| | - Taiping He
- Department of Nutrition, School of Public Health, Guangdong Medical University, Zhanjiang 524023, China;
- Correspondence: (T.H.); (H.G.); Tel.: +86-759-2388-523 (T.H.); +86-769-2289-6576 (H.G.)
| | - Honghui Guo
- Department of Nutrition, School of Public Health, Guangdong Medical University, Zhanjiang 524023, China;
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan 523808, China; (Y.X.); (Y.M.)
- Correspondence: (T.H.); (H.G.); Tel.: +86-759-2388-523 (T.H.); +86-769-2289-6576 (H.G.)
| |
Collapse
|
9
|
Nattagh-Eshtivani E, Barghchi H, Pahlavani N, Barati M, Amiri Y, Fadel A, Khosravi M, Talebi S, Arzhang P, Ziaei R, Ghavami A. Biological and pharmacological effects and nutritional impact of phytosterols: A comprehensive review. Phytother Res 2021; 36:299-322. [PMID: 34729825 DOI: 10.1002/ptr.7312] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 09/01/2021] [Accepted: 09/30/2021] [Indexed: 12/20/2022]
Abstract
Phytosterols (PSs), classified into plant sterols and stanols, are bioactive compounds found in foods of plant origin. PSs have been proposed to exert a wide number of pharmacological properties, including the potential to reduce total and low-density lipoprotein (LDL) cholesterol levels and thereby decreasing the risk of cardiovascular diseases. Other health-promoting effects of PSs include anti-obesity, anti-diabetic, anti-microbial, anti-inflammatory, and immunomodulatory effects. Also, anticancer effects have been strongly suggested, as phytosterol-rich diets may reduce the risk of cancer by 20%. The aim of this review is to provide a general overview of the available evidence regarding the beneficial physiological and pharmacological activities of PSs, with special emphasis on their therapeutic potential for human health and safety. Also, we will explore the factors that influence the physiologic response to PSs.
Collapse
Affiliation(s)
- Elyas Nattagh-Eshtivani
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Nutrition, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hanieh Barghchi
- Department of Nutrition, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Naseh Pahlavani
- Nutrition and Biochemistry Department, School of Medicine, Social Development and Health Promotion Research Center, Gonabad University of Medical Sciences, Gonabad, Iran.,Department of Clinical Biochemistry and Nutrition, Faculty of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Mehdi Barati
- Department of Immunology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Yasaman Amiri
- Medical School, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Abdulmannan Fadel
- School of Sport and Exercise Sciences, Liverpool John Moores University, Liverpool, UK
| | - Maryam Khosravi
- Department of Nutrition, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Saeedeh Talebi
- Department of Nutrition, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Pishva Arzhang
- Department of Biochemistry and Diet Therapy, Faculty of Nutritional Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Rahele Ziaei
- Department of Community Nutrition, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Abed Ghavami
- Department of Community Nutrition, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
10
|
Hai Q, Smith JD. Acyl-Coenzyme A: Cholesterol Acyltransferase (ACAT) in Cholesterol Metabolism: From Its Discovery to Clinical Trials and the Genomics Era. Metabolites 2021; 11:metabo11080543. [PMID: 34436484 PMCID: PMC8398989 DOI: 10.3390/metabo11080543] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 08/10/2021] [Accepted: 08/12/2021] [Indexed: 11/16/2022] Open
Abstract
The purification and cloning of the acyl-coenzyme A: cholesterol acyltransferase (ACAT) enzymes and the sterol O-acyltransferase (SOAT) genes has opened new areas of interest in cholesterol metabolism given their profound effects on foam cell biology and intestinal lipid absorption. The generation of mouse models deficient in Soat1 or Soat2 confirmed the importance of their gene products on cholesterol esterification and lipoprotein physiology. Although these studies supported clinical trials which used non-selective ACAT inhibitors, these trials did not report benefits, and one showed an increased risk. Early genetic studies have implicated common variants in both genes with human traits, including lipoprotein levels, coronary artery disease, and Alzheimer’s disease; however, modern genome-wide association studies have not replicated these associations. In contrast, the common SOAT1 variants are most reproducibly associated with testosterone levels.
Collapse
|
11
|
Lee JH, Song DY, Jun SH, Song SH, Shin CH, Ki CS, Lee K, Song J. High prevalence of increased sitosterol levels in hypercholesterolemic children suggest underestimation of sitosterolemia incidence. PLoS One 2020; 15:e0238079. [PMID: 32845916 PMCID: PMC7449458 DOI: 10.1371/journal.pone.0238079] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 08/10/2020] [Indexed: 01/26/2023] Open
Abstract
Background Sitosterolemia is an inherited lipid disorder which presents with elevated serum sitosterol and can result in an increased risk of premature cardiovascular disease. However, sitosterol cannot be accurately measured by routine diagnostic assays, meaning that sitosterolemia diagnosis can often be difficult, especially with many clinical features overlapping with familial hypercholesterolemia. With such complications resulting in increasing reports of misdiagnosis, the prevalence of sitosterolemia is predicted to be much higher than previously reported. Methods Gas chromatography-mass spectrometry was utilized to measure sitosterol levels of normocholesterolemic and hypercholesterolemic children. Subsequently, an epidemiologically determined cutoff level of sitosterol was calculated and applied to estimate the prevalence of children with increased sitosterol and identify potential sitosterolemia patients. Massively parallel sequencing was used to confirm the diagnosis in suspected patients. Results Samples from 109 normocholesterolemic and 220 hypercholesterolemic were tested for phytosterols. Sitosterol and campesterol levels were significantly increased in hypercholesterolemic children (mean 22.0±45.9 μmol/L for sitosterol and 26.0±32.8 μmol/L for campesterol) compared to normocholesterolemic children (mean 12.1±4.9 μmol/L for sistosterol and 14.8±6.7 μmol/L for campesterol). Via application of a cutoff of 35.9 μmol/L, the prevalence rates for increased and overtly increased sitosterol in hypercholesterolemic children were 6.4% and 1.4% respectively. Furthermore, 3 suspected sitosterolemia patients were identified, with 2 patients receiving molecular confirmation for sitosterolemia diagnosis. Conclusions Our findings reaffirm that the prevalence of sitosterolemia is probably much higher than previously reported, which also indicates the significant risk of misdiagnosis of sitosterolemia with familial hypercholesterolemia. Special lipid testing including sitosterol, especially in children with uncontrolled hypercholesterolemia, is recommended in children in order to identify potential sitosterolemia patients that would otherwise be neglected.
Collapse
Affiliation(s)
- Joon Hee Lee
- Department of Laboratory Medicine, Seoul National University College of Medicine, Seoul, Korea
- Department of Laboratory Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Da Young Song
- Department of Laboratory Medicine, Seoul National University College of Medicine, Seoul, Korea
- Department of Laboratory Medicine, Seoul National University Hospital, Seoul, Korea
| | - Sun-Hee Jun
- Department of Laboratory Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Sang Hoon Song
- Department of Laboratory Medicine, Seoul National University College of Medicine, Seoul, Korea
- Department of Laboratory Medicine, Seoul National University Hospital, Seoul, Korea
| | - Choong Ho Shin
- Department of Pediatrics, Seoul National University Children's Hospital, Seoul, Korea
| | | | - Kyunghoon Lee
- Department of Laboratory Medicine, Seoul National University College of Medicine, Seoul, Korea
- Department of Laboratory Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
- * E-mail: (KL); (JS)
| | - Junghan Song
- Department of Laboratory Medicine, Seoul National University College of Medicine, Seoul, Korea
- Department of Laboratory Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
- * E-mail: (KL); (JS)
| |
Collapse
|
12
|
Naudin CR, Maner-Smith K, Owens JA, Wynn GM, Robinson BS, Matthews JD, Reedy AR, Luo L, Wolfarth AA, Darby TM, Ortlund EA, Jones RM. Lactococcus lactis Subspecies cremoris Elicits Protection Against Metabolic Changes Induced by a Western-Style Diet. Gastroenterology 2020; 159:639-651.e5. [PMID: 32169430 DOI: 10.1053/j.gastro.2020.03.010] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 02/28/2020] [Accepted: 03/03/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS A Western-style diet, which is high in fat and sugar, can cause significant dyslipidemia and nonalcoholic fatty liver disease; the diet has an especially strong effect in women, regardless of total calorie intake. Dietary supplementation with beneficial microbes might reduce the detrimental effects of a Western-style diet. We assessed the effects of Lactococcus lactis subspecies (subsp) cremoris on weight gain, liver fat, serum cholesterol, and insulin resistance in female mice on a high-fat, high-carbohydrate diet. METHODS Female C57BL/6 mice were fed either a high-fat, high-carbohydrate (Western-style) diet that contained 40% fat (mostly milk fat) and 43% carbohydrate (mostly sucrose) or a calorie-matched-per-gram control diet. The diets of mice were supplemented with 1 × 109 colony-forming units of L lactis subsp cremoris ATCC 19257 or Lactobacillus rhamnosus GG ATCC 53103 (control bacteria) 3 times per week for 16 weeks. Body weights were measured, and fecal, blood, and liver tissues were collected and analyzed. Livers were analyzed for fat accumulation and inflammation, and blood samples were analyzed for cholesterol and glucose levels. Mice were housed within Comprehensive Lab Animal Monitoring System cages, and respiratory exchange ratio and activity were measured. Hepatic lipid profiles of L lactis subsp cremoris-supplemented mice were characterized by lipidomics mass spectrometry analysis. RESULTS Mice fed L lactis subsp cremoris while on the Western-style diet gained less weight, developed less hepatic steatosis and inflammation, and had a lower mean serum level of cholesterol and body mass index than mice fed the control bacteria. Mice fed the L lactis subsp cremoris had increased glucose tolerance while on the Western-style diet compared to mice fed control bacteria and had alterations in hepatic lipids, including oxylipins. CONCLUSIONS Dietary supplementation with L lactis subsp cremoris in female mice on a high-fat, high-carbohydrate (Western-style) diet caused them to gain less weight, develop less liver fat and inflammation, reduce serum cholesterol levels, and increase glucose tolerance compared with mice on the same diet fed control bacteria. L lactis subsp cremoris is safe for oral ingestion and might be developed for persons with metabolic and liver disorders caused by a Western-style diet.
