1
|
Liu Y, Ding F, Deng L, Zhang S, Wu L, Tong H. Discovery of selective ACAT2 antagonist via a combination strategy based on deep docking, pharmacophore modelling, and molecular dynamics simulation. J Enzyme Inhib Med Chem 2024; 39:2403736. [PMID: 39316789 PMCID: PMC11423527 DOI: 10.1080/14756366.2024.2403736] [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: 03/28/2024] [Revised: 08/18/2024] [Accepted: 08/29/2024] [Indexed: 09/26/2024] Open
Abstract
Acyl-CoA: cholesterol acyltransferase (ACAT), a pivotal enzyme in the absorption and metabolism of cholesterol, is primarily responsible for intracellular esterification. ACAT inhibition is expected to diminish plasma lipid levels by impeding intestinal cholesterol absorption, thereby preventing the progression of atherosclerotic lesions. A previous study shows that selective inhibition of ACAT2 significantly mitigated hypercholesterolaemia and atherosclerosis in mouse models. Therefore, the need for ACAT2 selective inhibitors becomes particularly urgent. In this study, we established a multilayer virtual screening workflow and subjected biologically evaluated representative compounds to enzyme inhibitory assays. The experimental results indicated that the two compounds, STL565001 (inhibition rate at 25 μM: 75.7 ± 27.8%, selectivity = 6) and STL528213 (inhibition rate at 25 μM: 87.8 ± 12.4%, selectivity = 13), demonstrated robust activity against ACAT2, displaying greater selectivity for ACAT2 than for ACAT1. The molecular mechanisms governing the inhibitory activities of the selected compounds were systematically elucidated using computational approaches. In addition, hotspot residues in ACAT2 that are crucial for ligand binding were successfully identified. In summary, we devised a multilayer screening scheme to expeditiously and efficiently identify compounds with enzyme inhibitory activity, offering novel scaffolds for subsequent drug design centred on ACAT2 targets.
Collapse
Affiliation(s)
- Yanfeng Liu
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
- Nanjing Lishui District Hospital of Traditional Chinese Medicine, Nanjing, China
- Warshel Institute for Computational Biology, School of Medicine, The Chinese University of Hong Kong, Shenzhen, China
| | - Feng Ding
- School of Chinese Medicine, Hong Kong Baptist University, Kowloon, Hong Kong SAR, China
| | - Liangying Deng
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Shuran Zhang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Lixing Wu
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
- Nanjing Lishui District Hospital of Traditional Chinese Medicine, Nanjing, China
| | - Huangjin Tong
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
2
|
Tu T, Zhang H, Xu H. Targeting sterol-O-acyltransferase 1 to disrupt cholesterol metabolism for cancer therapy. Front Oncol 2023; 13:1197502. [PMID: 37409263 PMCID: PMC10318190 DOI: 10.3389/fonc.2023.1197502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 06/05/2023] [Indexed: 07/07/2023] Open
Abstract
Cholesterol esterification is often dysregulated in cancer. Sterol O-acyl-transferase 1 (SOAT1) plays an important role in maintaining cellular cholesterol homeostasis by catalyzing the formation of cholesterol esters from cholesterol and long-chain fatty acids in cells. Many studies have implicated that SOAT1 plays a vital role in cancer initiation and progression and is an attractive target for novel anticancer therapy. In this review, we provide an overview of the mechanism and regulation of SOAT1 in cancer and summarize the updates of anticancer therapy targeting SOAT1.
Collapse
Affiliation(s)
- Teng Tu
- Department of Medical Oncology, Cancer Center and Laboratory of Molecular Targeted Therapy in Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - Hongying Zhang
- Laboratory of Oncogene, West China Hospital, Sichuan University, Chengdu, China
| | - Huanji Xu
- Department of Medical Oncology, Cancer Center and Laboratory of Molecular Targeted Therapy in Oncology, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
3
|
Bhattacharjee P, Rutland N, Iyer MR. Targeting Sterol O-Acyltransferase/Acyl-CoA:Cholesterol Acyltransferase (ACAT): A Perspective on Small-Molecule Inhibitors and Their Therapeutic Potential. J Med Chem 2022; 65:16062-16098. [PMID: 36473091 DOI: 10.1021/acs.jmedchem.2c01265] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Sterol O-acyltransferase (SOAT) is a membrane-bound enzyme that aids the esterification of cholesterol and fatty acids to cholesterol esters. SOAT has been studied extensively as a potential drug target, since its inhibition can serve as an alternative to statin therapy. Two SOAT isozymes that have discrete functions in the human body, namely, SOAT1 and SOAT2, have been characterized. Over three decades of research has focused on candidate SOAT1 inhibitors with unsatisfactory results in clinical trials. Recent research has focused on targeting SOAT2 selectively. In this perspective, we summarize the literature covering various SOAT inhibitory agents and discuss the design, structural requirements, and mode of action of SOAT inhibitors.
Collapse
Affiliation(s)
- Pinaki Bhattacharjee
- Section on Medicinal Chemistry, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, 5625 Fishers Lane, Rockville, Maryland 20852, United States
| | - Nicholas Rutland
- Section on Medicinal Chemistry, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, 5625 Fishers Lane, Rockville, Maryland 20852, United States
| | - Malliga R Iyer
- Section on Medicinal Chemistry, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, 5625 Fishers Lane, Rockville, Maryland 20852, United States
| |
Collapse
|
4
|
Amengual J, Ogando Y, Nikain C, Quezada A, Qian K, Vaisar T, Fisher EA. Short-Term Acyl-CoA:Cholesterol Acyltransferase Inhibition, Combined with Apoprotein A1 Overexpression, Promotes Atherosclerosis Inflammation Resolution in Mice. Mol Pharmacol 2020; 99:175-183. [PMID: 33384285 DOI: 10.1124/molpharm.120.000108] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 12/15/2020] [Indexed: 12/21/2022] Open
Abstract
Acyl-CoA:cholesterol acyltransferase (ACAT) mediates cellular cholesterol esterification. In atherosclerotic plaque macrophages, ACAT promotes cholesteryl ester accumulation, resulting in foam cell formation and atherosclerosis progression. Its complete inactivation in mice, however, showed toxic effects because of an excess of free cholesterol (FC) in macrophages, which can cause endoplasmic reticulum stress, cholesterol crystal formation, and inflammasome activation. Our previous studies showed that long-term partial ACAT inhibition, achieved by dietary supplementation with Fujirebio F1394, delays atherosclerosis progression in apoprotein E-deficient (Apoe -/-) mice by reducing plaque foam cell formation without inflammatory or toxic effects. Here, we determined whether short-term partial inhibition of ACAT, in combination with an enhanced systemic FC acceptor capacity, has synergistic benefits. Thus, we crossbred Apoe -/- with human apoprotein A1-transgenic (APOA1 tg/tg) mice, which have elevated cholesterol-effluxing high-density lipoprotein particles, and subjected Apoe -/- and APOA1 tg/tg/Apoe -/- mice to an atherogenic diet to develop advanced plaques. Then mice were either euthanized (baseline) or fed purified standard diet with or without F1394 for 4 more weeks. Plaques of APOA1 tg/tg/Apoe -/- mice fed F1394 showed a 60% reduction of macrophages accompanied by multiple other benefits, such as reduced inflammation and favorable changes in extracellular composition, in comparison with Apoe -/- baseline mice. In addition, there was no accumulation of cholesterol crystals or signs of toxicity. Overall, these results show that short-term partial ACAT inhibition, coupled to increased cholesterol efflux capacity, favorably remodels atherosclerosis lesions, supporting the potential of these combined therapies in the treatment of advanced atherosclerosis. SIGNIFICANCE STATEMENT: Short-term pharmacological inhibition of acyl-CoA:cholesterol acyltransferase-mediated cholesterol esterification, in combination with increased free cholesterol efflux acceptors, has positive effects in mice by 1) reducing the inflammatory state of the plaque macrophages and 2) favoring compositional changes associated with plaque stabilization. These effects occur without toxicity, showing the potential of these combined therapies in the treatment of advanced atherosclerosis.
