151
|
So MM, Mansukhani NA, Peters EB, Albaghdadi MS, Wang Z, Pérez CMR, Kibbe MR, Stupp SI. Peptide Amphiphile Nanostructures for Targeting of Atherosclerotic Plaque and Drug Delivery. ACTA ACUST UNITED AC 2018; 2. [PMID: 30666317 DOI: 10.1002/adbi.201700123] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Co-assembled peptide amphiphile nanofibers designed to target atherosclerotic plaque and enhance cholesterol efflux are shown to encapsulate and deliver a liver X receptor agonist to increase efflux from murine macrophages in vitro. Fluorescence microscopy reveals that the nanofibers, which display an apolipoprotein-mimetic peptide, localize at plaque sites in LDL receptor knockout mice with or without the encapsulated molecule, while nanofibers displaying a scrambled, non-targeting peptide sequence do not demonstrate comparable binding. These results show that nanofibers functionalized with apolipoprotein-mimetic peptides may be effective vehicles for intravascular targeted drug delivery to treat atherosclerosis.
Collapse
Affiliation(s)
- Miranda M So
- Simpson Querrey Institute for BioNanotechnology, Northwestern University, Chicago, IL 60611, USA
| | - Neel A Mansukhani
- Simpson Querrey Institute for BioNanotechnology, Northwestern University, Chicago, IL 60611, USA
| | - Erica B Peters
- Department of Surgery, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Mazen S Albaghdadi
- Simpson Querrey Institute for BioNanotechnology, Northwestern University, Chicago, IL 60611, USA
| | - Zheng Wang
- Division of Vascular Surgery, Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Charles M Rubert Pérez
- Simpson Querrey Institute for BioNanotechnology, Northwestern University, Chicago, IL 60611, USA
| | - Melina R Kibbe
- Simpson Querrey Institute for BioNanotechnology, Northwestern University, Chicago, IL 60611, USA
| | - Samuel I Stupp
- Simpson Querrey Institute for BioNanotechnology, Northwestern University, Chicago, IL 60611, USA
| |
Collapse
|
152
|
Guo Y, Yuan W, Yu B, Kuai R, Hu W, Morin EE, Garcia-Barrio MT, Zhang J, Moon JJ, Schwendeman A, Eugene Chen Y. Synthetic High-Density Lipoprotein-Mediated Targeted Delivery of Liver X Receptors Agonist Promotes Atherosclerosis Regression. EBioMedicine 2018; 28:225-233. [PMID: 29361501 PMCID: PMC5835545 DOI: 10.1016/j.ebiom.2017.12.021] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 12/06/2017] [Accepted: 12/18/2017] [Indexed: 11/30/2022] Open
Abstract
Targeting at enhancing reverse cholesterol transport (RCT) is apromising strategy for treating atherosclerosis via infusion of reconstitute high density lipoprotein (HDL) as cholesterol acceptors or increase of cholesterol efflux by activation of macrophage liver X receptors (LXRs). However, systemic activation of LXRs triggers excessive lipogenesis in the liver and infusion of HDL downregulates cholesterol efflux from macrophages. Here we describe an enlightened strategy using phospholipid reconstituted apoA-I peptide (22A)-derived synthetic HDL (sHDL) to deliver LXR agonists to the atheroma and examine their effect on atherosclerosis regression in vivo. A synthetic LXR agonist, T0901317 (T1317) was encapsulated in sHDL nanoparticles (sHDL-T1317). Similar to the T1317 compound, the sHDL-T1317 nanoparticles upregulated the expression of ATP-binding cassette transporters and increased cholesterol efflux in macrophages in vitro and in vivo. The sHDL nanoparticles accumulated in the atherosclerotic plaques of ApoE-deficient mice. Moreover, a 6-week low-dose LXR agonist-sHDL treatment induced atherosclerosis regression while avoiding lipid accumulation in the liver. These findings identify LXR agonist loaded sHDL nanoparticles as a promising therapeutic approach to treat atherosclerosis by targeting RCT in a multifaceted manner: sHDL itself serving as both a drug carrier and cholesterol acceptor and the LXR agonist mediating upregulation of ABC transporters in the aorta.
Collapse
Affiliation(s)
- Yanhong Guo
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, United States.
| | - Wenmin Yuan
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, United States
| | - Bilian Yu
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, United States
| | - Rui Kuai
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, United States
| | - Wenting Hu
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, United States
| | - Emily E Morin
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, United States
| | | | - Jifeng Zhang
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, United States
| | - James J Moon
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, United States; Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, United States; Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, United States
| | - Anna Schwendeman
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, United States; Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, United States.
| | - Y Eugene Chen
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, United States.
| |
Collapse
|
153
|
Ahsan F, Maertzdorf J, Guhlich-Bornhof U, Kaufmann SHE, Moura-Alves P. IL-36/LXR axis modulates cholesterol metabolism and immune defense to Mycobacterium tuberculosis. Sci Rep 2018; 8:1520. [PMID: 29367626 PMCID: PMC5784124 DOI: 10.1038/s41598-018-19476-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 12/27/2017] [Indexed: 12/14/2022] Open
Abstract
Mycobacterium tuberculosis (Mtb) is a life-threatening pathogen in humans. Bacterial infection of macrophages usually triggers strong innate immune mechanisms, including IL-1 cytokine secretion. The newer member of the IL-1 family, IL-36, was recently shown to be involved in cellular defense against Mtb. To unveil the underlying mechanism of IL-36 induced antibacterial activity, we analyzed its role in the regulation of cholesterol metabolism, together with the involvement of Liver X Receptor (LXR) in this process. We report that, in Mtb-infected macrophages, IL-36 signaling modulates cholesterol biosynthesis and efflux via LXR. Moreover, IL-36 induces the expression of cholesterol-converting enzymes and the accumulation of LXR ligands, such as oxysterols. Ultimately, both IL-36 and LXR signaling play a role in the regulation of antimicrobial peptides expression and in Mtb growth restriction. These data provide novel evidence for the importance of IL-36 and cholesterol metabolism mediated by LXR in cellular host defense against Mtb.
Collapse
Affiliation(s)
- Fadhil Ahsan
- Department of Immunology, Max Planck Institute for Infection Biology, Charitéplatz 1, Berlin, 10117, Germany
| | - Jeroen Maertzdorf
- Department of Immunology, Max Planck Institute for Infection Biology, Charitéplatz 1, Berlin, 10117, Germany
| | - Ute Guhlich-Bornhof
- Department of Immunology, Max Planck Institute for Infection Biology, Charitéplatz 1, Berlin, 10117, Germany
| | - Stefan H E Kaufmann
- Department of Immunology, Max Planck Institute for Infection Biology, Charitéplatz 1, Berlin, 10117, Germany.
| | - Pedro Moura-Alves
- Department of Immunology, Max Planck Institute for Infection Biology, Charitéplatz 1, Berlin, 10117, Germany.
| |
Collapse
|
154
|
Soni C, Schell SL, Fasnacht MJ, Chodisetti SB, Rahman ZS. Crucial role of Mer tyrosine kinase in the maintenance of SIGN-R1 + marginal zone macrophages. Immunol Cell Biol 2018; 96:298-315. [PMID: 29345385 DOI: 10.1111/imcb.12003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 12/11/2017] [Accepted: 12/12/2017] [Indexed: 01/07/2023]
Abstract
Mer Tyrosine Kinase receptor (Mer) is involved in anti-inflammatory efferocytosis. Here we report elevated spontaneous germinal center (Spt-GC) responses in Mer-deficient mice (Mer-/- ) that are associated with the loss of SIGN-R1+ marginal zone macrophages (MZMs). The dissipation of MZMs in Mer-/- mice occurs independently of reduced cellularity or delocalization of marginal zone B cells, sinusoidal cells or of CD169+ metallophillic macrophages. We find that MZM dissipation in Mer-/- mice contributes to apoptotic cell (AC) accumulation in Spt-GCs and dysregulation of the GC checkpoint, allowing an expansion of DNA-reactive B cells in GCs. We further observe that bone marrow derived macrophages from Mer-/- mice produce more TNFα, and are susceptible to cell death upon exposure to ACs compared to WT macrophages. Anti-TNFα Ab treatment of Mer-/- mice is, however, unable to reverse MZM loss, but results in reduced Spt-GC responses, indicating that TNFα promotes Spt-GC responses in Mer-/- mice. Contrary to an anti-TNFα Ab treatment, treatment of Mer-/- mice with a synthetic agonist for the transcription factor LXRα rescues a significant number of MZMs in vivo. Our data suggest that Mer-LXRα signaling plays an important role in the differentiation and maintenance of MZMs, which in turn regulate Spt-GC responses and tolerance.
Collapse
Affiliation(s)
- Chetna Soni
- Department of Microbiology and Immunology, Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Stephanie L Schell
- Department of Microbiology and Immunology, Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Melinda J Fasnacht
- Department of Microbiology and Immunology, Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Sathi Babu Chodisetti
- Department of Microbiology and Immunology, Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Ziaur Sm Rahman
- Department of Microbiology and Immunology, Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| |
Collapse
|
155
|
Liu Y, Wei Z, Ma X, Yang X, Chen Y, Sun L, Ma C, Miao QR, Hajjar DP, Han J, Duan Y. 25-Hydroxycholesterol activates the expression of cholesterol 25-hydroxylase in an LXR-dependent mechanism. J Lipid Res 2018; 59:439-451. [PMID: 29298812 DOI: 10.1194/jlr.m080440] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Revised: 12/06/2017] [Indexed: 12/13/2022] Open
Abstract
Cholesterol 25-hydroxylase (CH25H) catalyzes the production of 25-hydroxycholesterol (25-HC), an oxysterol that can play an important role in different biological processes. However, the mechanisms regulating CH25H expression have not been fully elucidated. In this study, we determined that CH25H is highly expressed in mouse liver and peritoneal macrophages. We identified several liver X receptor (LXR) response elements (LXREs) in the human CH25H promoter. In HepG2 cells, activation of LXR by 25-HC or other oxysterols and synthetic ligands [T0901317 (T317) and GW3965] induced CH25H protein expression, which was associated with increased CH25H mRNA expression. 25-HC or T317 activated CH25H transcription in an LXRE-dependent manner. Thus, high-expressing LXRα or LXRβ activated CH25H expression, and the activation was further enhanced by LXR ligands. In contrast, inhibition of LXRα/β expression attenuated 25-HC or T317-induced CH25H expression. Deficiency of interferon γ expression reduced, but did not block, LXR ligand-induced hepatic CH25H expression. Activation of LXR also substantially induced macrophage CH25H expression. In vivo, administration of GW3965 to mice increased CH25H expression in both liver and peritoneal macrophages. Taken together, our study demonstrates that 25-HC can activate CH25H expression in an LXR-dependent manner, which may be an important mechanism to exert the biological actions of 25-HC.
Collapse
Affiliation(s)
- Ying Liu
- Department of Biochemistry and Molecular Biology, College of Life Sciences and Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China.,Department of Biomedical Sciences, College of Biomedical Engineering, Hefei University of Technology, Hefei, China
| | - Zhuo Wei
- Department of Biochemistry and Molecular Biology, College of Life Sciences and Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China
| | - Xingzhe Ma
- Department of Immunology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
| | - Xiaoxiao Yang
- Department of Biomedical Sciences, College of Biomedical Engineering, Hefei University of Technology, Hefei, China
| | - Yuanli Chen
- Department of Biochemistry and Molecular Biology, College of Life Sciences and Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China
| | - Lei Sun
- Department of Biochemistry and Molecular Biology, College of Life Sciences and Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China
| | - Chuanrui Ma
- Department of Biochemistry and Molecular Biology, College of Life Sciences and Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China
| | - Qing R Miao
- Departments of Surgery and Pathology, Medical College of Wisconsin, Milwaukee, WI
| | - David P Hajjar
- Department of Pathology, Weill Cornell Medical College of Cornell University, New York, NY
| | - Jihong Han
- Department of Biomedical Sciences, College of Biomedical Engineering, Hefei University of Technology, Hefei, China .,College of Life Sciences and State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China
| | - Yajun Duan
- Department of Biomedical Sciences, College of Biomedical Engineering, Hefei University of Technology, Hefei, China .,College of Life Sciences and State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China
| |
Collapse
|
156
|
Gromovsky AD, Schugar RC, Brown AL, Helsley RN, Burrows AC, Ferguson D, Zhang R, Sansbury BE, Lee RG, Morton RE, Allende DS, Parks JS, Spite M, Brown JM. Δ-5 Fatty Acid Desaturase FADS1 Impacts Metabolic Disease by Balancing Proinflammatory and Proresolving Lipid Mediators. Arterioscler Thromb Vasc Biol 2018; 38:218-231. [PMID: 29074585 PMCID: PMC5746431 DOI: 10.1161/atvbaha.117.309660] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 10/08/2017] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Human genetic variants near the FADS (fatty acid desaturase) gene cluster (FADS1-2-3) are strongly associated with cardiometabolic traits including dyslipidemia, fatty liver, type 2 diabetes mellitus, and coronary artery disease. However, mechanisms underlying these genetic associations are unclear. APPROACH AND RESULTS Here, we specifically investigated the physiological role of the Δ-5 desaturase FADS1 in regulating diet-induced cardiometabolic phenotypes by treating hyperlipidemic LDLR (low-density lipoprotein receptor)-null mice with antisense oligonucleotides targeting the selective knockdown of Fads1. Fads1 knockdown resulted in striking reorganization of both ω-6 and ω-3 polyunsaturated fatty acid levels and their associated proinflammatory and proresolving lipid mediators in a highly diet-specific manner. Loss of Fads1 activity promoted hepatic inflammation and atherosclerosis, yet was associated with suppression of hepatic lipogenesis. Fads1 knockdown in isolated macrophages promoted classic M1 activation, whereas suppressing alternative M2 activation programs, and also altered systemic and tissue inflammatory responses in vivo. Finally, the ability of Fads1 to reciprocally regulate lipogenesis and inflammation may rely in part on its role as an effector of liver X receptor signaling. CONCLUSIONS These results position Fads1 as an underappreciated regulator of inflammation initiation and resolution, and suggest that endogenously synthesized arachidonic acid and eicosapentaenoic acid are key determinates of inflammatory disease progression and liver X receptor signaling.
Collapse
MESH Headings
- Animals
- Aorta/enzymology
- Aorta/pathology
- Aortic Diseases/enzymology
- Aortic Diseases/genetics
- Aortic Diseases/pathology
- Arachidonic Acid/metabolism
- Atherosclerosis/enzymology
- Atherosclerosis/genetics
- Atherosclerosis/pathology
- Cells, Cultured
- Delta-5 Fatty Acid Desaturase
- Disease Models, Animal
- Dyslipidemias/enzymology
- Dyslipidemias/genetics
- Dyslipidemias/pathology
- Eicosapentaenoic Acid/metabolism
- Fatty Acid Desaturases/genetics
- Fatty Acid Desaturases/metabolism
- Inflammation/enzymology
- Inflammation/genetics
- Inflammation/pathology
- Inflammation Mediators/metabolism
- Lipogenesis
- Liver/metabolism
- Liver X Receptors/metabolism
- Macrophage Activation
- Macrophages, Peritoneal/enzymology
- Macrophages, Peritoneal/pathology
- Mice, Inbred C57BL
- Mice, Knockout
- Oligonucleotides, Antisense/genetics
- Oligonucleotides, Antisense/metabolism
- Plaque, Atherosclerotic
- Receptors, LDL/deficiency
- Receptors, LDL/genetics
Collapse
Affiliation(s)
- Anthony D Gromovsky
- From the Department of Cellular and Molecular Medicine, Lerner Research Institute (A.D.G., R.C.S., A.L.B., R.N.H., A.C.B., D.F., R.Z., R.E.M., J.M.B.) and Department of Anatomical Pathology (D.S.A.), Cleveland Clinic, OH; Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (B.E.S., M.S.); Cardiovascular Group, Antisense Drug Discovery, Ionis Pharmaceuticals, Inc, Carlsbad, CA (R.G.L.); and Department of Internal Medicine-Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC (J.S.P.)
| | - Rebecca C Schugar
- From the Department of Cellular and Molecular Medicine, Lerner Research Institute (A.D.G., R.C.S., A.L.B., R.N.H., A.C.B., D.F., R.Z., R.E.M., J.M.B.) and Department of Anatomical Pathology (D.S.A.), Cleveland Clinic, OH; Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (B.E.S., M.S.); Cardiovascular Group, Antisense Drug Discovery, Ionis Pharmaceuticals, Inc, Carlsbad, CA (R.G.L.); and Department of Internal Medicine-Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC (J.S.P.)
| | - Amanda L Brown
- From the Department of Cellular and Molecular Medicine, Lerner Research Institute (A.D.G., R.C.S., A.L.B., R.N.H., A.C.B., D.F., R.Z., R.E.M., J.M.B.) and Department of Anatomical Pathology (D.S.A.), Cleveland Clinic, OH; Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (B.E.S., M.S.); Cardiovascular Group, Antisense Drug Discovery, Ionis Pharmaceuticals, Inc, Carlsbad, CA (R.G.L.); and Department of Internal Medicine-Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC (J.S.P.)
| | - Robert N Helsley
- From the Department of Cellular and Molecular Medicine, Lerner Research Institute (A.D.G., R.C.S., A.L.B., R.N.H., A.C.B., D.F., R.Z., R.E.M., J.M.B.) and Department of Anatomical Pathology (D.S.A.), Cleveland Clinic, OH; Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (B.E.S., M.S.); Cardiovascular Group, Antisense Drug Discovery, Ionis Pharmaceuticals, Inc, Carlsbad, CA (R.G.L.); and Department of Internal Medicine-Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC (J.S.P.)