Collapse
Affiliation(s)
- Crystal R Naudin
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Emory University, Atlanta, Georgia
| | - Kristal Maner-Smith
- Emory Integrated Metabolomics and Lipidomics Core, Emory University, Atlanta, Georgia
| | - Joshua A Owens
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Emory University, Atlanta, Georgia
| | - Grace M Wynn
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Emory University, Atlanta, Georgia
| | - Brian S Robinson
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia
| | - Jason D Matthews
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia
| | - April R Reedy
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia
| | - Liping Luo
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Emory University, Atlanta, Georgia
| | - Alexandra A Wolfarth
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia
| | - Trevor M Darby
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Emory University, Atlanta, Georgia
| | - Eric A Ortlund
- Department of Biochemistry, Emory University, Atlanta, Georgia, United States of America
| | - Rheinallt M Jones
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Emory University, Atlanta, Georgia.
| |
Collapse
|
13
|
Cedó L, Farràs M, Lee-Rueckert M, Escolà-Gil JC. Molecular Insights into the Mechanisms Underlying the Cholesterol- Lowering Effects of Phytosterols. Curr Med Chem 2019; 26:6704-6723. [DOI: 10.2174/0929867326666190822154701] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 01/18/2019] [Accepted: 02/22/2019] [Indexed: 12/11/2022]
Abstract
Dietary phytosterols, which comprise plant sterols and stanols, reduce plasma Low-Density Lipoprotein-Cholesterol (LDL-C) levels when given 2 g/day. Since this dose has not been reported to cause health-related side effects in long-term human studies, food products containing these plant compounds are used as potential therapeutic dietary options to reduce LDL-C and cardiovascular disease risk. Several mechanisms have been proposed to explain the cholesterol-lowering action of phytosterols. They may compete with dietary and biliary cholesterol for micellar solubilization in the intestinal lumen, impairing intestinal cholesterol absorption. Recent evidence indicates that phytosterols may also regulate other pathways. Impaired intestinal cholesterol absorption is usually associated with reduced cholesterol transport to the liver, which may reduce the incorporation of cholesterol into Very-Low- Density Lipoprotein (VLDL) particles, thereby lowering the rate of VLDL assembly and secretion. Impaired liver VLDL production may reduce the rate of LDL production. On the other hand, significant evidence supports a role for plant sterols in the Transintestinal Cholesterol Excretion (TICE) pathway, although the exact mechanisms by which they promote the flow of cholesterol from the blood to enterocytes and the intestinal lumen remains unknown. Dietary phytosterols may also alter the conversion of bile acids into secondary bile acids, and may lower the bile acid hydrophobic/hydrophilic ratio, thereby reducing intestinal cholesterol absorption. This article reviews the progress to date in research on the molecular mechanisms underlying the cholesterol-lowering effects of phytosterols.
Collapse
Affiliation(s)
- Lídia Cedó
- Institut d'Investigacions Biomediques (IIB) Sant Pau, Barcelona, Spain
| | - Marta Farràs
- Integrative Systems Medicine and Digestive Disease Division, Department of Surgery and Cancer, Imperial College London, London, United Kingdom
| | | | | |
Collapse
|
14
|
Mechanisms and regulation of cholesterol homeostasis. Nat Rev Mol Cell Biol 2019; 21:225-245. [DOI: 10.1038/s41580-019-0190-7] [Citation(s) in RCA: 450] [Impact Index Per Article: 75.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/24/2019] [Indexed: 12/14/2022]
|
15
|
Shibuya K, Kawamine K, Ozaki C, Ohgiya T, Edano T, Yoshinaka Y, Tsunenari Y. Discovery of Clinical Candidate 2-(4-(2-((1H-Benzo[d]imidazol-2-yl)thio)ethyl)piperazin-1-yl)-N-(6-methyl-2,4-bis(methylthio)pyridin-3-yl)acetamide Hydrochloride [K-604], an Aqueous-Soluble Acyl-CoA:Cholesterol O-Acyltransferase-1 Inhibitor. J Med Chem 2018; 61:10635-10650. [DOI: 10.1021/acs.jmedchem.8b01256] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Affiliation(s)
- Kimiyuki Shibuya
- Tokyo New Drug Research Laboratories, Pharmaceutical Division, Kowa Company, Ltd., 2-17-43, Noguchicho,
Higashimurayama, Tokyo 189-0022, Japan
| | - Katsumi Kawamine
- Tokyo New Drug Research Laboratories, Pharmaceutical Division, Kowa Company, Ltd., 2-17-43, Noguchicho,
Higashimurayama, Tokyo 189-0022, Japan
| | - Chiyoka Ozaki
- Tokyo New Drug Research Laboratories, Pharmaceutical Division, Kowa Company, Ltd., 2-17-43, Noguchicho,
Higashimurayama, Tokyo 189-0022, Japan
| | - Tadaaki Ohgiya
- Tokyo New Drug Research Laboratories, Pharmaceutical Division, Kowa Company, Ltd., 2-17-43, Noguchicho,
Higashimurayama, Tokyo 189-0022, Japan
| | - Toshiyuki Edano
- Tokyo New Drug Research Laboratories, Pharmaceutical Division, Kowa Company, Ltd., 2-17-43, Noguchicho,
Higashimurayama, Tokyo 189-0022, Japan
| | - Yasunobu Yoshinaka
- Tokyo New Drug Research Laboratories, Pharmaceutical Division, Kowa Company, Ltd., 2-17-43, Noguchicho,
Higashimurayama, Tokyo 189-0022, Japan
| | - Yoshihiko Tsunenari
- Tokyo New Drug Research Laboratories, Pharmaceutical Division, Kowa Company, Ltd., 2-17-43, Noguchicho,
Higashimurayama, Tokyo 189-0022, Japan
| |
Collapse
|
16
|
Shibuya K, Kawamine K, Miura T, Ozaki C, Edano T, Mizuno K, Yoshinaka Y, Tsunenari Y. Design, synthesis and pharmacology of aortic-selective acyl-CoA: Cholesterol O-acyltransferase (ACAT/SOAT) inhibitors. Bioorg Med Chem 2018; 26:4001-4013. [DOI: 10.1016/j.bmc.2018.06.024] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 06/14/2018] [Accepted: 06/16/2018] [Indexed: 10/28/2022]
|
17
|
A systemic study of lipid metabolism regulation in salmon fingerlings and early juveniles fed plant oil. Br J Nutr 2018; 120:653-664. [PMID: 30064538 DOI: 10.1017/s0007114518001885] [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] [Indexed: 01/09/2023]
Abstract
In salmon farming, the scarcity of fish oil has driven a shift towards the use of plant-based oil from vegetable or seed, leading to fish feed low in long-chain PUFA (LC-PUFA) and cholesterol. Atlantic salmon has the capacity to synthesise both LC-PUFA and cholesterol, but little is known about the regulation of synthesis and how it varies throughout salmon life span. Here, we present a systemic view of lipid metabolism pathways based on lipid analyses and transcriptomic data from salmon fed contrasting diets of plant or fish oil from first feeding. We analysed four tissues (stomach, pyloric caeca, hindgut and liver) at three life stages (initial feeding 0·16 g, 2·5 g fingerlings and 10 g juveniles). The strongest response to diets higher in plant oil was seen in pyloric caeca of fingerlings, with up-regulation of thirty genes in pathways for cholesterol uptake, transport and biosynthesis. In juveniles, only eleven genes showed differential expression in pyloric caeca. This indicates a higher requirement of dietary cholesterol in fingerlings, which could result in a more sensitive response to plant oil. The LC-PUFA elongation and desaturation pathway was down-regulated in pyloric caeca, probably regulated by srebp1 genes. In liver, cholesterol metabolism and elongation and desaturation genes were both higher on plant oil. Stomach and hindgut were not notably affected by dietary treatment. Plant oil also had a higher impact on fatty acid composition of fingerlings compared with juveniles, suggesting that fingerlings have less metabolic regulatory control when primed with plant oil diet compared with juveniles.