Collapse
Affiliation(s)
- Jaume Amengual
- Leon H. Charney Division of Cardiology, Department of Medicine, Cardiovascular Research Center, New York University Grossman School of Medicine, New York, New York (J.A., Y.O, C.N., A.Q., K.Q., E.A.F); Department of Food Science and Human Nutrition, University of Illinois Urbana Champaign, Champaign, Illinois (.J.A.); Division of Metabolism, Endocrinology and Nutrition, University of Washington, Seattle, Washington (T.V.); and Division of Biostatistics, Department of Population Health, New York University Grossman School of Medicine, New York, New York (K.Q.)
| | - Yoscar Ogando
- Leon H. Charney Division of Cardiology, Department of Medicine, Cardiovascular Research Center, New York University Grossman School of Medicine, New York, New York (J.A., Y.O, C.N., A.Q., K.Q., E.A.F); Department of Food Science and Human Nutrition, University of Illinois Urbana Champaign, Champaign, Illinois (.J.A.); Division of Metabolism, Endocrinology and Nutrition, University of Washington, Seattle, Washington (T.V.); and Division of Biostatistics, Department of Population Health, New York University Grossman School of Medicine, New York, New York (K.Q.)
| | - Cyrus Nikain
- Leon H. Charney Division of Cardiology, Department of Medicine, Cardiovascular Research Center, New York University Grossman School of Medicine, New York, New York (J.A., Y.O, C.N., A.Q., K.Q., E.A.F); Department of Food Science and Human Nutrition, University of Illinois Urbana Champaign, Champaign, Illinois (.J.A.); Division of Metabolism, Endocrinology and Nutrition, University of Washington, Seattle, Washington (T.V.); and Division of Biostatistics, Department of Population Health, New York University Grossman School of Medicine, New York, New York (K.Q.)
| | - Alexandra Quezada
- Leon H. Charney Division of Cardiology, Department of Medicine, Cardiovascular Research Center, New York University Grossman School of Medicine, New York, New York (J.A., Y.O, C.N., A.Q., K.Q., E.A.F); Department of Food Science and Human Nutrition, University of Illinois Urbana Champaign, Champaign, Illinois (.J.A.); Division of Metabolism, Endocrinology and Nutrition, University of Washington, Seattle, Washington (T.V.); and Division of Biostatistics, Department of Population Health, New York University Grossman School of Medicine, New York, New York (K.Q.)
| | - Kun Qian
- Leon H. Charney Division of Cardiology, Department of Medicine, Cardiovascular Research Center, New York University Grossman School of Medicine, New York, New York (J.A., Y.O, C.N., A.Q., K.Q., E.A.F); Department of Food Science and Human Nutrition, University of Illinois Urbana Champaign, Champaign, Illinois (.J.A.); Division of Metabolism, Endocrinology and Nutrition, University of Washington, Seattle, Washington (T.V.); and Division of Biostatistics, Department of Population Health, New York University Grossman School of Medicine, New York, New York (K.Q.)
| | - Tomas Vaisar
- Leon H. Charney Division of Cardiology, Department of Medicine, Cardiovascular Research Center, New York University Grossman School of Medicine, New York, New York (J.A., Y.O, C.N., A.Q., K.Q., E.A.F); Department of Food Science and Human Nutrition, University of Illinois Urbana Champaign, Champaign, Illinois (.J.A.); Division of Metabolism, Endocrinology and Nutrition, University of Washington, Seattle, Washington (T.V.); and Division of Biostatistics, Department of Population Health, New York University Grossman School of Medicine, New York, New York (K.Q.)
| | - Edward A Fisher
- Leon H. Charney Division of Cardiology, Department of Medicine, Cardiovascular Research Center, New York University Grossman School of Medicine, New York, New York (J.A., Y.O, C.N., A.Q., K.Q., E.A.F); Department of Food Science and Human Nutrition, University of Illinois Urbana Champaign, Champaign, Illinois (.J.A.); Division of Metabolism, Endocrinology and Nutrition, University of Washington, Seattle, Washington (T.V.); and Division of Biostatistics, Department of Population Health, New York University Grossman School of Medicine, New York, New York (K.Q.)
| |
Collapse
|
5
|
Hill J, Shao X, Wright JS, Stauff J, Sherman PS, Arteaga J, Wong KK, Viglianti BL, Scott PJH, Brooks AF. Synthesis and Evaluation of 11C- and 18F-Labeled SOAT1 Inhibitors as Macrophage Foam Cell Imaging Agents. ACS Med Chem Lett 2020; 11:1299-1304. [PMID: 32551015 PMCID: PMC7294710 DOI: 10.1021/acsmedchemlett.0c00127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 04/30/2020] [Indexed: 11/30/2022] Open
Abstract
PD-132301, an inhibitor of sterol O-acyltransferase 1 (SOAT1; also known as acyl-coenzyme A:cholesterol acyltransferase-1, ACAT1), is under clinical investigation for numerous adrenal disorders. Radiolabeled SOAT1 inhibitors could support drug discovery and help diagnose SOAT1-related disorders, such as atherosclerosis. We synthesized two radiolabeled SOAT1 inhibitors, [11C]PD-132301 and fluorine analogue [18F]1. Rat biodistribution studies were conducted with both agents and, as the most selective tracer, [11C]PD-132301 was advanced to preclinical positron emission tomography studies in (atherosclerotic) ApoE-/- mice. The uptake of [11C]PD-132301 in SOAT1-rich tissue warrants further investigation into the compound as an atherosclerosis and adrenal imaging agent.
Collapse
Affiliation(s)
- James
R. Hill
- Institute
for Molecular Bioscience, The University
of Queensland, Brisbane, Queensland 4072, Australia
- Department
of Radiology, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Xia Shao
- Department
of Radiology, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Jay S. Wright
- Department
of Radiology, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Jenelle Stauff
- Department
of Radiology, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Phillip S. Sherman
- Department
of Radiology, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Janna Arteaga
- Department
of Radiology, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Ka Kit Wong
- Department
of Radiology, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Benjamin L. Viglianti
- Department
of Radiology, University of Michigan, Ann Arbor, Michigan 48109, United States
- Nuclear
Medicine Service, Veterans Administration, Ann Arbor, Michigan 48105, United States
| | - Peter J. H. Scott
- Department
of Radiology, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Allen F. Brooks
- Department
of Radiology, University of Michigan, Ann Arbor, Michigan 48109, United States
| |
Collapse
|
6
|
Wang D, Yang Y, Lei Y, Tzvetkov NT, Liu X, Yeung AWK, Xu S, Atanasov AG. Targeting Foam Cell Formation in Atherosclerosis: Therapeutic Potential of Natural Products. Pharmacol Rev 2019; 71:596-670. [PMID: 31554644 DOI: 10.1124/pr.118.017178] [Citation(s) in RCA: 135] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Foam cell formation and further accumulation in the subendothelial space of the vascular wall is a hallmark of atherosclerotic lesions. Targeting foam cell formation in the atherosclerotic lesions can be a promising approach to treat and prevent atherosclerosis. The formation of foam cells is determined by the balanced effects of three major interrelated biologic processes, including lipid uptake, cholesterol esterification, and cholesterol efflux. Natural products are a promising source for new lead structures. Multiple natural products and pharmaceutical agents can inhibit foam cell formation and thus exhibit antiatherosclerotic capacity by suppressing lipid uptake, cholesterol esterification, and/or promoting cholesterol ester hydrolysis and cholesterol efflux. This review summarizes recent findings on these three biologic processes and natural products with demonstrated potential to target such processes. Discussed also are potential future directions for studying the mechanisms of foam cell formation and the development of foam cell-targeted therapeutic strategies.
Collapse
Affiliation(s)
- Dongdong Wang
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China (D.W., X.L.); Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzębiec, Poland (D.W., Y.Y., Y.L., A.G.A.); Department of Pharmacognosy, University of Vienna, Vienna, Austria (A.G.A.); Institute of Clinical Chemistry, University Hospital Zurich, Schlieren, Switzerland (D.W.); Institute of Molecular Biology "Roumen Tsanev," Department of Biochemical Pharmacology and Drug Design, Bulgarian Academy of Sciences, Sofia, Bulgaria (N.T.T.); Pharmaceutical Institute, University of Bonn, Bonn, Germany (N.T.T.); Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, Rochester, New York (S.X.); Oral and Maxillofacial Radiology, Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China (A.W.K.Y.); and Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia, Bulgaria (A.G.A.)
| | - Yang Yang
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China (D.W., X.L.); Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzębiec, Poland (D.W., Y.Y., Y.L., A.G.A.); Department of Pharmacognosy, University of Vienna, Vienna, Austria (A.G.A.); Institute of Clinical Chemistry, University Hospital Zurich, Schlieren, Switzerland (D.W.); Institute of Molecular Biology "Roumen Tsanev," Department of Biochemical Pharmacology and Drug Design, Bulgarian Academy of Sciences, Sofia, Bulgaria (N.T.T.); Pharmaceutical Institute, University of Bonn, Bonn, Germany (N.T.T.); Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, Rochester, New York (S.X.); Oral and Maxillofacial Radiology, Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China (A.W.K.Y.); and Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia, Bulgaria (A.G.A.)