| | - Amy C Burrows
- From the Department of Cellular and Molecular Medicine, Lerner Research Institute (A.D.G., R.C.S., A.L.B., R.N.H., A.C.B., D.F., R.Z., R.E.M., J.M.B.) and Department of Anatomical Pathology (D.S.A.), Cleveland Clinic, OH; Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (B.E.S., M.S.); Cardiovascular Group, Antisense Drug Discovery, Ionis Pharmaceuticals, Inc, Carlsbad, CA (R.G.L.); and Department of Internal Medicine-Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC (J.S.P.)
| | - Daniel Ferguson
- From the Department of Cellular and Molecular Medicine, Lerner Research Institute (A.D.G., R.C.S., A.L.B., R.N.H., A.C.B., D.F., R.Z., R.E.M., J.M.B.) and Department of Anatomical Pathology (D.S.A.), Cleveland Clinic, OH; Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (B.E.S., M.S.); Cardiovascular Group, Antisense Drug Discovery, Ionis Pharmaceuticals, Inc, Carlsbad, CA (R.G.L.); and Department of Internal Medicine-Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC (J.S.P.)
| | - Renliang Zhang
- From the Department of Cellular and Molecular Medicine, Lerner Research Institute (A.D.G., R.C.S., A.L.B., R.N.H., A.C.B., D.F., R.Z., R.E.M., J.M.B.) and Department of Anatomical Pathology (D.S.A.), Cleveland Clinic, OH; Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (B.E.S., M.S.); Cardiovascular Group, Antisense Drug Discovery, Ionis Pharmaceuticals, Inc, Carlsbad, CA (R.G.L.); and Department of Internal Medicine-Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC (J.S.P.)
| | - Brian E Sansbury
- From the Department of Cellular and Molecular Medicine, Lerner Research Institute (A.D.G., R.C.S., A.L.B., R.N.H., A.C.B., D.F., R.Z., R.E.M., J.M.B.) and Department of Anatomical Pathology (D.S.A.), Cleveland Clinic, OH; Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (B.E.S., M.S.); Cardiovascular Group, Antisense Drug Discovery, Ionis Pharmaceuticals, Inc, Carlsbad, CA (R.G.L.); and Department of Internal Medicine-Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC (J.S.P.)
| | - Richard G Lee
- From the Department of Cellular and Molecular Medicine, Lerner Research Institute (A.D.G., R.C.S., A.L.B., R.N.H., A.C.B., D.F., R.Z., R.E.M., J.M.B.) and Department of Anatomical Pathology (D.S.A.), Cleveland Clinic, OH; Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (B.E.S., M.S.); Cardiovascular Group, Antisense Drug Discovery, Ionis Pharmaceuticals, Inc, Carlsbad, CA (R.G.L.); and Department of Internal Medicine-Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC (J.S.P.)
| | - Richard E Morton
- From the Department of Cellular and Molecular Medicine, Lerner Research Institute (A.D.G., R.C.S., A.L.B., R.N.H., A.C.B., D.F., R.Z., R.E.M., J.M.B.) and Department of Anatomical Pathology (D.S.A.), Cleveland Clinic, OH; Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (B.E.S., M.S.); Cardiovascular Group, Antisense Drug Discovery, Ionis Pharmaceuticals, Inc, Carlsbad, CA (R.G.L.); and Department of Internal Medicine-Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC (J.S.P.)
| | - Daniela S Allende
- From the Department of Cellular and Molecular Medicine, Lerner Research Institute (A.D.G., R.C.S., A.L.B., R.N.H., A.C.B., D.F., R.Z., R.E.M., J.M.B.) and Department of Anatomical Pathology (D.S.A.), Cleveland Clinic, OH; Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (B.E.S., M.S.); Cardiovascular Group, Antisense Drug Discovery, Ionis Pharmaceuticals, Inc, Carlsbad, CA (R.G.L.); and Department of Internal Medicine-Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC (J.S.P.)
| | - John S Parks
- From the Department of Cellular and Molecular Medicine, Lerner Research Institute (A.D.G., R.C.S., A.L.B., R.N.H., A.C.B., D.F., R.Z., R.E.M., J.M.B.) and Department of Anatomical Pathology (D.S.A.), Cleveland Clinic, OH; Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (B.E.S., M.S.); Cardiovascular Group, Antisense Drug Discovery, Ionis Pharmaceuticals, Inc, Carlsbad, CA (R.G.L.); and Department of Internal Medicine-Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC (J.S.P.)
| | - Matthew Spite
- From the Department of Cellular and Molecular Medicine, Lerner Research Institute (A.D.G., R.C.S., A.L.B., R.N.H., A.C.B., D.F., R.Z., R.E.M., J.M.B.) and Department of Anatomical Pathology (D.S.A.), Cleveland Clinic, OH; Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (B.E.S., M.S.); Cardiovascular Group, Antisense Drug Discovery, Ionis Pharmaceuticals, Inc, Carlsbad, CA (R.G.L.); and Department of Internal Medicine-Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC (J.S.P.)
| | - J Mark Brown
- From the Department of Cellular and Molecular Medicine, Lerner Research Institute (A.D.G., R.C.S., A.L.B., R.N.H., A.C.B., D.F., R.Z., R.E.M., J.M.B.) and Department of Anatomical Pathology (D.S.A.), Cleveland Clinic, OH; Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (B.E.S., M.S.); Cardiovascular Group, Antisense Drug Discovery, Ionis Pharmaceuticals, Inc, Carlsbad, CA (R.G.L.); and Department of Internal Medicine-Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC (J.S.P.).
| |
Collapse
|
157
|
Nomura S, Endo-Umeda K, Fujii S, Makishima M, Hashimoto Y, Ishikawa M. Structural development of tetrachlorophthalimides as liver X receptor β (LXRβ)-selective agonists with improved aqueous solubility. Bioorg Med Chem Lett 2017; 28:796-801. [PMID: 29398545 DOI: 10.1016/j.bmcl.2017.12.024] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 12/08/2017] [Accepted: 12/11/2017] [Indexed: 10/18/2022]
Abstract
LXRβ-selective agonists are promising candidates to improve atherosclerosis without increasing plasma or hepatic TG levels. We have reported a series of tetrachlorophthalimide analogs as an LXRβ-selective agonist. However, they exhibited poor aqueous solubility probably due to its high hydrophobicity and highly rigid and plane structure. In this report, we present further structural development of tetrachloro(styrylphenyl)phthalimides as the LXRβ-selective agonists with improved aqueous solubility.
Collapse
Affiliation(s)
- Sayaka Nomura
- Institute of Molecular and Cellular Biosciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-0032, Japan
| | - Kaori Endo-Umeda
- Nihon University School of Medicine, 30-1 Oyaguchi-kamicho, Itabashi-ku, Tokyo 173-8610, Japan
| | - Shinya Fujii
- Institute of Molecular and Cellular Biosciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-0032, Japan
| | - Makoto Makishima
- Nihon University School of Medicine, 30-1 Oyaguchi-kamicho, Itabashi-ku, Tokyo 173-8610, Japan
| | - Yuichi Hashimoto
- Institute of Molecular and Cellular Biosciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-0032, Japan
| | - Minoru Ishikawa
- Institute of Molecular and Cellular Biosciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-0032, Japan.
| |
Collapse
|
158
|
Liberale L, Dallegri F, Carbone F, Montecucco F. Pathophysiological relevance of macrophage subsets in atherogenesis. Thromb Haemost 2017; 117:7-18. [PMID: 27683760 DOI: 10.1160/th16-08-0593] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Accepted: 09/12/2016] [Indexed: 12/14/2022]
Abstract
SummaryMacrophages are highly heterogeneous and plastic cells. They were shown to play a critical role in all stages of atherogenesis, from the initiation to the necrotic core formation and plaque rupture. Lesional macrophages primarily derive from blood monocyte, but local macrophage proliferation as well as differentiation from smooth muscle cells have also been described. Within atherosclerotic plaques, macrophages rapidly respond to changes in the microenvironment, shifting between pro- (M1) or anti-inflammatory (M2) functional phenotypes. Furthermore, different stimuli have been associated with differentiation of newly discovered M2 subtypes: IL-4/IL-13 (M2a), immunecomplex (M2b), IL-10/glucocorticoids (M2c), and adenosine receptor agonist (M2d). More recently, additional intraplaque macrophage phenotypes were also recognized in response to CXCL4 (M4), oxidized phospholipids (Mox), haemoglobin/haptoglobin complexes (HAmac/M(Hb)), and heme (Mhem). Such macrophage polarization was described as a progression among multiple phenotypes, which reflect the activity of different transcriptional factors and the cross-talk between intracellular signalling. Finally, the distribution of macrophage subsets within different plaque areas was markedly associated with cardiovascular (CV) vulnerability. The aim of this review is to update the current knowledge on the role of macrophage subsets in atherogenesis. In addition, the molecular mechanisms underlying macrophage phenotypic shift will be summarised and discussed. Finally, the role of intraplaque macrophages as predictors of CV events and the therapeutic potential of these cells will be discussed.
Collapse
|
159
|
Emerging role of various signaling pathways in the pathogenesis and therapeutics of atherosclerosis. ACTA ACUST UNITED AC 2017. [DOI: 10.1016/j.rvm.2017.05.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
160
|
Zhang L, Rajbhandari P, Priest C, Sandhu J, Wu X, Temel R, Castrillo A, de Aguiar Vallim TQ, Sallam T, Tontonoz P. Inhibition of cholesterol biosynthesis through RNF145-dependent ubiquitination of SCAP. eLife 2017; 6:e28766. [PMID: 29068315 PMCID: PMC5656429 DOI: 10.7554/elife.28766] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 10/05/2017] [Indexed: 12/25/2022] Open
Abstract
Cholesterol homeostasis is maintained through concerted action of the SREBPs and LXRs. Here, we report that RNF145, a previously uncharacterized ER membrane ubiquitin ligase, participates in crosstalk between these critical signaling pathways. RNF145 expression is induced in response to LXR activation and high-cholesterol diet feeding. Transduction of RNF145 into mouse liver inhibits the expression of genes involved in cholesterol biosynthesis and reduces plasma cholesterol levels. Conversely, acute suppression of RNF145 via shRNA-mediated knockdown, or chronic inactivation of RNF145 by genetic deletion, potentiates the expression of cholesterol biosynthetic genes and increases cholesterol levels both in liver and plasma. Mechanistic studies show that RNF145 triggers ubiquitination of SCAP on lysine residues within a cytoplasmic loop essential for COPII binding, potentially inhibiting its transport to Golgi and subsequent processing of SREBP-2. These findings define an additional mechanism linking hepatic sterol levels to the reciprocal actions of the SREBP-2 and LXR pathways.
Collapse
Affiliation(s)
- Li Zhang
- Department of Pathology and Laboratory MedicineHoward Hughes Medical Institute, University of California, Los AngelesLos AngelesUnited States
| | - Prashant Rajbhandari
- Department of Pathology and Laboratory MedicineHoward Hughes Medical Institute, University of California, Los AngelesLos AngelesUnited States
| | - Christina Priest
- Department of Pathology and Laboratory MedicineHoward Hughes Medical Institute, University of California, Los AngelesLos AngelesUnited States
| | - Jaspreet Sandhu
- Department of Pathology and Laboratory MedicineHoward Hughes Medical Institute, University of California, Los AngelesLos AngelesUnited States
| | - Xiaohui Wu
- Department of Medicine, Division of CardiologyUniversity of California, Los AngelesLos AngelesUnited States
| | - Ryan Temel
- Saha Cardiovascular Research CenterUniversity of KentuckyLexingtonUnited States
- Department of Pharmacology and Nutritional SciencesUniversity of KentuckyLexingtonUnited States
| | - Antonio Castrillo
- Instituto de Investigaciones Biomédicas Alberto SolsCSIC-Universidad Autónoma de Madrid, Unidad de Biomedicina-Universidad de Las Palmas de Gran Canaria (Unidad asociada al CSIC)Las Palmas de Gran CanariaSpain
- Instituto Universitario de Investigaciones Biomédicas y SanitariasUniversidad de Las Palmas de Gran CanariaLas Palmas de Gran CanariaSpain
| | - Thomas Q de Aguiar Vallim
- Department of Medicine, Division of CardiologyUniversity of California, Los AngelesLos AngelesUnited States
| | - Tamer Sallam
- Department of Medicine, Division of CardiologyUniversity of California, Los AngelesLos AngelesUnited States
| | - Peter Tontonoz
- Department of Pathology and Laboratory MedicineHoward Hughes Medical Institute, University of California, Los AngelesLos AngelesUnited States
| |
Collapse
|
161
|
Functional diversity of macrophages in vascular biology and disease. Vascul Pharmacol 2017; 99:13-22. [PMID: 29074468 DOI: 10.1016/j.vph.2017.10.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Accepted: 10/19/2017] [Indexed: 12/24/2022]
Abstract
Atherosclerosis is a multifactorial chronic inflammatory disease and is largely responsible for cardiovascular disease, the most common cause of global mortality. The hallmark of atherogenesis is immune activation following lipid accumulation in the arterial wall. In particular, macrophages play a non-redundant role in both the progression and regression of inflammation in the atherosclerotic lesion. Macrophages are remarkably heterogeneous phagocytes that perform versatile functions in health and disease. Their functional diversity in vascular biology is only partially mapped. Targeting macrophages is often highlighted as a therapeutic approach for cancer, metabolic and inflammatory diseases. Future strategies for therapeutic intervention in atherosclerosis may benefit from attempts to reduce local proliferation of pro-inflammatory macrophage subsets or enhance resolution of inflammation. Thus, characterisation of macrophage subsets during atherosclerosis would empower clinical interventions. Therefore, it would be of fundamental importance to understand how pathological factors modulate macrophage activity in order to exploit their use in the treatment of atherosclerosis and other diseases.
Collapse
|
162
|
Ju X, Huang P, Chen M, Wang Q. Liver X receptors as potential targets for cancer therapeutics. Oncol Lett 2017; 14:7676-7680. [PMID: 29250170 DOI: 10.3892/ol.2017.7259] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Accepted: 09/07/2017] [Indexed: 11/06/2022] Open
Abstract
Liver X receptors (LXRs) are important members of the nuclear receptor family that were originally determined to function in cholesterol transport and the regulation of immune responses. Synthetic LXR ligands have been developed to treat various diseases including diabetes, Alzheimer's disease and atherosclerosis. Previous studies have suggested that LXRs are also involved in numerous types of cancer and are therefore potential targets for cancer therapeutics. The present review summarizes LXR ligands and their mechanisms of action, the effects of LXRs in different types of cancer and their potential applications in clinical treatment. Together, the studies discussed in the present review indicate that LXRs may be potential targets for cancer therapeutics.
Collapse
Affiliation(s)
- Xiaoli Ju
- Department of Pathology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Pan Huang
- Department of Pathology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Miao Chen
- Department of Pathology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China.,Department of Pathology, The Affiliated People's Hospital, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Qiang Wang
- Institute of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| |
Collapse
|
163
|
Nagenborg J, Goossens P, Biessen EAL, Donners MMPC. Heterogeneity of atherosclerotic plaque macrophage origin, phenotype and functions: Implications for treatment. Eur J Pharmacol 2017; 816:14-24. [PMID: 28989084 DOI: 10.1016/j.ejphar.2017.10.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Revised: 10/02/2017] [Accepted: 10/04/2017] [Indexed: 01/01/2023]
Abstract
Macrophages are key players in atherosclerotic lesions, regulating the local inflammatory milieu and plaque stability by the secretion of many inflammatory molecules, growth factors and cytokines. Monocytes have long been considered to be the main source of plaque macrophages. However, recent findings provide evidence for proliferation of local macrophages or transdifferentiation from other vascular cells as alternative sources. Recent years of research focused on the further identification and characterisation of macrophage phenotypes and functions. In this review we describe the advances in our understanding of monocyte and macrophage heterogeneity and its implications for specific therapeutic interventions, aiming to reduce the ever growing significant risk of cardiovascular events without any detrimental side effects on the patient's immune response.