Collapse
|
18
|
Fumeron F, Bard JM, Lecerf JM. Interindividual variability in the cholesterol-lowering effect of supplementation with plant sterols or stanols. Nutr Rev 2018; 75:134-145. [PMID: 28158760 DOI: 10.1093/nutrit/nuw059] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Accepted: 10/21/2016] [Indexed: 01/29/2023] Open
Abstract
Low-density lipoprotein cholesterol (LDL-C) plays a causal role in atherosclerosis. One way to reduce LDL-C levels is to inhibit cholesterol absorption. Plant sterols and stanols compete with cholesterol for absorption in the intestine and induce an average decrease in LDL-C by 5% to 15% in a dose-dependent manner, but not in all individuals. This review focuses on the interindividual variability in response to dietary supplementation with plant sterols and stanols. Dietary plant sterols and stanols have no significant effects on LDL-C in substantial numbers of individuals. Higher responses, in absolute value and percentage of LDL-C, are observed in individuals with higher cholesterol absorption and a lower rate of cholesterol synthesis. Some data provide evidence of the influence of genetics on the response to plant sterols and stanols. Further studies in large populations are required to extend these conclusions about genetic influences.
Collapse
Affiliation(s)
- Frédéric Fumeron
- INSERM, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France; UPMC Université Paris 6, Sorbonne Universités, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France; Univ Paris Diderot, Sorbonne Paris Cité, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France. Université de Nantes (EA 2160), Institut Universitaire Mer et Littoral (IUML) FR3473, CNRS et CRNH (Centre de recherche en Nutrition Humaine), Nantes, France; Institut de Cancérologie de l'Ouest, Saint-Herblain, France. Service de Nutrition, Institut Pasteur de Lille, Lille, France
| | - Jean-Marie Bard
- INSERM, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France; UPMC Université Paris 6, Sorbonne Universités, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France; Univ Paris Diderot, Sorbonne Paris Cité, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France. Université de Nantes (EA 2160), Institut Universitaire Mer et Littoral (IUML) FR3473, CNRS et CRNH (Centre de recherche en Nutrition Humaine), Nantes, France; Institut de Cancérologie de l'Ouest, Saint-Herblain, France. Service de Nutrition, Institut Pasteur de Lille, Lille, France
| | - Jean-Michel Lecerf
- INSERM, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France; UPMC Université Paris 6, Sorbonne Universités, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France; Univ Paris Diderot, Sorbonne Paris Cité, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France. Université de Nantes (EA 2160), Institut Universitaire Mer et Littoral (IUML) FR3473, CNRS et CRNH (Centre de recherche en Nutrition Humaine), Nantes, France; Institut de Cancérologie de l'Ouest, Saint-Herblain, France. Service de Nutrition, Institut Pasteur de Lille, Lille, France
| |
Collapse
|
19
|
Changes in Intestinal Gene Expression of Zebrafish (Danio rerio) Related to Sterol Uptake and Excretion upon β-Sitosterol Administration. FISHES 2018. [DOI: 10.3390/fishes3010001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
|
20
|
Jesch ED, Carr TP. Food Ingredients That Inhibit Cholesterol Absorption. Prev Nutr Food Sci 2017; 22:67-80. [PMID: 28702423 PMCID: PMC5503415 DOI: 10.3746/pnf.2017.22.2.67] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 05/23/2017] [Indexed: 12/12/2022] Open
Abstract
Cholesterol is a vital component of the human body. It stabilizes cell membranes and is the precursor of bile acids, vitamin D and steroid hormones. However, cholesterol accumulation in the bloodstream (hypercholesterolemia) can cause atherosclerotic plaques within artery walls, leading to heart attacks and strokes. The efficiency of cholesterol absorption in the small intestine is of great interest because human and animal studies have linked cholesterol absorption with plasma concentration of total and low density lipoprotein cholesterol. Cholesterol absorption is highly regulated and influenced by particular compounds in the food supply. Therefore, it is desirable to learn more about natural food components that inhibit cholesterol absorption so that food ingredients and dietary supplements can be developed for consumers who wish to manage their plasma cholesterol levels by non-pharmacological means. Food components thus far identified as inhibitors of cholesterol absorption include phytosterols, soluble fibers, phospholipids, and stearic acid.
Collapse
Affiliation(s)
- Elliot D Jesch
- Department of Food, Nutrition and Packaging Sciences, Clemson University, Clemson, SC 29634, USA
| | - Timothy P Carr
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE 68588, USA
| |
Collapse
|
21
|
Schött HF, Baumgartner S, Husche C, Luister A, Friedrichs S, Miller CM, McCarthy FO, Plat J, Laufs U, Weingärtner O, Lütjohann D. Oxidation of sitosterol and transport of its 7-oxygenated products from different tissues in humans and ApoE knockout mice. J Steroid Biochem Mol Biol 2017; 169:145-151. [PMID: 27132025 DOI: 10.1016/j.jsbmb.2016.04.011] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 04/18/2016] [Accepted: 04/20/2016] [Indexed: 12/24/2022]
Abstract
The most common phytosterols in the human diet are sitosterol and campesterol, which originate exclusively from plant derived food. These phytosterols are taken up by NPC1L1 transport from the intestine into the enterocytes together with cholesterol and other xenosterols. Phytosterols are selectively pumped back from the enterocytes into the intestinal lumen and on the liver site from hepatocytes into bile by heterodimeric ABCG5/G8 transporters. Like cholesterol, both phytosterols are prone to ring and side chain oxidation. It could be shown that oxyphytosterols, found in atherosclerotic tissue, are most likely of in situ oxidation (Schött et al.; Biochem. Biophys. Res. Commun. 2014 Apr 11;446(3):805-10). However, up to now, the entire mechanism of phytosterol oxidation is not clearly understood. Here, we provide further information about the oxidation of sitosterol and the transport of its oxidation products out of tissue. Our survey includes data of 104 severe aortic stenosis patients that underwent an elective aortic valve cusp replacement. We studied their phytosterol concentrations, as well as absolute and substrate corrected oxyphytosterol levels in plasma and valve cusp tissue. In addition, we also examined phytosterol and oxyphytosterol concentrations in plasma and tissues (from brain and liver) of 10 male ApoE knockout mice. The ratio of 7-oxygenated-sitosterol-to-sitosterol exceeds the ratio for 7-oxygenated-campesterol-to-campesterol in plasma and tissue of both humans and mice. This finding indicates that sitosterol is oxidized to a higher amount than campesterol and that a selective oxidative mechanism might exist which can differentiate between certain phytosterols. Secondly, the concentrations of oxyphytosterols found in plasma and tissue support the idea that oxysitosterols are preferably transported out of individual tissues. Selective oxidation of sitosterol and preferred transport of sitosterol oxidation products out of tissue seem to be a metabolic pathway of forced sitosterol clearance from tissue compartments.
