1
|
Xue Q, Ma Y, Shao H. Bacillus amyloliquefaciens Protect Against Atherosclerosis Through Alleviating Foam Cell Formation and Macrophage Polarization. Curr Microbiol 2024; 81:263. [PMID: 38997545 DOI: 10.1007/s00284-024-03775-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 11/01/2023] [Indexed: 07/14/2024]
Abstract
This study was to investigate the therapeutic effect of Bacillus amyloliquefaciens (Ba) on atherosclerosis (AS). THP-1 monocyte was differentiated to THP-1 macrophage (THP-M) through phorbol 12-myristate 13-acetate. After pre-treatment by 108 cfu/ml Ba lasting 6 h, THP-M was induced with 100 mg/l ox-LDL lasting 48 h to form macrophage foam cell (THP-F). RT-qPCR and flow cytometry were employed to determine the polarization of THP-M and THP-F. ApoE-/- mice with high-fat and high-cholesterol diet were used for constructing an AS model to evaluate the effect of Ba on AS. Our in vitro results showed that Ba vegetative cells pre-treatment distinctly inhibited the levels of iNOS and CD16/CD32 (M1 macrophage markers), and increased the levels of FIZZ1, Ym1, Arg1, CD163, and CD206 (M2 macrophage markers), indicating that Ba pre-treatment promoted anti-inflammatory M2-like polarization both in THP-M and THP-F. Meanwhile, it also suppressed cholesterol uptake, esterification, and hydrolysis, and efflux by THP-M and THP-F. Additionally, our animal experiments demonstrated that Ba vegetative cells treatment suppressed high cholesterol, hyperglycemia, hyperlipidemia, and the release of inflammatory factors (TNF-α, IL-6 and IL-1β) in ApoE-/- AS mice. In a word, our results indicated that Ba may protect against AS through alleviating foam cell formation and macrophage polarization through targeting certain stages of AS.
Collapse
Affiliation(s)
- Qi Xue
- Department of Cardiology, Zhejiang Provincial People's Hospital, No. 158 Shangtang Road, Hangzhou, 310014, Zhejiang, China
| | - Yuan Ma
- Department of Cardiology, Zhejiang Provincial People's Hospital, No. 158 Shangtang Road, Hangzhou, 310014, Zhejiang, China
| | - Hong Shao
- Department of Cardiology, Zhejiang Provincial People's Hospital, No. 158 Shangtang Road, Hangzhou, 310014, Zhejiang, China.
| |
Collapse
|
2
|
Fazliyeva R, Makhov P, Uzzo RG, Kolenko VM. Targeting NPC1 in Renal Cell Carcinoma. Cancers (Basel) 2024; 16:517. [PMID: 38339268 PMCID: PMC10854724 DOI: 10.3390/cancers16030517] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 01/05/2024] [Accepted: 01/20/2024] [Indexed: 02/12/2024] Open
Abstract
Rapidly proliferating cancer cells have a greater requirement for cholesterol than normal cells. Tumor cells are largely dependent on exogenous lipids given that their growth requirements are not fully met by endogenous pathways. Our current study shows that ccRCC cells have redundant mechanisms of cholesterol acquisition. We demonstrate that all major lipoproteins (i.e., LDL, HDL, and VLDL) have a comparable ability to support the growth of ccRCC cells and are equally effective in counteracting the antitumor activities of TKIs. The intracellular trafficking of exogenous lipoprotein-derived cholesterol appears to be distinct from the movement of endogenously synthesized cholesterol. De novo synthetized cholesterol is transported from the endoplasmic reticulum directly to the plasma membrane and to the acyl-CoA: cholesterol acyltransferase, whereas lipoprotein-derived cholesterol is distributed through the NPC1-dependent endosomal trafficking system. Expression of NPC1 is increased in ccRCC at mRNA and protein levels, and high expression of NPC1 is associated with poor prognosis. Our current findings show that ccRCC cells are particularly sensitive to the inhibition of endolysosomal cholesterol export and underline the therapeutic potential of targeting NPC1 in ccRCC.
Collapse
Affiliation(s)
- Rushaniya Fazliyeva
- Nuclear Dynamics and Cancer Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA;
| | - Peter Makhov
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA;
| | - Robert G. Uzzo
- Department of Urology, Fox Chase Cancer Center, Philadelphia, PA 19111, USA;
| | - Vladimir M. Kolenko
- Nuclear Dynamics and Cancer Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA;
| |
Collapse
|
3
|
Wang S, Link F, Han M, Chaudhary R, Asimakopoulos A, Liebe R, Yao Y, Hammad S, Dropmann A, Krizanac M, Rubie C, Feiner LK, Glanemann M, Ebert MPA, Weiskirchen R, Henis YI, Ehrlich M, Dooley S. The Interplay of TGF-β1 and Cholesterol Orchestrating Hepatocyte Cell Fate, EMT, and Signals for HSC Activation. Cell Mol Gastroenterol Hepatol 2023; 17:567-587. [PMID: 38154598 PMCID: PMC10883985 DOI: 10.1016/j.jcmgh.2023.12.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 12/21/2023] [Accepted: 12/22/2023] [Indexed: 12/30/2023]
Abstract
BACKGROUND & AIMS Transforming growth factor-β1 (TGF-β1) plays important roles in chronic liver diseases, including metabolic dysfunction-associated steatotic liver disease (MASLD). MASLD involves various biological processes including dysfunctional cholesterol metabolism and contributes to progression to metabolic dysfunction-associated steatohepatitis and hepatocellular carcinoma. However, the reciprocal regulation of TGF-β1 signaling and cholesterol metabolism in MASLD is yet unknown. METHODS Changes in transcription of genes associated with cholesterol metabolism were assessed by RNA sequencing of murine hepatocyte cell line (alpha mouse liver 12/AML12) and mouse primary hepatocytes treated with TGF-β1. Functional assays were performed on AML12 cells (untreated, TGF-β1 treated, or subjected to cholesterol enrichment [CE] or cholesterol depletion [CD]), and on mice injected with adenovirus-associated virus 8-control/TGF-β1. RESULTS TGF-β1 inhibited messenger RNA expression of several cholesterol metabolism regulatory genes, including rate-limiting enzymes of cholesterol biosynthesis in AML12 cells, mouse primary hepatocytes, and adenovirus-associated virus-TGF-β1-treated mice. Total cholesterol levels and lipid droplet accumulation in AML12 cells and liver tissue also were reduced upon TGF-β1 treatment. Smad2/3 phosphorylation after 2 hours of TGF-β1 treatment persisted after CE or CD and was mildly increased after CD, whereas TGF-β1-mediated AKT phosphorylation (30 min) was inhibited by CE. Furthermore, CE protected AML12 cells from several effects mediated by 72 hours of incubation with TGF-β1, including epithelial-mesenchymal transition, actin polymerization, and apoptosis. CD mimicked the outcome of long-term TGF-β1 administration, an effect that was blocked by an inhibitor of the type I TGF-β receptor. In addition, the supernatant of CE- or CD-treated AML12 cells inhibited or promoted, respectively, the activation of LX-2 hepatic stellate cells. CONCLUSIONS TGF-β1 inhibits cholesterol metabolism whereas cholesterol attenuates TGF-β1 downstream effects in hepatocytes.
Collapse
Affiliation(s)
- Sai Wang
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Frederik Link
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Mei Han
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; Department of Internal Medicine, The Second Hospital of Dalian Medical University, Dalian, China
| | - Roohi Chaudhary
- Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel; Department of Neurobiology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Anastasia Asimakopoulos
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry, RWTH Aachen University Hospital, Aachen, Germany
| | - Roman Liebe
- Clinic of Gastroenterology, Hepatology and Infectious Diseases, Otto-von-Guericke-University, Magdeburg, Germany
| | - Ye Yao
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Seddik Hammad
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Anne Dropmann
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Marinela Krizanac
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry, RWTH Aachen University Hospital, Aachen, Germany
| | - Claudia Rubie
- Department of General, Visceral, Vascular and Pediatric Surgery, Saarland University, Homburg/Saar, Germany
| | - Laura Kim Feiner
- Department of General, Visceral, Vascular and Pediatric Surgery, Saarland University, Homburg/Saar, Germany
| | - Matthias Glanemann
- Department of General, Visceral, Vascular and Pediatric Surgery, Saarland University, Homburg/Saar, Germany
| | - Matthias P A Ebert
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; Mannheim Institute for Innate Immunoscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; Clinical Cooperation Unit Healthy Metabolism, Center of Preventive Medicine and Digital Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry, RWTH Aachen University Hospital, Aachen, Germany
| | - Yoav I Henis
- Department of Neurobiology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Marcelo Ehrlich
- Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Steven Dooley
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.
| |
Collapse
|
4
|
Owaki R, Aoki H, Toriuchi K, Inoue Y, Hayashi H, Takeshita S, Kakita H, Yamada Y, Aoyama M. AMPK activators suppress cholesterol accumulation in macrophages via suppression of the mTOR pathway. Exp Cell Res 2023; 432:113784. [PMID: 37730144 DOI: 10.1016/j.yexcr.2023.113784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 09/13/2023] [Accepted: 09/14/2023] [Indexed: 09/22/2023]
Abstract
Atherosclerosis is a persistent inflammatory state that contributes significantly to cardiovascular disease, a primary cause of mortality worldwide. Enhanced lipid uptake by macrophages and their transformation into foam cells play a key role in the development of atherosclerosis. Recent studies using in vivo mouse models indicated that activation of AMPK has anti-atherosclerotic effects by upregulating the expression of cholesterol efflux transporters in foam cells and promoting cholesterol efflux. However, the pathway downstream of AMPK that contributes to elevated expression of cholesterol efflux transporters remains unclear. In this study, we found that activation of AMPK by AICAR and metformin inhibits foam cell formation via suppression of mTOR in macrophages. Specifically, activation of AMPK indirectly reduced the phosphorylation level of mTOR at Ser2448 and promoted the expression of cholesterol efflux transporters and cholesterol efflux. These inhibitory effects on foam cell formation were counteracted by mTOR activators. Metformin, a more nonspecific AMPK activator than AICAR, appears to inhibit foam cell formation via anti-inflammatory effects in addition to suppression of the mTOR pathway. The results of this study suggest that the development of new drugs targeting AMPK activation and mTOR inhibition may lead to beneficial results in the prevention and treatment of atherosclerosis.