Collapse
Affiliation(s)
- Jan Nagenborg
- Department of Pathology, CARIM, Maastricht University, 6200 MD Maastricht, the Netherlands
| | - Pieter Goossens
- Department of Pathology, CARIM, Maastricht University, 6200 MD Maastricht, the Netherlands
| | - Erik A L Biessen
- Department of Pathology, CARIM, Maastricht University, 6200 MD Maastricht, the Netherlands
| | - Marjo M P C Donners
- Department of Pathology, CARIM, Maastricht University, 6200 MD Maastricht, the Netherlands.
| |
Collapse
|
164
|
Wang J, Xu P, Xie X, Li J, Zhang J, Wang J, Hong F, Li J, Zhang Y, Song Y, Zheng X, Zhai Y. DBZ (Danshensu Bingpian Zhi), a Novel Natural Compound Derivative, Attenuates Atherosclerosis in Apolipoprotein E-Deficient Mice. J Am Heart Assoc 2017; 6:e006297. [PMID: 28971954 PMCID: PMC5721843 DOI: 10.1161/jaha.117.006297] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 07/07/2017] [Indexed: 01/01/2023]
Abstract
BACKGROUND DBZ (Danshensu Bingpian Zhi), a synthetic derivative of a natural compound found in traditional Chinese medicine, has been reported to suppress lipopolysaccharide-induced macrophage activation and lipid accumulation in vitro. The aim of this study was to assess whether DBZ could attenuate atherosclerosis at early and advanced stages. METHODS AND RESULTS The effects of DBZ on the development of atherosclerosis were studied using apolipoprotein E-deficient (apoE-/-) mice. For early treatment, 5-week-old apoE-/- mice were fed a Western diet and treated daily by oral gavage with or without DBZ or atorvastatin for 10 weeks. For advanced treatment, 5-week-old apoE-/- mice were fed a Western diet for 10 weeks to induce atherosclerosis, and then they were randomly divided into 4 groups and subjected to the treatment of vehicle, 20 mg/kg per day DBZ, 40 mg/kg per day DBZ, or 10 mg/kg per day atorvastatin for the subsequent 10 weeks. We showed that early treatment of apoE-/- mice with DBZ markedly reduced atherosclerotic lesion formation by inhibiting inflammation and decreasing macrophage infiltration into the vessel wall. Treatment with DBZ also attenuated the progression of preestablished diet-induced atherosclerotic plaques in apoE-/- mice. In addition, we showed that DBZ may affect LXR (liver X receptor) function and that treatment of macrophages with DBZ suppressed lipopolysaccharide-stimulated cell migration and oxidized low-density lipoprotein-induced foam cell formation. CONCLUSIONS DBZ potentially has antiatherosclerotic effects that involve the inhibition of inflammation, macrophage migration, leukocyte adhesion, and foam cell formation. These results suggest that DBZ may be used as a therapeutic agent for the prevention and treatment of atherosclerosis.
Collapse
Affiliation(s)
- Jing Wang
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing, China
| | - Pengfei Xu
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing, China
| | - Xinni Xie
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing, China
- State key laboratory of environmental chemistry and ecotoxicology Research Center for Eco-Environmental Science Chinese Academy of Science, Beijing, China
| | - Jiao Li
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing, China
| | - Jun Zhang
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing, China
| | - Jialin Wang
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing, China
- Department of Biology Science and Technology, Baotou Teacher's College, Baotou, China
- State key laboratory of environmental chemistry and ecotoxicology Research Center for Eco-Environmental Science Chinese Academy of Science, Beijing, China
| | - Fan Hong
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing, China
| | - Jian Li
- Beijing Hospital, National Center of Gerontology, Beijing, China
| | - Youyi Zhang
- Key Laboratory for Cell Proliferation and Regulation Biology of State Education Ministry and College of Life Sciences, Beijing Normal University, Beijing, China
- Institute of Vascular Medicine, Peking University Third Hospital and Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, Beijing, China
- Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education and Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing, China
| | - Yao Song
- Institute of Vascular Medicine, Peking University Third Hospital and Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, Beijing, China
- Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education and Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing, China
| | - Xiaohui Zheng
- Key Laboratory of Resource Biology and Biotechnology in Western China and College of Life Sciences Northwest University, Xi'an, China
| | - Yonggong Zhai
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing, China
| |
Collapse
|
165
|
Liver X receptors agonist T0901317 downregulates matrix metalloproteinase-9 expression in non-small-cell lung cancer by repressing nuclear factor-κB. Anticancer Drugs 2017; 28:952-958. [DOI: 10.1097/cad.0000000000000532] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
166
|
Yu M, Amengual J, Menon A, Kamaly N, Zhou F, Xu X, Saw PE, Lee SJ, Si K, Ortega CA, Choi WI, Lee IH, Bdour Y, Shi J, Mahmoudi M, Jon S, Fisher EA, Farokhzad OC. Targeted Nanotherapeutics Encapsulating Liver X Receptor Agonist GW3965 Enhance Antiatherogenic Effects without Adverse Effects on Hepatic Lipid Metabolism in Ldlr -/- Mice. Adv Healthc Mater 2017; 6:10.1002/adhm.201700313. [PMID: 28730752 PMCID: PMC5656530 DOI: 10.1002/adhm.201700313] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Revised: 04/27/2017] [Indexed: 11/11/2022]
Abstract
The pharmacological manipulation of liver X receptors (LXRs) has been an attractive therapeutic strategy for atherosclerosis treatment as they control reverse cholesterol transport and inflammatory response. This study presents the development and efficacy of nanoparticles (NPs) incorporating the synthetic LXR agonist GW3965 (GW) in targeting atherosclerotic lesions. Collagen IV (Col IV) targeting ligands are employed to functionalize the NPs to improve targeting to the atherosclerotic plaque, and formulation parameters such as the length of the polyethylene glycol (PEG) coating molecules are systematically optimized. In vitro studies indicate that the GW-encapsulated NPs upregulate the LXR target genes and downregulate proinflammatory mediator in macrophages. The Col IV-targeted NPs encapsulating GW (Col IV-GW-NPs) successfully reaches atherosclerotic lesions when administered for 5 weeks to mice with preexisting lesions, substantially reducing macrophage content (≈30%) compared to the PBS group, which is with greater efficacy versus nontargeting NPs encapsulating GW (GW-NPs) (≈18%). In addition, mice administered the Col IV-GW-NPs do not demonstrate increased hepatic lipid biosynthesis or hyperlipidemia during the treatment period, unlike mice injected with the free GW. These findings suggest a new form of LXR-based therapeutics capable of enhanced delivery of the LXR agonist to atherosclerotic lesions without altering hepatic lipid metabolism.
Collapse
Affiliation(s)
- Mikyung Yu
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Jaume Amengual
- Division of Cardiology, Department of Medicine, Marc and Ruti Bell Program in Vascular Biology, New York University School of Medicine, New York, NY, 10016, USA
| | - Arjun Menon
- Division of Cardiology, Department of Medicine, Marc and Ruti Bell Program in Vascular Biology, New York University School of Medicine, New York, NY, 10016, USA
| | - Nazila Kamaly
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- Department of Micro and Nanotechnology, Technical University of Denmark, DTU Nanotech, 2800 Kgs. Lyngby, Denmark
| | - Felix Zhou
- Division of Cardiology, Department of Medicine, Marc and Ruti Bell Program in Vascular Biology, New York University School of Medicine, New York, NY, 10016, USA
| | - Xiaoding Xu
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Phei Er Saw
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Seung-Joo Lee
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, 240 Longwood Ave., Boston, MA, 02115, USA
| | - Kevin Si
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Carleena Angelica Ortega
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Won Il Choi
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- Center for Convergence Bioceramic Materials, Convergence R&D Division, Korea Institute of Ceramic Engineering and Technology, 202, Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-guCheongju, Chungbuk, 28160, Republic of Korea
| | - In-Hyun Lee
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Yazan Bdour
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Jinjun Shi
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Morteza Mahmoudi
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Sangyong Jon
- KAIST Institute for the BioCentury, Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Republic of Korea
| | - Edward A Fisher
- Division of Cardiology, Department of Medicine, Marc and Ruti Bell Program in Vascular Biology, New York University School of Medicine, New York, NY, 10016, USA
| | - Omid C Farokhzad
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- College of Pharmacy, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| |
Collapse
|
167
|
Xu Y, Niu Y, Gao Y, Wang F, Qin W, Lu Y, Hu J, Peng L, Liu J, Xiong W. Borapetoside E, a Clerodane Diterpenoid Extracted from Tinospora crispa, Improves Hyperglycemia and Hyperlipidemia in High-Fat-Diet-Induced Type 2 Diabetes Mice. JOURNAL OF NATURAL PRODUCTS 2017; 80:2319-2327. [PMID: 28742368 DOI: 10.1021/acs.jnatprod.7b00365] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
An insidious increase in the incidence of obesity, insulin resistance, and hyperlipidemia has led to an epidemic of type 2 diabetes worldwide. Tinospora crispa (T. crispa) is a familiar plant traditionally used in herbal medicine for diabetes; however, the major active ingredients of this plant are still unclear. In this study, we identified the therapeutic effects of borapetoside E, a small molecule extracted from T. crispa, in high-fat-diet (HFD)-induced obesity in mice. The therapeutic effects of borapetoside E in HFD-induced obese mice were assessed physiologically, histologically, and biochemically following intraperitoneal injection. Furthermore, we analyzed the expression of glucose and lipid metabolism-related genes and proteins in borapetoside E-treated obese mice. Compared with vehicle-treated mice, borapetoside E markedly improved hyperglycemia, insulin resistance, hepatic steatosis, hyperlipidemia, and oxygen consumption in obese mice, and the effects were comparable to or better than the drug metformin. In addition, borapetoside E suppressed the expression of sterol regulatory element binding proteins (SREBPs) and their downstream target genes related to lipid synthesis in the liver and adipose tissue. Borapetoside E showed beneficial effects in vivo, demonstrating that borapetoside E may be a potential therapy for the treatment of diet-induced type 2 diabetes and related metabolic syndromes.
Collapse
Affiliation(s)
- Yuhui Xu
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences , Kunming 650201, People's Republic of China
- University of the Chinese Academy of Sciences , Beijing 100049, People's Republic of China
| | - Yanfen Niu
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences , Kunming 650201, People's Republic of China
- University of the Chinese Academy of Sciences , Beijing 100049, People's Republic of China
- Engineering Research Center, Kunming Medical University , Kunming 650500, People's Republic of China
| | - Yuan Gao
- BioBioPha Co., Ltd , Kunming 650201, People's Republic of China
- Deparment of Chemical Engineering, Yibin University , Yibin 644000, People's Republic of China
| | - Fang Wang
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences , Kunming 650201, People's Republic of China
- University of the Chinese Academy of Sciences , Beijing 100049, People's Republic of China
| | - Wanying Qin
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences , Kunming 650201, People's Republic of China
- University of the Chinese Academy of Sciences , Beijing 100049, People's Republic of China
| | - Yanting Lu
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences , Kunming 650201, People's Republic of China
- University of the Chinese Academy of Sciences , Beijing 100049, People's Republic of China
| | - Jing Hu
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences , Kunming 650201, People's Republic of China
- University of the Chinese Academy of Sciences , Beijing 100049, People's Republic of China
| | - Li Peng
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences , Kunming 650201, People's Republic of China
- University of the Chinese Academy of Sciences , Beijing 100049, People's Republic of China
| | - Jikai Liu
- School of Pharmaceutical Sciences, South-Central University for Nationalities , Wuhan 430074, People's Republic of China
| | - Wenyong Xiong
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences , Kunming 650201, People's Republic of China
- University of the Chinese Academy of Sciences , Beijing 100049, People's Republic of China
| |
Collapse
|
168
|
Lai C, Cheng H, Lin C, Huang S, Chen T, Chung C, Chang C, Wang H, Chuu C. Activation of liver X receptor suppresses angiogenesis
via
induction of ApoD. FASEB J 2017; 31:5568-5576. [DOI: 10.1096/fj.201700374r] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 08/07/2017] [Indexed: 01/25/2023]
Affiliation(s)
- Chih‐Jen Lai
- Institute of Cellular and System MedicineNational Health Research Institutes Miaoli Taiwan
- Institute of BiotechnologyNational Tsing Hua University Hsinchu Taiwan
| | - Hsu‐Chen Cheng
- Department of Life SciencesNational Chung Hsing University Taichung Taiwan
| | - Ching‐Yu Lin
- Institute of Cellular and System MedicineNational Health Research Institutes Miaoli Taiwan
| | - Shih‐Han Huang
- Institute of Cellular and System MedicineNational Health Research Institutes Miaoli Taiwan
- Department of Life SciencesNational Central University Taoyuan Taiwan
| | - Ting‐Huan Chen
- Institute of Cellular and System MedicineNational Health Research Institutes Miaoli Taiwan
- Institute of BiotechnologyNational Tsing Hua University Hsinchu Taiwan
| | - Chi‐Jung Chung
- Department of Health Risk ManagementChina Medical University Taichung Taiwan
| | - Chung‐Ho Chang
- Institute of Cellular and System MedicineNational Health Research Institutes Miaoli Taiwan
- Department of Internal MedicineChanghua Christian Hospital Changhua Taiwan
| | - Horng‐Dar Wang
- Institute of BiotechnologyNational Tsing Hua University Hsinchu Taiwan
| | - Chih‐Pin Chuu
- Institute of Cellular and System MedicineNational Health Research Institutes Miaoli Taiwan
- Biotechnology CenterNational Chung Hsing University Taichung Taiwan
- Graduate Institute of Basic Medical ScienceChina Medical University Taichung Taiwan
- Graduate Program for AgingChina Medical University Taichung Taiwan
| |
Collapse
|
169
|
Gu M, Zhang Y, Liu C, Wang D, Feng L, Fan S, Yang B, Tong Q, Ji G, Huang C. Morin, a novel liver X receptor α/β dual antagonist, has potent therapeutic efficacy for nonalcoholic fatty liver diseases. Br J Pharmacol 2017. [PMID: 28646531 DOI: 10.1111/bph.13933] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND AND PURPOSE Morin is a natural occurring flavonoid in many dietary plants and has a wide range of beneficial effects on metabolism; however, the mechanism underlying its action remains elusive. EXPERIMENTAL APPROACH A reporter assay and the time-resolved FRET assay were used to identify morin as a dual antagonist of liver X receptor (LXR)-α and -β. Morin (100 mg. 100 g-1 diet) was administered to high-fat diet-induced obese or LXRβ-/- mice. The pharmacological effects and mechanism of action of morin were evaluated by Western blot and RT-PCR analyses. KEY RESULTS From the in vitro assays, morin was shown to be a dual antagonist of LXRα and LXRβ. In vivo, morin blunted the development of liver hepatic steatosis, reduced body weight gains, lowered triglyceride levels and improved glucose and insulin tolerance in mice fed a high-fat diet. Mechanistically, morin inhibited 3T3-L1 adipocyte differentiation and lipid formation in human hepatic HepG2 cells and suppressed the mRNA expression of genes downstream of LXR. Consistently, the effects of morin on metabolic disorders were attenuated in LXRβ-/- mice. CONCLUSION AND IMPLICATIONS Our data reveal that morin is a dual antagonist of LXRα and LXRβ and suggest that morin may alleviate hepatic steatosis and other associated metabolic disorders via the suppression of LXR signalling and, therefore, shows promise as a novel therapy or nutraceutical for nonalcoholic fatty liver disease.
Collapse
Affiliation(s)
- Ming Gu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yu Zhang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,School of Life Science and Technology, Shanghai Tech University, Shanghai, China
| | - Chuhe Liu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Dongshan Wang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Li Feng
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Shengjie Fan
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Brown Foundation Institute of Molecular Medicine and Program in Neuroscience, Graduate School of Biological Sciences, University of Texas McGovern Medical School, Houston, TX, USA
| | - Baican Yang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qingchun Tong
- Brown Foundation Institute of Molecular Medicine and Program in Neuroscience, Graduate School of Biological Sciences, University of Texas McGovern Medical School, Houston, TX, USA
| | - Guang Ji
- Institute of Digestive Disease, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Cheng Huang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
170
|
Li Z, Martin M, Zhang J, Huang HY, Bai L, Zhang J, Kang J, He M, Li J, Maurya MR, Gupta S, Zhou G, Sangwung P, Xu YJ, Lei T, Huang HD, Jain M, Jain MK, Subramaniam S, Shyy JYJ. Krüppel-Like Factor 4 Regulation of Cholesterol-25-Hydroxylase and Liver X Receptor Mitigates Atherosclerosis Susceptibility. Circulation 2017; 136:1315-1330. [PMID: 28794002 DOI: 10.1161/circulationaha.117.027462] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 07/31/2017] [Indexed: 12/30/2022]
Abstract
BACKGROUND Atherosclerosis is a multifaceted inflammatory disease involving cells in the vascular wall (eg, endothelial cells [ECs]), as well as circulating and resident immunogenic cells (eg, monocytes/macrophages). Acting as a ligand for liver X receptor (LXR), but an inhibitor of SREBP2 (sterol regulatory element-binding protein 2), 25-hydroxycholesterol, and its catalyzing enzyme cholesterol-25-hydroxylase (Ch25h) are important in regulating cellular inflammatory status and cholesterol biosynthesis in both ECs and monocytes/macrophages. METHODS Bioinformatic analyses were used to investigate RNA-sequencing data to identify cholesterol oxidation and efflux genes regulated by Krüppel-like factor 4 (KLF4). In vitro experiments involving cultured ECs and macrophages and in vivo methods involving mice with Ch25h ablation were then used to explore the atheroprotective role of KLF4-Ch25h/LXR. RESULTS Vasoprotective stimuli increased the expression of Ch25h and LXR via KLF4. The KLF4-Ch25h/LXR homeostatic axis functions through suppressing inflammation, evidenced by the reduction of inflammasome activity in ECs and the promotion of M1 to M2 phenotypic transition in macrophages. The increased atherosclerosis in apolipoprotein E-/-/Ch25h-/- mice further demonstrates the beneficial role of the KLF4-Ch25h/LXR axis in vascular function and disease. CONCLUSIONS KLF4 transactivates Ch25h and LXR, thereby promoting the synergistic effects between ECs and macrophages to protect against atherosclerosis susceptibility.