| | - Yingnan Lei
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China (D.W., X.L.); Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzębiec, Poland (D.W., Y.Y., Y.L., A.G.A.); Department of Pharmacognosy, University of Vienna, Vienna, Austria (A.G.A.); Institute of Clinical Chemistry, University Hospital Zurich, Schlieren, Switzerland (D.W.); Institute of Molecular Biology "Roumen Tsanev," Department of Biochemical Pharmacology and Drug Design, Bulgarian Academy of Sciences, Sofia, Bulgaria (N.T.T.); Pharmaceutical Institute, University of Bonn, Bonn, Germany (N.T.T.); Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, Rochester, New York (S.X.); Oral and Maxillofacial Radiology, Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China (A.W.K.Y.); and Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia, Bulgaria (A.G.A.)
| | - Nikolay T Tzvetkov
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China (D.W., X.L.); Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzębiec, Poland (D.W., Y.Y., Y.L., A.G.A.); Department of Pharmacognosy, University of Vienna, Vienna, Austria (A.G.A.); Institute of Clinical Chemistry, University Hospital Zurich, Schlieren, Switzerland (D.W.); Institute of Molecular Biology "Roumen Tsanev," Department of Biochemical Pharmacology and Drug Design, Bulgarian Academy of Sciences, Sofia, Bulgaria (N.T.T.); Pharmaceutical Institute, University of Bonn, Bonn, Germany (N.T.T.); Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, Rochester, New York (S.X.); Oral and Maxillofacial Radiology, Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China (A.W.K.Y.); and Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia, Bulgaria (A.G.A.)
| | - Xingde Liu
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China (D.W., X.L.); Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzębiec, Poland (D.W., Y.Y., Y.L., A.G.A.); Department of Pharmacognosy, University of Vienna, Vienna, Austria (A.G.A.); Institute of Clinical Chemistry, University Hospital Zurich, Schlieren, Switzerland (D.W.); Institute of Molecular Biology "Roumen Tsanev," Department of Biochemical Pharmacology and Drug Design, Bulgarian Academy of Sciences, Sofia, Bulgaria (N.T.T.); Pharmaceutical Institute, University of Bonn, Bonn, Germany (N.T.T.); Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, Rochester, New York (S.X.); Oral and Maxillofacial Radiology, Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China (A.W.K.Y.); and Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia, Bulgaria (A.G.A.)
| | - Andy Wai Kan Yeung
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China (D.W., X.L.); Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzębiec, Poland (D.W., Y.Y., Y.L., A.G.A.); Department of Pharmacognosy, University of Vienna, Vienna, Austria (A.G.A.); Institute of Clinical Chemistry, University Hospital Zurich, Schlieren, Switzerland (D.W.); Institute of Molecular Biology "Roumen Tsanev," Department of Biochemical Pharmacology and Drug Design, Bulgarian Academy of Sciences, Sofia, Bulgaria (N.T.T.); Pharmaceutical Institute, University of Bonn, Bonn, Germany (N.T.T.); Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, Rochester, New York (S.X.); Oral and Maxillofacial Radiology, Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China (A.W.K.Y.); and Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia, Bulgaria (A.G.A.)
| | - Suowen Xu
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China (D.W., X.L.); Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzębiec, Poland (D.W., Y.Y., Y.L., A.G.A.); Department of Pharmacognosy, University of Vienna, Vienna, Austria (A.G.A.); Institute of Clinical Chemistry, University Hospital Zurich, Schlieren, Switzerland (D.W.); Institute of Molecular Biology "Roumen Tsanev," Department of Biochemical Pharmacology and Drug Design, Bulgarian Academy of Sciences, Sofia, Bulgaria (N.T.T.); Pharmaceutical Institute, University of Bonn, Bonn, Germany (N.T.T.); Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, Rochester, New York (S.X.); Oral and Maxillofacial Radiology, Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China (A.W.K.Y.); and Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia, Bulgaria (A.G.A.)
| | - Atanas G Atanasov
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China (D.W., X.L.); Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzębiec, Poland (D.W., Y.Y., Y.L., A.G.A.); Department of Pharmacognosy, University of Vienna, Vienna, Austria (A.G.A.); Institute of Clinical Chemistry, University Hospital Zurich, Schlieren, Switzerland (D.W.); Institute of Molecular Biology "Roumen Tsanev," Department of Biochemical Pharmacology and Drug Design, Bulgarian Academy of Sciences, Sofia, Bulgaria (N.T.T.); Pharmaceutical Institute, University of Bonn, Bonn, Germany (N.T.T.); Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, Rochester, New York (S.X.); Oral and Maxillofacial Radiology, Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China (A.W.K.Y.); and Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia, Bulgaria (A.G.A.)
| |
Collapse
|
7
|
Liu Y, Zhang X, Zhan Y, Li B, Lu W, Nan F. Design and synthesis of further simplified pyripyropene A based ACAT2 selective inhibitors. Tetrahedron 2019. [DOI: 10.1016/j.tet.2019.01.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
8
|
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
|
9
|
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]
|
10
|
Qiao LS, Zhang XB, Jiang LD, Zhang YL, Li GY. Identification of potential ACAT-2 selective inhibitors using pharmacophore, SVM and SVR from Chinese herbs. Mol Divers 2016; 20:933-944. [PMID: 27329301 DOI: 10.1007/s11030-016-9684-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2016] [Accepted: 06/06/2016] [Indexed: 12/13/2022]
Abstract
Acyl-coenzyme A cholesterol acyltransferase (ACAT) plays an important role in maintaining cellular and organismal cholesterol homeostasis. Two types of ACAT isozymes with different functions exist in mammals, named ACAT-1 and ACAT-2. Numerous studies showed that ACAT-2 selective inhibitors are effective for the treatment of hypercholesterolemia and atherosclerosis. However, as a typical endoplasmic reticulum protein, ACAT-2 protein has not been purified and revealed, so combinatorial ligand-based methods might be the optimal strategy for discovering the ACAT-2 selective inhibitors. In this study, selective pharmacophore models of ACAT-1 inhibitors and ACAT-2 inhibitors were built, respectively. The optimal pharmacophore model for each subtype was identified and utilized as queries for the Traditional Chinese Medicine Database screening. A total of 180 potential ACAT-2 selective inhibitors were obtained, which were identified using an ACAT-2 pharmacophore and not by our ACAT-1 model. Selective SVM model and bioactive SVR model were generated for further identification of the obtained ACAT-2 inhibitors. Ten compounds were finally obtained with predicted inhibitory activities toward ACAT-2. Hydrogen bond acceptor, 2D autocorrelations, GETAWAY descriptors, and BCUT descriptors were identified as key structural features for selectivity and activity of ACAT-2 inhibitors. This study provides a reasonable ligand-based approach to discover potential ACAT-2 selective inhibitors from Chinese herbs, which could help in further screening and development of ACAT-2 selective inhibitors.
Collapse
Affiliation(s)
- Lian-Sheng Qiao
- Key Laboratory of TCM Foundation and New Drug Research, School of Chinese Material Medica, Beijing University of Chinese Medicine, Beijing, 100102, China
| | - Xian-Bao Zhang
- Key Laboratory of TCM Foundation and New Drug Research, School of Chinese Material Medica, Beijing University of Chinese Medicine, Beijing, 100102, China
| | - Lu-di Jiang
- Key Laboratory of TCM Foundation and New Drug Research, School of Chinese Material Medica, Beijing University of Chinese Medicine, Beijing, 100102, China
| | - Yan-Ling Zhang
- Key Laboratory of TCM Foundation and New Drug Research, School of Chinese Material Medica, Beijing University of Chinese Medicine, Beijing, 100102, China.
| | - Gong-Yu Li
- Key Laboratory of TCM Foundation and New Drug Research, School of Chinese Material Medica, Beijing University of Chinese Medicine, Beijing, 100102, China
| |
Collapse
|
11
|
Kroiss M, Fassnacht M. Inhibition of Cholesterol Esterification in the Adrenal Gland by ATR101/PD132301-2, A Promising Case of Drug Repurposing. Endocrinology 2016; 157:1719-21. [PMID: 27149038 DOI: 10.1210/en.2016-1210] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Matthias Kroiss
- Division of Endocrinology and Diabetology, Department of Internal Medicine, University Hospital Würzburg, University of Würzburg; and Clinical Chemistry and Laboratory Medicine, University Hospital Würzburg; and Comprehensive Cancer Center Mainfranken, University of Würzburg, 97080 Würzburg, Germany
| | - Martin Fassnacht
- Division of Endocrinology and Diabetology, Department of Internal Medicine, University Hospital Würzburg, University of Würzburg; and Clinical Chemistry and Laboratory Medicine, University Hospital Würzburg; and Comprehensive Cancer Center Mainfranken, University of Würzburg, 97080 Würzburg, Germany
| |
Collapse
|
12
|
Zhan Y, Zhang XW, Xiong Y, Li BL, Nan FJ. Design and synthesis of simple, yet potent and selective non-ring-A pyripyropene A-based inhibitors of acyl-coenzyme A: cholesterol acyltransferase 2 (ACAT2). Org Biomol Chem 2016; 14:747-751. [PMID: 26584338 DOI: 10.1039/c5ob02019k] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
A series of pyripyropene A-based compounds were designed and synthesized by opening the upper section of the A-ring, which significantly simplifies the structure and synthesis from commercially available starting materials. Representative compound (-)-3 exhibited potent activity against ACAT2 and greater selectivity for ACAT2 than for ACAT1.