Collapse
Affiliation(s)
- Reina Owaki
- Department of Pathobiology, Nagoya City University Graduate School of Pharmaceutical Sciences, 3-1 Tanabe-dori, Mizuho-ku, Nagoya, Aichi, 467-8603, Japan
| | - Hiromasa Aoki
- Department of Pathobiology, Nagoya City University Graduate School of Pharmaceutical Sciences, 3-1 Tanabe-dori, Mizuho-ku, Nagoya, Aichi, 467-8603, Japan
| | - Kohki Toriuchi
- Department of Pathobiology, Nagoya City University Graduate School of Pharmaceutical Sciences, 3-1 Tanabe-dori, Mizuho-ku, Nagoya, Aichi, 467-8603, Japan
| | - Yasumichi Inoue
- Department of Cell Signaling, Nagoya City University Graduate School of Pharmaceutical Sciences, 3-1 Tanabe-dori, Mizuho-ku, Nagoya, Aichi, 467-8603, Japan
| | - Hidetoshi Hayashi
- Department of Cell Signaling, Nagoya City University Graduate School of Pharmaceutical Sciences, 3-1 Tanabe-dori, Mizuho-ku, Nagoya, Aichi, 467-8603, Japan
| | - Satoru Takeshita
- Department of Pathobiology, Nagoya City University Graduate School of Pharmaceutical Sciences, 3-1 Tanabe-dori, Mizuho-ku, Nagoya, Aichi, 467-8603, Japan; Department of Perinatal and Neonatal Medicine, Aichi Medical University, 1-1 Yazakokarimata, Nagakute, Aichi, 480-1195, Japan
| | - Hiroki Kakita
- Department of Pathobiology, Nagoya City University Graduate School of Pharmaceutical Sciences, 3-1 Tanabe-dori, Mizuho-ku, Nagoya, Aichi, 467-8603, Japan; Department of Perinatal and Neonatal Medicine, Aichi Medical University, 1-1 Yazakokarimata, Nagakute, Aichi, 480-1195, Japan
| | - Yasumasa Yamada
- Department of Perinatal and Neonatal Medicine, Aichi Medical University, 1-1 Yazakokarimata, Nagakute, Aichi, 480-1195, Japan
| | - Mineyoshi Aoyama
- Department of Pathobiology, Nagoya City University Graduate School of Pharmaceutical Sciences, 3-1 Tanabe-dori, Mizuho-ku, Nagoya, Aichi, 467-8603, Japan.
| |
Collapse
|
5
|
Strahlhofer-Augsten M, Schliefsteiner C, Cvitic S, George M, Lang-Olip I, Hirschmugl B, Marsche G, Lang U, Novakovic B, Saffery R, Desoye G, Wadsack C. The Distinct Role of the HDL Receptor SR-BI in Cholesterol Homeostasis of Human Placental Arterial and Venous Endothelial Cells. Int J Mol Sci 2022; 23:ijms23105364. [PMID: 35628180 PMCID: PMC9141204 DOI: 10.3390/ijms23105364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 04/28/2022] [Accepted: 04/30/2022] [Indexed: 11/17/2022] Open
Abstract
As opposed to adults, high-density lipoprotein (HDL) is the main cholesterol carrying lipoprotein in fetal circulation. The major HDL receptor, scavenger receptor class B type I (SR-BI), contributes to local cholesterol homeostasis. Arterial endothelial cells (ECA) from human placenta are enriched with cholesterol compared to venous endothelial cells (ECV). Moreover, umbilical venous and arterial plasma cholesterol levels differ markedly. We tested the hypothesis that the uptake of HDL-cholesteryl esters differs between ECA and ECV because of the differential expression of SR-BI. We aimed to identify the key regulators underlying these differences and the functional consequences. Immunohistochemistry was used for visualization of SR-BI in situ. ECA and ECV were isolated from the chorionic plate of human placenta and used for RT-qPCR, Western Blot, and HDL uptake assays with 3H- and 125I-labeled HDL. DNA was extracted for the methylation profiling of the SR-BI promoter. SR-BI regulation was studied by exposing ECA and ECV to differential oxygen concentrations or shear stress. Our results show elevated SR-BI expression and protein abundance in ECA compared to ECV in situ and in vitro. Immunohistochemistry demonstrated that SR-BI is mainly expressed on the apical side of placental endothelial cells in situ, allowing interaction with mature HDL circulating in the fetal blood. This was functionally linked to a higher increase of selective cholesterol ester uptake from fetal HDL in ECA than in ECV, and resulted in increased cholesterol availability in ECA. SR-BI expression on ECV tended to decrease with shear stress, which, together with heterogeneous immunostaining, suggests that SR-BI expression is locally regulated in the placental vasculature. In addition, hypomethylation of several CpG sites within the SR-BI promoter region might contribute to differential expression of SR-BI between chorionic arteries and veins. Therefore, SR-BI contributes to a local cholesterol homeostasis in ECA and ECV of the human feto-placental vasculature.
Collapse
Affiliation(s)
- Manuela Strahlhofer-Augsten
- Research Unit, Department of Obstetrics and Gynecology, Medical University of Graz, 8036 Graz, Austria; (M.S.-A.); (C.S.); (S.C.); (B.H.); (G.D.)
- BioBank Graz, Medical University of Graz, 8036 Graz, Austria
| | - Carolin Schliefsteiner
- Research Unit, Department of Obstetrics and Gynecology, Medical University of Graz, 8036 Graz, Austria; (M.S.-A.); (C.S.); (S.C.); (B.H.); (G.D.)
| | - Silvija Cvitic
- Research Unit, Department of Obstetrics and Gynecology, Medical University of Graz, 8036 Graz, Austria; (M.S.-A.); (C.S.); (S.C.); (B.H.); (G.D.)
- Research Unit of Analytical Mass Spectrometry, Cell Biology and Biochemistry of Inborn Errors of Metabolism, Department of Paediatrics and Adolescent Medicine, Medical University of Graz, 8036 Graz, Austria
| | - Meekha George
- Otto Loewi Research Center, Division of Pathophysiology and Immunology, Medical University of Graz, 8010 Graz, Austria;
| | - Ingrid Lang-Olip
- Gottfried Schatz Research Center, Divison of Cell Biology, Histology and Embryology, Medical University of Graz, 8036 Graz, Austria;
| | - Birgit Hirschmugl
- Research Unit, Department of Obstetrics and Gynecology, Medical University of Graz, 8036 Graz, Austria; (M.S.-A.); (C.S.); (S.C.); (B.H.); (G.D.)
| | - Gunther Marsche
- Otto Loewi Research Center, Division of Pharmacology, Medical University of Graz, 8010 Graz, Austria;
| | - Uwe Lang
- Research Unit, Department of Obstetrics and Gynecology, Medical University of Graz, 8036 Graz, Austria; (M.S.-A.); (C.S.); (S.C.); (B.H.); (G.D.)
| | - Boris Novakovic
- Molecular Immunity, Infection and Immunity Theme, Murdoch Children’s Research Institute, Parkville, VIC 3052, Australia; (B.N.); (R.S.)
| | - Richard Saffery
- Molecular Immunity, Infection and Immunity Theme, Murdoch Children’s Research Institute, Parkville, VIC 3052, Australia; (B.N.); (R.S.)
| | - Gernot Desoye
- Research Unit, Department of Obstetrics and Gynecology, Medical University of Graz, 8036 Graz, Austria; (M.S.-A.); (C.S.); (S.C.); (B.H.); (G.D.)
| | - Christian Wadsack
- Research Unit, Department of Obstetrics and Gynecology, Medical University of Graz, 8036 Graz, Austria; (M.S.-A.); (C.S.); (S.C.); (B.H.); (G.D.)