Collapse
Affiliation(s)
- Zhao Li
- From Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, China (Z.L., Jin Zhang, L.B., Jiao Zhang, M.H., J.L., T.L., J.Y.-J.S.); Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education of China (Z.L., Jin Zhang, L.B., Jiao Zhang, M.H., J.L., T.L., J.Y.-J.S.); Division of Cardiology, Department of Medicine, University of California, San Diego, La Jolla (M.M., Jin Zhang, J.K., M.H., Y.-J.X., M.J., J.Y.-J.S.);Department of Bioengineering, University of California, San Diego, La Jolla (M.R.M., S.G.); Division of Biochemistry and Molecular Biology, University of California, Riverside (M.M.); Institute of Bioinformatics and Systems Biology and Department of Biological Science and Technology, National Chiao Tung University, Hsin-Chu, Taiwan (H.-Y.H., H.-D.H.); and Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH (G.Z., P.S., M.K.J.)
| | - Marcy Martin
- From Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, China (Z.L., Jin Zhang, L.B., Jiao Zhang, M.H., J.L., T.L., J.Y.-J.S.); Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education of China (Z.L., Jin Zhang, L.B., Jiao Zhang, M.H., J.L., T.L., J.Y.-J.S.); Division of Cardiology, Department of Medicine, University of California, San Diego, La Jolla (M.M., Jin Zhang, J.K., M.H., Y.-J.X., M.J., J.Y.-J.S.);Department of Bioengineering, University of California, San Diego, La Jolla (M.R.M., S.G.); Division of Biochemistry and Molecular Biology, University of California, Riverside (M.M.); Institute of Bioinformatics and Systems Biology and Department of Biological Science and Technology, National Chiao Tung University, Hsin-Chu, Taiwan (H.-Y.H., H.-D.H.); and Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH (G.Z., P.S., M.K.J.)
| | - Jin Zhang
- From Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, China (Z.L., Jin Zhang, L.B., Jiao Zhang, M.H., J.L., T.L., J.Y.-J.S.); Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education of China (Z.L., Jin Zhang, L.B., Jiao Zhang, M.H., J.L., T.L., J.Y.-J.S.); Division of Cardiology, Department of Medicine, University of California, San Diego, La Jolla (M.M., Jin Zhang, J.K., M.H., Y.-J.X., M.J., J.Y.-J.S.);Department of Bioengineering, University of California, San Diego, La Jolla (M.R.M., S.G.); Division of Biochemistry and Molecular Biology, University of California, Riverside (M.M.); Institute of Bioinformatics and Systems Biology and Department of Biological Science and Technology, National Chiao Tung University, Hsin-Chu, Taiwan (H.-Y.H., H.-D.H.); and Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH (G.Z., P.S., M.K.J.)
| | - Hsi-Yuan Huang
- From Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, China (Z.L., Jin Zhang, L.B., Jiao Zhang, M.H., J.L., T.L., J.Y.-J.S.); Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education of China (Z.L., Jin Zhang, L.B., Jiao Zhang, M.H., J.L., T.L., J.Y.-J.S.); Division of Cardiology, Department of Medicine, University of California, San Diego, La Jolla (M.M., Jin Zhang, J.K., M.H., Y.-J.X., M.J., J.Y.-J.S.);Department of Bioengineering, University of California, San Diego, La Jolla (M.R.M., S.G.); Division of Biochemistry and Molecular Biology, University of California, Riverside (M.M.); Institute of Bioinformatics and Systems Biology and Department of Biological Science and Technology, National Chiao Tung University, Hsin-Chu, Taiwan (H.-Y.H., H.-D.H.); and Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH (G.Z., P.S., M.K.J.)
| | - Liang Bai
- From Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, China (Z.L., Jin Zhang, L.B., Jiao Zhang, M.H., J.L., T.L., J.Y.-J.S.); Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education of China (Z.L., Jin Zhang, L.B., Jiao Zhang, M.H., J.L., T.L., J.Y.-J.S.); Division of Cardiology, Department of Medicine, University of California, San Diego, La Jolla (M.M., Jin Zhang, J.K., M.H., Y.-J.X., M.J., J.Y.-J.S.);Department of Bioengineering, University of California, San Diego, La Jolla (M.R.M., S.G.); Division of Biochemistry and Molecular Biology, University of California, Riverside (M.M.); Institute of Bioinformatics and Systems Biology and Department of Biological Science and Technology, National Chiao Tung University, Hsin-Chu, Taiwan (H.-Y.H., H.-D.H.); and Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH (G.Z., P.S., M.K.J.)
| | - Jiao Zhang
- From Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, China (Z.L., Jin Zhang, L.B., Jiao Zhang, M.H., J.L., T.L., J.Y.-J.S.); Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education of China (Z.L., Jin Zhang, L.B., Jiao Zhang, M.H., J.L., T.L., J.Y.-J.S.); Division of Cardiology, Department of Medicine, University of California, San Diego, La Jolla (M.M., Jin Zhang, J.K., M.H., Y.-J.X., M.J., J.Y.-J.S.);Department of Bioengineering, University of California, San Diego, La Jolla (M.R.M., S.G.); Division of Biochemistry and Molecular Biology, University of California, Riverside (M.M.); Institute of Bioinformatics and Systems Biology and Department of Biological Science and Technology, National Chiao Tung University, Hsin-Chu, Taiwan (H.-Y.H., H.-D.H.); and Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH (G.Z., P.S., M.K.J.)
| | - Jian Kang
- From Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, China (Z.L., Jin Zhang, L.B., Jiao Zhang, M.H., J.L., T.L., J.Y.-J.S.); Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education of China (Z.L., Jin Zhang, L.B., Jiao Zhang, M.H., J.L., T.L., J.Y.-J.S.); Division of Cardiology, Department of Medicine, University of California, San Diego, La Jolla (M.M., Jin Zhang, J.K., M.H., Y.-J.X., M.J., J.Y.-J.S.);Department of Bioengineering, University of California, San Diego, La Jolla (M.R.M., S.G.); Division of Biochemistry and Molecular Biology, University of California, Riverside (M.M.); Institute of Bioinformatics and Systems Biology and Department of Biological Science and Technology, National Chiao Tung University, Hsin-Chu, Taiwan (H.-Y.H., H.-D.H.); and Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH (G.Z., P.S., M.K.J.)
| | - Ming He
- From Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, China (Z.L., Jin Zhang, L.B., Jiao Zhang, M.H., J.L., T.L., J.Y.-J.S.); Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education of China (Z.L., Jin Zhang, L.B., Jiao Zhang, M.H., J.L., T.L., J.Y.-J.S.); Division of Cardiology, Department of Medicine, University of California, San Diego, La Jolla (M.M., Jin Zhang, J.K., M.H., Y.-J.X., M.J., J.Y.-J.S.);Department of Bioengineering, University of California, San Diego, La Jolla (M.R.M., S.G.); Division of Biochemistry and Molecular Biology, University of California, Riverside (M.M.); Institute of Bioinformatics and Systems Biology and Department of Biological Science and Technology, National Chiao Tung University, Hsin-Chu, Taiwan (H.-Y.H., H.-D.H.); and Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH (G.Z., P.S., M.K.J.)
| | - Jie Li
- From Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, China (Z.L., Jin Zhang, L.B., Jiao Zhang, M.H., J.L., T.L., J.Y.-J.S.); Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education of China (Z.L., Jin Zhang, L.B., Jiao Zhang, M.H., J.L., T.L., J.Y.-J.S.); Division of Cardiology, Department of Medicine, University of California, San Diego, La Jolla (M.M., Jin Zhang, J.K., M.H., Y.-J.X., M.J., J.Y.-J.S.);Department of Bioengineering, University of California, San Diego, La Jolla (M.R.M., S.G.); Division of Biochemistry and Molecular Biology, University of California, Riverside (M.M.); Institute of Bioinformatics and Systems Biology and Department of Biological Science and Technology, National Chiao Tung University, Hsin-Chu, Taiwan (H.-Y.H., H.-D.H.); and Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH (G.Z., P.S., M.K.J.)
| | - Mano R Maurya
- From Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, China (Z.L., Jin Zhang, L.B., Jiao Zhang, M.H., J.L., T.L., J.Y.-J.S.); Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education of China (Z.L., Jin Zhang, L.B., Jiao Zhang, M.H., J.L., T.L., J.Y.-J.S.); Division of Cardiology, Department of Medicine, University of California, San Diego, La Jolla (M.M., Jin Zhang, J.K., M.H., Y.-J.X., M.J., J.Y.-J.S.);Department of Bioengineering, University of California, San Diego, La Jolla (M.R.M., S.G.); Division of Biochemistry and Molecular Biology, University of California, Riverside (M.M.); Institute of Bioinformatics and Systems Biology and Department of Biological Science and Technology, National Chiao Tung University, Hsin-Chu, Taiwan (H.-Y.H., H.-D.H.); and Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH (G.Z., P.S., M.K.J.)
| | - Shakti Gupta
- From Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, China (Z.L., Jin Zhang, L.B., Jiao Zhang, M.H., J.L., T.L., J.Y.-J.S.); Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education of China (Z.L., Jin Zhang, L.B., Jiao Zhang, M.H., J.L., T.L., J.Y.-J.S.); Division of Cardiology, Department of Medicine, University of California, San Diego, La Jolla (M.M., Jin Zhang, J.K., M.H., Y.-J.X., M.J., J.Y.-J.S.);Department of Bioengineering, University of California, San Diego, La Jolla (M.R.M., S.G.); Division of Biochemistry and Molecular Biology, University of California, Riverside (M.M.); Institute of Bioinformatics and Systems Biology and Department of Biological Science and Technology, National Chiao Tung University, Hsin-Chu, Taiwan (H.-Y.H., H.-D.H.); and Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH (G.Z., P.S., M.K.J.)
| | - Guangjin Zhou
- From Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, China (Z.L., Jin Zhang, L.B., Jiao Zhang, M.H., J.L., T.L., J.Y.-J.S.); Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education of China (Z.L., Jin Zhang, L.B., Jiao Zhang, M.H., J.L., T.L., J.Y.-J.S.); Division of Cardiology, Department of Medicine, University of California, San Diego, La Jolla (M.M., Jin Zhang, J.K., M.H., Y.-J.X., M.J., J.Y.-J.S.);Department of Bioengineering, University of California, San Diego, La Jolla (M.R.M., S.G.); Division of Biochemistry and Molecular Biology, University of California, Riverside (M.M.); Institute of Bioinformatics and Systems Biology and Department of Biological Science and Technology, National Chiao Tung University, Hsin-Chu, Taiwan (H.-Y.H., H.-D.H.); and Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH (G.Z., P.S., M.K.J.)
| | - Panjamaporn Sangwung
- From Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, China (Z.L., Jin Zhang, L.B., Jiao Zhang, M.H., J.L., T.L., J.Y.-J.S.); Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education of China (Z.L., Jin Zhang, L.B., Jiao Zhang, M.H., J.L., T.L., J.Y.-J.S.); Division of Cardiology, Department of Medicine, University of California, San Diego, La Jolla (M.M., Jin Zhang, J.K., M.H., Y.-J.X., M.J., J.Y.-J.S.);Department of Bioengineering, University of California, San Diego, La Jolla (M.R.M., S.G.); Division of Biochemistry and Molecular Biology, University of California, Riverside (M.M.); Institute of Bioinformatics and Systems Biology and Department of Biological Science and Technology, National Chiao Tung University, Hsin-Chu, Taiwan (H.-Y.H., H.-D.H.); and Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH (G.Z., P.S., M.K.J.)
| | - Yong-Jiang Xu
- From Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, China (Z.L., Jin Zhang, L.B., Jiao Zhang, M.H., J.L., T.L., J.Y.-J.S.); Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education of China (Z.L., Jin Zhang, L.B., Jiao Zhang, M.H., J.L., T.L., J.Y.-J.S.); Division of Cardiology, Department of Medicine, University of California, San Diego, La Jolla (M.M., Jin Zhang, J.K., M.H., Y.-J.X., M.J., J.Y.-J.S.);Department of Bioengineering, University of California, San Diego, La Jolla (M.R.M., S.G.); Division of Biochemistry and Molecular Biology, University of California, Riverside (M.M.); Institute of Bioinformatics and Systems Biology and Department of Biological Science and Technology, National Chiao Tung University, Hsin-Chu, Taiwan (H.-Y.H., H.-D.H.); and Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH (G.Z., P.S., M.K.J.)
| | - Ting Lei
- From Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, China (Z.L., Jin Zhang, L.B., Jiao Zhang, M.H., J.L., T.L., J.Y.-J.S.); Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education of China (Z.L., Jin Zhang, L.B., Jiao Zhang, M.H., J.L., T.L., J.Y.-J.S.); Division of Cardiology, Department of Medicine, University of California, San Diego, La Jolla (M.M., Jin Zhang, J.K., M.H., Y.-J.X., M.J., J.Y.-J.S.);Department of Bioengineering, University of California, San Diego, La Jolla (M.R.M., S.G.); Division of Biochemistry and Molecular Biology, University of California, Riverside (M.M.); Institute of Bioinformatics and Systems Biology and Department of Biological Science and Technology, National Chiao Tung University, Hsin-Chu, Taiwan (H.-Y.H., H.-D.H.); and Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH (G.Z., P.S., M.K.J.)
| | - Hsien-Da Huang
- From Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, China (Z.L., Jin Zhang, L.B., Jiao Zhang, M.H., J.L., T.L., J.Y.-J.S.); Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education of China (Z.L., Jin Zhang, L.B., Jiao Zhang, M.H., J.L., T.L., J.Y.-J.S.); Division of Cardiology, Department of Medicine, University of California, San Diego, La Jolla (M.M., Jin Zhang, J.K., M.H., Y.-J.X., M.J., J.Y.-J.S.);Department of Bioengineering, University of California, San Diego, La Jolla (M.R.M., S.G.); Division of Biochemistry and Molecular Biology, University of California, Riverside (M.M.); Institute of Bioinformatics and Systems Biology and Department of Biological Science and Technology, National Chiao Tung University, Hsin-Chu, Taiwan (H.-Y.H., H.-D.H.); and Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH (G.Z., P.S., M.K.J.)
| | - Mohit Jain
- From Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, China (Z.L., Jin Zhang, L.B., Jiao Zhang, M.H., J.L., T.L., J.Y.-J.S.); Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education of China (Z.L., Jin Zhang, L.B., Jiao Zhang, M.H., J.L., T.L., J.Y.-J.S.); Division of Cardiology, Department of Medicine, University of California, San Diego, La Jolla (M.M., Jin Zhang, J.K., M.H., Y.-J.X., M.J., J.Y.-J.S.);Department of Bioengineering, University of California, San Diego, La Jolla (M.R.M., S.G.); Division of Biochemistry and Molecular Biology, University of California, Riverside (M.M.); Institute of Bioinformatics and Systems Biology and Department of Biological Science and Technology, National Chiao Tung University, Hsin-Chu, Taiwan (H.-Y.H., H.-D.H.); and Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH (G.Z., P.S., M.K.J.)
| | - Mukesh K Jain
- From Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, China (Z.L., Jin Zhang, L.B., Jiao Zhang, M.H., J.L., T.L., J.Y.-J.S.); Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education of China (Z.L., Jin Zhang, L.B., Jiao Zhang, M.H., J.L., T.L., J.Y.-J.S.); Division of Cardiology, Department of Medicine, University of California, San Diego, La Jolla (M.M., Jin Zhang, J.K., M.H., Y.-J.X., M.J., J.Y.-J.S.);Department of Bioengineering, University of California, San Diego, La Jolla (M.R.M., S.G.); Division of Biochemistry and Molecular Biology, University of California, Riverside (M.M.); Institute of Bioinformatics and Systems Biology and Department of Biological Science and Technology, National Chiao Tung University, Hsin-Chu, Taiwan (H.-Y.H., H.-D.H.); and Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH (G.Z., P.S., M.K.J.)
| | - Shankar Subramaniam
- From Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, China (Z.L., Jin Zhang, L.B., Jiao Zhang, M.H., J.L., T.L., J.Y.-J.S.); Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education of China (Z.L., Jin Zhang, L.B., Jiao Zhang, M.H., J.L., T.L., J.Y.-J.S.); Division of Cardiology, Department of Medicine, University of California, San Diego, La Jolla (M.M., Jin Zhang, J.K., M.H., Y.-J.X., M.J., J.Y.-J.S.);Department of Bioengineering, University of California, San Diego, La Jolla (M.R.M., S.G.); Division of Biochemistry and Molecular Biology, University of California, Riverside (M.M.); Institute of Bioinformatics and Systems Biology and Department of Biological Science and Technology, National Chiao Tung University, Hsin-Chu, Taiwan (H.-Y.H., H.-D.H.); and Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH (G.Z., P.S., M.K.J.)
| | - John Y-J Shyy
- From Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, China (Z.L., Jin Zhang, L.B., Jiao Zhang, M.H., J.L., T.L., J.Y.-J.S.); Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education of China (Z.L., Jin Zhang, L.B., Jiao Zhang, M.H., J.L., T.L., J.Y.-J.S.); Division of Cardiology, Department of Medicine, University of California, San Diego, La Jolla (M.M., Jin Zhang, J.K., M.H., Y.-J.X., M.J., J.Y.-J.S.);Department of Bioengineering, University of California, San Diego, La Jolla (M.R.M., S.G.); Division of Biochemistry and Molecular Biology, University of California, Riverside (M.M.); Institute of Bioinformatics and Systems Biology and Department of Biological Science and Technology, National Chiao Tung University, Hsin-Chu, Taiwan (H.-Y.H., H.-D.H.); and Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH (G.Z., P.S., M.K.J.).
| |
Collapse
|
171
|
The transcription factor MafB promotes anti-inflammatory M2 polarization and cholesterol efflux in macrophages. Sci Rep 2017; 7:7591. [PMID: 28790455 PMCID: PMC5548719 DOI: 10.1038/s41598-017-07381-8] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Accepted: 06/28/2017] [Indexed: 12/21/2022] Open
Abstract
Macrophages play pivotal roles in the progression and regression of atherosclerosis. Accumulating evidence suggests that macrophage polarization into an anti-inflammatory M2 state is a key characteristic of atherosclerotic plaques undergoing regression. However, the molecular mechanisms underlying this potential association of the M2 polarization with atherosclerosis regression remain poorly understood. Further, human genetic factors that facilitate these anti-atherogenic processes remain largely unknown. We report that the transcription factor MafB plays pivotal roles in promoting macrophage M2 polarization. Further, MafB promotes cholesterol efflux from macrophage foam cells by directly up-regulating its key cellular mediators. Notably, MafB expression is significantly up-regulated in response to various metabolic and immunological stimuli that promote macrophage M2 polarization or cholesterol efflux, and thereby MafB mediates their beneficial effects, in both liver x receptor (LXR)-dependent and independent manners. In contrast, MafB is strongly down-regulated upon elevated pro-inflammatory signaling or by pro-inflammatory and pro-atherogenic microRNAs, miR-155 and miR-33. Using an integrative systems biology approach, we also revealed that M2 polarization and cholesterol efflux do not necessarily represent inter-dependent events, but MafB is broadly involved in both the processes. These findings highlight physiological protective roles that MafB may play against atherosclerosis progression.