Collapse
Affiliation(s)
- Yang Zhan
- State Key Laboratory of Drug Research, The National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
| | | | | | | | | |
Collapse
|
13
|
Ohshiro T, Ohtawa M, Nagamitsu T, Matsuda D, Yagyu H, Davis MA, Rudel LL, Ishibashi S, Tomoda H. New pyripyropene A derivatives, highly SOAT2-selective inhibitors, improve hypercholesterolemia and atherosclerosis in atherogenic mouse models. J Pharmacol Exp Ther 2015; 355:299-307. [PMID: 26338984 PMCID: PMC4613958 DOI: 10.1124/jpet.115.227348] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Accepted: 08/28/2015] [Indexed: 12/26/2022] Open
Abstract
Sterol O-acyltransferase 2 (SOAT2; also known as ACAT2) is considered as a new therapeutic target for the treatment or prevention of hypercholesterolemia and atherosclerosis. Fungal pyripyropene A (PPPA: 1,7,11-triacyl type), the first SOAT2-selective inhibitor, proved orally active in vivo using atherogenic mouse models. The purpose of the present study was to demonstrate that the PPPA derivatives (PRDs) prove more effective in the mouse models than PPPA. Among 196 semisynthetic PPPA derivatives, potent, SOAT2-selective, and stable PRDs were selected. In vivo antiatherosclerotic activity of selected PRDs was tested in apolipoprotein E knockout (Apoe(-/-)) mice or low-density lipoprotein receptor knockout (Ldlr(-/-)) mice fed a cholesterol-enriched diet (0.2% cholesterol and 21% fat) for 12 weeks. During the PRD treatments, no detrimental side effects were observed. Among three PRDs, Apoe(-/-) mice treated with PRD125 (1-,11-O-benzylidene type) at 1 mg/kg/day had significantly lower total plasma cholesterol concentration by 57.9 ± 9.3%; further, the ratio of cholesteryl oleate to cholesteryl linoleate in low-density lipoprotein was lower by 55.6 ± 7.5%, respectively. The hepatic cholesteryl ester levels and SOAT2 activity in the small intestines and livers of the PRD-treated mice were selectively lowered. The atherosclerotic lesion areas in the aortae of PRD125-treated mice were significantly lower at 62.2 ± 13.1%, respectively. Furthermore, both PRDs were also orally active in atherogenic Ldlr(-/-) mice. Among the PRDs tested, PRD125 was the most potent in both mouse models. These results suggest that SOAT2-selective inhibitors such as PRD125 have a high potential as poststatin agents for treatment and/or prevention in patients with atherosclerosis and hypercholesterolemia.
Collapse
Affiliation(s)
- Taichi Ohshiro
- Graduate School of Pharmaceutical Sciences, Kitasato University, Tokyo, Japan (T.O., M.O., T.N., D.M., H.T.); Department of Medicine, Jichi Medical University, Tochigi, Japan (T.O., H.Y., S.I.); and Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina (T.O., M.A.D., L.L.R.)
| | - Masaki Ohtawa
- Graduate School of Pharmaceutical Sciences, Kitasato University, Tokyo, Japan (T.O., M.O., T.N., D.M., H.T.); Department of Medicine, Jichi Medical University, Tochigi, Japan (T.O., H.Y., S.I.); and Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina (T.O., M.A.D., L.L.R.)
| | - Tohru Nagamitsu
- Graduate School of Pharmaceutical Sciences, Kitasato University, Tokyo, Japan (T.O., M.O., T.N., D.M., H.T.); Department of Medicine, Jichi Medical University, Tochigi, Japan (T.O., H.Y., S.I.); and Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina (T.O., M.A.D., L.L.R.)
| | - Daisuke Matsuda
- Graduate School of Pharmaceutical Sciences, Kitasato University, Tokyo, Japan (T.O., M.O., T.N., D.M., H.T.); Department of Medicine, Jichi Medical University, Tochigi, Japan (T.O., H.Y., S.I.); and Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina (T.O., M.A.D., L.L.R.)
| | - Hiroaki Yagyu
- Graduate School of Pharmaceutical Sciences, Kitasato University, Tokyo, Japan (T.O., M.O., T.N., D.M., H.T.); Department of Medicine, Jichi Medical University, Tochigi, Japan (T.O., H.Y., S.I.); and Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina (T.O., M.A.D., L.L.R.)
| | - Matthew A Davis
- Graduate School of Pharmaceutical Sciences, Kitasato University, Tokyo, Japan (T.O., M.O., T.N., D.M., H.T.); Department of Medicine, Jichi Medical University, Tochigi, Japan (T.O., H.Y., S.I.); and Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina (T.O., M.A.D., L.L.R.)
| | - Lawrence L Rudel
- Graduate School of Pharmaceutical Sciences, Kitasato University, Tokyo, Japan (T.O., M.O., T.N., D.M., H.T.); Department of Medicine, Jichi Medical University, Tochigi, Japan (T.O., H.Y., S.I.); and Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina (T.O., M.A.D., L.L.R.)
| | - Shun Ishibashi
- Graduate School of Pharmaceutical Sciences, Kitasato University, Tokyo, Japan (T.O., M.O., T.N., D.M., H.T.); Department of Medicine, Jichi Medical University, Tochigi, Japan (T.O., H.Y., S.I.); and Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina (T.O., M.A.D., L.L.R.)
| | - Hiroshi Tomoda
- Graduate School of Pharmaceutical Sciences, Kitasato University, Tokyo, Japan (T.O., M.O., T.N., D.M., H.T.); Department of Medicine, Jichi Medical University, Tochigi, Japan (T.O., H.Y., S.I.); and Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina (T.O., M.A.D., L.L.R.)
| |
Collapse
|
14
|
Abstract
INTRODUCTION Acyltransferase (AT) catalyzes the transfer of an acyl moiety from acyl-coenzyme A (acyl-CoA) to an acceptor. ATs play important roles in the maintenance of homeostasis in the human body and have been linked to various diseases; therefore, several ATs have been proposed as potential targets for the treatment or prevention of such diseases. The AT family includes acyl-CoA:cholesterol AT (ACAT), diacylglycerol AT (DGAT), and monoacylglycerol AT (MGAT) for the metabolism of lipids. Furthermore, recent molecular biological studies revealed the existence of their isozymes with distinct functions in the body. AREAS COVERED This review summarized patent filings published between 2010 and the present date that claimed isozyme-selective inhibitors of ACAT, DGAT and MGAT, which are involved in neutral lipid metabolism. EXPERT OPINION Isozymes of ACAT, DGAT and MGAT play distinct functions in neutral lipid metabolism in the human body and have been considered as potential therapeutic targets. Accordingly, isozyme-selective inhibitors that could be used in the treatment or prevention of lipid metabolism disorders were searched for. Of these, pyripyropene A derivatives, ACAT2-selective inhibitors, may be potential therapeutics for the treatment of atherosclerosis, homozygous familial hypercholesterolemia and nonalcoholic fatty liver disease.