- Correspondence:
| |
Collapse
|
6
|
Pavlík V, Machalová V, Čepa M, Šínová R, Šafránková B, Kulhánek J, Drmota T, Kubala L, Huerta-Ángeles G, Velebný V, Nešporová K. Retinoic Acid Grafted to Hyaluronic Acid Activates Retinoid Gene Expression and Removes Cholesterol from Cellular Membranes. Biomolecules 2022; 12:biom12020200. [PMID: 35204701 PMCID: PMC8961547 DOI: 10.3390/biom12020200] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 01/12/2022] [Accepted: 01/20/2022] [Indexed: 02/05/2023] Open
Abstract
All-trans-retinoic acid (atRA) is a potent ligand that regulates gene expression and is used to treat several skin disorders. Hyaluronic acid (HA) was previously conjugated with atRA (HA-atRA) to obtain a novel amphiphilic compound. HA-atRA forms micelles that incorporate hydrophobic molecules and facilitate their transport through the skin. The aim of this study was to determine the influence of HA-atRA on gene expression in skin cells and to compare it with that of unbound atRA. Gene expression was investigated using microarrays and a luciferase system with a canonical atRA promoter. HA-atRA upregulated gene expression similarly to atRA. However, HA-atRA activated the expression of cholesterol metabolism genes, unlike atRA. Further investigation using HPLC and filipin III staining suggested that the treated cells induced cholesterol synthesis to replenish the cholesterol removed from the cells by HA-atRA. HA modified with oleate (HA-C18:1) removed cholesterol from the cells similarly to HA-atRA, suggesting that the cholesterol removal stemmed from the amphiphilic nature of the two derivatives. HA-atRA induces retinoid signaling. Thus, HA-atRA could be used to treat skin diseases, such as acne and psoriasis, where the combined action of atRA signaling and anti-inflammatory cholesterol removal may be potentially beneficial.
Collapse
Affiliation(s)
- Vojtěch Pavlík
- R&D Department, Contipro, a.s., 562 04 Dolní Dobrouč, Czech Republic; (V.M.); (M.Č.); (R.Š.); (B.Š.); (J.K.); (T.D.); (G.H.-Á.); (V.V.); (K.N.)
- Third Faculty of Medicine, Charles University, 100 00 Prague, Czech Republic
- Correspondence:
| | - Veronika Machalová
- R&D Department, Contipro, a.s., 562 04 Dolní Dobrouč, Czech Republic; (V.M.); (M.Č.); (R.Š.); (B.Š.); (J.K.); (T.D.); (G.H.-Á.); (V.V.); (K.N.)
| | - Martin Čepa
- R&D Department, Contipro, a.s., 562 04 Dolní Dobrouč, Czech Republic; (V.M.); (M.Č.); (R.Š.); (B.Š.); (J.K.); (T.D.); (G.H.-Á.); (V.V.); (K.N.)
| | - Romana Šínová
- R&D Department, Contipro, a.s., 562 04 Dolní Dobrouč, Czech Republic; (V.M.); (M.Č.); (R.Š.); (B.Š.); (J.K.); (T.D.); (G.H.-Á.); (V.V.); (K.N.)
- Institute of Experimental Biology, Faculty of Science, Masaryk University, 625 00 Brno, Czech Republic;
| | - Barbora Šafránková
- R&D Department, Contipro, a.s., 562 04 Dolní Dobrouč, Czech Republic; (V.M.); (M.Č.); (R.Š.); (B.Š.); (J.K.); (T.D.); (G.H.-Á.); (V.V.); (K.N.)
| | - Jaromír Kulhánek
- R&D Department, Contipro, a.s., 562 04 Dolní Dobrouč, Czech Republic; (V.M.); (M.Č.); (R.Š.); (B.Š.); (J.K.); (T.D.); (G.H.-Á.); (V.V.); (K.N.)
| | - Tomáš Drmota
- R&D Department, Contipro, a.s., 562 04 Dolní Dobrouč, Czech Republic; (V.M.); (M.Č.); (R.Š.); (B.Š.); (J.K.); (T.D.); (G.H.-Á.); (V.V.); (K.N.)
| | - Lukáš Kubala
- Institute of Experimental Biology, Faculty of Science, Masaryk University, 625 00 Brno, Czech Republic;
- International Clinical Research Center, St. Anne’s University Hospital, 656 91 Brno, Czech Republic
| | - Gloria Huerta-Ángeles
- R&D Department, Contipro, a.s., 562 04 Dolní Dobrouč, Czech Republic; (V.M.); (M.Č.); (R.Š.); (B.Š.); (J.K.); (T.D.); (G.H.-Á.); (V.V.); (K.N.)
| | - Vladimír Velebný
- R&D Department, Contipro, a.s., 562 04 Dolní Dobrouč, Czech Republic; (V.M.); (M.Č.); (R.Š.); (B.Š.); (J.K.); (T.D.); (G.H.-Á.); (V.V.); (K.N.)
| | - Kristina Nešporová
- R&D Department, Contipro, a.s., 562 04 Dolní Dobrouč, Czech Republic; (V.M.); (M.Č.); (R.Š.); (B.Š.); (J.K.); (T.D.); (G.H.-Á.); (V.V.); (K.N.)
| |
Collapse
|
7
|
Casado ME, Huerta L, Marcos-Díaz A, Ortiz AI, Kraemer FB, Lasunción MA, Busto R, Martín-Hidalgo A. Hormone-sensitive lipase deficiency affects the expression of SR-BI, LDLr, and ABCA1 receptors/transporters involved in cellular cholesterol uptake and efflux and disturbs fertility in mouse testis. Biochim Biophys Acta Mol Cell Biol Lipids 2021; 1866:159043. [PMID: 34461308 DOI: 10.1016/j.bbalip.2021.159043] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 08/18/2021] [Accepted: 08/19/2021] [Indexed: 12/14/2022]
Abstract
Hormone-sensitive lipase (HSL) hydrolyse acylglycerols, cholesteryl and retinyl esters. HSL is a key lipase in mice testis, as HSL deficiency results in male sterility. The present work study the effects of the deficiency and lack of HSL on the localization and expression of SR-BI, LDLr, and ABCA1 receptors/transporters involved in uptake and efflux of cholesterol in mice testis, to determine the impact of HSL gene dosage on testis morphology, lipid homeostasis and fertility. The results of this work show that the lack of HSL in mice alters testis morphology and spermatogenesis, decreasing sperm counts, sperm motility and increasing the amount of Leydig cells and lipid droplets. They also show that there are differences in the localization of HSL, SR-BI, LDLr and ABCA1 in HSL+/+, HSL+/- and HSL-/- mice. The deficiency or lack of HSL has effects on protein and mRNA expression of genes involved in lipid metabolisms in mouse testis. HSL-/- testis have augmented expression of SR-BI, LDLr, ABCA1 and LXRβ, a critical sterol sensor that regulate multiple genes involved in lipid metabolism; whereas LDLr expression decreased in HSL+/- mice. Plin2, Abca1 and Ldlr mRNA levels increased; and LXRα (Nr1h3) and LXRβ (Nr1h2) decreased in testis from HSL-/- compared with HSL+/+; with no differences in Scarb1. Together these data suggest that HSL deficiency or lack in mice testis induces lipid homeostasis alterations that affect the cellular localization and expression of key receptors/transporter involved in cellular cholesterol uptake and efflux (SR-BI, LDRr, ABCA1); alters normal cellular function and impact fertility.
Collapse
Affiliation(s)
- María Emilia Casado
- Servicio de Bioquímica-Investigación, Hospital Universitario Ramón y Cajal, Instituto Ramón y Cajal de Investigación Sanitaria (IRyCIS), E-28034 Madrid, Spain; CIBER de Fisiopatología de la Obesidad y Nutrición (CIBERobn), ISCIII, Spain
| | - Lydia Huerta
- Servicio de Bioquímica-Investigación, Hospital Universitario Ramón y Cajal, Instituto Ramón y Cajal de Investigación Sanitaria (IRyCIS), E-28034 Madrid, Spain; CIBER de Fisiopatología de la Obesidad y Nutrición (CIBERobn), ISCIII, Spain
| | - Ana Marcos-Díaz
- Servicio de Bioquímica-Investigación, Hospital Universitario Ramón y Cajal, Instituto Ramón y Cajal de Investigación Sanitaria (IRyCIS), E-28034 Madrid, Spain; CIBER de Fisiopatología de la Obesidad y Nutrición (CIBERobn), ISCIII, Spain
| | - Ana Isabel Ortiz
- Unidad de Cirugía Experimental y Animalario, Hospital Universitario Ramón y Cajal, Instituto Ramón y Cajal de Investigación Sanitaria (IRyCIS), E-28034 Madrid, Spain
| | - Fredric B Kraemer
- Division of Endocrinology, Stanford University, United States of America; VA Palo Alto Health Care System, Palo Alto, CA, United States of America
| | - Miguel Angel Lasunción
- Servicio de Bioquímica-Investigación, Hospital Universitario Ramón y Cajal, Instituto Ramón y Cajal de Investigación Sanitaria (IRyCIS), E-28034 Madrid, Spain; CIBER de Fisiopatología de la Obesidad y Nutrición (CIBERobn), ISCIII, Spain
| | - Rebeca Busto
- Servicio de Bioquímica-Investigación, Hospital Universitario Ramón y Cajal, Instituto Ramón y Cajal de Investigación Sanitaria (IRyCIS), E-28034 Madrid, Spain; CIBER de Fisiopatología de la Obesidad y Nutrición (CIBERobn), ISCIII, Spain
| | - Antonia Martín-Hidalgo
- Servicio de Bioquímica-Investigación, Hospital Universitario Ramón y Cajal, Instituto Ramón y Cajal de Investigación Sanitaria (IRyCIS), E-28034 Madrid, Spain; CIBER de Fisiopatología de la Obesidad y Nutrición (CIBERobn), ISCIII, Spain.