Collapse
|
172
|
Mistriotis P, Andreadis ST. Vascular aging: Molecular mechanisms and potential treatments for vascular rejuvenation. Ageing Res Rev 2017; 37:94-116. [PMID: 28579130 DOI: 10.1016/j.arr.2017.05.006] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2017] [Revised: 05/22/2017] [Accepted: 05/25/2017] [Indexed: 12/14/2022]
Abstract
Aging is the main risk factor contributing to vascular dysfunction and the progression of vascular diseases. In this review, we discuss the causes and mechanisms of vascular aging at the tissue and cellular level. We focus on Endothelial Cell (EC) and Smooth Muscle Cell (SMC) aging due to their critical role in mediating the defective vascular phenotype. We elaborate on two categories that contribute to cellular dysfunction: cell extrinsic and intrinsic factors. Extrinsic factors reflect systemic or environmental changes which alter EC and SMC homeostasis compromising vascular function. Intrinsic factors induce EC and SMC transformation resulting in cellular senescence. Replenishing or rejuvenating the aged/dysfunctional vascular cells is critical to the effective repair of the vasculature. As such, this review also elaborates on recent findings which indicate that stem cell and gene therapies may restore the impaired vascular cell function, reverse vascular aging, and prolong lifespan.
Collapse
Affiliation(s)
- Panagiotis Mistriotis
- Bioengineering Laboratory, Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, Amherst, NY 14260-4200, USA
| | - Stelios T Andreadis
- Bioengineering Laboratory, Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, Amherst, NY 14260-4200, USA; Department of Biomedical Engineering, University at Buffalo, The State University of New York, Amherst, NY 14260-4200, USA; Center of Excellence in Bioinformatics and Life Sciences, Buffalo, NY 14203, USA.
| |
Collapse
|
173
|
|
174
|
Huang Y, Liu H, Zhang Y, Li J, Wang C, Zhou L, Jia Y, Li X. Synthesis and Biological Evaluation of Ginsenoside Compound K Derivatives as a Novel Class of LXRα Activator. Molecules 2017; 22:molecules22071232. [PMID: 28737726 PMCID: PMC6152260 DOI: 10.3390/molecules22071232] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2017] [Revised: 07/17/2017] [Accepted: 07/18/2017] [Indexed: 12/20/2022] Open
Abstract
Compound K is one of the active metabolites of Panaxnotoginseng saponins, which could attenuate the formation of atherosclerosis in mice modelsvia activating LXRα. We synthesized and evaluated a series of ginsenoside compound K derivatives modified with short chain fatty acids. All of the structures of this class of ginsenoside compound K derivative exhibited comparable or better biological activity than ginsenoside compound K. Especially structure 1 exhibited the best potency (cholesteryl ester content: 41.51%; expression of ABCA1 mRNA: 319%) and low cytotoxicity.
Collapse
Affiliation(s)
- Yan Huang
- Institute of Materia Medica and Department of Pharmaceutics, College of Pharmacy, Third Military Medical University, Shapingba, Chongqing 400038, China.
| | - Hongmei Liu
- Institute of Materia Medica and Department of Pharmaceutics, College of Pharmacy, Third Military Medical University, Shapingba, Chongqing 400038, China.
| | - Yingxian Zhang
- Institute of Materia Medica and Department of Pharmaceutics, College of Pharmacy, Third Military Medical University, Shapingba, Chongqing 400038, China.
| | - Jin Li
- Institute of Materia Medica and Department of Pharmaceutics, College of Pharmacy, Third Military Medical University, Shapingba, Chongqing 400038, China.
| | - Chenping Wang
- Institute of Materia Medica and Department of Pharmaceutics, College of Pharmacy, Third Military Medical University, Shapingba, Chongqing 400038, China.
| | - Li Zhou
- Department of Pharmacy, Xinqiao Hospital & The Second Affiliated Hospital, Third Military Medical University, Shapingba, Chongqing 400037, China.
| | - Yi Jia
- Institute of Materia Medica and Department of Pharmaceutics, College of Pharmacy, Third Military Medical University, Shapingba, Chongqing 400038, China.
| | - Xiaohui Li
- Institute of Materia Medica and Department of Pharmaceutics, College of Pharmacy, Third Military Medical University, Shapingba, Chongqing 400038, China.
| |
Collapse
|
175
|
Ducheix S, Montagner A, Polizzi A, Lasserre F, Régnier M, Marmugi A, Benhamed F, Bertrand-Michel J, Mselli-Lakhal L, Loiseau N, Martin PG, Lobaccaro JM, Ferrier L, Postic C, Guillou H. Dietary oleic acid regulates hepatic lipogenesis through a liver X receptor-dependent signaling. PLoS One 2017; 12:e0181393. [PMID: 28732092 PMCID: PMC5521785 DOI: 10.1371/journal.pone.0181393] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 06/22/2017] [Indexed: 12/13/2022] Open
Abstract
Olive oil consumption is beneficial for health as it is associated with a decreased prevalence of cancer and cardiovascular diseases. Oleic acid is, by far, the most abundant component of olive oil. Since it can be made through de novo synthesis in animals, it is not an essential fatty acid. While it has become clear that dietary oleic acid regulates many biological processes, the signaling pathway involved in these regulations remains poorly defined. In this work we tested the impact of an oleic acid-rich diet on hepatic gene expression. We were particularly interested in addressing the contribution of Liver X Receptors (LXR) in the control of genes involved in hepatic lipogenesis, an essential process in whole body energy homeostasis. We used wild-type mice and transgenic mice deficient for both α and β Liver X Receptor isoforms (LXR-/-) fed a control or an oleate enriched diet. We observed that hepatic-lipid accumulation was enhanced as well as the expression of lipogenic genes in the liver of wild-type mice fed the oleate enriched diet. In contrast, none of these changes occurred in the liver of LXR-/- mice. Strikingly, oleate-rich diet reduced cholesterolemia in wild-type mice and induced signs of liver inflammation and damage in LXR-/- mice but not in wild-type mice. This work suggests that dietary oleic acid reduces cholesterolemia while promoting LXR-dependent hepatic lipogenesis without detrimental effects to the liver.
Collapse
Affiliation(s)
- Simon Ducheix
- INRA, ToxAlim, Toulouse, France.,Université de Toulouse, INP, UPS, ToxAlim, Toulouse, France
| | - Alexandra Montagner
- INRA, ToxAlim, Toulouse, France.,Université de Toulouse, INP, UPS, ToxAlim, Toulouse, France
| | - Arnaud Polizzi
- INRA, ToxAlim, Toulouse, France.,Université de Toulouse, INP, UPS, ToxAlim, Toulouse, France
| | - Frédéric Lasserre
- INRA, ToxAlim, Toulouse, France.,Université de Toulouse, INP, UPS, ToxAlim, Toulouse, France
| | - Marion Régnier
- INRA, ToxAlim, Toulouse, France.,Université de Toulouse, INP, UPS, ToxAlim, Toulouse, France
| | - Alice Marmugi
- INRA, ToxAlim, Toulouse, France.,Université de Toulouse, INP, UPS, ToxAlim, Toulouse, France
| | - Fadila Benhamed
- INSERM, U1016, Institut Cochin, Paris, France.,CNRS, UMR8104, Paris, France
| | | | - Laila Mselli-Lakhal
- INRA, ToxAlim, Toulouse, France.,Université de Toulouse, INP, UPS, ToxAlim, Toulouse, France
| | - Nicolas Loiseau
- INRA, ToxAlim, Toulouse, France.,Université de Toulouse, INP, UPS, ToxAlim, Toulouse, France
| | - Pascal G Martin
- INRA, ToxAlim, Toulouse, France.,Université de Toulouse, INP, UPS, ToxAlim, Toulouse, France
| | - Jean-Marc Lobaccaro
- Clermont Université, Université Blaise Pascal, Génétique Reproduction et Développement, Clermont-Ferrand, France.,CNRS, UMR 6293, GReD, Aubière, France.,INSERM, U1103, GReD, Aubière, France.,Centre de Recherche en Nutrition Humaine d'Auvergne, Clermont-Ferrand, France
| | - Laurent Ferrier
- INRA, ToxAlim, Toulouse, France.,Université de Toulouse, INP, UPS, ToxAlim, Toulouse, France
| | - Catherine Postic
- INSERM, U1016, Institut Cochin, Paris, France.,CNRS, UMR8104, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Hervé Guillou
- INRA, ToxAlim, Toulouse, France.,Université de Toulouse, INP, UPS, ToxAlim, Toulouse, France
| |
Collapse
|
176
|
Fessler MB. The challenges and promise of targeting the Liver X Receptors for treatment of inflammatory disease. Pharmacol Ther 2017; 181:1-12. [PMID: 28720427 DOI: 10.1016/j.pharmthera.2017.07.010] [Citation(s) in RCA: 103] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The Liver X Receptors (LXRs) are oxysterol-activated transcription factors that upregulate a suite of genes that together promote coordinated mobilization of excess cholesterol from cells and from the body. The LXRs, like other nuclear receptors, are anti-inflammatory, inhibiting signal-dependent induction of pro-inflammatory genes by nuclear factor-κB, activating protein-1, and other transcription factors. Synthetic LXR agonists have been shown to ameliorate atherosclerosis and a wide range of inflammatory disorders in preclinical animal models. Although this has suggested potential for application to human disease, systemic LXR activation is complicated by hepatic steatosis and hypertriglyceridemia, consequences of lipogenic gene induction in the liver by LXRα. The past several years have seen the development of multiple advanced LXR therapeutics aiming to avoid hepatic lipogenesis, including LXRβ-selective agonists, tissue-selective agonists, and transrepression-selective agonists. Although several synthetic LXR agonists have made it to phase I clinical trials, none have progressed due to unforeseen adverse reactions or undisclosed reasons. Nonetheless, several sophisticated pharmacologic strategies, including structure-guided drug design, cell-specific drug targeting, as well as non-systemic drug routes have been initiated and remain to be comprehensively explored. In addition, recent studies have identified potential utility for targeting the LXRs during therapy with other agents, such as glucocorticoids and rexinoids. Despite the pitfalls encountered to date in translation of LXR agonists to human disease, it appears likely that this accelerating field will ultimately yield effective and safe applications for LXR targeting in humans.
Collapse
Affiliation(s)
- Michael B Fessler
- National Institute of Environmental Health Sciences, 111 T.W. Alexander Drive, P.O. Box 12233, MD D2-01, Research Triangle Park, NC 27709, United States.
| |
Collapse
|
177
|
Schulman IG. Liver X receptors link lipid metabolism and inflammation. FEBS Lett 2017; 591:2978-2991. [PMID: 28555747 DOI: 10.1002/1873-3468.12702] [Citation(s) in RCA: 141] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Accepted: 05/23/2017] [Indexed: 12/14/2022]
Abstract
The response of immune cells to pathogens is often associated with changes in the flux through basic metabolic pathways. Indeed, in many cases changes in metabolism appear to be necessary for a robust immune response. The Liver X receptors (LXRs) are members of the nuclear hormone receptor superfamily that regulate gene networks controlling cholesterol and lipid metabolism. In immune cells, particularly in macrophages, LXRs also inhibit proinflammatory gene expression. This Review will highlight recent studies that connect LXR-dependent control of lipid metabolism to regulation of the immune response.
Collapse
Affiliation(s)
- Ira G Schulman
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA, USA
| |
Collapse
|
178
|
Combined use of metformin and atorvastatin attenuates atherosclerosis in rabbits fed a high-cholesterol diet. Sci Rep 2017; 7:2169. [PMID: 28526884 PMCID: PMC5438352 DOI: 10.1038/s41598-017-02080-w] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 04/10/2017] [Indexed: 12/16/2022] Open
Abstract
Statins are widely used to reduce cardiovascular risk. Unfortunately, some patients still experience cardiovascular events though prescribed with high-intensity statins. Metformin, an anti-diabetic drug, was reported to possess anti-atherosclerotic effects. Therefore, the experiments were designed to evaluate whether combined use of metformin and atorvastatin can achieve additional benefits. In rabbits fed a high-cholesterol diet, we evaluated the effects of the combination therapy on atherosclerotic plaques, lipid profiles, blood glucose levels, liver and kidney functions. Effects of combination therapy on cholesterol efflux and the expression of related transporters were studied in vitro. Our results showed that the combination therapy induced a more significant decrease in atherosclerotic lesion area than atorvastatin without additional lipid-lowering effect. The combination therapy significantly increased the percentage of large high-density lipoprotein subfraction. The intravenous glucose tolerance test showed that atorvastatin-treated rabbits had an increased area under the curve for time-dependent glucose levels after a bolus injection of glucose, which was completely reversed by metformin treatment. In cultured macrophages, co-treatment with metformin and atorvastatin promoted cholesterol efflux and up-regulated expression of ATP-binding cassette transporters A1 and G1. Taken together, our results suggest that atorvastatin/metformin combination therapy may achieve additional anti-atherosclerotic benefits likely through increasing cholesterol efflux in macrophages.
Collapse
|
179
|
Cho S, Namkoong K, Shin M, Park J, Yang E, Ihm J, Thu VT, Kim HK, Han J. Cardiovascular Protective Effects and Clinical Applications of Resveratrol. J Med Food 2017; 20:323-334. [PMID: 28346848 DOI: 10.1089/jmf.2016.3856] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Resveratrol is a naturally occurring phenol that is generated by plant species following injury or attack by bacterial and fungal pathogens. This compound was first described as the French Paradox in 1992. Later in 2003, resveratrol was reported to activate sirtuins in yeast cells. Recent experimental studies have found that resveratrol offers a variety of benefits that include both anticarcinogenic and anti-inflammatory effects in addition to the ability to reverse obesity, attenuate hyperglycemia and hyperinsulinemia, protect heart and endothelial function, and increase the life span. Multiple molecular targets are associated with the cardioprotective capabilities of resveratrol, and therefore, resveratrol has potential for a wide range of new therapeutic strategies for atherosclerosis, ischemia/reperfusion, metabolic syndrome, cardiac failure, and inflammatory alterations during aging. Expectations for application in human patients, however, suffer from a lack of sufficient clinical evidence in support of these beneficial effects. This article reviews recently reported basic research results that describe the beneficial effects of resveratrol in an attempt to condense the evidence observed in clinical trials and provide support for the future development of novel clinical therapeutics in patients with cardiovascular diseases.