Collapse
Affiliation(s)
- Taichi Ohshiro
- Kitasato University, Graduate School of Pharmaceutical Sciences , 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641 , Japan
| | | |
Collapse
|
15
|
Liefhebber JMP, Hague CV, Zhang Q, Wakelam MJO, McLauchlan J. Modulation of triglyceride and cholesterol ester synthesis impairs assembly of infectious hepatitis C virus. J Biol Chem 2014; 289:21276-88. [PMID: 24917668 PMCID: PMC4118089 DOI: 10.1074/jbc.m114.582999] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
In hepatitis C virus infection, replication of the viral genome and virion assembly are linked to cellular metabolic processes. In particular, lipid droplets, which store principally triacylglycerides (TAGs) and cholesterol esters (CEs), have been implicated in production of infectious virus. Here, we examine the effect on productive infection of triacsin C and YIC-C8-434, which inhibit synthesis of TAGs and CEs by targeting long-chain acyl-CoA synthetase and acyl-CoA:cholesterol acyltransferase, respectively. Our results present high resolution data on the acylglycerol and cholesterol ester species that were affected by the compounds. Moreover, triacsin C, which blocks both triglyceride and cholesterol ester synthesis, cleared most of the lipid droplets in cells. By contrast, YIC-C8-434, which only abrogates production of cholesterol esters, induced an increase in size of droplets. Although both compounds slightly reduced viral RNA synthesis, they significantly impaired assembly of infectious virions in infected cells. In the case of triacsin C, reduced stability of the viral core protein, which forms the virion nucleocapsid and is targeted to the surface of lipid droplets, correlated with lower virion assembly. In addition, the virus particles that were released from cells had reduced specific infectivity. YIC-C8-434 did not alter the association of core with lipid droplets but appeared to decrease production of infectious virus particles, suggesting a block in virion assembly. Thus, the compounds have antiviral properties, indicating that targeting synthesis of lipids stored in lipid droplets might be an option for therapeutic intervention in treating chronic hepatitis C virus infection.
Collapse
Affiliation(s)
- Jolanda M P Liefhebber
- From the Medical Research Council-University of Glasgow Centre for Virus Research, 8 Church Street, Glasgow G11 5JR, Scotland, United Kingdom and
| | - Charlotte V Hague
- From the Medical Research Council-University of Glasgow Centre for Virus Research, 8 Church Street, Glasgow G11 5JR, Scotland, United Kingdom and
| | - Qifeng Zhang
- The Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, United Kingdom
| | - Michael J O Wakelam
- The Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, United Kingdom
| | - John McLauchlan
- From the Medical Research Council-University of Glasgow Centre for Virus Research, 8 Church Street, Glasgow G11 5JR, Scotland, United Kingdom and
| |
Collapse
|
16
|
Pedrelli M, Davoodpour P, Degirolamo C, Gomaraschi M, Graham M, Ossoli A, Larsson L, Calabresi L, Gustafsson JÅ, Steffensen KR, Eriksson M, Parini P. Hepatic ACAT2 knock down increases ABCA1 and modifies HDL metabolism in mice. PLoS One 2014; 9:e93552. [PMID: 24695360 PMCID: PMC3973598 DOI: 10.1371/journal.pone.0093552] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2013] [Accepted: 03/06/2014] [Indexed: 11/18/2022] Open
Abstract
OBJECTIVES ACAT2 is the exclusive cholesterol-esterifying enzyme in hepatocytes and enterocytes. Hepatic ABCA1 transfers unesterified cholesterol (UC) to apoAI, thus generating HDL. By changing the hepatic UC pool available for ABCA1, ACAT2 may affect HDL metabolism. The aim of this study was to reveal whether hepatic ACAT2 influences HDL metabolism. DESIGN WT and LXRα/β double knockout (DOKO) mice were fed a western-type diet for 8 weeks. Animals were i.p. injected with an antisense oligonucleotide targeted to hepatic ACAT2 (ASO6), or with an ASO control. Injections started 4 weeks after, or concomitantly with, the beginning of the diet. RESULTS ASO6 reduced liver cholesteryl esters, while not inducing UC accumulation. ASO6 increased hepatic ABCA1 protein independently of the diet conditions. ASO6 affected HDL lipids (increased UC) only in DOKO, while it increased apoE-containing HDL in both genotypes. In WT mice ASO6 led to the appearance of large HDL enriched in apoAI and apoE. CONCLUSIONS The use of ASO6 revealed a new pathway by which the liver may contribute to HDL metabolism in mice. ACAT2 seems to be a hepatic player affecting the cholesterol fluxes fated to VLDL or to HDL, the latter via up-regulation of ABCA1.
Collapse
Affiliation(s)
- Matteo Pedrelli
- Division of Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
- Molecular Nutrition Unit, Department of Bioscience and Nutrition, Karolinska Institutet, Stockholm, Sweden
| | - Padideh Davoodpour
- Division of Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Chiara Degirolamo
- Division of Lipid Science, Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, North Carolina, United States of America
| | - Monica Gomaraschi
- Department of Pharmacological Sciences, University of Milan, Milan, Italy
| | - Mark Graham
- Cardiovascular Group, Department of Antisense Drug Discovery, Isis Pharmaceuticals, Inc., Carlsbad, California, United States of America
| | - Alice Ossoli
- Department of Pharmacological Sciences, University of Milan, Milan, Italy
| | - Lilian Larsson
- Division of Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Laura Calabresi
- Department of Pharmacological Sciences, University of Milan, Milan, Italy
| | - Jan-Åke Gustafsson
- Molecular Nutrition Unit, Department of Bioscience and Nutrition, Karolinska Institutet, Stockholm, Sweden
- Center for Nuclear Receptors and Cell Signaling, University of Houston, Houston, Texas, United States of America
| | - Knut R. Steffensen
- Molecular Nutrition Unit, Department of Bioscience and Nutrition, Karolinska Institutet, Stockholm, Sweden
| | - Mats Eriksson
- Molecular Nutrition Unit, Department of Bioscience and Nutrition, Karolinska Institutet, Stockholm, Sweden
- Department of Medicine, Karolinska Institute, Stockholm, Sweden
| | - Paolo Parini
- Division of Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
- Molecular Nutrition Unit, Department of Bioscience and Nutrition, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
17
|
Goo YH, Son SH, Kreienberg PB, Paul A. Novel lipid droplet-associated serine hydrolase regulates macrophage cholesterol mobilization. Arterioscler Thromb Vasc Biol 2013; 34:386-96. [PMID: 24357060 DOI: 10.1161/atvbaha.113.302448] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
OBJECTIVE Lipid-laden macrophages or foam cells are characterized by massive cytosolic lipid droplet (LD) deposition containing mostly cholesterol ester (CE) derived from the lipoproteins cleared from the arterial wall. Cholesterol efflux from foam cells is considered to be atheroprotective. Because cholesterol is effluxed as free cholesterol, CE accumulation in LDs may limit free cholesterol efflux. Our objective was to identify proteins that regulate cholesterol trafficking through LDs. APPROACH AND RESULTS In a proteomic analysis of the LD fraction of RAW 264.7 macrophages, we identified an evolutionarily conserved protein with a canonical GXSXG lipase catalytic motif and a predicted α/β-hydrolase fold, the RIKEN cDNA 1110057K04 gene, which we named LD-associated hydrolase (LDAH). LDAH association with LDs was confirmed by immunoblotting and immunocytochemistry. LDAH was labeled with a probe specific for active serine hydrolases. LDAH showed relatively weak in vitro CE hydrolase activity. However, cholesterol measurements in intact cells supported a significant role of LDAH in CE homeostasis because LDAH upregulation and downregulation decreased and increased, respectively, intracellular cholesterol and CE in human embryonic kidney-293 cells and RAW 264.7 macrophages. Mutation of the putative nucleophilic serine impaired active hydrolase probe binding, in vitro CE hydrolase activity, and cholesterol-lowering effect in cells, whereas this mutant still localized to the LD. LDAH upregulation increased CE hydrolysis and cholesterol efflux from macrophages, and, interestingly, LDAH is highly expressed in macrophage-rich areas within mouse and human atherosclerotic lesions. CONCLUSIONS The data identify a candidate target to promote reverse cholesterol transport from atherosclerotic lesions.