| |
Collapse
|
8
|
Wang H, Zhang P, Chen X, Liu W, Fu Z, Liu M. Activin a inhibits foam cell formation and up-regulates ABCA1 and ABCG1 expression through Alk4-Smad signaling pathway in RAW 264.7 macrophages. Steroids 2021; 174:108887. [PMID: 34237315 DOI: 10.1016/j.steroids.2021.108887] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 06/23/2021] [Accepted: 06/26/2021] [Indexed: 12/14/2022]
Abstract
BACKGROUND Activin A has been reported to play important roles in the pathogenesis of atherosclerosis. The purpose of this study is to investigate the effects of activin A on oxidized low-density lipoprotein (ox-LDL)-induced foam cell formation and explore the underlying molecular mechanisms in murine macrophage-like cell line RAW 264.7. METHODS The effects of activin A on Dil-labeled ox-LDL uptake were examined by confocal microscopy and flow cytometry analysis. The mRNA and protein levels of cholesterol receptors were analyzed by RT-qPCR and western blot analysis, respectively. To investigate whether activin receptor-like kinase 4 (Alk4) is required for activin A-mediated cellular effects, cells were pre-treated with SB-431542. The involvement of Smad2, Smad3 and Smad4 was confirmed by transfection with specific small interfering RNAs (siRNAs). RESULTS Activin A inhibits ox-ldl-induced foam cell formation and class A scavenger receptors (SR-A) expression, while up-regulates ATP-binding cassette transporter A1 (ABCA1) and ABCG1 expression in RAW 264.7 macrophages. Pre-treatment with SB-431542 abolished activin A-mediated anti-atherogenic effect. Knockdown of Smad2 reversed activin A-induced inhibition of ox-LDL uptake and SR-A expression. However, knockdown of Smad3 or Smad4 did not have such effect. Meanwhile, knockdown of either Smad2, Smad3 or Smad4 reversed the activin A-induced up-regulation of ABCA1 and ABCG1. CONCLUSIONS Our study provides novel evidence that activin A may exert anti-atherogenic effects through Alk4-Smad signaling pathway in RAW 264.7 macrophages.
Collapse
Affiliation(s)
- Hao Wang
- Department of Geriatrics, Peking University First Hospital, Beijing 100034, People's Republic of China
| | - Peng Zhang
- Department of Geriatrics, Peking University First Hospital, Beijing 100034, People's Republic of China; Division of Cardiology, Xiamen Cardiovascular Hospital, Xiamen University, Xiamen 361000, People's Republic of China
| | - Xiahuan Chen
- Department of Geriatrics, Peking University First Hospital, Beijing 100034, People's Republic of China
| | - Wenwen Liu
- Department of Geriatrics, Peking University First Hospital, Beijing 100034, People's Republic of China
| | - Zhifang Fu
- Department of Geriatrics, Peking University First Hospital, Beijing 100034, People's Republic of China
| | - Meilin Liu
- Department of Geriatrics, Peking University First Hospital, Beijing 100034, People's Republic of China.
| |
Collapse
|
9
|
TGF-β/activin signaling promotes CDK7 inhibitor resistance in triple-negative breast cancer cells through upregulation of multidrug transporters. J Biol Chem 2021; 297:101162. [PMID: 34481843 PMCID: PMC8498470 DOI: 10.1016/j.jbc.2021.101162] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Revised: 08/23/2021] [Accepted: 08/31/2021] [Indexed: 01/10/2023] Open
Abstract
Cyclin-dependent kinase 7 (CDK7) is a master regulatory kinase that drives cell cycle progression and stimulates expression of oncogenes in a myriad of cancers. Inhibitors of CDK7 (CDK7i) are currently in clinical trials; however, as with many cancer therapies, patients will most likely experience recurrent disease due to acquired resistance. Identifying targets underlying CDK7i resistance will facilitate prospective development of new therapies that can circumvent such resistance. Here we utilized triple-negative breast cancer as a model to discern mechanisms of resistance as it has been previously shown to be highly responsive to CDK7 inhibitors. After generating cell lines with acquired resistance, high-throughput RNA sequencing revealed significant upregulation of genes associated with efflux pumps and transforming growth factor-beta (TGF-β) signaling pathways. Genetic silencing or pharmacological inhibition of ABCG2, an efflux pump associated with multidrug resistance, resensitized resistant cells to CDK7i, indicating a reliance on these transporters. Expression of activin A (INHBA), a member of the TGF-β family of ligands, was also induced, whereas its intrinsic inhibitor, follistatin (FST), was repressed. In resistant cells, increased phosphorylation of SMAD3, a downstream mediator, confirmed an increase in activin signaling, and phosphorylated SMAD3 directly bound the ABCG2 promoter regulatory region. Finally, pharmacological inhibition of TGF-β/activin receptors or genetic silencing of SMAD4, a transcriptional partner of SMAD3, reversed the upregulation of ABCG2 in resistant cells and phenocopied ABCG2 inhibition. This study reveals that inhibiting the TGF-β/Activin-ABCG2 pathway is a potential avenue for preventing or overcoming resistance to CDK7 inhibitors.
Collapse
|
10
|
Yang X, Lin P, Wang J, Liu N, Yin F, Shen N, Guo S. Purification, characterization and anti-atherosclerotic effects of the polysaccharides from the fruiting body of Cordyceps militaris. Int J Biol Macromol 2021; 181:890-904. [PMID: 33878353 DOI: 10.1016/j.ijbiomac.2021.04.083] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 04/01/2021] [Accepted: 04/14/2021] [Indexed: 12/16/2022]
Abstract
Hyperlipidemia is one major cause of atherosclerosis, which is a basic pathological change of cardiovascular diseases. Polysaccharide is a water-soluble component with lipid-lowering effects. In this study, alkaline-extracted polysaccharides were obtained from the fruiting body of C. militaris. Polysaccharides were purified via anion exchange and size exclusion chromatography. Their structural characteristics were investigated via chemical and spectroscopic methods. CM3I was mainly composed of →4)α-D-Glcp(1 → glycosyls and differed from starch due to the presence of →4,6)β-D-Glcp(1 → glycosyls. CM3II was characterized by its backbone, which was composed of →4)-β-D-Manp(1 → 6)-α-D-Manp(1 → 6)-β-D-Manp(1 → linked glycosyls, and especially the presence of O-methyl. Moreover, CM3II exhibited powerful anti-atherosclerotic effects via lowering plasma lipid levels in apolipoprotein E-deficient mice. The underlying mechanisms were attributed to its promoting effect on LXRα and inhibitory effect on SREBP-2. Collectively, CM3I and CM3II are different from the previously reported polysaccharides from C. militaris, and CM3II has a potential application in hypolipidemia and anti-atherosclerosis.
Collapse
Affiliation(s)
- Xiaoqian Yang
- Institute of Lipid metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Weifang Medical University, Weifang 261053, China
| | - Ping Lin
- Institute of Lipid metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Weifang Medical University, Weifang 261053, China
| | - Jin Wang
- Institute of Lipid metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Weifang Medical University, Weifang 261053, China
| | - Na Liu
- Institute of Lipid metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Weifang Medical University, Weifang 261053, China
| | - Fan Yin
- Institute of Lipid metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Weifang Medical University, Weifang 261053, China
| | - Nuo Shen
- Institute of Lipid metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Weifang Medical University, Weifang 261053, China
| | - Shoudong Guo
- Institute of Lipid metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Weifang Medical University, Weifang 261053, China.
| |
Collapse
|
11
|
Lei S, Chen J, Song C, Li J, Zuo A, Xu D, Li T, Guo Y. CTRP9 alleviates foam cells apoptosis by enhancing cholesterol efflux. Mol Cell Endocrinol 2021; 522:111138. [PMID: 33352225 DOI: 10.1016/j.mce.2020.111138] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Revised: 12/11/2020] [Accepted: 12/17/2020] [Indexed: 12/27/2022]
Abstract
The apoptosis of foam cells leads to instability of atherosclerotic plaques. This study was designed to explore the protective role of CTRP9 in foam cell apoptosis. In our experiment, CTRP9 alleviated foam cell apoptosis. Meanwhile, CTRP9 upregulated the expression of proteins important for cholesterol efflux, such as LXRα, CYP27A1, ABCG1 and ABCA1, and improved cholesterol efflux in foam cells. Moreover, CTRP9 inhibited Wnt3a and β-catenin expression and β-catenin nuclear translocation in foam cells. In addition, adenovirus overexpression of Wnt3a abolished the effect of CTRP9 on macrophage apoptosis. Mechanistically, the AMPK inhibitor abolished the effect of CTRP9 on foam cell apoptosis, and downregulation of AdipoR1 by siRNA abrogated the activation of AMPK and the effect of CTRP9 on foam cell apoptosis. We concluded that CTRP9 achieved these protective effects on foam cells through the AdipoR1/AMPK pathway.