Collapse
Affiliation(s)
- Sanghyun Cho
- 1 National Research Laboratory for Mitochondrial Signaling, Cardiovascular and Metabolic Disease Center, Department of Health Sciences and Technology, BK21 Project Team, Department of Physiology, College of Medicine, Inje University , Busan, Korea
| | - Kyung Namkoong
- 1 National Research Laboratory for Mitochondrial Signaling, Cardiovascular and Metabolic Disease Center, Department of Health Sciences and Technology, BK21 Project Team, Department of Physiology, College of Medicine, Inje University , Busan, Korea
| | - Minji Shin
- 1 National Research Laboratory for Mitochondrial Signaling, Cardiovascular and Metabolic Disease Center, Department of Health Sciences and Technology, BK21 Project Team, Department of Physiology, College of Medicine, Inje University , Busan, Korea
| | - Jueun Park
- 1 National Research Laboratory for Mitochondrial Signaling, Cardiovascular and Metabolic Disease Center, Department of Health Sciences and Technology, BK21 Project Team, Department of Physiology, College of Medicine, Inje University , Busan, Korea
| | - Eunyeong Yang
- 1 National Research Laboratory for Mitochondrial Signaling, Cardiovascular and Metabolic Disease Center, Department of Health Sciences and Technology, BK21 Project Team, Department of Physiology, College of Medicine, Inje University , Busan, Korea
| | - Jinsoo Ihm
- 1 National Research Laboratory for Mitochondrial Signaling, Cardiovascular and Metabolic Disease Center, Department of Health Sciences and Technology, BK21 Project Team, Department of Physiology, College of Medicine, Inje University , Busan, Korea
| | - Vu Thi Thu
- 1 National Research Laboratory for Mitochondrial Signaling, Cardiovascular and Metabolic Disease Center, Department of Health Sciences and Technology, BK21 Project Team, Department of Physiology, College of Medicine, Inje University , Busan, Korea.,2 Key Laboratory of Enzyme and Protein Technology, Faculty of Biology, VNU University of Science , Hanoi, Vietnam
| | - Hyoung Kyu Kim
- 1 National Research Laboratory for Mitochondrial Signaling, Cardiovascular and Metabolic Disease Center, Department of Health Sciences and Technology, BK21 Project Team, Department of Physiology, College of Medicine, Inje University , Busan, Korea.,3 Department of Integrated Biomedical Science, College of Medicine, Inje University , Busan, Korea
| | - Jin Han
- 1 National Research Laboratory for Mitochondrial Signaling, Cardiovascular and Metabolic Disease Center, Department of Health Sciences and Technology, BK21 Project Team, Department of Physiology, College of Medicine, Inje University , Busan, Korea
| |
Collapse
|
180
|
Amersfoort J, Kuiper J. T cell metabolism in metabolic disease-associated autoimmunity. Immunobiology 2017; 222:925-936. [PMID: 28363498 DOI: 10.1016/j.imbio.2017.03.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Revised: 03/06/2017] [Accepted: 03/13/2017] [Indexed: 12/29/2022]
Abstract
This review discusses the relevant metabolic pathways and their regulators which show potential for T cell metabolism-based immunotherapy in diseases hallmarked by both metabolic disease and autoimmunity. Multiple therapeutic approaches using existing pharmaceuticals are possible from a rationale in which T cell metabolism forms the hub in dampening the T cell component of autoimmunity in metabolic diseases. Future research into the effects of a metabolically aberrant micro-environment on T cell metabolism and its potential as a therapeutic target for immunomodulation could lead to novel treatment strategies for metabolic disease-associated autoimmunity.
Collapse
Affiliation(s)
- Jacob Amersfoort
- Division of Biopharmaceutics, LACDR, Leiden University, Leiden, The Netherlands.
| | - Johan Kuiper
- Division of Biopharmaceutics, LACDR, Leiden University, Leiden, The Netherlands
| |
Collapse
|
181
|
Mendelsohn AR, Larrick JW. Preclinical Reversal of Atherosclerosis by FDA-Approved Compound that Transforms Cholesterol into an Anti-Inflammatory "Prodrug". Rejuvenation Res 2017; 19:252-5. [PMID: 27241174 DOI: 10.1089/rej.2016.1849] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Although atherosclerosis is treatable with lipid-lowering drugs, not all patients respond. Hydroxypropyl-beta-cyclodextrin (CD) is an FDA-approved compound for solubilizing, capturing, and delivering lipophilic drugs in humans. Zimmer et al. report that CD mediates regression of atherosclerotic plaques in two mouse models by solubilizing cholesterol crystals (CCs), and promoting metabolism of CCs into water-soluble 27-hydroxycholesterol, which, in turn, activates anti-inflammatory LXR receptor target genes, promotes active and passive efflux of cholesterol from macrophages, and increases metabolic processing of cholesterol. In effect, CD inverts the role of its cargo, cholesterol, from inflammatory to anti-inflammatory by converting cholesterol into a "prodrug" that when modified to 27-hydroxycholesterol reduces atherosclerosis. This mechanism defines a new class of pharmaceuticals, "inverters": compounds that cause innate biomolecules to act opposite to their normal function. However, chronic CD treatment in animal models damages auditory cells, which must be addressed before CD can be developed as a systemic drug for atherosclerosis.
Collapse
Affiliation(s)
- Andrew R Mendelsohn
- 1 Panorama Research Institute , Sunnyvale, California.,2 Regenerative Sciences Institute , Sunnyvale, California
| | - James W Larrick
- 1 Panorama Research Institute , Sunnyvale, California.,2 Regenerative Sciences Institute , Sunnyvale, California
| |
Collapse
|
182
|
Human paraoxonase 1 overexpression in mice stimulates HDL cholesterol efflux and reverse cholesterol transport. PLoS One 2017; 12:e0173385. [PMID: 28278274 PMCID: PMC5344486 DOI: 10.1371/journal.pone.0173385] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 02/20/2017] [Indexed: 11/19/2022] Open
Abstract
This study was aimed to investigate the effect of human PON1 overexpression in mice on cholesterol efflux and reverse cholesterol transport. PON1 overexpression in PON1-Tg mice induced a significant 3-fold (p<0.0001) increase in plasma paraoxonase activity and a significant ~30% (p<0.0001) increase in the capacity of HDL to mediate cholesterol efflux from J774 macrophages compared to wild-type mice. It also caused a significant 4-fold increase (p<0.0001) in the capacity of macrophages to transfer cholesterol to apoA-1, a significant 2-fold (p<0.0003) increase in ABCA1 mRNA and protein expression, and a significant increase in the expression of PPARγ (p<0.0003 and p<0.04, respectively) and LXRα (p<0.0001 and p<0.01, respectively) mRNA and protein compared to macrophages from wild-type mice. Moreover, transfection of J774 macrophages with human PON1 also increased ABCA1, PPARγ and LXRα protein expression and stimulates macrophages cholesterol efflux to apo A1. In vivo measurements showed that the overexpression of PON1 significantly increases the fecal elimination of macrophage-derived cholesterol in PON1-Tg mice. Overall, our results suggested that the overexpression of PON1 in mice may contribute to the regulation of the cholesterol homeostasis by improving the capacity of HDL to mediate cholesterol efflux and by stimulating reverse cholesterol transport.
Collapse
|
183
|
Heitel P, Achenbach J, Moser D, Proschak E, Merk D. DrugBank screening revealed alitretinoin and bexarotene as liver X receptor modulators. Bioorg Med Chem Lett 2017; 27:1193-1198. [DOI: 10.1016/j.bmcl.2017.01.066] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Revised: 01/20/2017] [Accepted: 01/23/2017] [Indexed: 12/12/2022]
|
184
|
Unsworth AJ, Kriek N, Bye AP, Naran K, Sage T, Flora GD, Gibbins JM. PPARγ agonists negatively regulate αIIbβ3 integrin outside-in signaling and platelet function through up-regulation of protein kinase A activity. J Thromb Haemost 2017; 15:356-369. [PMID: 27896950 PMCID: PMC5396324 DOI: 10.1111/jth.13578] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Indexed: 12/31/2022]
Abstract
Essentials peroxisome proliferator-activated receptor γ (PPARγ) agonists inhibit platelet function. PPARγ agonists negatively regulate outside-in signaling via integrin αIIbβ3. PPARγ agonists disrupt the interaction of Gα13 with integrin β3. This is attributed to an upregulation of protein kinase A activity. SUMMARY Background Agonists for the peroxisome proliferator-activated receptor (PPARγ) have been shown to have inhibitory effects on platelet activity following stimulation by GPVI and GPCR agonists. Objectives Profound effects on thrombus formation led us to suspect a role for PPARγ agonists in the regulation of integrin αIIbβ3 mediated signaling. Both GPVI and GPCR signaling pathways lead to αIIbβ3 activation, and signaling through αIIbβ3 plays a critical role in platelet function and normal hemostasis. Methods The effects of PPARγ agonists on the regulation of αIIbβ3 outside-in signaling was determined by monitoring the ability of platelets to adhere and spread on fibrinogen and undergo clot retraction. Effects on signaling components downstream of αIIbβ3 activation were also determined following adhesion to fibrinogen by Western blotting. Results Treatment of platelets with PPARγ agonists inhibited platelet adhesion and spreading on fibrinogen and diminished clot retraction. A reduction in phosphorylation of several components of αIIbβ3 signaling, including the integrin β3 subunit, Syk, PLCγ2, focal adhesion kinase (FAK) and Akt, was also observed as a result of reduced interaction of the integrin β3 subunit with Gα13. Studies of VASP phosphorylation revealed that this was because of an increase in PKA activity following treatment with PPARγ receptor agonists. Conclusions This study provides further evidence for antiplatelet actions of PPARγ agonists, identifies a negative regulatory role for PPARγ agonists in the control of integrin αIIbβ3 outside-in signaling, and provides a molecular basis by which the PPARγ agonists negatively regulate platelet activation and thrombus formation.
Collapse
Affiliation(s)
- A. J. Unsworth
- Institute for Cardiovascular and Metabolic ResearchSchool of Biological SciencesUniversity of ReadingReadingUK
| | - N. Kriek
- Institute for Cardiovascular and Metabolic ResearchSchool of Biological SciencesUniversity of ReadingReadingUK
| | - A. P. Bye
- Institute for Cardiovascular and Metabolic ResearchSchool of Biological SciencesUniversity of ReadingReadingUK
| | - K. Naran
- Institute for Cardiovascular and Metabolic ResearchSchool of Biological SciencesUniversity of ReadingReadingUK
| | - T. Sage
- Institute for Cardiovascular and Metabolic ResearchSchool of Biological SciencesUniversity of ReadingReadingUK
| | - G. D. Flora
- Institute for Cardiovascular and Metabolic ResearchSchool of Biological SciencesUniversity of ReadingReadingUK
| | - J. M. Gibbins
- Institute for Cardiovascular and Metabolic ResearchSchool of Biological SciencesUniversity of ReadingReadingUK
| |
Collapse
|
185
|
Nelson JK, Koenis DS, Scheij S, Cook ECL, Moeton M, Santos A, Lobaccaro JMA, Baron S, Zelcer N. EEPD1 Is a Novel LXR Target Gene in Macrophages Which Regulates ABCA1 Abundance and Cholesterol Efflux. Arterioscler Thromb Vasc Biol 2017; 37:423-432. [PMID: 28082258 PMCID: PMC5321112 DOI: 10.1161/atvbaha.116.308434] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Accepted: 01/02/2017] [Indexed: 11/16/2022]
Abstract
Supplemental Digital Content is available in the text. Objective— The sterol-responsive nuclear receptors, liver X receptors α (LXRα, NR1H3) and β (LXRβ, NR1H2), are key determinants of cellular cholesterol homeostasis. LXRs are activated under conditions of high cellular sterol load and induce expression of the cholesterol efflux transporters ABCA1 and ABCG1 to promote efflux of excess cellular cholesterol. However, the full set of genes that contribute to LXR-stimulated cholesterol efflux is unknown, and their identification is the objective of this study. Approach and Results— We systematically compared the global transcriptional response of macrophages to distinct classes of LXR ligands. This allowed us to identify both common and ligand-specific transcriptional responses in macrophages. Among these, we identified endonuclease–exonuclease–phosphatase family domain containing 1 (EEPD1/KIAA1706) as a direct transcriptional target of LXRs in human and murine macrophages. EEPD1 specifically localizes to the plasma membrane owing to the presence of a myristoylation site in its N terminus. Accordingly, the first 10 amino acids of EEPD1 are sufficient to confer plasma membrane localization in the context of a chimeric protein with GFP. Functionally, we report that silencing expression of EEPD1 blunts maximal LXR-stimulated Apo AI-dependent efflux and demonstrate that this is the result of reduced abundance of ABCA1 protein in human and murine macrophages. Conclusions— In this study, we identify EEPD1 as a novel LXR-regulated gene in macrophages and propose that it promotes cellular cholesterol efflux by controlling cellular levels and activity of ABCA1.
Collapse
Affiliation(s)
- Jessica Kristine Nelson
- From the Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, The Netherlands (J.K.N., D.S.K., S.S., E.C.L.C., M.M., A.S., N.Z.); and Université Clermont Auvergne, CNRS, Inserm, GReD, Clermont-Ferrand, France (J.-M.A.L., S.B.)
| | - Duco Steven Koenis
- From the Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, The Netherlands (J.K.N., D.S.K., S.S., E.C.L.C., M.M., A.S., N.Z.); and Université Clermont Auvergne, CNRS, Inserm, GReD, Clermont-Ferrand, France (J.-M.A.L., S.B.)
| | - Saskia Scheij
- From the Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, The Netherlands (J.K.N., D.S.K., S.S., E.C.L.C., M.M., A.S., N.Z.); and Université Clermont Auvergne, CNRS, Inserm, GReD, Clermont-Ferrand, France (J.-M.A.L., S.B.)
| | - Emma Clare Laura Cook
- From the Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, The Netherlands (J.K.N., D.S.K., S.S., E.C.L.C., M.M., A.S., N.Z.); and Université Clermont Auvergne, CNRS, Inserm, GReD, Clermont-Ferrand, France (J.-M.A.L., S.B.)
| | - Martina Moeton
- From the Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, The Netherlands (J.K.N., D.S.K., S.S., E.C.L.C., M.M., A.S., N.Z.); and Université Clermont Auvergne, CNRS, Inserm, GReD, Clermont-Ferrand, France (J.-M.A.L., S.B.)
| | - Ana Santos
- From the Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, The Netherlands (J.K.N., D.S.K., S.S., E.C.L.C., M.M., A.S., N.Z.); and Université Clermont Auvergne, CNRS, Inserm, GReD, Clermont-Ferrand, France (J.-M.A.L., S.B.)
| | - Jean-Marc Adolphe Lobaccaro
- From the Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, The Netherlands (J.K.N., D.S.K., S.S., E.C.L.C., M.M., A.S., N.Z.); and Université Clermont Auvergne, CNRS, Inserm, GReD, Clermont-Ferrand, France (J.-M.A.L., S.B.)
| | - Silvere Baron
- From the Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, The Netherlands (J.K.N., D.S.K., S.S., E.C.L.C., M.M., A.S., N.Z.); and Université Clermont Auvergne, CNRS, Inserm, GReD, Clermont-Ferrand, France (J.-M.A.L., S.B.)
| | - Noam Zelcer
- From the Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, The Netherlands (J.K.N., D.S.K., S.S., E.C.L.C., M.M., A.S., N.Z.); and Université Clermont Auvergne, CNRS, Inserm, GReD, Clermont-Ferrand, France (J.-M.A.L., S.B.).
| |
Collapse
|
186
|
Downing LE, Edgar D, Ellison PA, Ricketts ML. Mechanistic insight into nuclear receptor-mediated regulation of bile acid metabolism and lipid homeostasis by grape seed procyanidin extract (GSPE). Cell Biochem Funct 2017; 35:12-32. [DOI: 10.1002/cbf.3247] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Revised: 12/13/2016] [Accepted: 12/16/2016] [Indexed: 12/18/2022]
Affiliation(s)
- Laura E. Downing
- Department of Agriculture, Nutrition and Veterinary Sciences; University of Nevada Reno; Reno Nevada USA
| | - Daniel Edgar
- Department of Biochemistry and Molecular Biology; University of Nevada Reno; Reno Nevada USA
| | - Patricia A. Ellison
- Department of Biochemistry and Molecular Biology; University of Nevada Reno; Reno Nevada USA
| | - Marie-Louise Ricketts
- Department of Agriculture, Nutrition and Veterinary Sciences; University of Nevada Reno; Reno Nevada USA
| |
Collapse
|
187
|
Komati R, Spadoni D, Zheng S, Sridhar J, Riley KE, Wang G. Ligands of Therapeutic Utility for the Liver X Receptors. Molecules 2017; 22:molecules22010088. [PMID: 28067791 PMCID: PMC5373669 DOI: 10.3390/molecules22010088] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 12/29/2016] [Accepted: 12/30/2016] [Indexed: 12/21/2022] Open
Abstract
Liver X receptors (LXRs) have been increasingly recognized as a potential therapeutic target to treat pathological conditions ranging from vascular and metabolic diseases, neurological degeneration, to cancers that are driven by lipid metabolism. Amidst intensifying efforts to discover ligands that act through LXRs to achieve the sought-after pharmacological outcomes, several lead compounds are already being tested in clinical trials for a variety of disease interventions. While more potent and selective LXR ligands continue to emerge from screening of small molecule libraries, rational design, and empirical medicinal chemistry approaches, challenges remain in minimizing undesirable effects of LXR activation on lipid metabolism. This review provides a summary of known endogenous, naturally occurring, and synthetic ligands. The review also offers considerations from a molecular modeling perspective with which to design more specific LXRβ ligands based on the interaction energies of ligands and the important amino acid residues in the LXRβ ligand binding domain.
Collapse
Affiliation(s)
- Rajesh Komati
- Department of Chemistry and RCMI Cancer Research Center, Xavier University of Louisiana, New Orleans, LA 70125, USA.
| | - Dominick Spadoni
- Department of Chemistry and RCMI Cancer Research Center, Xavier University of Louisiana, New Orleans, LA 70125, USA.
| | - Shilong Zheng
- Department of Chemistry and RCMI Cancer Research Center, Xavier University of Louisiana, New Orleans, LA 70125, USA.
| | - Jayalakshmi Sridhar
- Department of Chemistry and RCMI Cancer Research Center, Xavier University of Louisiana, New Orleans, LA 70125, USA.
| | - Kevin E Riley
- Department of Chemistry and RCMI Cancer Research Center, Xavier University of Louisiana, New Orleans, LA 70125, USA.
| | - Guangdi Wang
- Department of Chemistry and RCMI Cancer Research Center, Xavier University of Louisiana, New Orleans, LA 70125, USA.
| |
Collapse
|
188
|
Åstrand OAH, Viktorsson EÖ, Kristensen AL, Ekeberg D, Røberg-Larsen H, Wilson SR, Gabrielsen M, Sylte I, Rustan AC, Thoresen GH, Rongved P, Kase ET. Synthesis, in vitro and in vivo biological evaluation of new oxysterols as modulators of the liver X receptors. J Steroid Biochem Mol Biol 2017; 165:323-330. [PMID: 27471149 DOI: 10.1016/j.jsbmb.2016.07.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Revised: 07/06/2016] [Accepted: 07/24/2016] [Indexed: 11/24/2022]
Abstract
Liver X Receptor (LXR) modulators have shown potential as drugs since they target genes affecting metabolism and fatty acid synthesis. LXR antagonists are of particular interest since they are able to reduce the synthesis of complex fatty acids and glucose uptake. Based on molecular modeling, five new cholesterol mimics were synthesized, where four contained a hydroxyl group in the 22-S-position. The new compounds were screened in vitro against several genes affecting lipid metabolism. The compound that performed best in vitro was a dimethylamide derivative of 22(S)-hydroxycholesterol and it was chosen for in vivo testing. However, the blood plasma analysis from the in vivo tests revealed a concentration lower than needed to give any response, indicating either rapid metabolism or low bioavailability.