Collapse
Affiliation(s)
- Hans-Frieder Schött
- Institute for Clinical Chemistry and Clinical Pharmacology, University Clinics Bonn, Germany; Department of Human Biology, Maastricht University, Maastricht, The Netherlands
| | - Sabine Baumgartner
- Department of Human Biology, Maastricht University, Maastricht, The Netherlands
| | - Constanze Husche
- Institute for Clinical Chemistry and Clinical Pharmacology, University Clinics Bonn, Germany
| | - Alexandra Luister
- Klinik für Innere Medizin III, Kardiologie, Angiologie und Internistische Intensivmedizin, Universitätsklinikum des Saarlandes, Homburg/Saar, Germany
| | - Silvia Friedrichs
- Institute for Clinical Chemistry and Clinical Pharmacology, University Clinics Bonn, Germany
| | - Charlotte M Miller
- Department of Chemistry and Analytical and Biological Chemistry Research Facility, University College Cork, Cork, Ireland
| | - Florence O McCarthy
- Department of Chemistry and Analytical and Biological Chemistry Research Facility, University College Cork, Cork, Ireland
| | - Jogchum Plat
- Department of Human Biology, Maastricht University, Maastricht, The Netherlands
| | - Ulrich Laufs
- Klinik für Innere Medizin III, Kardiologie, Angiologie und Internistische Intensivmedizin, Universitätsklinikum des Saarlandes, Homburg/Saar, Germany
| | - Oliver Weingärtner
- Klinik für Innere Medizin III, Kardiologie, Angiologie und Internistische Intensivmedizin, Universitätsklinikum des Saarlandes, Homburg/Saar, Germany; Department of Cardiology, Klinikum Oldenburg, Carl von Ossietzky University, European Medical School Oldenburg-Groningen, Oldenburg, Germany
| | - Dieter Lütjohann
- Institute for Clinical Chemistry and Clinical Pharmacology, University Clinics Bonn, Germany.
| |
Collapse
|
22
|
Nghiem-Rao TH, Patel SB. Investigating Sitosterolemia to Understand Lipid Physiology. ACTA ACUST UNITED AC 2017; 8:649-658. [PMID: 29928317 DOI: 10.2217/clp.13.60] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The cholesterol molecule is at the center of the pathophysiology of many vascular diseases. Whole-body cholesterol pools are maintained by a balance of endogenous synthesis, dietary absorption and elimination from our bodies. While the cellular aspects of cholesterol metabolism received significant impetus from the seminal work of Goldstein and Brown investigating LDL receptor trafficking, how dietary cholesterol was absorbed and eliminated was relatively neglected. The identification of the molecular defect a rare human disorder, Sitosterolemia, led to elucidation of a key mechanism of how we regulate the excretory pathway in the liver and in the intestine. Two proteins, ABCG5 and ABCG8, constitute a heterodimeric transporter that facilitates the extrusion of sterols from the cell into the biliary lumen, with a preference for xenosterols. This mechanism explained how dietary xenosterols are prevented from accumulating in our bodies. In addition, this disease has also highlighted the potential harm of xenosterols; macrothrombocytopenia, liver disease and endocrine disruption are seen when xenosterols accumulate. Mouse models of this disease suggest that there are more dramatic alterations of physiology, suggesting that these highly conserved mechanisms have evolved to prevent these xenosterols from accumulating in our bodies.
Collapse
Affiliation(s)
| | - Shailendra B Patel
- Medical College of Wisconsin, and the Clement J. Zablocki Veterans Affairs Medical Center, Milwaukee, WI
| |
Collapse
|
23
|
Guo D, Zhang X, Li Q, Qian L, Xu J, Lu M, Hu X, Zhu M, Chang CCY, Song B, Chang T, Xiong Y, Li B. The ACAT2 expression of human leukocytes is responsible for the excretion of lipoproteins containing cholesteryl/steryl esters. Acta Biochim Biophys Sin (Shanghai) 2016; 48:990-997. [PMID: 27688150 PMCID: PMC5091290 DOI: 10.1093/abbs/gmw095] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Revised: 08/24/2016] [Accepted: 06/30/2016] [Indexed: 12/19/2022] Open
Abstract
Acyl-coenzymeA:cholesterol acyltransferase 2 (ACAT2) is abundantly expressed in intestine and fetal liver of healthy human. Our previous studies have shown that in monocytic cells the low-level expression of human ACAT2 gene with specific CpG-hypomethylated promoter is regulated by the CCAAT/enhancer binding protein (C/EBP) transcription factors. In this study, we further report that the ACAT2 gene expression is attributable to the C/EBPs in the human leukocytes and correlated with the excretion of fluorescent lipoproteins containing the ACAT2-catalyzed NBD22-steryl esters. Moreover, this lipoprotein excretion can be inhibited by the ACAT2 isoform-selective inhibitor pyripyropene A (PPPA) in a dose-dependent manner, and employed to determine the half maximum inhibitory concentration (IC50) values of PPPA. Significantly, it is found that the differentiation-inducing factor all-trans retinoic acid, but not the proinflammatory cytokine tumor necrosis factor-α, enhances this ACAT2-dependent lipoprotein excretion. These data demonstrate that the ACAT2 expression of human leukocytes is responsible for the excretion of lipoproteins containing cholesteryl/steryl esters (CE/SE), and suggest that the excretion of lipoproteins containing the ACAT2-catalyzed CS/SE may avoid cytotoxicity through decreasing the excess intracellular cholesterols/sterols (especially various oxysterols), which is essential for the action of the human leukocytes.
Collapse
Affiliation(s)
- Dongqing Guo
- Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Xiaowei Zhang
- Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Qin Li
- Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Lei Qian
- Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Jiajia Xu
- Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Ming Lu
- Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Xihan Hu
- Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Ming Zhu
- Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Catherine C Y Chang
- Department of Biochemistry, Geisel School of Medicine at Dartmouth, Hanover NH 03755, USA
| | - Baoliang Song
- Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
- College of Life Sciences, The Institute for Advanced Studies, Wuhan University, Wuhan 430072, China
| | - Tayuan Chang
- Department of Biochemistry, Geisel School of Medicine at Dartmouth, Hanover NH 03755, USA
| | - Ying Xiong
- Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Boliang Li
- Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| |
Collapse
|
24
|
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
|
25
|
Yoo EG. Sitosterolemia: a review and update of pathophysiology, clinical spectrum, diagnosis, and management. Ann Pediatr Endocrinol Metab 2016; 21:7-14. [PMID: 27104173 PMCID: PMC4835564 DOI: 10.6065/apem.2016.21.1.7] [Citation(s) in RCA: 102] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 03/15/2016] [Indexed: 12/28/2022] Open
Abstract
Sitosterolemia is an autosomal recessive disorder characterized by increased plant sterol levels, xanthomas, and accelerated atherosclerosis. Although it was originally reported in patients with normolipemic xanthomas, severe hypercholesterolemia have been reported in patients with sitosterolemia, especially in children. Sitosterolemia is caused by increased intestinal absorption and decreased biliary excretion of sterols resulting from biallelic mutations in either ABCG5 or ABCG8, which encode the sterol efflux transporter ABCG5 and ABCG8. Patients with sitosterolemia show extreme phenotypic heterogeneity, ranging from almost asymptomatic individuals to those with severe hypercholesterolemia leading to accelerated atherosclerosis and premature cardiac death. Hematologic manifestations include hemolytic anemia with stomatocytosis, macrothrombocytopenia, splenomegaly, and abnormal bleeding. The mainstay of therapy includes dietary restriction of both cholesterol and plant sterols and the sterol absorption inhibitor, ezetimibe. Foods rich in plant sterols include vegetable oils, wheat germs, nuts, seeds, avocado, shortening, margarine and chocolate. Hypercholesterolemia in patients with sitosterolemia is dramatically responsive to low cholesterol diet and bile acid sequestrants. Plant sterol assay should be performed in patients with normocholesterolemic xanthomas, hypercholesterolemia with unexpectedly good response to dietary modifications or to cholesterol absorption inhibitors, or hypercholesterolemia with poor response to statins, or those with unexplained hemolytic anemia and macrothrombocytopenia. Because prognosis can be improved by proper management, it is important to find these patients out and diagnose correctly. This review article aimed to summarize recent publications on sitosterolemia, and to suggest clinical indications for plant sterol assay.
Collapse
Affiliation(s)
- Eun-Gyong Yoo
- Department of Pediatrics, CHA Bundang Medical Center, CHA University, Seongnam, Korea
| |
Collapse
|
26
|
Ceglarek U, Kresse K, Becker S, Fiedler GM, Thiery J, Quante M, Wieland R, Bartels M, Aust G. Circulating sterols as predictors of early allograft dysfunction and clinical outcome in patients undergoing liver transplantation. Metabolomics 2016; 12:182. [PMID: 27840599 PMCID: PMC5078158 DOI: 10.1007/s11306-016-1129-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Accepted: 10/15/2016] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Sensitive and specific assessment of the hepatic graft metabolism after liver transplantation (LTX) is essential for early detection of postoperative dysfunction implying the need for consecutive therapeutic interventions. OBJECTIVES Here, we assessed circulating liver metabolites of the cholesterol pathway, amino acids and acylcarnitines and evaluated their predictive value on early allograft dysfunction (EAD) and clinical outcome in the context of LTX. METHODS The metabolites were quantified in the plasma of 40 liver graft recipients one day pre- and 10 days post-LTX by liquid chromatography/tandem mass spectrometry (LC-MS/MS). Plant sterols as well as cholesterol and its precursors were determined in the free and esterified form; lanosterol in the free form only. Metabolites and esterification ratios were compared to the model for early allograft function scoring (MEAF) which is calculated at day 3 post-LTX from routine parameters defining EAD. RESULTS The hepatic esterification ratio of all sterols, but not amino acids and acylcarnitine concentrations, showed substantial metabolic disturbances post-LTX and correlated to the MEAF. In ROC analysis, the low esterification ratio of β-sitosterol and stigmasterol from day 1 and of the other sterols from day 3 were predictive for a high MEAF, i.e. EAD. Additionally, the ratio of esterified β-sitosterol and free lanosterol were predictive for all days and the esterification ratio of the other sterols at day 3 or 4 post-LTX for 3-month mortality. CONCLUSION Low ratios of circulating esterified sterols are associated with a high risk of EAD and impaired clinical outcome in the early postoperative phase following LTX.