Collapse
Affiliation(s)
- Young-Hwa Goo
- From the Center for Cardiovascular Sciences, Albany Medical College, NY (Y.-H.G., S.-H.S., A.P.); and the Institute for Vascular Health and Disease, Albany, NY (P.B.K.)
| | | | | | | |
Collapse
|
18
|
Cao XZ, Mi TY, Li L, Vermeer MA, Zhang CC, Huang N, Manoj JK. HPLC-FLD determination of NBD-cholesterol, its ester and other metabolites in cellular lipid extracts. Biomed Chromatogr 2013; 27:910-5. [PMID: 23526237 DOI: 10.1002/bmc.2881] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2012] [Revised: 01/22/2013] [Accepted: 01/23/2013] [Indexed: 01/01/2023]
Abstract
22-[N(-7-Nitrobenz-2-oxa-1,3-diazol-4-yl)amino]-23,24-bisnor-5-cholen-3β-ol (NBD-cholesterol), a fluorescent cholesterol analog, was an extragenous cholesterol tracer used to study cholesterol absorption and metabolism in cultured cells. In order to measure free intracellular cholesterol and its esters, a precise and sensitive method employing high-performance liquid chromatography/fluorescence detection (HPLC-FLD) was developed for the first time. Method validation showed a limit of detection at 30 ng/mL. The calibration curve was linear within the range of 0.0625-10.0 µg/mL (r(2) = 0.999). Accuracy and precision were highlighted by good recovery and low variations. Apart from NBD-cholesteryl oleate, two additional cellular metabolites of NBD-cholesterol, probably an isomer and an oxidation product, were determined in the lipid extracts of Caco-2 human colon adenocarcinoma cells according to mass spectrometry. In AC29 mouse malignant mesothelioma cells overexpressing acyl-CoA:cholesterol acyltransferase-1 (ACAT1) or ACAT2, only the oxidized metabolite was detected. Using the newly developed method, YIC-C8-434, a known ACAT inhibitor, was shown to inhibit ACAT activity in Caco-2 cells, as well as in AC29/ACAT1 or AC29/ACAT2 cells. In conclusion, the sensitive and specific HPLC-FLD method is a powerful tool for simultaneous quantification of intracellular NBD-cholesterol and its oleoyl-ester.
Collapse
Affiliation(s)
- Xiu-zhen Cao
- Unilever R&D Shanghai, 66 Lin Xin Road, Shanghai, 200335, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
19
|
Rong JX, Blachford C, Feig JE, Bander I, Mayne J, Kusunoki J, Miller C, Davis M, Wilson M, Dehn S, Thorp E, Tabas I, Taubman MB, Rudel LL, Fisher EA. ACAT inhibition reduces the progression of preexisting, advanced atherosclerotic mouse lesions without plaque or systemic toxicity. Arterioscler Thromb Vasc Biol 2012; 33:4-12. [PMID: 23139293 DOI: 10.1161/atvbaha.112.252056] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Acyl-CoA:cholesterol acyltransferase (ACAT) converts cholesterol to cholesteryl esters in plaque foam cells. Complete deficiency of macrophage ACAT has been shown to increase atherosclerosis in hypercholesterolemic mice because of cytotoxicity from free cholesterol accumulation, whereas we previously showed that partial ACAT inhibition by Fujirebio compound F1394 decreased early atherosclerosis development. In this report, we tested F1394 effects on preestablished, advanced lesions of apolipoprotein-E-deficient mice. METHODS AND RESULTS Apolipoprotein-E-deficient mice on Western diet for 14 weeks developed advanced plaques, and were either euthanized (Baseline), or continued on Western diet with or without F1394 and euthanized after 14 more weeks. F1394 was not associated with systemic toxicity. Compared with the baseline group, lesion size progressed in both groups; however, F1394 significantly retarded plaque progression and reduced plaque macrophage, free and esterified cholesterol, and tissue factor contents compared with the untreated group. Apoptosis of plaque cells was not increased, consistent with the decrease in lesional free cholesterol. There was no increase in plaque necrosis and unimpaired efferocytosis (phagocytic clearance of apoptotic cells). The effects of F1394 were independent of changes in plasma cholesterol levels. CONCLUSIONS Partial ACAT inhibition by F1394 lowered plaque cholesterol content and had other antiatherogenic effects in advanced lesions in apolipoprotein-E-deficient mice without overt systemic or plaque toxicity, suggesting the continued potential of ACAT inhibition for the clinical treatment of atherosclerosis, in spite of recent trial data.
Collapse
Affiliation(s)
- James X Rong
- Marc and Ruti Bell Vascular Biology and Disease Research Program of the Leon H. Charney Division of Cardiology and the Department of Medicine (Cardiology), New York University School of Medicine, Smilow 7, 522 First Ave, New York, NY 10029, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Yang Y, Wang YF, Yang XF, Wang ZH, Lian YT, Yang Y, Li XW, Gao X, Chen J, Shu YW, Cheng LX, Liao YH, Liu K. Specific Kv1.3 blockade modulates key cholesterol-metabolism-associated molecules in human macrophages exposed to ox-LDL. J Lipid Res 2012; 54:34-43. [PMID: 23099443 DOI: 10.1194/jlr.m023846] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Cholesterol-metabolism-associated molecules, including scavenger receptor class A (SR-A), lectin-like oxidized low-density lipoprotein receptor-1 (LOX-1), CD36, ACAT1, ABCA1, ABCG1, and scavenger receptor class B type I, can modulate cholesterol metabolism in the transformation from macrophages to foam cells. Voltage-gated potassium channel Kv1.3 has increasingly been demonstrated to play an important role in the modulation of macrophage function. Here, we investigate the role of Kv1.3 in modulating cholesterol-metabolism-associated molecules in human acute monocytic leukemia cell-derived macrophages (THP-1 macrophages) and human monocyte-derived macrophages exposed to oxidized LDL (ox-LDL). Human Kv1.3 and Kv1.5 channels (hKv1.3 and hKv1.5) are expressed in macrophages and form a heteromultimeric channel. The hKv1.3-E314 antibody that we had generated as a specific hKv1.3 blocker inhibited outward delayed rectifier potassium currents, whereas the hKv1.5-E313 antibody that we had generated as a specific hKv1.5 blocker failed. Accordingly, the hKv1.3-E314 antibody reduced percentage of cholesterol ester and enhanced apoA-I-mediated cholesterol efflux in THP-1 macrophages and human monocyte-derived macrophages exposed to ox-LDL. The hKv1.3-E314 antibody downregulated SR-A, LOX-1, and ACAT1 expression and upregulated ABCA1 expression in THP-1 macrophages and human monocyte-derived macrophages. Our results reveal that specific Kv1.3 blockade represents a novel strategy modulating cholesterol metabolism in macrophages, which benefits the treatment of atherosclerotic lesions.
Collapse
Affiliation(s)
- Yong Yang
- Department of Cardiology, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Isoform-specific inhibitors of ACATs: recent advances and promising developments. Future Med Chem 2011; 3:2039-61. [DOI: 10.4155/fmc.11.158] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Acyl-CoA:cholesterol acyltransferase (ACAT) is a promising therapeutic target for cardiovascular diseases. Although a number of synthetic ACAT inhibitors have been developed, they have failed to show efficacy in clinical trials. Now, the presence of two ACAT isoforms with distinct functions, ACAT1 and ACAT2, has been discovered. Thus, the selectivity of ACAT inhibitors toward the two isoforms is important for their development as novel anti-atherosclerotic agents. The selectivity study indicated that fungal pyripyropene A (PPPA) is only an ACAT2-specific inhibitor. Furthermore, PPPA proved orally active in atherogenic mouse models, indicating it possessed cholesterol-lowering and atheroprotective activities. Certain PPPA derivatives, semi-synthetically prepared, possessed more potent and selective in vitro activity than PPPA against ACAT2. This review covers these studies and describes the future prospects of ACAT2-specific inhibitors.
Collapse
|
22
|
McLaren JE, Michael DR, Ashlin TG, Ramji DP. Cytokines, macrophage lipid metabolism and foam cells: implications for cardiovascular disease therapy. Prog Lipid Res 2011; 50:331-47. [PMID: 21601592 DOI: 10.1016/j.plipres.2011.04.002] [Citation(s) in RCA: 273] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2011] [Revised: 04/14/2011] [Accepted: 04/14/2011] [Indexed: 12/23/2022]
Abstract
Cardiovascular disease is the biggest killer globally and the principal contributing factor to the pathology is atherosclerosis; a chronic, inflammatory disorder characterized by lipid and cholesterol accumulation and the development of fibrotic plaques within the walls of large and medium arteries. Macrophages are fundamental to the immune response directed to the site of inflammation and their normal, protective function is harnessed, detrimentally, in atherosclerosis. Macrophages contribute to plaque development by internalizing native and modified lipoproteins to convert them into cholesterol-rich foam cells. Foam cells not only help to bridge the innate and adaptive immune response to atherosclerosis but also accumulate to create fatty streaks, which help shape the architecture of advanced plaques. Foam cell formation involves the disruption of normal macrophage cholesterol metabolism, which is governed by a homeostatic mechanism that controls the uptake, intracellular metabolism, and efflux of cholesterol. It has emerged over the last 20 years that an array of cytokines, including interferon-γ, transforming growth factor-β1, interleukin-1β, and interleukin-10, are able to manipulate these processes. Foam cell targeting, anti-inflammatory therapies, such as agonists of nuclear receptors and statins, are known to regulate the actions of pro- and anti-atherogenic cytokines indirectly of their primary pharmacological function. A clear understanding of macrophage foam cell biology will hopefully enable novel foam cell targeting therapies to be developed for use in the clinical intervention of atherosclerosis.