Collapse
Affiliation(s)
- Shengyun Lei
- Department of General Medicine, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, 250012, Jinan, Shandong, China; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, 250012, Jinan, China
| | - Jiying Chen
- Department of General Medicine, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, 250012, Jinan, Shandong, China; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, 250012, Jinan, China
| | - Chengxiang Song
- Department of Cardiology, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, 250012, Jinan, Shandong, China; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, 250012, Jinan, China
| | - Jun Li
- Department of General Medicine, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, 250012, Jinan, Shandong, China; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, 250012, Jinan, China
| | - Anju Zuo
- Department of General Medicine, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, 250012, Jinan, Shandong, China; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, 250012, Jinan, China
| | - Dan Xu
- Department of General Medicine, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, 250012, Jinan, Shandong, China; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, 250012, Jinan, China
| | - Tingting Li
- Department of General Medicine, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, 250012, Jinan, Shandong, China; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, 250012, Jinan, China.
| | - Yuan Guo
- Department of General Medicine, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, 250012, Jinan, Shandong, China; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, 250012, Jinan, China.
| |
Collapse
|
12
|
Castaño D, Rattanasopa C, Monteiro-Cardoso VF, Corlianò M, Liu Y, Zhong S, Rusu M, Liehn EA, Singaraja RR. Lipid efflux mechanisms, relation to disease and potential therapeutic aspects. Adv Drug Deliv Rev 2020; 159:54-93. [PMID: 32423566 DOI: 10.1016/j.addr.2020.04.013] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 04/29/2020] [Accepted: 04/30/2020] [Indexed: 02/06/2023]
Abstract
Lipids are hydrophobic and amphiphilic molecules involved in diverse functions such as membrane structure, energy metabolism, immunity, and signaling. However, altered intra-cellular lipid levels or composition can lead to metabolic and inflammatory dysfunction, as well as lipotoxicity. Thus, intra-cellular lipid homeostasis is tightly regulated by multiple mechanisms. Since most peripheral cells do not catabolize cholesterol, efflux (extra-cellular transport) of cholesterol is vital for lipid homeostasis. Defective efflux contributes to atherosclerotic plaque development, impaired β-cell insulin secretion, and neuropathology. Of these, defective lipid efflux in macrophages in the arterial walls leading to foam cell and atherosclerotic plaque formation has been the most well studied, likely because a leading global cause of death is cardiovascular disease. Circulating high density lipoprotein particles play critical roles as acceptors of effluxed cellular lipids, suggesting their importance in disease etiology. We review here mechanisms and pathways that modulate lipid efflux, the role of lipid efflux in disease etiology, and therapeutic options aimed at modulating this critical process.
Collapse
|
13
|
Protective Effect of Shenqi Maixintong Capsule against ox-LDL-Induced Injury in RAW264.7 Macrophages. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020. [DOI: 10.1155/2020/6708152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Shenqi Maixintong capsule (SMC), a traditional Chinese medicine compound, exerts various therapeutic effects, including nourishing one’s vitality, improving blood circulation, regulating vital energy, and promoting coronary circulation. Studies have demonstrated that SMC could effectively alleviate atherosclerosis (AS) and delay its development. However, the effect of SMC on lipid metabolism remains unclear. Herein, we investigated the mechanism of SMC on lipid metabolism of RAW264.7 cells induced by oxidized low-density lipoprotein (ox-LDL). Drug-containing serum was prepared by intragastric administration of SD (Sprague Dawley) rats. RAW264.7 cells were transformed into foam cells with 25 mg/L ox-LDL. SMC-treated rat serum increased the survival rate of ox-LDL-induced RAW264.7 cells. Oil Red staining, total cholesterol, and free cholesterol detection data showed that the intermediated dose of SMC had the best effect on reducing the lipid accumulation, lipid droplets, intracellular cholesterol, and total cholesterol content of RAW264.7 cells. The results of western blotting showed that the expression of ABCA1, ABCG1, LXRα, PPARγ, and peroxisome proliferation-activated receptor alpha (PPARα) was increased by SMC, and proprotein convertase subtilisin/kexin type 9 (PCSK9) was reduced, which promoted reverse cholesterol transport (RCT) of RAW264.7 cells and inhibited foam cell formation. Furthermore, SOD and MDA data indicated that SMC could reduce the senescence of RAW264.7 cells. These findings suggest that SMC might prevent ox-LDL-induced damage of macrophages in AS patients by improving cell viability and slowing down lipid accumulation and senescence.
Collapse
|
14
|
Liu H, Jiang X, Gao X, Tian W, Xu C, Wang R, Xu Y, Wei L, Cao F, Li W. Identification of N-benzothiazolyl-2-benzenesulfonamides as novel ABCA1 expression upregulators. RSC Med Chem 2020; 11:411-418. [PMID: 33479646 DOI: 10.1039/c9md00556k] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 02/11/2020] [Indexed: 11/21/2022] Open
Abstract
ATP binding cassette transporter A1 (ABCA1) is a critical transporter that mediates cellular cholesterol efflux from macrophages to apolipoprotein A-I (ApoA-I). Therefore, increasing the expression level of ABCA1 is anti-atherogenic and ABCA1 expression upregulators have become novel choices for atherosclerosis treatment. In this study, a series of N-benzothiazolyl-2-benzenesulfonamides, based on the structure of WY06 discovered in our laboratory, were designed and synthesized as novel ABCA1 expression upregulators. Based on an in vitro ABCA1 upregulatory cell model, ABCA1 upregulation of target compounds was evaluated. Compounds 6c, 6d, and 6i have good upregulated ABCA1 expression activities, with EC50 values of 0.97, 0.37, and 0.41 μM, respectively. A preliminary structure-activity relationship is summarized. Replacing the methoxy group on the benzothiazole moiety of WY06 with a fluorine or chlorine atom and exchanging the ester group with a cyano group resulted in more potent ABCA1 upregulating activity. Moreover, compound 6i increased ABCA1 mRNA and protein expression and significantly promoted cholesterol efflux in RAW264.7 cells. In conclusion, N-benzothiazolyl-2-benzenesulfonamides were identified as novel ABCA1 expression upregulators.
Collapse
Affiliation(s)
- Hongtao Liu
- Department of Pharmacy , Hebei General Hospital , Shijiazhuang 05005 , China.,Hebei Key Laboratory of Organic Functional Molecules , College of Chemistry and Materials Science , Hebei Normal University , Shijiazhuang , 050024 , China .
| | - Xinhai Jiang
- NHC Key Laboratory of Biotechnology of Antibiotics , Institute of Medicinal Biotechnology , Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS&PUMC) , Beijing 100050 , China .
| | - Xinfeng Gao
- Hebei Key Laboratory of Organic Functional Molecules , College of Chemistry and Materials Science , Hebei Normal University , Shijiazhuang , 050024 , China .
| | - Wenhua Tian
- Hebei Key Laboratory of Organic Functional Molecules , College of Chemistry and Materials Science , Hebei Normal University , Shijiazhuang , 050024 , China .
| | - Chen Xu
- Hebei Key Laboratory of Organic Functional Molecules , College of Chemistry and Materials Science , Hebei Normal University , Shijiazhuang , 050024 , China .
| | - Ruizhi Wang
- Hebei Key Laboratory of Organic Functional Molecules , College of Chemistry and Materials Science , Hebei Normal University , Shijiazhuang , 050024 , China .
| | - Yanni Xu
- NHC Key Laboratory of Biotechnology of Antibiotics , Institute of Medicinal Biotechnology , Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS&PUMC) , Beijing 100050 , China .
| | - Liping Wei
- Department of Cardiology , Tianjin Union Medical Center , Nankai University Affiliated Hospital , 190 Jieyuan Road, Hongqiao , Tianjin 300121 , P. R. China
| | - Feng Cao
- Department of Cardiology & National Clinical Research Center of Geriatrics Disease , Chinese PLA General Hospital , Beijing 100853 , China
| | - Wenyan Li
- Hebei Key Laboratory of Organic Functional Molecules , College of Chemistry and Materials Science , Hebei Normal University , Shijiazhuang , 050024 , China .
| |
Collapse
|
15
|
Li T, Hu S, Pang X, Wang J, Yin J, Li F, Wang J, Yang X, Xia B, Liu Y, Song W, Guo S. The marine-derived furanone reduces intracellular lipid accumulation in vitro by targeting LXRα and PPARα. J Cell Mol Med 2020; 24:3384-3398. [PMID: 31981312 PMCID: PMC7131916 DOI: 10.1111/jcmm.15012] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 12/30/2019] [Accepted: 01/10/2020] [Indexed: 12/22/2022] Open
Abstract
Recent studies have demonstrated that commercially available lipid-lowering drugs cause various side effects; therefore, searching for anti-hyperlipidaemic compounds with lower toxicity is a research hotspot. This study was designed to investigate whether the marine-derived compound, 5-hydroxy-3-methoxy-5-methyl-4-butylfuran-2(5H)-one, has an anti-hyperlipidaemic activity, and the potential underlying mechanism in vitro. Results showed that the furanone had weaker cytotoxicity compared to positive control drugs. In RAW 264.7 cells, the furanone significantly lowered ox-LDL-induced lipid accumulation (~50%), and its triglyceride (TG)-lowering effect was greater than that of liver X receptor (LXR) agonist T0901317. In addition, it significantly elevated the protein levels of peroxisome proliferator-activated receptors (PPARα) and ATP-binding cassette (ABC) transporters, which could be partially inhibited by LXR antagonists, GSK2033 and SR9243. In HepG2 cells, it significantly decreased oleic acid-induced lipid accumulation, enhanced the protein levels of low-density lipoprotein receptor (LDLR), ABCG5, ABCG8 and PPARα, and reduced the expression of sterol regulatory element-binding protein 2 (~32%). PPARα antagonists, GW6471 and MK886, could significantly inhibit the furanone-induced lipid-lowering effect. Furthermore, the furanone showed a significantly lower activity on the activation of the expression of lipogenic genes compared to T0901317. Taken together, the furanone exhibited a weak cytotoxicity but had powerful TC- and TG-lowering effects most likely through targeting LXRα and PPARα, respectively. These findings indicate that the furanone has a potential application for the treatment of dyslipidaemia.