Collapse
Affiliation(s)
- Uta Ceglarek
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, University of Leipzig, 04103 Leipzig, Germany
- LIFE–Leipzig Research Center for Civilization Diseases, University Leipzig, Philipp-Rosenthal-Strasse 27, 04103 Leipzig, Germany
| | - Kathleen Kresse
- Research Laboratories and Clinic of Visceral, Transplantation, Thoracic, and Vascular Surgery, University Hospital Leipzig, University of Leipzig, Liebigstraße 19, 04103 Leipzig, Germany
| | - Susen Becker
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, University of Leipzig, 04103 Leipzig, Germany
- LIFE–Leipzig Research Center for Civilization Diseases, University Leipzig, Philipp-Rosenthal-Strasse 27, 04103 Leipzig, Germany
| | - Georg Martin Fiedler
- Department of Laboratory Medicine, Institute of Clinical Chemistry, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Joachim Thiery
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, University of Leipzig, 04103 Leipzig, Germany
- LIFE–Leipzig Research Center for Civilization Diseases, University Leipzig, Philipp-Rosenthal-Strasse 27, 04103 Leipzig, Germany
| | - Markus Quante
- Research Laboratories and Clinic of Visceral, Transplantation, Thoracic, and Vascular Surgery, University Hospital Leipzig, University of Leipzig, Liebigstraße 19, 04103 Leipzig, Germany
| | - Robert Wieland
- Research Laboratories and Clinic of Visceral, Transplantation, Thoracic, and Vascular Surgery, University Hospital Leipzig, University of Leipzig, Liebigstraße 19, 04103 Leipzig, Germany
| | - Michael Bartels
- Research Laboratories and Clinic of Visceral, Transplantation, Thoracic, and Vascular Surgery, University Hospital Leipzig, University of Leipzig, Liebigstraße 19, 04103 Leipzig, Germany
| | - Gabriela Aust
- Research Laboratories and Clinic of Visceral, Transplantation, Thoracic, and Vascular Surgery, University Hospital Leipzig, University of Leipzig, Liebigstraße 19, 04103 Leipzig, Germany
| |
Collapse
|
27
|
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
|
28
|
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
|
29
|
Abstract
Phytosterolemia (sitosterolemia) is a rare autosomal recessive sterol storage disease caused by mutations in either of the adenosine triphosphate (ATP) binding cassette transporter genes; (ABC) G5 or ABCG8, leading to impaired elimination of plant sterols and stanols, with their increased accumulation in the blood and tissues. Thus the disease is characterized by substantially elevated serum plant sterols and stanols, with moderate to high plasma cholesterol levels, and increased risk of premature atherosclerosis. Hematologic abnormalities including macrothrombocytopenia, stomatocytosis and hemolysis are frequently observed in sitosterolemia patients. Currently, ezetimibe, a sterol absorption inhibitor, is used as the routine treatment for sitosterolemia, with reported improvement in plant sterol levels and hemolytic parameters. This review summarizes the research related to the health impact of plant sterols and stanols on sitosterolemia.
Collapse
Affiliation(s)
- Bridget O. Ajagbe
- University of Manitoba, Department of Human Nutritional Sciences, Winnipeg, MB, Canada, R3T 2N2, and University of Manitoba, Richardson Centre for Functional Foods and Nutraceuticals, Winnipeg, MB, Canada, R3T 6C5
| | - Rgia A. Othman
- University of Manitoba, Department of Human Nutritional Sciences, Winnipeg, MB, Canada, R3T 2N2, and University of Manitoba, Richardson Centre for Functional Foods and Nutraceuticals, Winnipeg, MB, Canada, R3T 6C5
| | - Semone B. Myrie
- University of Manitoba, Department of Human Nutritional Sciences, Winnipeg, MB, Canada, R3T 2N2, and University of Manitoba, Richardson Centre for Functional Foods and Nutraceuticals, Winnipeg, MB, Canada, R3T 6C5
| |
Collapse
|
30
|
ABCG5/G8 deficiency in mice reduces dietary triacylglycerol and cholesterol transport into the lymph. Lipids 2015; 50:371-9. [PMID: 25676339 DOI: 10.1007/s11745-015-3995-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Accepted: 02/01/2015] [Indexed: 10/24/2022]
Abstract
The adenosine triphosphate-binding cassette (ABC) transporter G5/G8 is critical in protecting the body from accumulating dietary plant sterols. Expressed in the liver and small intestine, it transports plant sterols into the biliary and intestinal lumens, thus promoting their excretion. The extent to which G5/G8 regulates cholesterol absorption remains unclear. G5/G8 is also implicated in reducing the absorption of dietary triacylglycerols (TAG) by unknown mechanisms. We hypothesized that G5/G8 suppresses the production of chylomicrons, and its deficiency would enhance the absorption of both dietary TAG and cholesterol. The aim of this study was to investigate the effects of G5/G8 deficiency on lipid uptake and secretion into the lymph under steady-state conditions. Surprisingly, compared with wild-type mice (WT) (n = 9), G5/G8 KO (n = 13) lymph fistula mice given a continuous intraduodenal infusion of [3H]-TAG and [14C]-cholesterol showed a significant (P < 0.05) reduction in lymphatic transport of both [(3)H]-TAG and [(14)C]-cholesterol, concomitant with a significant (P < 0.05) increase of [(3)H]-TAG and [(14)C]-cholesterol accumulated in the intestinal lumen. There was no difference in the total amount of radiolabeled lipids retained in the intestinal mucosa between the two groups. G5/G8 KO mice given a bolus of TAG showed reduced intestinal TAG secretion compared with WT, suggesting an independent role for G5/G8 in facilitating intestinal TAG transport. Our data demonstrate that G5/G8 deficiency reduces the uptake and secretion of both dietary TAG and cholesterol by the intestine, suggesting a novel role for the sterol transporter in the formation and secretion of chylomicrons.
Collapse
|
31
|
Liu Y, Lei L, Wang X, Ma KY, Li YM, Wang L, Man SW, Huang Y, Chen ZY. Plasma cholesterol-raising potency of dietary free cholesterol versus cholesteryl ester and effect of β-sitosterol. Food Chem 2015; 169:277-82. [DOI: 10.1016/j.foodchem.2014.07.123] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Revised: 07/24/2014] [Accepted: 07/25/2014] [Indexed: 11/30/2022]
|
32
|
Wang X, Huang W, Lei L, Liu Y, Ma KY, Li YM, Wang L, Huang Y, Chen ZY. Blockage of hydroxyl group partially abolishes the cholesterol-lowering activity of β-sitosterol. J Funct Foods 2015. [DOI: 10.1016/j.jff.2014.11.019] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
33
|
Liu Y, Guan L, Zhao Y, Lei L, Wang X, Ma KY, Li YM, Wang L, Man SW, Wang J, Huang Y, Chen ZY. Fatty acid moieties have little effect on cholesterol-lowering potency of plant sterol esters. EUR J LIPID SCI TECH 2014. [DOI: 10.1002/ejlt.201400444] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Yuwei Liu
- Food & Nutritional Sciences Programme, School of Life Sciences; The Chinese University of Hong Kong; Shatin NT, Hong Kong, P. R. China
| | - Lei Guan
- R&D; Nestle; Beijing P. R. China
| | | | - Lin Lei
- Food & Nutritional Sciences Programme, School of Life Sciences; The Chinese University of Hong Kong; Shatin NT, Hong Kong, P. R. China
| | - Xiaobo Wang
- Food & Nutritional Sciences Programme, School of Life Sciences; The Chinese University of Hong Kong; Shatin NT, Hong Kong, P. R. China
| | - Ka Ying Ma
- Food & Nutritional Sciences Programme, School of Life Sciences; The Chinese University of Hong Kong; Shatin NT, Hong Kong, P. R. China
| | - Yuk Man Li
- Food & Nutritional Sciences Programme, School of Life Sciences; The Chinese University of Hong Kong; Shatin NT, Hong Kong, P. R. China
| | - Lijun Wang
- Food & Nutritional Sciences Programme, School of Life Sciences; The Chinese University of Hong Kong; Shatin NT, Hong Kong, P. R. China
| | - Sun Wa Man
- Food & Nutritional Sciences Programme, School of Life Sciences; The Chinese University of Hong Kong; Shatin NT, Hong Kong, P. R. China
| | | | - Yu Huang
- School of Biomedical Sciences; Chinese University of Hong Kong; Shatin NT, Hong Kong, P. R. China
| | - Zhen-Yu Chen
- Food & Nutritional Sciences Programme, School of Life Sciences; The Chinese University of Hong Kong; Shatin NT, Hong Kong, P. R. China
| |
Collapse
|
34
|
Escolà-Gil JC, Quesada H, Julve J, Martín-Campos JM, Cedó L, Blanco-Vaca F. Sitosterolemia: Diagnosis, Investigation, and Management. Curr Atheroscler Rep 2014; 16:424. [DOI: 10.1007/s11883-014-0424-2] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
35
|
Chen G, Wang H, Zhang X, Yang ST. Nutraceuticals and Functional Foods in the Management of Hyperlipidemia. Crit Rev Food Sci Nutr 2014; 54:1180-201. [DOI: 10.1080/10408398.2011.629354] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
36
|
Othman RA, Myrie SB, Jones PJH. Non-cholesterol sterols and cholesterol metabolism in sitosterolemia. Atherosclerosis 2013; 231:291-9. [PMID: 24267242 DOI: 10.1016/j.atherosclerosis.2013.09.038] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2013] [Revised: 09/05/2013] [Accepted: 09/30/2013] [Indexed: 12/14/2022]
Abstract
Sitosterolemia (STSL) is a rare autosomal recessive disease, manifested by extremely elevated plant sterols (PS) in plasma and tissue, leading to xanthoma and premature atherosclerotic disease. Therapeutic approaches include limiting PS intake, interrupting enterohepatic circulation of bile acid using bile acid binding resins such as cholestyramine, and/or ileal bypass, and inhibiting intestinal sterol absorption by ezetimibe (EZE). The objective of this review is to evaluate sterol metabolism in STSL and the impact of the currently available treatments on sterol trafficking in this disease. The role of PS in initiation of xanthomas and premature atherosclerosis is also discussed. Blocking sterols absorption with EZE has revolutionized STSL patient treatment as it reduces circulating levels of non-cholesterol sterols in STSL. However, none of the available treatments including EZE have normalized plasma PS concentrations. Future studies are needed to: (i) explore where cholesterol and non-cholesterol sterols accumulate, (ii) assess to what extent these sterols in tissues can be mobilized after blocking their absorption, and (iii) define the factors governing sterol flux.