Collapse
Affiliation(s)
- Ove Alexander Høgmoen Åstrand
- Department of Pharmaceutical Chemistry, School of Pharmacy, University of Oslo, PO Box 1068 Blindern, N-0316 Oslo, Norway
| | - Elvar Örn Viktorsson
- Department of Pharmaceutical Chemistry, School of Pharmacy, University of Oslo, PO Box 1068 Blindern, N-0316 Oslo, Norway
| | - Aleksander Lim Kristensen
- Department of Pharmaceutical Chemistry, School of Pharmacy, University of Oslo, PO Box 1068 Blindern, N-0316 Oslo, Norway
| | - Dag Ekeberg
- Department of Chemistry, Biotechnology and Food Sciences, Norwegian University of Life Sciences, PO Box 5003, N-1432, Aas, Norway
| | - Hanne Røberg-Larsen
- Department of Chemistry, University of Oslo, PO Box 1033 Blindern, NO-0315 Oslo, Norway
| | - Steven Ray Wilson
- Department of Chemistry, University of Oslo, PO Box 1033 Blindern, NO-0315 Oslo, Norway
| | - Mari Gabrielsen
- Department of Medical Biology, Faculty of Health Sciences, UiT The Arctic University of Norway, Tromsø, Norway
| | - Ingebrigt Sylte
- Department of Medical Biology, Faculty of Health Sciences, UiT The Arctic University of Norway, Tromsø, Norway
| | - Arild Christian Rustan
- Department of Pharmaceutical Biosciences, School of Pharmacy, University of Oslo, PO Box 1068 Blindern, N-0316 Oslo, Norway
| | - G Hege Thoresen
- Department of Pharmaceutical Biosciences, School of Pharmacy, University of Oslo, PO Box 1068 Blindern, N-0316 Oslo, Norway; Department of Pharmacology, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Pål Rongved
- Department of Pharmaceutical Chemistry, School of Pharmacy, University of Oslo, PO Box 1068 Blindern, N-0316 Oslo, Norway
| | - Eili Tranheim Kase
- Department of Pharmaceutical Biosciences, School of Pharmacy, University of Oslo, PO Box 1068 Blindern, N-0316 Oslo, Norway.
| |
Collapse
|
189
|
Unsworth AJ, Bye AP, Gibbins JM. Platelet-Derived Inhibitors of Platelet Activation. PLATELETS IN THROMBOTIC AND NON-THROMBOTIC DISORDERS 2017. [PMCID: PMC7123044 DOI: 10.1007/978-3-319-47462-5_37] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
190
|
Bedi S, Hines GV, Lozada-Fernandez VV, de Jesus Piva C, Kaliappan A, Rider SD, Hostetler HA. Fatty acid binding profile of the liver X receptor α. J Lipid Res 2016; 58:393-402. [PMID: 28011707 PMCID: PMC5282955 DOI: 10.1194/jlr.m072447] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Revised: 12/16/2016] [Indexed: 12/12/2022] Open
Abstract
Liver X receptor (LXR)α is a nuclear receptor that responds to oxysterols and cholesterol overload by stimulating cholesterol efflux, transport, conversion to bile acids, and excretion. LXRα binds to and is regulated by synthetic (T-0901317, GW3695) and endogenous (oxysterols) ligands. LXRα activity is also modulated by FAs, but the ligand binding specificity of FA and acyl-CoA derivatives for LXRα remains unknown. We investigated whether LXRα binds FA or FA acyl-CoA with affinities that mimic in vivo concentrations, examined the effect of FA chain length and the degree of unsaturation on binding, and investigated whether FAs regulate LXRα activation. Saturated medium-chain FA (MCFA) displayed binding affinities in the low nanomolar concentration range, while long-chain fatty acyl-CoA did not bind or bound weakly to LXRα. Circular dichroic spectra and computational docking experiments confirmed that MCFA bound to the LXRα ligand binding pocket similar to the known synthetic agonist of LXRα (T0901317), but with limited change to the conformation of the receptor. Transactivation assays showed that MCFA activated LXRα, whereas long-chain FA caused no effect. Our results suggest that LXRα functions as a receptor for saturated FA or acyl-CoA of C10 and C12 in length.
Collapse
Affiliation(s)
- Shimpi Bedi
- Department of Biochemistry and Molecular Biology, Boonshoft School of Medicine, Wright State University, Dayton, OH 45435
| | - Genesis Victoria Hines
- Department of Biochemistry and Molecular Biology, Boonshoft School of Medicine, Wright State University, Dayton, OH 45435
| | - Valery V Lozada-Fernandez
- Department of Biochemistry and Molecular Biology, Boonshoft School of Medicine, Wright State University, Dayton, OH 45435
| | - Camila de Jesus Piva
- Department of Biochemistry and Molecular Biology, Boonshoft School of Medicine, Wright State University, Dayton, OH 45435
| | - Alagammai Kaliappan
- Department of Biochemistry and Molecular Biology, Boonshoft School of Medicine, Wright State University, Dayton, OH 45435
| | - S Dean Rider
- Department of Biochemistry and Molecular Biology, Boonshoft School of Medicine, Wright State University, Dayton, OH 45435
| | - Heather A Hostetler
- Department of Biochemistry and Molecular Biology, Boonshoft School of Medicine, Wright State University, Dayton, OH 45435
| |
Collapse
|
191
|
Cao H, Yu S, Chen D, Jing C, Wang Z, Ma R, Liu S, Ni J, Feng J, Wu J. Liver X receptor agonist T0901317 reverses resistance of A549 human lung cancer cells to EGFR-TKI treatment. FEBS Open Bio 2016; 7:35-43. [PMID: 28097086 PMCID: PMC5221460 DOI: 10.1002/2211-5463.12147] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Revised: 09/30/2016] [Accepted: 10/14/2016] [Indexed: 12/13/2022] Open
Abstract
Epidermal growth factor receptor‐tyrosine kinase inhibitor (EGFR‐TKI) is effective in lung cancer patients carrying sensitive EGFR mutations. In this study, we investigated if liver X receptor (LXR) agonist T0901317 could reverse the resistance of lung cancer cell lines A549 and H1650 to EGFR‐TKI treatment. We found that T0901317 could make natural EGFR‐TKI‐resistant A549 human lung cancer cells sensitive to EGFR‐TKI treatment and that this was dependent on LXRβ expression. However, T0901317 does not have a similar effect on another natural EGFR‐TKI‐resistant cell line H1650.
Collapse
Affiliation(s)
- Haixia Cao
- Research Center for Clinical Oncology Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research Nanjing Medical University Affiliated Cancer Hospital Jiangsu Province China
| | - Shaorong Yu
- Research Center for Clinical Oncology Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research Nanjing Medical University Affiliated Cancer Hospital Jiangsu Province China; Department of Oncology Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research Nanjing Medical University Affiliated Cancer Hospital Jiangsu Province China
| | - Dan Chen
- Research Center for Clinical Oncology Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research Nanjing Medical University Affiliated Cancer Hospital Jiangsu Province China
| | - Changwen Jing
- Research Center for Clinical Oncology Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research Nanjing Medical University Affiliated Cancer Hospital Jiangsu Province China
| | - Zhuo Wang
- Research Center for Clinical Oncology Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research Nanjing Medical University Affiliated Cancer Hospital Jiangsu Province China
| | - Rong Ma
- Research Center for Clinical Oncology Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research Nanjing Medical University Affiliated Cancer Hospital Jiangsu Province China
| | - Siwen Liu
- Research Center for Clinical Oncology Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research Nanjing Medical University Affiliated Cancer Hospital Jiangsu Province China
| | - Jie Ni
- The Fourth Clinical School of Nanjing Medical University Jiangsu Province China
| | - Jifeng Feng
- Research Center for Clinical Oncology Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research Nanjing Medical University Affiliated Cancer Hospital Jiangsu Province China; Department of Oncology Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research Nanjing Medical University Affiliated Cancer Hospital Jiangsu Province China
| | - Jianzhong Wu
- Research Center for Clinical Oncology Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research Nanjing Medical University Affiliated Cancer Hospital Jiangsu Province China
| |
Collapse
|
192
|
Coisne C, Tilloy S, Monflier E, Wils D, Fenart L, Gosselet F. Cyclodextrins as Emerging Therapeutic Tools in the Treatment of Cholesterol-Associated Vascular and Neurodegenerative Diseases. Molecules 2016; 21:E1748. [PMID: 27999408 PMCID: PMC6273856 DOI: 10.3390/molecules21121748] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Revised: 12/13/2016] [Accepted: 12/14/2016] [Indexed: 12/20/2022] Open
Abstract
Cardiovascular diseases, like atherosclerosis, and neurodegenerative diseases affecting the central nervous system (CNS) are closely linked to alterations of cholesterol metabolism. Therefore, innovative pharmacological approaches aiming at counteracting cholesterol imbalance display promising therapeutic potential. However, these approaches need to take into account the existence of biological barriers such as intestinal and blood-brain barriers which participate in the organ homeostasis and are major defense systems against xenobiotics. Interest in cyclodextrins (CDs) as medicinal agents has increased continuously based on their ability to actively extract lipids from cell membranes and to provide suitable carrier system for drug delivery. Many novel CD derivatives are constantly generated with the objective to improve CD bioavailability, biocompatibility and therapeutic outcomes. Newly designed drug formulation complexes incorporating CDs as drug carriers have demonstrated better efficiency in treating cardiovascular and neurodegenerative diseases. CD-based therapies as cholesterol-sequestrating agent have recently demonstrated promising advances with KLEPTOSE® CRYSMEB in atherosclerosis as well as with the 2-hydroxypropyl-β-cyclodextrin (HPβCD) in clinical trials for Niemann-Pick type C disease. Based on this success, many investigations evaluating the therapeutical beneficial of CDs in Alzheimer's, Parkinson's and Huntington's diseases are currently on-going.
Collapse
Affiliation(s)
- Caroline Coisne
- Laboratoire de la barrière hémato-encéphalique (LBHE), University Artois, EA 2465, Lens, F-62300, France.
| | - Sébastien Tilloy
- Unité de Catalyse et de Chimie du Solide (UCCS), University Artois, CNRS, UMR 8181, Lens, F-62300, France.
| | - Eric Monflier
- Unité de Catalyse et de Chimie du Solide (UCCS), University Artois, CNRS, UMR 8181, Lens, F-62300, France.
| | - Daniel Wils
- ROQUETTE, Nutrition & Health R & D, 62136 Lestrem, France.
| | - Laurence Fenart
- Laboratoire de la barrière hémato-encéphalique (LBHE), University Artois, EA 2465, Lens, F-62300, France.
| | - Fabien Gosselet
- Laboratoire de la barrière hémato-encéphalique (LBHE), University Artois, EA 2465, Lens, F-62300, France.
| |
Collapse
|
193
|
Zimmer S, Grebe A, Bakke SS, Bode N, Halvorsen B, Ulas T, Skjelland M, De Nardo D, Labzin LI, Kerksiek A, Hempel C, Heneka MT, Hawxhurst V, Fitzgerald ML, Trebicka J, Björkhem I, Gustafsson JÅ, Westerterp M, Tall AR, Wright SD, Espevik T, Schultze JL, Nickenig G, Lütjohann D, Latz E. Cyclodextrin promotes atherosclerosis regression via macrophage reprogramming. Sci Transl Med 2016; 8:333ra50. [PMID: 27053774 DOI: 10.1126/scitranslmed.aad6100] [Citation(s) in RCA: 258] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2015] [Accepted: 02/18/2016] [Indexed: 12/12/2022]
Abstract
Atherosclerosis is an inflammatory disease linked to elevated blood cholesterol concentrations. Despite ongoing advances in the prevention and treatment of atherosclerosis, cardiovascular disease remains the leading cause of death worldwide. Continuous retention of apolipoprotein B-containing lipoproteins in the subendothelial space causes a local overabundance of free cholesterol. Because cholesterol accumulation and deposition of cholesterol crystals (CCs) trigger a complex inflammatory response, we tested the efficacy of the cyclic oligosaccharide 2-hydroxypropyl-β-cyclodextrin (CD), a compound that increases cholesterol solubility in preventing and reversing atherosclerosis. We showed that CD treatment of murine atherosclerosis reduced atherosclerotic plaque size and CC load and promoted plaque regression even with a continued cholesterol-rich diet. Mechanistically, CD increased oxysterol production in both macrophages and human atherosclerotic plaques and promoted liver X receptor (LXR)-mediated transcriptional reprogramming to improve cholesterol efflux and exert anti-inflammatory effects. In vivo, this CD-mediated LXR agonism was required for the antiatherosclerotic and anti-inflammatory effects of CD as well as for augmented reverse cholesterol transport. Because CD treatment in humans is safe and CD beneficially affects key mechanisms of atherogenesis, it may therefore be used clinically to prevent or treat human atherosclerosis.
Collapse
Affiliation(s)
- Sebastian Zimmer
- Medizinische Klinik und Poliklinik II, University Hospital Bonn, 53105 Bonn, Germany
| | - Alena Grebe
- Institute of Innate Immunity, University Hospital Bonn, 53127 Bonn, Germany
| | - Siril S Bakke
- Institute of Innate Immunity, University Hospital Bonn, 53127 Bonn, Germany. German Center of Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany. Centre of Molecular Inflammation Research, Norwegian University of Science and Technology, 7489 Trondheim, Norway
| | - Niklas Bode
- Medizinische Klinik und Poliklinik II, University Hospital Bonn, 53105 Bonn, Germany
| | - Bente Halvorsen
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, 0424 Oslo, Norway
| | - Thomas Ulas
- Genomics and Immunoregulation, Life and Medical Sciences Institute, University of Bonn, 53115 Bonn, Germany
| | - Mona Skjelland
- Department of Neurology, Oslo University Hospital Rikshospitalet, 0424 Oslo, Norway
| | - Dominic De Nardo
- Institute of Innate Immunity, University Hospital Bonn, 53127 Bonn, Germany. Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia. Department of Medical Biology, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Larisa I Labzin
- Institute of Innate Immunity, University Hospital Bonn, 53127 Bonn, Germany
| | - Anja Kerksiek
- Institute of Clinical Chemistry und Clinical Pharmacology, University Hospital Bonn, 53105 Bonn, Germany
| | | | - Michael T Heneka
- Clinic and Polyclinic for Neurology, University Hospital Bonn, 53105 Bonn, Germany
| | - Victoria Hawxhurst
- Lipid Metabolism Unit, Center for Computational and Integrative Biology, Boston, MA 02114, USA
| | - Michael L Fitzgerald
- Lipid Metabolism Unit, Center for Computational and Integrative Biology, Boston, MA 02114, USA
| | - Jonel Trebicka
- Medizinische Klinik und Poliklinik I, University Hospital Bonn, 53105 Bonn, Germany. Faculty of Health Sciences, University of Southern Denmark Campusvej 55, DK-5230 Odense M, Denmark
| | - Ingemar Björkhem
- Division of Clinical Chemistry, Karolinska Institutet, Huddinge University Hospital, 141 86 Huddinge, Sweden
| | - Jan-Åke Gustafsson
- Center for Nuclear Receptors and Cell Signaling, University of Houston, Houston, TX 77004, USA
| | - Marit Westerterp
- Department of Medicine, Columbia University, New York, NY 10032, USA
| | - Alan R Tall
- Department of Medicine, Columbia University, New York, NY 10032, USA
| | | | - Terje Espevik
- Centre of Molecular Inflammation Research, Norwegian University of Science and Technology, 7489 Trondheim, Norway
| | - Joachim L Schultze
- German Center of Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany. Genomics and Immunoregulation, Life and Medical Sciences Institute, University of Bonn, 53115 Bonn, Germany
| | - Georg Nickenig
- Medizinische Klinik und Poliklinik II, University Hospital Bonn, 53105 Bonn, Germany
| | - Dieter Lütjohann
- Institute of Clinical Chemistry und Clinical Pharmacology, University Hospital Bonn, 53105 Bonn, Germany
| | - Eicke Latz
- Institute of Innate Immunity, University Hospital Bonn, 53127 Bonn, Germany. German Center of Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany. Centre of Molecular Inflammation Research, Norwegian University of Science and Technology, 7489 Trondheim, Norway. Department of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, MA 01605, USA.
| |
Collapse
|
194
|
Kick EK, Busch BB, Martin R, Stevens WC, Bollu V, Xie Y, Boren BC, Nyman MC, Nanao MH, Nguyen L, Plonowski A, Schulman IG, Yan G, Zhang H, Hou X, Valente MN, Narayanan R, Behnia K, Rodrigues AD, Brock B, Smalley J, Cantor GH, Lupisella J, Sleph P, Grimm D, Ostrowski J, Wexler RR, Kirchgessner T, Mohan R. Discovery of Highly Potent Liver X Receptor β Agonists. ACS Med Chem Lett 2016; 7:1207-1212. [PMID: 27994765 PMCID: PMC5150697 DOI: 10.1021/acsmedchemlett.6b00234] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Accepted: 09/27/2016] [Indexed: 12/13/2022] Open
Abstract
![]()
Introducing a uniquely substituted
phenyl sulfone into a series
of biphenyl imidazole liver X receptor (LXR) agonists afforded a dramatic
potency improvement for induction of ATP binding cassette transporters,
ABCA1 and ABCG1, in human whole blood. The agonist series demonstrated
robust LXRβ activity (>70%) with low partial LXRα agonist
activity (<25%) in cell assays, providing a window between desired
blood cell ABCG1 gene induction in cynomolgus monkeys and modest elevation
of plasma triglycerides for agonist 15. The addition
of polarity to the phenyl sulfone also reduced binding to the plasma
protein, human α-1-acid glycoprotein. Agonist 15 was selected for clinical development based on the favorable combination
of in vitro properties, excellent pharmacokinetic
parameters, and a favorable lipid profile.