Collapse
Affiliation(s)
- Ting Li
- Institute of Lipid Metabolism and AtherosclerosisSchool of PharmacyInnovative Drug Research CentreWeifang Medical UniversityWeifangChina
| | - Shu‐Mei Hu
- Institute of Lipid Metabolism and AtherosclerosisSchool of PharmacyInnovative Drug Research CentreWeifang Medical UniversityWeifangChina
| | - Xiao‐Yan Pang
- Institute of Lipid Metabolism and AtherosclerosisSchool of PharmacyInnovative Drug Research CentreWeifang Medical UniversityWeifangChina
| | - Jun‐feng Wang
- CAS Key Laboratory of Tropical Marine Bio‐resources and Ecology/Guangdong Key Laboratory of Marine Materia Medica/RNAM Center for Marine MicrobiologySouth China Sea Institute of OceanologyChinese Academy of SciencesGuangzhouChina
| | - Jia‐Yu Yin
- Institute of Lipid Metabolism and AtherosclerosisSchool of PharmacyInnovative Drug Research CentreWeifang Medical UniversityWeifangChina
| | - Fa‐Hui Li
- Institute of Lipid Metabolism and AtherosclerosisSchool of PharmacyInnovative Drug Research CentreWeifang Medical UniversityWeifangChina
| | - Jin Wang
- Institute of Lipid Metabolism and AtherosclerosisSchool of PharmacyInnovative Drug Research CentreWeifang Medical UniversityWeifangChina
| | - Xiao‐Qian Yang
- Institute of Lipid Metabolism and AtherosclerosisSchool of PharmacyInnovative Drug Research CentreWeifang Medical UniversityWeifangChina
| | - Bin Xia
- Institute of Lipid Metabolism and AtherosclerosisSchool of PharmacyInnovative Drug Research CentreWeifang Medical UniversityWeifangChina
| | - Yong‐Hong Liu
- CAS Key Laboratory of Tropical Marine Bio‐resources and Ecology/Guangdong Key Laboratory of Marine Materia Medica/RNAM Center for Marine MicrobiologySouth China Sea Institute of OceanologyChinese Academy of SciencesGuangzhouChina
| | - Wei‐Guo Song
- Institute of Lipid Metabolism and AtherosclerosisSchool of PharmacyInnovative Drug Research CentreWeifang Medical UniversityWeifangChina
| | - Shou‐Dong Guo
- Institute of Lipid Metabolism and AtherosclerosisSchool of PharmacyInnovative Drug Research CentreWeifang Medical UniversityWeifangChina
| |
Collapse
|
16
|
Tan YL, Ou HX, Zhang M, Gong D, Zhao ZW, Chen LY, Xia XD, Mo ZC, Tang CK. Tanshinone IIA Promotes Macrophage Cholesterol Efflux and Attenuates Atherosclerosis of apoE-/- Mice by Omentin-1/ABCA1 Pathway. Curr Pharm Biotechnol 2019; 20:422-432. [PMID: 30947667 DOI: 10.2174/1389201020666190404125213] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 03/22/2019] [Accepted: 03/26/2019] [Indexed: 12/16/2022]
Abstract
BACKGROUND Tanshinone IIA (Tan IIA) and Omentin-1 have a protective role in the cardiovascular system. However, if and how Tan IIA and Omentin-1 regulate cholesterol metabolism in macrophages has not been fully elucidated. OBJECTIVE To investigate the possible mechanisms of Tan IIA and Omentin-1 on preventing macrophage cholesterol accumulation and atherosclerosis development. METHODS The effect of Tan IIA on the protein and mRNA levels of Omentin-1 and ATP-binding cassette transporter A1 (ABCA1) in macrophages was examined by Western blot and qRT-PCR assay, respectively. Cholesterol efflux was assessed by liquid scintillation counting (LSC). Cellular lipid droplet was measured by Oil Red O staining, and intracellular lipid content was detected by high performance liquid chromatography (HPLC). In addition, the serum lipid profile of apoE-/- mice was measured by enzymatic method. The size of atherosclerotic lesion areas and content of lipids and collagen in the aortic of apoE-/- mice were examined by Sudan IV, Oil-red O, and Masson staining, respectively. RESULTS Tan IIA up-regulated expression of Omentin-1 and ABCA1 in THP-1 macrophages, promoting ABCA1-mediated cholesterol efflux and consequently decreasing cellular lipid content. Consistently, Tan IIA increased reverse cholesterol transport in apoE-/- mice. Plasma levels of high-density lipoprotein cholesterol (HDL-C), ABCA1 expression and atherosclerotic plaque collagen content were increased while plasma levels of low-density lipoprotein cholesterol (LDL-C) and atherosclerotic plaque sizes were reduced in Tan IIA-treated apoE-/- mice. These beneficial effects were, however, essentially blocked by knockdown of Omentin-1. CONCLUSION Our results revealed that Tan IIA promotes cholesterol efflux and ameliorates lipid accumulation in macrophages most likely via the Omentin-1/ABCA1 pathway, reducing the development of aortic atherosclerosis.
Collapse
Affiliation(s)
- Yu-Lin Tan
- Institute of Cardiovascular Disease, Key Laboratory for Arterosclerology of Hunan Province, Medical Research Center, Hunan Province Cooperative innovation Center for Molecular Target New Drug Study, University of South China, Hengyang 421001, Hunan, China.,Key Laboratory for Natural Cardiovascular Medicine Research of Hunan Province, Institute of Pathology Research, Department of Pathophysiology, Key Disciplines of Immunology, XiangNan University, Chenzhou 423000, China
| | - Han-Xiao Ou
- Cooperative Innovation Base of Basic and Clinic Medicine, University of South China & Yueyan Maternity-Child Health Hospital, Department of Genetics and Eugenics, Yueyan Maternity-Child Health Hospital, 414000, Hunan, China.,Clinical Anatomy & Reproductive Medicine Application Institute, Department of Histology and Embryology, School of Medicine, University of South China
| | - Min Zhang
- Institute of Cardiovascular Disease, Key Laboratory for Arterosclerology of Hunan Province, Medical Research Center, Hunan Province Cooperative innovation Center for Molecular Target New Drug Study, University of South China, Hengyang 421001, Hunan, China
| | - Duo Gong
- Institute of Cardiovascular Disease, Key Laboratory for Arterosclerology of Hunan Province, Medical Research Center, Hunan Province Cooperative innovation Center for Molecular Target New Drug Study, University of South China, Hengyang 421001, Hunan, China
| | - Zhen-Wang Zhao
- Institute of Cardiovascular Disease, Key Laboratory for Arterosclerology of Hunan Province, Medical Research Center, Hunan Province Cooperative innovation Center for Molecular Target New Drug Study, University of South China, Hengyang 421001, Hunan, China
| | - Ling-Yan Chen
- Institute of Cardiovascular Disease, Key Laboratory for Arterosclerology of Hunan Province, Medical Research Center, Hunan Province Cooperative innovation Center for Molecular Target New Drug Study, University of South China, Hengyang 421001, Hunan, China
| | - Xiao-Dan Xia
- Institute of Cardiovascular Disease, Key Laboratory for Arterosclerology of Hunan Province, Medical Research Center, Hunan Province Cooperative innovation Center for Molecular Target New Drug Study, University of South China, Hengyang 421001, Hunan, China
| | - Zhong-Cheng Mo
- Cooperative Innovation Base of Basic and Clinic Medicine, University of South China & Yueyan Maternity-Child Health Hospital, Department of Genetics and Eugenics, Yueyan Maternity-Child Health Hospital, 414000, Hunan, China.,Clinical Anatomy & Reproductive Medicine Application Institute, Department of Histology and Embryology, School of Medicine, University of South China
| | - Chao-Ke Tang
- Institute of Cardiovascular Disease, Key Laboratory for Arterosclerology of Hunan Province, Medical Research Center, Hunan Province Cooperative innovation Center for Molecular Target New Drug Study, University of South China, Hengyang 421001, Hunan, China
| |
Collapse
|
17
|
Thelenota ananas saponin extracts attenuate the atherosclerosis in apoE−/− mice by modulating lipid metabolism. J Funct Foods 2019. [DOI: 10.1016/j.jff.2019.04.065] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
|
18
|
Lv Y, Yang J, Gao A, Sun S, Zheng X, Chen X, Wan W, Tang C, Xie W, Li S, Guo D, Peng T, Zhao G, Zhong L. Sortilin promotes macrophage cholesterol accumulation and aortic atherosclerosis through lysosomal degradation of ATP-binding cassette transporter A1 protein. Acta Biochim Biophys Sin (Shanghai) 2019; 51:471-483. [PMID: 30950489 DOI: 10.1093/abbs/gmz029] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Indexed: 11/13/2022] Open
Abstract
Sortilin is closely associated with hyperlipidemia and the risk of atherosclerosis (AS). The role of sortilin and the underlying mechanism in peripheral macrophage are not fully understood. In this study, we investigated the effect of macrophage sortilin on ATP-binding cassette transporter A1 (ABCA1) expression, ABCA1-mediated cholesterol efflux, and aortic AS. Macrophage sortilin expression was upregulated by oxidized low-density lipoproteins (ox-LDLs) in both concentration- and time-dependent manners. Its expression reached the peak level when cells were incubated with 50 μg/ml ox-LDL for 24 h. Overexpression of sortilin in macrophage reduced cholesterol efflux, leading to an increase in intracellular total cholesterol, free cholesterol, and cholesterol ester. Sortilin was found to bind with ABCA1 protein and suppress macrophage ABCA1 expression, resulting in a decrease in cholesterol efflux from macrophages. The inhibitory effect of sortilin in cholesterol efflux was partially reversed by treatment with chloroquine, a lysosomal inhibitor. On the contrary, the ABCA1 protein level and ABCA1-mediated cholesterol efflux is increased by sortilin short hairpin RNA transfection. The fecal and biliary cholesterol 3H-sterol from cholesterol-laden mouse peritoneal macrophage was reduced by sortilin overexpression through lentivirus vector (LV)-sortilin in low-density lipoprotein receptor knockout mice, which was prevented by co-treatment with chloroquine. Treatment with LV-sortilin reduced plasma high-density lipoprotein and increased plasma ox-LDL levels. Accordingly, aortic lipid deposition and plaque area were exacerbated, and ABCA1 expression was reduced in mice in response to infection with LV-sortilin alone. These effects of LV-sortilin were partially reversed by chloroquine. Sortilin enhances lysosomal degradation of ABCA1 protein and suppresses ABCA1-mediated cholesterol efflux from macrophages, leading to foam cell formation and AS development.