Collapse
Affiliation(s)
- Rgia A Othman
- Department of Human Nutritional Sciences, University of Manitoba, Winnipeg, Manitoba, Canada; Richardson Centre for Functional Foods and Nutraceuticals, University of Manitoba, Winnipeg, Manitoba, Canada
| | | | | |
Collapse
|
37
|
Gandhi H, Pal P, Giridhar R, Yadav MR. An HPTLC method for quantification of cholesteryl esters from human plasma and rat liver microsomes. Biomed Chromatogr 2013. [DOI: 10.1002/bmc.3075] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Hardik Gandhi
- Pharmacy Department, Faculty of Technology and Engineering; The M. S. University of Baroda; Vadodara 390 001 Gujarat India
| | - Palash Pal
- Pharmacy Department, Faculty of Technology and Engineering; The M. S. University of Baroda; Vadodara 390 001 Gujarat India
| | - Rajani Giridhar
- Pharmacy Department, Faculty of Technology and Engineering; The M. S. University of Baroda; Vadodara 390 001 Gujarat India
| | - Mange Ram Yadav
- Pharmacy Department, Faculty of Technology and Engineering; The M. S. University of Baroda; Vadodara 390 001 Gujarat India
| |
Collapse
|
38
|
Jahreis G, Wohlgemuth S, Grünz G, Martin L, Knieling M, Engel R, Türk M, Keller S. Dietary crystalline common-, micro-, nanoscale and emulsified nanoscale sitosterol reduce equally the cholesterol pool in guinea pigs, but varying nanosystems result in different sterol concentrations in serosal jejunum. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2013; 9:1027-35. [DOI: 10.1016/j.nano.2013.03.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2012] [Revised: 03/13/2013] [Accepted: 03/19/2013] [Indexed: 10/27/2022]
|
39
|
Chaput D, Kirouac LH, Bell-Temin H, Stevens SM, Padmanabhan J. SILAC-based proteomic analysis to investigate the impact of amyloid precursor protein expression in neuronal-like B103 cells. Electrophoresis 2013; 33:3728-37. [PMID: 23161580 DOI: 10.1002/elps.201200251] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2012] [Revised: 08/27/2012] [Accepted: 08/27/2012] [Indexed: 11/05/2022]
Abstract
Alzheimer's disease (AD) is the most prevalent form of dementia in the elderly. Amyloid plaque formation through aggregation of the amyloid beta peptide derived from amyloid precursor protein (APP) is considered one of the hallmark processes leading to AD pathology; however, the precise role of APP in plaque formation and AD pathogenesis is yet to be determined. Using stable isotope labeling by amino acids in cell culture (SILAC) and MS, protein expression profiles of APP null, rat neuronal-like B103 cells were compared to B103-695 cells that express the APP isoform, APP-695. A total of 2979 unique protein groups were identified among three biological replicates and significant protein expression changes were identified in a total of 102 nonredundant proteins. Some of the top biological functions associated with the differentially expressed proteins identified include cellular assembly, organization and morphology, cell cycle, lipid metabolism, protein folding, and PTMs. We report several novel biological pathways influenced by APP-695 expression in neuronal-like cells and provide additional framework for investigating altered molecular mechanisms associated with APP expression and processing and contribution to AD pathology.
Collapse
Affiliation(s)
- Dale Chaput
- Department of Cell Biology, Microbiology, and Molecular Biology, University of South Florida, Tampa, FL, USA
| | | | | | | | | |
Collapse
|
40
|
The role of the gut in reverse cholesterol transport--focus on the enterocyte. Prog Lipid Res 2013; 52:317-28. [PMID: 23608233 DOI: 10.1016/j.plipres.2013.04.003] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2013] [Revised: 03/01/2013] [Accepted: 04/10/2013] [Indexed: 11/20/2022]
Abstract
In the arterial intima, macrophages become cholesterol-enriched foam cells and atherosclerotic lesions are generated. This atherogenic process can be attenuated, prevented, or even reversed by HDL particles capable of initiating a multistep pathway known as the macrophage-specific reverse cholesterol transport. The macrophage-derived cholesterol released to HDL is taken up by the liver, secreted into the bile, and ultimately excreted in the feces. Importantly, the absorptive epithelial cells lining the lumen of the small intestine, the enterocytes, express several membrane-associated proteins which mediate the influx of luminal cholesterol and its subsequent efflux at their apical and basolateral sides. Moreover, generation of intestinal HDL and systemic effects of the gut microbiota recently revealed a direct link between the gut and the cholesterol cargo of peripheral macrophages. This review summarizes experimental evidence establishing that the reverse cholesterol transport pathway which initiates in macrophages is susceptible to modulation in the small intestine. We also describe four paths which govern cholesterol passage across the enterocyte and define a role for the gut in the regulation of reverse cholesterol transport. Understanding the concerted function of these paths may be useful when designing therapeutic strategies aimed at removing cholesterol from the foam cells which occupy atherosclerotic lesions.
Collapse
|
41
|
van der Wulp MYM, Verkade HJ, Groen AK. Regulation of cholesterol homeostasis. Mol Cell Endocrinol 2013; 368:1-16. [PMID: 22721653 DOI: 10.1016/j.mce.2012.06.007] [Citation(s) in RCA: 129] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2012] [Revised: 05/17/2012] [Accepted: 06/11/2012] [Indexed: 12/28/2022]
Abstract
Hypercholesterolemia is an important risk factor for cardiovascular disease. It is caused by a disturbed balance between cholesterol secretion into the blood versus uptake. The pathways involved are regulated via a complex interplay of enzymes, transport proteins, transcription factors and non-coding RNA's. The last two decades insight into underlying mechanisms has increased vastly but there are still a lot of unknowns, particularly regarding intracellular cholesterol transport. After decades of concentration on the liver, in recent years the intestine has come into focus as an important control point in cholesterol homeostasis. This review will discuss current knowledge of cholesterol physiology, with emphasis on cholesterol absorption, cholesterol synthesis and fecal excretion, and new (possible) therapeutic options for hypercholesterolemia.