Collapse
Affiliation(s)
| | - Brett B. Busch
- Exelixis Inc., 210 East Grand
Avenue, South San Francisco, California 94080, United States
| | - Richard Martin
- Exelixis Inc., 210 East Grand
Avenue, South San Francisco, California 94080, United States
| | - William C. Stevens
- Exelixis Inc., 210 East Grand
Avenue, South San Francisco, California 94080, United States
| | - Venkataiah Bollu
- Exelixis Inc., 210 East Grand
Avenue, South San Francisco, California 94080, United States
| | - Yinong Xie
- Exelixis Inc., 210 East Grand
Avenue, South San Francisco, California 94080, United States
| | - Brant C. Boren
- Exelixis Inc., 210 East Grand
Avenue, South San Francisco, California 94080, United States
| | - Michael C. Nyman
- Exelixis Inc., 210 East Grand
Avenue, South San Francisco, California 94080, United States
| | - Max H. Nanao
- Exelixis Inc., 210 East Grand
Avenue, South San Francisco, California 94080, United States
| | - Lam Nguyen
- Exelixis Inc., 210 East Grand
Avenue, South San Francisco, California 94080, United States
| | - Artur Plonowski
- Exelixis Inc., 210 East Grand
Avenue, South San Francisco, California 94080, United States
| | - Ira G. Schulman
- Exelixis Inc., 210 East Grand
Avenue, South San Francisco, California 94080, United States
| | - Grace Yan
- Exelixis Inc., 210 East Grand
Avenue, South San Francisco, California 94080, United States
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Raju Mohan
- Exelixis Inc., 210 East Grand
Avenue, South San Francisco, California 94080, United States
| |
Collapse
|
195
|
Villa GR, Hulce JJ, Zanca C, Bi J, Ikegami S, Cahill GL, Gu Y, Lum KM, Masui K, Yang H, Rong X, Hong C, Turner KM, Liu F, Hon GC, Jenkins D, Martini M, Armando AM, Quehenberger O, Cloughesy TF, Furnari FB, Cavenee WK, Tontonoz P, Gahman TC, Shiau AK, Cravatt BF, Mischel PS. An LXR-Cholesterol Axis Creates a Metabolic Co-Dependency for Brain Cancers. Cancer Cell 2016; 30:683-693. [PMID: 27746144 PMCID: PMC5479636 DOI: 10.1016/j.ccell.2016.09.008] [Citation(s) in RCA: 244] [Impact Index Per Article: 27.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Revised: 05/19/2016] [Accepted: 09/17/2016] [Indexed: 12/11/2022]
Abstract
Small-molecule inhibitors targeting growth factor receptors have failed to show efficacy for brain cancers, potentially due to their inability to achieve sufficient drug levels in the CNS. Targeting non-oncogene tumor co-dependencies provides an alternative approach, particularly if drugs with high brain penetration can be identified. Here we demonstrate that the highly lethal brain cancer glioblastoma (GBM) is remarkably dependent on cholesterol for survival, rendering these tumors sensitive to Liver X receptor (LXR) agonist-dependent cell death. We show that LXR-623, a clinically viable, highly brain-penetrant LXRα-partial/LXRβ-full agonist selectively kills GBM cells in an LXRβ- and cholesterol-dependent fashion, causing tumor regression and prolonged survival in mouse models. Thus, a metabolic co-dependency provides a pharmacological means to kill growth factor-activated cancers in the CNS.
Collapse
Affiliation(s)
- Genaro R Villa
- Department of Molecular and Medical Pharmacology, David Geffen UCLA School of Medicine, Los Angeles, CA 90095, USA; Medical Scientist Training Program, David Geffen UCLA School of Medicine, Los Angeles, CA 90095, USA; Ludwig Institute for Cancer Research, University of California San Diego, La Jolla, CA 92093, USA
| | - Jonathan J Hulce
- Department of Chemical Physiology, The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Ciro Zanca
- Ludwig Institute for Cancer Research, University of California San Diego, La Jolla, CA 92093, USA
| | - Junfeng Bi
- Ludwig Institute for Cancer Research, University of California San Diego, La Jolla, CA 92093, USA
| | - Shiro Ikegami
- Ludwig Institute for Cancer Research, University of California San Diego, La Jolla, CA 92093, USA
| | - Gabrielle L Cahill
- Ludwig Institute for Cancer Research, University of California San Diego, La Jolla, CA 92093, USA
| | - Yuchao Gu
- Department of Molecular and Medical Pharmacology, David Geffen UCLA School of Medicine, Los Angeles, CA 90095, USA; Ludwig Institute for Cancer Research, University of California San Diego, La Jolla, CA 92093, USA
| | - Kenneth M Lum
- Department of Chemical Physiology, The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Kenta Masui
- Department of Pathology, Tokyo Women's Medical University, Tokyo 162-8666, Japan
| | - Huijun Yang
- Ludwig Institute for Cancer Research, University of California San Diego, La Jolla, CA 92093, USA
| | - Xin Rong
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Cynthia Hong
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Kristen M Turner
- Ludwig Institute for Cancer Research, University of California San Diego, La Jolla, CA 92093, USA
| | - Feng Liu
- Ludwig Institute for Cancer Research, University of California San Diego, La Jolla, CA 92093, USA
| | - Gary C Hon
- Ludwig Institute for Cancer Research, University of California San Diego, La Jolla, CA 92093, USA
| | - David Jenkins
- Small Molecule Discovery Program, Ludwig Institute for Cancer Research, University of California San Diego, La Jolla, CA 92093, USA
| | - Michael Martini
- Department of Chemical Physiology, The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Aaron M Armando
- Department of Pharmacology, UCSD School of Medicine, La Jolla, CA 92093, USA
| | - Oswald Quehenberger
- Department of Pharmacology, UCSD School of Medicine, La Jolla, CA 92093, USA; Department of Medicine, UCSD School of Medicine, La Jolla, CA 92093, USA
| | - Timothy F Cloughesy
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Frank B Furnari
- Ludwig Institute for Cancer Research, University of California San Diego, La Jolla, CA 92093, USA; Department of Pathology, UCSD School of Medicine, La Jolla, CA 92093, USA; Moores Cancer Center, UCSD School of Medicine, La Jolla, CA 92093, USA
| | - Webster K Cavenee
- Ludwig Institute for Cancer Research, University of California San Diego, La Jolla, CA 92093, USA; Department of Medicine, UCSD School of Medicine, La Jolla, CA 92093, USA; Moores Cancer Center, UCSD School of Medicine, La Jolla, CA 92093, USA
| | - Peter Tontonoz
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Howard Hughes Medical Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Timothy C Gahman
- Small Molecule Discovery Program, Ludwig Institute for Cancer Research, University of California San Diego, La Jolla, CA 92093, USA
| | - Andrew K Shiau
- Small Molecule Discovery Program, Ludwig Institute for Cancer Research, University of California San Diego, La Jolla, CA 92093, USA
| | - Benjamin F Cravatt
- Department of Chemical Physiology, The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA 92037, USA.
| | - Paul S Mischel
- Ludwig Institute for Cancer Research, University of California San Diego, La Jolla, CA 92093, USA; Department of Pathology, UCSD School of Medicine, La Jolla, CA 92093, USA; Moores Cancer Center, UCSD School of Medicine, La Jolla, CA 92093, USA.
| |
Collapse
|
196
|
Hoekstra M, Van Berkel TJ. Functionality of High-Density Lipoprotein as Antiatherosclerotic Therapeutic Target. Arterioscler Thromb Vasc Biol 2016; 36:e87-e94. [DOI: 10.1161/atvbaha.116.308262] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Affiliation(s)
- Menno Hoekstra
- From the Division of Biopharmaceutics, Cluster BioTherapeutics, Leiden Academic Centre for Drug Research, Gorlaeus Laboratories, The Netherlands
| | - Theo J.C. Van Berkel
- From the Division of Biopharmaceutics, Cluster BioTherapeutics, Leiden Academic Centre for Drug Research, Gorlaeus Laboratories, The Netherlands
| |
Collapse
|
197
|
Hu W, Zhang W, Chen Y, Rana U, Teng RJ, Duan Y, Liu Z, Zhao B, Foeckler J, Weiler H, Kallinger RE, Thomas MJ, Zhang K, Han J, Miao QR. Nogo-B receptor deficiency increases liver X receptor alpha nuclear translocation and hepatic lipogenesis through an adenosine monophosphate-activated protein kinase alpha-dependent pathway. Hepatology 2016; 64:1559-1576. [PMID: 27480224 PMCID: PMC5074877 DOI: 10.1002/hep.28747] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Revised: 06/30/2016] [Accepted: 07/14/2016] [Indexed: 01/01/2023]
Abstract
UNLABELLED Nogo-B receptor (NgBR) was identified as a specific receptor for binding Nogo-B and is essential for the stability of Niemann-Pick type C2 protein (NPC2) and NPC2-dependent cholesterol trafficking. Here, we report that NgBR expression levels decrease in the fatty liver and that NgBR plays previously unrecognized roles in regulating hepatic lipogenesis through NPC2-independent pathways. To further elucidate the pathophysiological role of NgBR in mammals, we generated NgBR liver-specific knockout mice and investigated the roles of NgBR in hepatic lipid homeostasis. The results showed that NgBR knockout in mouse liver did not decrease NPC2 levels or increase NPC2-dependent intracellular cholesterol levels. However, NgBR deficiency still resulted in remarkable cellular lipid accumulation that was associated with increased free fatty acids and triglycerides in hepatocytes in vitro and in mouse livers in vivo. Mechanistically, NgBR deficiency specifically promotes the nuclear translocation of the liver X receptor alpha (LXRα) and increases the expression of LXRα-targeted lipogenic genes. LXRα knockout attenuates the accumulation of free fatty acids and triglycerides caused by NgBR deficiency. In addition, we elucidated the mechanisms by which NgBR bridges the adenosine monophosphate-activated protein kinase alpha signaling pathway with LXRα nuclear translocation and LXRα-mediated lipogenesis. CONCLUSION NgBR is a specific negative regulator for LXRα-dependent hepatic lipogenesis. Loss of NgBR may be a potential trigger for inducing hepatic steatosis. (Hepatology 2016;64:1559-1576).
Collapse
Affiliation(s)
- Wenquan Hu
- Department of Surgery and Pathology, Medical College of Wisconsin
,State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China
| | - Wenwen Zhang
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China
| | - Yuanli Chen
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China
| | - Ujala Rana
- Department of Surgery and Pathology, Medical College of Wisconsin
| | - Ru-jeng Teng
- Department of Pediatrics, Children’s Research Institute, Medical College of Wisconsin
| | - Yajun Duan
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China
| | - Zhong Liu
- Department of Surgery and Pathology, Medical College of Wisconsin
| | - Baofeng Zhao
- Department of Surgery and Pathology, Medical College of Wisconsin
| | | | | | | | - Michael J. Thomas
- Department of Pharmacology and Toxicology, Medical College of Wisconsin
| | - Kezhong Zhang
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI 48201, USA
| | - Jihong Han
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China. .,College of Biomedical Engineering, Hefei University of Technology, Hefei, China.
| | - Qing Robert Miao
- Departments of Surgery and Pathology, Children's Research Institute, Medical College of Wisconsin, Milwaukee, WI.
| |
Collapse
|
198
|
Immune cell screening of a nanoparticle library improves atherosclerosis therapy. Proc Natl Acad Sci U S A 2016; 113:E6731-E6740. [PMID: 27791119 DOI: 10.1073/pnas.1609629113] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Immunological complexity in atherosclerosis warrants targeted treatment of specific inflammatory cells that aggravate the disease. With the initiation of large phase III trials investigating immunomodulatory drugs for atherosclerosis, cardiovascular disease treatment enters a new era. We here propose a radically different approach: implementing and evaluating in vivo a combinatorial library of nanoparticles with distinct physiochemical properties and differential immune cell specificities. The library's nanoparticles are based on endogenous high-density lipoprotein, which can preferentially deliver therapeutic compounds to pathological macrophages in atherosclerosis. Using the apolipoprotein E-deficient (Apoe-/-) mouse model of atherosclerosis, we quantitatively evaluated the library's immune cell specificity by combining immunological techniques and in vivo positron emission tomography imaging. Based on this screen, we formulated a liver X receptor agonist (GW3965) and abolished its liver toxicity while still preserving its therapeutic function. Screening the immune cell specificity of nanoparticles can be used to develop tailored therapies for atherosclerosis and other inflammatory diseases.
Collapse
|
199
|
Cholesterol efflux capacity: An introduction for clinicians. Am Heart J 2016; 180:54-63. [PMID: 27659883 DOI: 10.1016/j.ahj.2016.07.005] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Accepted: 07/07/2016] [Indexed: 12/28/2022]
Abstract
Epidemiologic studies have shown an inverse correlation between high-density lipoprotein (HDL) cholesterol (HDL-C) levels and cardiovascular disease outcomes. However, the hypothesis of a causal relationship between HDL-C and cardiovascular disease has been challenged by genetic and clinical studies. Serum cholesterol efflux capacity (CEC) is an important measure of HDL function in humans. Recent large clinical studies have shown a correlation between in vitro CEC and cardiovascular disease prevalence and incidence, which appears to be independent of HDL-C concentration. The present review summarizes recent large clinical studies and introduces important methodological considerations. Further studies are required to standardize and establish the reproducibility of this measure of HDL function and clarify whether modulating CEC will emerge as a useful therapeutic target.
Collapse
|
200
|
Nomura S, Endo-Umeda K, Makishima M, Hashimoto Y, Ishikawa M. Development of Tetrachlorophthalimides as Liver X Receptor β (LXRβ)-Selective Agonists. ChemMedChem 2016; 11:2347-2360. [PMID: 27690261 DOI: 10.1002/cmdc.201600305] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 08/23/2016] [Indexed: 11/10/2022]
Abstract
Liver X receptor (LXR) agonists are candidates for the treatment of atherosclerosis via induction of ABCA1 (ATP-binding cassette A1) gene expression, which contributes to reverse cholesterol transport (RCT) and to cholesterol efflux from the liver and intestine. However, LXR agonists also induce genes involved in lipogenesis, such as SREBP-1c (sterol regulatory binding element protein 1c) and FAS (fatty acid synthase), thereby causing an undesirable increase in plasma and hepatic triglyceride (TG) levels. Recent studies indicate that LXRα contributes to lipogenesis in liver, and selective LXRβ activation improves RCT in mice. Therefore, LXRβ-selective agonists are promising candidates to improve atherosclerosis without increasing plasma or hepatic TG levels. However, the ligand-binding domains in the two LXR isoforms α/β share high sequence identity, and few LXR ligands show subtype selectivity. In this study we identified a tetrachlorophthalimide analogue as an LXRβ-selective agonist. Structural development led to (E)-4,5,6,7-tetrachloro-2-(2-styrylphenyl)isoindoline-1,3-dione (24 a), which shows potent and selective LXRβ agonistic activity in reporter gene assays. In binding assays, compound 24 a bound to LXRβ preferentially over LXRα. It also induced the expression of ABCA1 mRNA but not SREBP-1c mRNA in cells. Compound 24 a appears to be a promising lead compound for therapeutic agents to treat atherosclerosis without the side effects induced by LXRα/β dual agonists.
Collapse
Affiliation(s)
- Sayaka Nomura
- Institute of Molecular and Cellular Biosciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo, 113-0032, Japan
| | - Kaori Endo-Umeda
- Nihon University School of Medicine, 30-1 Oyaguchi-kamicho, Itabashi-ku, Tokyo, 173-8610, Japan
| | - Makoto Makishima
- Nihon University School of Medicine, 30-1 Oyaguchi-kamicho, Itabashi-ku, Tokyo, 173-8610, Japan
| | - Yuichi Hashimoto
- Institute of Molecular and Cellular Biosciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo, 113-0032, Japan
| | - Minoru Ishikawa
- Institute of Molecular and Cellular Biosciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo, 113-0032, Japan.
| |
Collapse
|