Collapse
Affiliation(s)
- James E McLaren
- Cardiff School of Biosciences, Cardiff University, Museum Avenue, Cardiff CF10 3AX, UK
| | | | | | | |
Collapse
|
23
|
Ohshiro T, Matsuda D, Sakai K, Degirolamo C, Yagyu H, Rudel LL, Omura S, Ishibashi S, Tomoda H. Pyripyropene A, an acyl-coenzyme A:cholesterol acyltransferase 2-selective inhibitor, attenuates hypercholesterolemia and atherosclerosis in murine models of hyperlipidemia. Arterioscler Thromb Vasc Biol 2011; 31:1108-15. [PMID: 21393580 DOI: 10.1161/atvbaha.111.223552] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Pyripyropene A (PPPA) of fungal origin is the first compound that has been found to strongly and selectively inhibit acyl-coenzyme A:cholesterol acyltransferase 2 (ACAT2) isozyme activity in vitro. The purpose of the present study was to investigate in vivo efficacy of the ACAT2-selective inhibitor in atherosclerosis. METHODS AND RESULTS PPPA treatment (10 to 100 mg/kg) caused 30.5±4.7% to 55.8±3.3% inhibition of the cholesterol absorption from the mouse intestine. When PPPA (10 to 50 mg/kg per day) was orally administered to apolipoprotein E-knockout mice for 12 weeks, the levels of plasma cholesterol, very-low-density lipoprotein (VLDL), and low-density lipoprotein (LDL) and hepatic cholesterol content were lowered. Furthermore, the ratio of cholesteryl oleate (exclusively synthesized in hepatic ACAT2) to cholesteryl linoleate in VLDL- and LDL-derived cholesteryl ester decreased, indicating that hepatic ACAT2 activity was inhibited by PPPA. PPPA-treated mice had reduced atherogenic lesion areas that were lowered by 26.2±3.7% to 46±3.8% in the aortae and by 18.9±3.6% to 37.6±6.0% in the hearts. CONCLUSIONS Our findings indicate that ACAT2-selective inhibition in the intestine and the liver can be effective against atherosclerosis and that PPPA appears to be a potential antiatherogenic lead compound. This study is the first demonstration of the in vivo efficacy of PPPA, an ACAT2-selective inhibitor, in atherosclerosis. PPPA-treated atherogenic mice showed a decrease in intestinal cholesterol absorption and cholesterol and cholesteryl oleate levels in both LDL and VLDL, resulting in protection of atherosclerosis development.
Collapse
Affiliation(s)
- Taichi Ohshiro
- Department of Microbial Chemistry, Graduate School of Pharmaceutical Sciences, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Seneff S, Wainwright G, Mascitelli L. Is the metabolic syndrome caused by a high fructose, and relatively low fat, low cholesterol diet? Arch Med Sci 2011; 7:8-20. [PMID: 22291727 PMCID: PMC3258689 DOI: 10.5114/aoms.2011.20598] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2010] [Revised: 08/15/2010] [Accepted: 08/23/2010] [Indexed: 01/24/2023] Open
Abstract
The metabolic syndrome (MetS) is manifested by a lipid triad which includes elevated serum triglycerides, small LDL particles, and low high-density lipoprotein (HDL) cholesterol, by central obesity (central adiposity), insulin resistance, glucose intolerance and elevated blood pressure, and it is associated with an increased risk of type 2 diabetes and coronary heart disease. We have developed a new hypothesis regarding MetS as a consequence of a high intake in carbohydrates and food with a high glycemic index, particularly fructose, and relatively low intake of cholesterol and saturated fat. We support our arguments through animal studies which have shown that exposure of the liver to increased quantities of fructose leads to rapid stimulation of lipogenesis and accumulation of triglycerides. The adipocytes store triglycerides in lipid droplets, leading to adipocyte hypertrophy. Adipocyte hypertrophy is associated with macrophage accumulation in adipose tissue. An important modulator of obesity-associated macrophage responses in white adipose tissue is the death of adipocytes. Excess exposure to fructose intake determines the liver to metabolize high doses of fructose, producing increased levels of fructose end products, like glyceraldehyde and dihydroxyacetone phosphate, that can converge with the glycolytic pathway. Fructose also leads to increased levels of advanced glycation end products. The macrophages exposed to advanced glycation end products become dysfunctional and, on entry into the artery wall, contribute to plaque formation and thrombosis.
Collapse
Affiliation(s)
- Stephanie Seneff
- Department of Electrical Engineering and Computer Science, MIT, Cambridge, MA, USA
| | | | - Luca Mascitelli
- Medical Service, Comando Brigata Alpina “Julia”, Udine, Italy
| |
Collapse
|
25
|
Chhabria MT, Mahajan BM. Update on patented cholesterol absorption inhibitors. Expert Opin Ther Pat 2009; 19:1083-107. [PMID: 19552506 DOI: 10.1517/13543770903036826] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
BACKGROUND Atherosclerosis is one of the most life-threatening diseases primarily associated with hypercholesterolemia and is characterized by increased serum cholesterol level. Cholesterol originates from both its de novo synthesis within the hepatic cells and its absorption into the intestine in the form of dietary or bile cholesterol. Interventions influencing both of these processes are promising therapeutic options to lower the cholesterol level. Hydroxymethyl glutaryl-CoA reductase inhibitors, commonly known as statins, effectively block the rate determining step in the biosynthesis of cholesterol. Ezetimibe is the first new class of drugs used to treat hypercholesterolemia by inhibition of cholesterol absorption through Niemann Pick C1 Like 1 membrane of enterocytes. Therefore, combination therapy of ezetimibe and statins offers an efficacious new approach for the prevention and treatment of hypercholesterolemia. OBJECTIVES The present review focuses on updates on ezetimibe and patented profile of novel cholesterol absorption inhibitors followed by critical analysis of different targets such as cholesterol esterase inhibitors, bile acid transport inhibitors or phospholipase-A(2) inhibitors, etc.which play an important role in the lipid absorption. CONCLUSION The discovery of ezetimibe has opened a new door for the management of hyper-cholesterolemia in combination with statins. There are newer analogues that are under clinical trials, among which darapladib, FM-VP4 and A-002 are promising compounds.
Collapse
Affiliation(s)
- M T Chhabria
- L. M. College of Pharmacy, Department of Pharmaceutical Chemistry, Navrangpura, Ahmedabad - 380 009, Gujarat, India.
| | | |
Collapse
|
26
|
Hans CP, Zerfaoui M, Naura AS, Troxclair D, Strong JP, Matrougui K, Boulares AH. Thieno[2,3-c]isoquinolin-5-one, a potent poly(ADP-ribose) polymerase inhibitor, promotes atherosclerotic plaque regression in high-fat diet-fed apolipoprotein E-deficient mice: effects on inflammatory markers and lipid content. J Pharmacol Exp Ther 2009; 329:150-8. [PMID: 19124646 DOI: 10.1124/jpet.108.145938] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
We recently showed that poly(ADP-ribose) polymerase (PARP) is activated within atherosclerotic plaques in an animal model of atherosclerosis. Pharmacological inhibition of PARP or reduced expression in heterozygous animals interferes with atherogenesis and may promote factors of plaque stability, possibly reflecting changes in inflammatory and cellular factors consistent with plaque stability. The current study addresses the hypothesis that pharmacological inhibition of PARP promotes atherosclerotic plaque regression. Using a high-fat diet-induced atherosclerosis apolipoprotein E(-/-) mouse model, we demonstrate that administration of the potent PARP inhibitor, thieno[2,3-c]isoquinolin-5-one (TIQ-A), when combined with a regular diet regimen during treatment, induced regression of established plaques. Plaque regression was associated with a reduction in total cholesterol and low-density lipoproteins. Furthermore, plaques of TIQ-A-treated mice were highly enriched with collagen and smooth muscle cells, displayed thick fibrous caps, and exhibited a marked reduction in CD68-positive macrophage recruitment and associated foam cell presence. These changes correlated with a significant decrease in expression of monocyte chemoattractant protein-1 and intercellular cell adhesion molecule-1, potentially as a result of a robust reduction in tumor necrosis factor expression. The PARP inhibitor appeared to affect cholesterol metabolism by affecting acyl-coenzymeA/cholesterol acyltransferase-1 expression but exerted no effect on cholesterol influx or efflux as assessed by an examination of the ATP-binding cassette transporter-1 and the scavenger receptor-A expression levels in the different experimental groups. In accordance, PARP inhibition may prove beneficial not only in preventing atherogenesis but also in promoting regression of preexisting plaques.