Collapse
|
42
|
Alemany L, Laparra JM, Barberá R, Alegría A. Relative expression of cholesterol transport-related proteins and inflammation markers through the induction of 7-ketosterol-mediated stress in Caco-2 cells. Food Chem Toxicol 2013; 56:247-53. [PMID: 23454145 DOI: 10.1016/j.fct.2013.02.040] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2013] [Revised: 02/14/2013] [Accepted: 02/20/2013] [Indexed: 01/30/2023]
Abstract
Human diets contain sterol oxidation products that can induce cytotoxic effects, mainly caused by cholesterol oxides. However, phytosterol oxides effects have been less extensively investigated. This study evaluates the production of inflammatory biomarkers (IL-1β, IL-8, IL-10, TNFα) and the influence of gene expression transporters and enzymes related to cholesterol absorption and metabolism (NPC1L1, ABCG5/8, HMGCoA, ACAT) produced by 7-ketosterols (stigmasterol/cholesterol) in Caco-2 cells. These effects were linked to intracellular signaling pathways by using several inhibitors. Results showed 7-ketostigmasterol to have a greater proinflammatory potential than 7-ketocholesterol. In non-pre-treated cells, only efflux transporters were down-regulated by 7-ketosterols, showing a greater influence upon ABCG5 expression. Cell-pre-incubation with bradykinin induced changes in ABCG expression levels after 7-ketostigmasterol-incubation; however, the energetic metabolism inhibition reduced NPC1L1 expression only in 7-ketocholesterol-incubated cells. In non-pre-treated cells, HMG-CoA was up-regulated by both 7-ketosterols. However, exposure to inhibitors down-regulated the expression levels, mainly in 7-ketocholesterol-incubated cells. While ACAT expression values in non-pre-treated cells were unchanged, exposure to inhibitors caused down-regulation of mRNA levels. These results suggest that internalization and excretion of 7-ketostigmasterol is probably influenced by [Ca]i, which also could mediate HMGCoA activity in POPs metabolism. However, energetic metabolism and reducing equivalents exert different influences upon the 7-ketosterol internalization.
Collapse
Affiliation(s)
- L Alemany
- Nutrition and Food Chemistry, Faculty of Pharmacy, University of Valencia, Avda. Vicente Andrés Estellés s/n, 46100 Burjassot, Valencia, Spain
| | | | | | | |
Collapse
|
43
|
McDaniel AL, Alger HM, Sawyer JK, Kelley KL, Kock ND, Brown JM, Temel RE, Rudel LL. Phytosterol feeding causes toxicity in ABCG5/G8 knockout mice. THE AMERICAN JOURNAL OF PATHOLOGY 2013; 182:1131-8. [PMID: 23380580 DOI: 10.1016/j.ajpath.2012.12.014] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2012] [Revised: 12/12/2012] [Accepted: 12/17/2012] [Indexed: 10/27/2022]
Abstract
Plant sterols, or phytosterols, are very similar in structure to cholesterol and are abundant in typical diets. The reason for poor absorption of plant sterols by the body is still unknown. Mutations in the ABC transporters G5 and G8 are known to cause an accumulation of plant sterols in blood and tissues (sitosterolemia). To determine the significance of phytosterol exclusion from the body, we fed wild-type and ABCG5/G8 knockout mice a diet enriched with plant sterols. The high-phytosterol diet was extremely toxic to the ABCG5/G8 knockout mice but had no adverse effects on wild-type mice. ABCG5/G8 knockout mice died prematurely and developed a phenotype that included high levels of plant sterols in many tissues, liver abnormalities, and severe cardiac lesions. This study is the first to report such toxic effects of phytosterol accumulation in ABCG5/G8 knockout mice. We believe these new data support the conclusion that plant sterols are excluded from the body because they are toxic when present at high levels.
Collapse
Affiliation(s)
- Allison L McDaniel
- Section on Lipid Sciences, Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157, USA
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Nguyen TM, Sawyer JK, Kelley KL, Davis MA, Kent CR, Rudel LL. ACAT2 and ABCG5/G8 are both required for efficient cholesterol absorption in mice: evidence from thoracic lymph duct cannulation. J Lipid Res 2012; 53:1598-609. [PMID: 22669916 PMCID: PMC3540850 DOI: 10.1194/jlr.m026823] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2012] [Revised: 05/24/2012] [Indexed: 11/20/2022] Open
Abstract
The metabolic fate of newly absorbed cholesterol and phytosterol is orchestrated through adenosine triphosphate-binding cassette transporter G5 and G8 heterodimer (G5G8), and acyl CoA:cholesterol acyltransferase 2 (ACAT2). We hypothesized that intestinal G5G8 limits sterol absorption by reducing substrate availability for ACAT2 esterification and have attempted to define the roles of these two factors using gene deletion studies in mice. Male ACAT2(-/-), G5G8(-/-), ACAT2(-/-)G5G8(-/-) (DKO), and wild-type (WT) control mice were fed a diet with 20% of energy as palm oil and 0.2% (w/w) cholesterol. Sterol absorption efficiency was directly measured by monitoring the appearance of [(3)H]sitosterol and [(14)C]cholesterol tracers in lymph after thoracic lymph duct cannulation. The average percentage (± SEM) absorption of [(14)C]cholesterol after 8 h of lymph collection was 40.55 ± 0.76%, 19.41 ± 1.52%, 32.13 ± 1.60%, and 21.27 ± 1.35% for WT, ACAT2(-/-), G5G8(-/-), and DKO mice, respectively. [(3)H]sitosterol absorption was <2% in WT and ACAT2(-/-) mice, whereas it was up to 6.8% in G5G8(-/-) and DKO mice. G5G8(-/-) mice also produced chylomicrons with ∼70% less cholesterol ester mass than WT mice. In contrast to expectations, the data demonstrated that the absence of G5G8 led to decreased intestinal cholesterol esterification and reduced cholesterol transport efficiency. Intestinal G5G8 appeared to limit the absorption of phytosterols; ACAT2 more efficiently esterified cholesterol than phytosterols. The data indicate that handling of sterols by the intestine involves both G5G8 and ACAT2 but that an additional factor (possibly Niemann-Pick C1-like 1) may be key in determining absorption efficiency.
Collapse
Affiliation(s)
- Tam M. Nguyen
- Department of Pathology, Wake Forest School of Medicine, Winston-Salem, NC 27157
| | - Janet K. Sawyer
- Department of Pathology, Wake Forest School of Medicine, Winston-Salem, NC 27157
| | - Kathryn L. Kelley
- Department of Pathology, Wake Forest School of Medicine, Winston-Salem, NC 27157
| | - Matthew A. Davis
- Department of Pathology, Wake Forest School of Medicine, Winston-Salem, NC 27157
| | - Carol R. Kent
- Department of Pathology, Wake Forest School of Medicine, Winston-Salem, NC 27157
| | - Lawrence L. Rudel
- Department of Pathology, Wake Forest School of Medicine, Winston-Salem, NC 27157
| |
Collapse
|
45
|
Brauner R, Johannes C, Ploessl F, Bracher F, Lorenz RL. Phytosterols reduce cholesterol absorption by inhibition of 27-hydroxycholesterol generation, liver X receptor α activation, and expression of the basolateral sterol exporter ATP-binding cassette A1 in Caco-2 enterocytes. J Nutr 2012; 142:981-9. [PMID: 22535758 DOI: 10.3945/jn.111.157198] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Phytosterol-enriched foods are increasingly marketed to lower cholesterol levels and atherosclerosis in the general population. Phytosterols reduce cholesterol absorption, but the molecular mechanism is controversial. We therefore investigated the phytosterol effects on cholesterol metabolism in human enterocyte, hepatocyte, and macrophage models relevant for sterol absorption, reverse transport, and excretion. Isomolar sitosterol (50 μmol/L) was less effectively taken up by enterocytes than cholesterol but suppressed apical cholesterol uptake by 50% (P < 0.01) and basolateral secretion by two-thirds (P < 0.01) whether added in micelles or ethanol or complexed to cyclodextrin. In contrast, enterocytes handled nanomolar (3)H-sitosterol similarly to cholesterol. Enterocytes selectively oxidized all sterols to 27-hydroxy- and 27-carboxy-sterols. Conversion rates were much lower for sitosterol (0.05 ± 0.02 nmol/mg protein) and campesterol (0.48 ± 0.10) compared with cholesterol (3.73 ± 0.60) (P < 0.001). 27-Hydroxycholesterol (27OH-C) activated liver-X-receptor alpha (LXRα) (P < 0.01) and stimulated ATP-binding cassette transporter (ABC) A1 expression (P < 0.001) and basolateral systemic cholesterol secretion from enterocytes (P < 0.05). In co-incubations, phytosterols inhibited 27OH-C generation by sterol 27-hydroxylase (P < 0.001) and reduced LXRα-mediated ABCA1 expression (P < 0.01) and basolateral systemic cholesterol secretion. In contrast, ABCG8 transcription and apical sterol resecretion was unchanged by LXRα activation in human enterocytes. Exogenous LXRα agonists reverted sterol selectivity and phytosterol cholesterol interaction. Due to constitutive apical expression of ABCG5/G8 and LXRα-enhanced basolateral expression of ABCA1 in enterocytes, interference of phytosterols with the generation of the dominating LXRα-agonist 27OH-C blocks the self-priming component of cholesterol absorption. This local LXRα antagonism of dietary phytosterols contributes to sterol selectivity and reduces fractional cholesterol absorption and preloading of nascent HDL with dietary cholesterol.