Collapse
Affiliation(s)
- Yuncheng Lv
- Clinical Anatomy and Reproductive Medicine Application Institute, University of South China, Hengyang, China
| | - Jing Yang
- Clinical Medical Research Institute of the First Affiliated Hospital, University of South China, Hengyang, China
| | - Anbo Gao
- Clinical Anatomy and Reproductive Medicine Application Institute, University of South China, Hengyang, China
| | - Sha Sun
- Clinical Anatomy and Reproductive Medicine Application Institute, University of South China, Hengyang, China
| | - Xilong Zheng
- Department of Biochemistry and Molecular Biology, The Libin Cardiovascular Institute of Alberta, The University of Calgary, Health Sciences Center, Calgary, Canada
| | - Xi Chen
- Clinical Anatomy and Reproductive Medicine Application Institute, University of South China, Hengyang, China
| | - Wei Wan
- Clinical Anatomy and Reproductive Medicine Application Institute, University of South China, Hengyang, China
| | - Chaoke Tang
- Institute of Cardiovascular Research, Medical Research Center, University of South China, Hengyang, China
| | - Wei Xie
- Clinical Anatomy and Reproductive Medicine Application Institute, University of South China, Hengyang, China
| | - Suyun Li
- Clinical Anatomy and Reproductive Medicine Application Institute, University of South China, Hengyang, China
| | - Dongming Guo
- Clinical Anatomy and Reproductive Medicine Application Institute, University of South China, Hengyang, China
| | - Tianhong Peng
- Clinical Anatomy and Reproductive Medicine Application Institute, University of South China, Hengyang, China
| | - Guojun Zhao
- Department of Histology and Embryology, Guilin Medical University, Guilin, China
| | - Liyuan Zhong
- Clinical Anatomy and Reproductive Medicine Application Institute, University of South China, Hengyang, China
| |
Collapse
|
19
|
Yu XH, Zhang DW, Zheng XL, Tang CK. Cholesterol transport system: An integrated cholesterol transport model involved in atherosclerosis. Prog Lipid Res 2018; 73:65-91. [PMID: 30528667 DOI: 10.1016/j.plipres.2018.12.002] [Citation(s) in RCA: 136] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 10/30/2018] [Accepted: 12/01/2018] [Indexed: 02/07/2023]
Abstract
Atherosclerosis, the pathological basis of most cardiovascular disease (CVD), is closely associated with cholesterol accumulation in the arterial intima. Excessive cholesterol is removed by the reverse cholesterol transport (RCT) pathway, representing a major antiatherogenic mechanism. In addition to the RCT, other pathways are required for maintaining the whole-body cholesterol homeostasis. Thus, we propose a working model of integrated cholesterol transport, termed the cholesterol transport system (CTS), to describe body cholesterol metabolism. The novel model not only involves the classical view of RCT but also contains other steps, such as cholesterol absorption in the small intestine, low-density lipoprotein uptake by the liver, and transintestinal cholesterol excretion. Extensive studies have shown that dysfunctional CTS is one of the major causes for hypercholesterolemia and atherosclerosis. Currently, several drugs are available to improve the CTS efficiently. There are also several therapeutic approaches that have entered into clinical trials and shown considerable promise for decreasing the risk of CVD. In recent years, a variety of novel findings reveal the molecular mechanisms for the CTS and its role in the development of atherosclerosis, thereby providing novel insights into the understanding of whole-body cholesterol transport and metabolism. In this review, we summarize the latest advances in this area with an emphasis on the therapeutic potential of targeting the CTS in CVD patients.
Collapse
Affiliation(s)
- Xiao-Hua Yu
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Medical Research Experiment Center, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan 421001, China
| | - Da-Wei Zhang
- Department of Pediatrics and Group on the Molecular and Cell Biology of Lipids, University of Alberta, Alberta, Canada
| | - Xi-Long Zheng
- Department of Biochemistry and Molecular Biology, Libin Cardiovascular Institute of Alberta, Cumming School of Medicine, University of Calgary, Health Sciences Center, 3330 Hospital Dr NW, Calgary, Alberta T2N 4N1, Canada
| | - Chao-Ke Tang
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Medical Research Experiment Center, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan 421001, China.
| |
Collapse
|
20
|
Yu XH, He LH, Gao JH, Zhang DW, Zheng XL, Tang CK. Pregnancy-associated plasma protein-A in atherosclerosis: Molecular marker, mechanistic insight, and therapeutic target. Atherosclerosis 2018; 278:250-258. [DOI: 10.1016/j.atherosclerosis.2018.10.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 09/10/2018] [Accepted: 10/04/2018] [Indexed: 12/20/2022]
|
21
|
Ren K, Jiang T, Zhao GJ. Quercetin induces the selective uptake of HDL-cholesterol via promoting SR-BI expression and the activation of the PPARγ/LXRα pathway. Food Funct 2018; 9:624-635. [PMID: 29292466 DOI: 10.1039/c7fo01107e] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Reverse cholesterol transport (RCT) is the process to deliver cholesterol to the liver for further excretion and involves scavenger receptor class B type I (SR-BI)-mediated selective lipid uptake (SLU) from high-density lipoprotein cholesterol (HDL-C). The up-regulation of hepatic SR-BI expression accelerates HDL-C clearance in circulation and impedes the development of atherosclerosis (AS). In the present study, we explored the modulation of hepatic SR-BI expression and SR-BI-mediated SLU by quercetin, a natural flavonoid compound in the diet with a favorable role in cardiovascular disorders. We found that quercetin significantly increased the expression level of SR-BI in HepG2 cells in a concentration- and time-dependent manner. Besides, quercetin had stimulatory effects on the binding of 1,1'-dioctadecyl-3,3,3',3'-tetramethylindocarbocyanine perchlorate (Dil)-labeled HDL to hepatocytes and 125I/3H-CE-HDL association. Treatment with small interfering RNA (siRNA) or SR-BI specific inhibitor, BLT-1, inhibited quercetin-induced Dil-HDL binding and selective HDL-C uptake. Treatment with quercetin increased both proliferator-activated receptor γ (PPARγ) and liver X receptor α (LXRα) levels. Additionally, the quercetin-induced expression of SR-BI, Dil-HDL binding and the selective uptake of HDL-C were significantly attenuated by treatment with PPARγ siRNA, LXRα siRNA, and their antagonists, respectively. In C57BL/6 mice, quercetin administration potently increased SR-BI, PPARγ and LXRα levels and lipid accumulation in the liver. Altogether, our results suggest that quercetin-induced up-regulation of SR-BI and subsequent lipid uptake in hepatocytes might contribute to its beneficial effects on cholesterol homeostasis and atherogenesis.
Collapse
Affiliation(s)
- Kun Ren
- Department of Histology and Embryology, Guilin Medical University, Guilin, Guangxi 541004, China.
| | | | | |
Collapse
|
22
|
Li Y, Shen S, Ding S, Wang L. Toll-like receptor 2 downregulates the cholesterol efflux by activating the nuclear factor-κB pathway in macrophages and may be a potential therapeutic target for the prevention of atherosclerosis. Exp Ther Med 2018; 15:198-204. [PMID: 29375683 PMCID: PMC5766071 DOI: 10.3892/etm.2017.5404] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Accepted: 06/08/2017] [Indexed: 02/06/2023] Open
Abstract
Atherosclerosis is a chronic inflammatory disease, which is triggered by lipid retention. Toll-like receptor 2 (TLR2) is a novel target for therapeutic intervention in atherosclerosis. In addition, nuclear factor-κB (NF-κB) serves important roles in stress response and inflammation. The present study investigated whether TLR2 is involved in the activation of cholesterol efflux in macrophages by regulating the NF-κB pathway. The human monocytic THP-1 cell line and murine macrophage RAW264.7 cell line were treated with 50 µg/ml oxidized low-density lipoprotein (ox-LDL) for 48 h in order to obtain macrophage foam cells. The cholesterol efflux of the cell lines under exogenous TLR2 treatment was assessed by liquid scintillation counting. Furthermore, the protein and mRNA expression levels of ATP binding cassette transporter A1 (ABCA1), ABCG1 and scavenger receptor B1 (SR-B1) were examined by western blot and quantitative polymerase chain reaction assays, respectively. To detect the effect of NF-κB on cholesterol efflux, the cells were divided into three groups, including the control, 10 ng/ml lipopolysaccharides (LPS; 24 h) and 10 ng/ml LPS + 50 µM pyrrolidinedithiocarbamate (PDTC; 24 h) groups. The results indicated that ox-LDL induced foam cell formation in the THP-1 and RAW264.7 cells, while TLR2 significantly decreased the cholesterol efflux in dose- and time-dependent manners. Accordingly, TLR2 reduced ABCA1, ABCG1 and SR-B1 expression at the transcriptional and translational levels in a dose-dependent manner. In addition, application of PDTC (an NF-κB specific inhibitor) markedly suppressed the LPS-induced downregulation of cholesterol efflux. These data revealed that TLR2 may be involved in the activation of cholesterol efflux in macrophages by regulating the NF-κB signaling pathway.