Collapse
Affiliation(s)
- Chetan P Hans
- Department of Pharmacology and Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | | | | | | | | | | | | |
Collapse
|
27
|
Takahashi K, Kasai M, Ohta M, Shoji Y, Kunishiro K, Kanda M, Kurahashi K, Shirahase H. Novel Indoline-Based Acyl-CoA:Cholesterol Acyltransferase Inhibitor with Antiperoxidative Activity: Improvement of Physicochemical Properties and Biological Activities by Introduction of Carboxylic Acid. J Med Chem 2008; 51:4823-33. [DOI: 10.1021/jm800248r] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Kenji Takahashi
- Research Laboratories, Kyoto Pharmaceutical Industries, Ltd., 38 Nishinokyo Tsukinowa-cho, Nakagyo-ku, Kyoto 604-8444, Japan, Radioisotope Research Center, Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Masayasu Kasai
- Research Laboratories, Kyoto Pharmaceutical Industries, Ltd., 38 Nishinokyo Tsukinowa-cho, Nakagyo-ku, Kyoto 604-8444, Japan, Radioisotope Research Center, Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Masaru Ohta
- Research Laboratories, Kyoto Pharmaceutical Industries, Ltd., 38 Nishinokyo Tsukinowa-cho, Nakagyo-ku, Kyoto 604-8444, Japan, Radioisotope Research Center, Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Yoshimichi Shoji
- Research Laboratories, Kyoto Pharmaceutical Industries, Ltd., 38 Nishinokyo Tsukinowa-cho, Nakagyo-ku, Kyoto 604-8444, Japan, Radioisotope Research Center, Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Kazuyoshi Kunishiro
- Research Laboratories, Kyoto Pharmaceutical Industries, Ltd., 38 Nishinokyo Tsukinowa-cho, Nakagyo-ku, Kyoto 604-8444, Japan, Radioisotope Research Center, Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Mamoru Kanda
- Research Laboratories, Kyoto Pharmaceutical Industries, Ltd., 38 Nishinokyo Tsukinowa-cho, Nakagyo-ku, Kyoto 604-8444, Japan, Radioisotope Research Center, Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Kazuyoshi Kurahashi
- Research Laboratories, Kyoto Pharmaceutical Industries, Ltd., 38 Nishinokyo Tsukinowa-cho, Nakagyo-ku, Kyoto 604-8444, Japan, Radioisotope Research Center, Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Hiroaki Shirahase
- Research Laboratories, Kyoto Pharmaceutical Industries, Ltd., 38 Nishinokyo Tsukinowa-cho, Nakagyo-ku, Kyoto 604-8444, Japan, Radioisotope Research Center, Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto 606-8501, Japan
| |
Collapse
|
28
|
Tomoda H, Doi T. Discovery and combinatorial synthesis of fungal metabolites beauveriolides, novel antiatherosclerotic agents. Acc Chem Res 2008; 41:32-9. [PMID: 17803269 DOI: 10.1021/ar700117b] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
For discovery of a new type of antiatherosclerotic agents, a cell-based assay of lipid droplet accumulation using primary mouse peritoneal macrophages was conducted as a model of macrophage-derived foam cell accumulation, which occurs in the early stage of atherosclerogenesis. During the screening of microbial metabolites for inhibitors of lipid droplet accumulation, 13-membered cyclodepsipeptides, known beauveriolide I and new beauveriolide III, were isolated from the culture broth of fungal Beauveria sp. FO-6979, a soil isolate, by solvent extraction, ODS column chromatography, silica gel column chromatography, and preparative HPLC. The structure including the absolute stereochemistry of beauveriolide III was elucidated as cyclo-[(3 S,4 S)-3-hydroxy-4-methyloctanoyl- l-phenylalanyl- l-alanyl- d-alloisoleucyl] by spectral analyses, amino acid analyses, and synthetic methods. Furthermore, the absolute stereochemistry was confirmed by the total synthesis of beauveriolides. Study on the mechanism of action revealed that beauveriolides inhibited macrophage acyl-CoA:cholesterol acyltransferase (ACAT) activity to block the synthesis of cholesteryl ester (CE), leading to a reduction of lipid droplets in macrophages. There are two ACAT isozymes in mammals, ACAT1 and ACAT2. ACAT1 is ubiquitously expressed in most tissues and cells including macrophages, while ACAT2 is expressed predominantly in the liver (hepatocytes) and the intestine (enterocytes). Interestingly, beauveriolides inhibited both ACAT1 and ACAT2 to a similar extent in an enzyme assay that utilized microsomes but inhibited ACAT1 selectively in intact cell-based assays. Beauveriolides proved orally active in both low-density lipoprotein receptor and apolipoprotein E knockout mice, reducing the atheroma lesion of heart and aorta without any side effects such as diarrhea or cytotoxicity to adrenal tissues as observed for many synthetic ACAT inhibitors. To obtain more potent inhibitors, a focused library of beauveriolide analogues was prepared by combinatorial chemistry in which solid-phase assembly of linear depsipeptides was carried out using a 2-chlorotrityl linker, followed by solution-phase cyclization, yielding 104 beauveriolide analogues. Among them, diphenyl derivatives were found to show 10 times more potent inhibition of CE synthesis in macrophages than beauveriolide III. Furthermore, most analogues showed selective ACAT1 inhibition or inhibition of both ACAT1 and ACAT2, but interestingly certain analogues gave selective ACAT2 inhibition. These data indicated that subtle structural differences of the inhibitors could discriminate the active sites of the ACAT1 and ACAT2 isozymes. Efforts of further analogue synthesis would make it possible to obtain highly selective ACAT1/ACAT2 inhibitors.
Collapse
Affiliation(s)
- Hiroshi Tomoda
- School of Pharmacy, Kitasato University, Shirokane, Minato-ku, Tokyo 108-8641, Japan.
| | | |
Collapse
|
29
|
Lipid homeostasis in macrophages – Implications for atherosclerosis. REVIEWS OF PHYSIOLOGY BIOCHEMISTRY AND PHARMACOLOGY 2008; 160:93-125. [DOI: 10.1007/112_2008_802] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
30
|
Tomoda H, Omura S. Potential therapeutics for obesity and atherosclerosis: Inhibitors of neutral lipid metabolism from microorganisms. Pharmacol Ther 2007; 115:375-89. [PMID: 17614133 DOI: 10.1016/j.pharmthera.2007.05.008] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2007] [Accepted: 05/16/2007] [Indexed: 11/24/2022]
Abstract
Diacylglycerol acyltransferase (DGAT) and acyl-CoA: cholesterol acyltransferase (ACAT) are the enzymes that catalyze the final reactions of triacylgycerol (TG) and cholesteryl ester (CE) synthesis, and accumulation of TG and CE in adipocytes and arteries causes obesity and atherosclerosis, respectively. Therefore, DGAT and ACAT have been viewed as potential therapeutic targets for these diseases. From the screening program for DGAT inhibitors, new compounds were discovered from fungal and plant extracts, and are expected to provide leads for drug development. From the screening programs for ACAT inhibitors and lipid droplet synthesis inhibitors, new compounds with chemical structures different from those of known synthetic inhibitors were discovered from the cultures of fungal and actinomycete strains. Among them, fungal beauveriolide III rather selectively inhibited ACAT1 isozyme, while fungal pyripyropene A was found to be a highly selective inhibitor of ACAT2 isozyme. Both inhibitors proved orally active in in vivo models. Furthermore, a library of beauveriolide and pyripyropene analogs was prepared by combinatorial and semisynthetic methods, respectively. The future prospects of these inhibitors are discussed.
Collapse
|
31
|
Rau O, Zettl H, Popescu L, Steinhilber D, Schubert-Zsilavecz M. [New targets and drugs for treatment of lipid metabolism disorders]. PHARMAZIE IN UNSERER ZEIT 2007; 36:142-8. [PMID: 17425000 DOI: 10.1002/pauz.200600213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Affiliation(s)
- Oliver Rau
- Institut für Pharmazeutische Chemie der Johann Wolfgang Goethe-Universität, Frankfurt
| | | | | | | | | |
Collapse
|