Collapse
|
46
|
Vanmierlo T, Weingärtner O, van der Pol S, Husche C, Kerksiek A, Friedrichs S, Sijbrands E, Steinbusch H, Grimm M, Hartmann T, Laufs U, Böhm M, de Vries HE, Mulder M, Lütjohann D. Dietary intake of plant sterols stably increases plant sterol levels in the murine brain. J Lipid Res 2012; 53:726-35. [PMID: 22279184 DOI: 10.1194/jlr.m017244] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Plant sterols such as sitosterol and campesterol are frequently administered as cholesterol-lowering supplements in food. Recently, it has been shown in mice that, in contrast to the structurally related cholesterol, circulating plant sterols can enter the brain. We questioned whether the accumulation of plant sterols in murine brain is reversible. After being fed a plant sterol ester-enriched diet for 6 weeks, C57BL/6NCrl mice displayed significantly increased concentrations of plant sterols in serum, liver, and brain by 2- to 3-fold. Blocking intestinal sterol uptake for the next 6 months while feeding the mice with a plant stanol ester-enriched diet resulted in strongly decreased plant sterol levels in serum and liver, without affecting brain plant sterol levels. Relative to plasma concentrations, brain levels of campesterol were higher than sitosterol, suggesting that campesterol traverses the blood-brain barrier more efficiently. In vitro experiments with brain endothelial cell cultures showed that campesterol crossed the blood-brain barrier more efficiently than sitosterol. We conclude that, over a 6-month period, plant sterol accumulation in murine brain is virtually irreversible.
Collapse
Affiliation(s)
- Tim Vanmierlo
- Institute of Clinical Chemistry and Clinical Pharmacology, University of Bonn, Bonn, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Nguyen TM, Sawyer JK, Kelley KL, Davis MA, Rudel LL. Cholesterol esterification by ACAT2 is essential for efficient intestinal cholesterol absorption: evidence from thoracic lymph duct cannulation. J Lipid Res 2012; 53:95-104. [PMID: 22045928 PMCID: PMC3243485 DOI: 10.1194/jlr.m018820] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2011] [Revised: 10/11/2011] [Indexed: 01/21/2023] Open
Abstract
The hypothesis tested in this study was that cholesterol esterification by ACAT2 would increase cholesterol absorption efficiency by providing cholesteryl ester (CE) for incorporation into chylomicrons. The assumption was that absorption would be proportional to Acat2 gene dosage. Male ACAT2⁺/⁺, ACAT2⁺/⁻, and ACAT2⁻/⁻ mice were fed a diet containing 20% of energy as palm oil with 0.2% (w/w) cholesterol. Cholesterol absorption efficiency was measured by fecal dual-isotope and thoracic lymph duct cannulation (TLDC) methods using [³H]sitosterol and [¹⁴C]cholesterol tracers. Excellent agreement among individual mice was found for cholesterol absorption measured by both techniques. Cholesterol absorption efficiency in ACAT2⁻/⁻ mice was 16% compared with 46-47% in ACAT2⁺/⁺ and ACAT2⁺/⁻ mice. Chylomicrons from ACAT2⁺/⁺ and ACAT2⁺/⁻ mice carried ∼80% of total sterol mass as CE, whereas ACAT2⁻/⁻ chylomicrons carried >90% of sterol mass in the unesterified form. The total percentage of chylomicron mass as CE was reduced from 12% in the presence of ACAT2 to ∼1% in ACAT2⁻/⁻ mice. Altogether, the data demonstrate that ACAT2 increases cholesterol absorption efficiency by providing CE for chylomicron transport, but one copy of the Acat2 gene, providing ∼50% of ACAT2 mRNA and enzyme activity, was as effective as two copies in promoting cholesterol absorption.
Collapse
Affiliation(s)
| | - Janet K. Sawyer
- Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, NC
| | - Kathryn L. Kelley
- Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, NC
| | - Matthew A. Davis
- Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, NC
| | - Lawrence L. Rudel
- Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, NC
| |
Collapse
|
48
|
Cheng Y, Tang K, Wu S, Liu L, Qiang C, Lin X, Liu B. Astragalus polysaccharides lowers plasma cholesterol through mechanisms distinct from statins. PLoS One 2011; 6:e27437. [PMID: 22110652 PMCID: PMC3217967 DOI: 10.1371/journal.pone.0027437] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2011] [Accepted: 10/17/2011] [Indexed: 11/18/2022] Open
Abstract
To determine the efficacy and underlying mechanism of Astragalus polysaccharides (APS) on plasma lipids in hypercholesterolemia hamsters. The effect of APS (0.25g/kg/d) on plasma and liver lipids, fecal bile acids and neutral sterol, cholesterol absorption and synthesis, HMG-CoA reductase activity, and gene and protein expressions in the liver and small intestine was investigated in twenty-four hypercholesterolemia hamsters. Treatment periods lasted for three months. APS significantly lowered plasma total cholesterol by 45.8%, triglycerides by 30%, and low-density lipoprotein-cholesterol by 47.4%, comparable to simvastatin. Further examinations revealed that APS reduced total cholesterol and triglycerides in the liver, increased fecal bile acid and neutral sterol excretion, inhibited cholesterol absorption, and by contrast, increased hepatic cholesterol synthesis and HMG-CoA reductase activity. Plasma total cholesterol or low-density lipoprotein-cholesterol levels were significantly correlated with cholesterol absorption rates. APS up-regulated cholesterol-7α-hydroxylase and LDL-receptor gene expressions. These new findings identify APS as a potential natural cholesterol lowering agent, working through mechanisms distinct from statins.
Collapse
Affiliation(s)
- Yunjiu Cheng
- Division of Cardiology, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Kai Tang
- Division of Cardiology, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Suhua Wu
- Division of Cardiology, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- * E-mail:
| | - Lijuan Liu
- Division of Cardiology, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Cancan Qiang
- Division of Cardiology, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Xiaoxiong Lin
- Division of Cardiology, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Bingqing Liu
- Division of Cardiology, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
49
|
Lupattelli G, De Vuono S, Mannarino E. Patterns of cholesterol metabolism: pathophysiological and therapeutic implications for dyslipidemias and the metabolic syndrome. Nutr Metab Cardiovasc Dis 2011; 21:620-627. [PMID: 21855307 DOI: 10.1016/j.numecd.2011.04.010] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2011] [Revised: 04/08/2011] [Accepted: 04/27/2011] [Indexed: 11/17/2022]
Abstract
Investigating cholesterol metabolism, which derives from balancing cholesterol synthesis and absorption, opens new perspectives in the pathogenesis of dyslipidemias and the metabolic syndrome (MS). Cholesterol metabolism is studied by measuring plasma levels of campesterol, sitosterol and cholestanol, that is, plant sterols which are recognised as surrogate cholesterol-absorption markers and lathosterol or squalene, that is, cholesterol precursors, which are considered surrogate cholesterol-synthesis markers. This article presents current knowledge on cholesterol synthesis and absorption, as evaluated by means of cholesterol precursors and plant sterols, and discusses patterns of cholesterol balance in the main forms of primary hyperlipidaemia and MS. Understanding the mechanism(s) underlying these patterns of cholesterol synthesis and absorption will help to predict the response to hypolipidemic treatment, which can then be tailored to ensure the maximum clinical benefit for patients.
Collapse
Affiliation(s)
- G Lupattelli
- Internal Medicine, Angiology and Atherosclerosis, Department of Clinical and Experimental Medicine, University of Perugia, Italy.
| | | | | |
Collapse
|
50
|
Brufau G, Kuipers F, Lin Y, Trautwein EA, Groen AK. A reappraisal of the mechanism by which plant sterols promote neutral sterol loss in mice. PLoS One 2011; 6:e21576. [PMID: 21738715 PMCID: PMC3128081 DOI: 10.1371/journal.pone.0021576] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2011] [Accepted: 06/03/2011] [Indexed: 11/23/2022] Open
Abstract
Dietary plant sterols (PS) reduce serum total and LDL-cholesterol in hyperlipidemic animal models and in humans. This hypocholesterolemic effect is generally ascribed to inhibition of cholesterol absorption. However, whether this effect fully explains the reported strong induction of neutral sterol excretion upon plant sterol feeding is not known. Recent data demonstrate that the intestine directly mediates plasma cholesterol excretion into feces, i.e., without involvement of the hepato-biliary route.
Collapse
Affiliation(s)
- Gemma Brufau
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.
| | | | | | | | | |
Collapse
|