Collapse
Affiliation(s)
- Yongqiang Li
- Department of Cardiology, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, P.R. China
| | - Shuxin Shen
- Department of Cardiology, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, P.R. China
| | - Shoukun Ding
- Department of Cardiology, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, P.R. China
| | - Lixia Wang
- Department of Cardiology, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, P.R. China
| |
Collapse
|
23
|
Li Y, Shen S, Ding S, Wang L. LincRNA DYN-LRB2-2 upregulates cholesterol efflux by decreasing TLR2 expression in macrophages. J Cell Biochem 2017; 119:1911-1921. [PMID: 28815701 DOI: 10.1002/jcb.26352] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 08/11/2017] [Indexed: 12/30/2022]
Abstract
This study is designed to determine whether lincRNA-DYNLRB2-2 could promote cholesterol efflux through regulating the expression of TLR2. THP-1 and RAW264.7 cells were incubated with oxLDL for 48 h to induce the formation of foam cells, and ORO staining was performed and intracellular cholesterol contents were measured by HPLC assay. qRT-PCR and Western blotting were performed to detect mRNA and protein expression levels, respectively. Lentiviral vector LV-DYNLRB2-2 and lincRNA-DYNLRB2-2 siRNA was constructed to explore its potential role. The cholesterol efflux was assessed by liquid scintillation counting. The effects of TRL2 were determined in apoE-/- mice that fed a high fat diet and were randomly divided into three groups and infected with LV-Mock, LV-Sh-TRL2, or LV-TRL2. Atherosclerosis was observed in the aortic sinus and the levels of cytokines and serum biochemical parameters were measured. Ox-LDL induced foam cell formation in the THP-1 and RAW264.7 cells. LincRNA DYN-LRB2-2 was upregulated in oxLDL-treated THP-1 and Raw264.7 cells. LincRNA-DYNLRB2-2 plays important role in regulating the cholesterol efflux, ABCA1 expression level and anti-inflammatory processes in THP-1 and RAW264.7 cells. Further study indicated that lincRNA-DYNLRB2-2 negatively regulated TRL2 expression and TRL2 overexpression reversed the effects of lincRNA-DYNLRB2-2 on cholesterol efflux and ABCA1 expression level in THP-1 and RAW264.7 cells. Besides, we found TRL2 plays important role in lipid accumulation, plaque formation and regulating serum inflammatory cytokines level in apoE-/- mice with a high fat diet. LincRNA DYN-LRB2-2 upregulates cholesterol efflux by decreasing TLR2 expression in macrophages.
Collapse
Affiliation(s)
- Yongqiang Li
- Department of Cardiology, Henan Provincial People's Hospital, Zhengzhou, Henan, People's Republic of China
| | - Shuxin Shen
- Department of Cardiology, Henan Provincial People's Hospital, Zhengzhou, Henan, People's Republic of China
| | - Shoukun Ding
- Department of Cardiology, Henan Provincial People's Hospital, Zhengzhou, Henan, People's Republic of China
| | - Lixia Wang
- Department of Cardiology, Henan Provincial People's Hospital, Zhengzhou, Henan, People's Republic of China
| |
Collapse
|
24
|
Ooi BK, Goh BH, Yap WH. Oxidative Stress in Cardiovascular Diseases: Involvement of Nrf2 Antioxidant Redox Signaling in Macrophage Foam Cells Formation. Int J Mol Sci 2017; 18:ijms18112336. [PMID: 29113088 PMCID: PMC5713305 DOI: 10.3390/ijms18112336] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 10/11/2017] [Accepted: 10/23/2017] [Indexed: 02/07/2023] Open
Abstract
Oxidative stress is an important risk factor contributing to the pathogenesis of cardiovascular diseases. Oxidative stress that results from excessive reactive oxygen species (ROS) production accounts for impaired endothelial function, a process which promotes atherosclerotic lesion or fatty streaks formation (foam cells). Nuclear factor erythroid 2-related factor 2 (Nrf2) is a transcription factor involved in cellular redox homeostasis. Upon exposure to oxidative stress, Nrf2 is dissociated from its inhibitor Keap-1 and translocated into the nucleus, where it results in the transcriptional activation of cell defense genes. Nrf2 has been demonstrated to be involved in the protection against foam cells formation by regulating the expression of antioxidant proteins (HO-1, Prxs, and GPx1), ATP-binding cassette (ABC) efflux transporters (ABCA1 and ABCG1) and scavenger receptors (scavenger receptor class B (CD36), scavenger receptor class A (SR-A) and lectin-type oxidized LDL receptor (LOX-1)). However, Nrf2 has also been reported to exhibit pro-atherogenic effects. A better understanding on the mechanism of Nrf2 in oxidative stress-induced cardiac injury, as well as the regulation of cholesterol uptake and efflux, are required before it can serve as a novel therapeutic target for cardiovascular diseases prevention and treatment.
Collapse
Affiliation(s)
- Bee Kee Ooi
- School of Biosciences, Taylor's University, Subang Jaya, Selangor Darul Ehsan 47500, Malaysia.
| | - Bey Hing Goh
- School of Pharmacy, Monash University Malaysia, Bandar Sunway, Selangor Darul Ehsan 47500, Malaysia.
| | - Wei Hsum Yap
- School of Biosciences, Taylor's University, Subang Jaya, Selangor Darul Ehsan 47500, Malaysia.
| |
Collapse
|
25
|
Oxidative Stress in Cardiovascular Diseases: Involvement of Nrf2 Antioxidant Redox Signaling in Macrophage Foam Cells Formation. Int J Mol Sci 2017. [PMID: 29113088 DOI: 10.3390/ijms18112336.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Oxidative stress is an important risk factor contributing to the pathogenesis of cardiovascular diseases. Oxidative stress that results from excessive reactive oxygen species (ROS) production accounts for impaired endothelial function, a process which promotes atherosclerotic lesion or fatty streaks formation (foam cells). Nuclear factor erythroid 2-related factor 2 (Nrf2) is a transcription factor involved in cellular redox homeostasis. Upon exposure to oxidative stress, Nrf2 is dissociated from its inhibitor Keap-1 and translocated into the nucleus, where it results in the transcriptional activation of cell defense genes. Nrf2 has been demonstrated to be involved in the protection against foam cells formation by regulating the expression of antioxidant proteins (HO-1, Prxs, and GPx1), ATP-binding cassette (ABC) efflux transporters (ABCA1 and ABCG1) and scavenger receptors (scavenger receptor class B (CD36), scavenger receptor class A (SR-A) and lectin-type oxidized LDL receptor (LOX-1)). However, Nrf2 has also been reported to exhibit pro-atherogenic effects. A better understanding on the mechanism of Nrf2 in oxidative stress-induced cardiac injury, as well as the regulation of cholesterol uptake and efflux, are required before it can serve as a novel therapeutic target for cardiovascular diseases prevention and treatment.
Collapse
|
26
|
Massafra V, Milona A, Vos HR, Burgering BMT, van Mil SWC. Quantitative liver proteomics identifies FGF19 targets that couple metabolism and proliferation. PLoS One 2017; 12:e0171185. [PMID: 28178326 PMCID: PMC5298232 DOI: 10.1371/journal.pone.0171185] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 01/18/2017] [Indexed: 12/14/2022] Open
Abstract
Fibroblast growth factor 19 (FGF19) is a gut-derived peptide hormone that is produced following activation of Farnesoid X Receptor (FXR). FGF19 is secreted and signals to the liver, where it contributes to the homeostasis of bile acid (BA), lipid and carbohydrate metabolism. FGF19 is a promising therapeutic target for the metabolic syndrome and cholestatic diseases, but enthusiasm for its use has been tempered by FGF19-mediated induction of proliferation and hepatocellular carcinoma. To inform future rational design of FGF19-variants, we have conducted temporal quantitative proteomic and gene expression analyses to identify FGF19-targets related to metabolism and proliferation. Mice were fasted for 16 hours, and injected with human FGF19 (1 mg/kg body weight) or vehicle. Liver protein extracts (containing “light” lysine) were mixed 1:1 with a spike-in protein extract from 13C6-lysine metabolically labelled mouse liver (containing “heavy” lysine) and analysed by LC-MS/MS. Our analyses provide a resource of FGF19 target proteins in the liver. 189 proteins were upregulated (≥ 1.5 folds) and 73 proteins were downregulated (≤ -1.5 folds) by FGF19. FGF19 treatment decreased the expression of proteins involved in fatty acid (FA) synthesis, i.e., Fabp5, Scd1, and Acsl3 and increased the expression of Acox1, involved in FA oxidation. As expected, FGF19 increased the expression of proteins known to drive proliferation (i.e., Tgfbi, Vcam1, Anxa2 and Hdlbp). Importantly, many of the FGF19 targets (i.e., Pdk4, Apoa4, Fas and Stat3) have a dual function in both metabolism and cell proliferation. Therefore, our findings challenge the development of FGF19-variants that fully uncouple metabolic benefit from mitogenic potential.
Collapse
Affiliation(s)
- Vittoria Massafra
- Center for Molecular Medicine, UMC Utrecht, Utrecht, The Netherlands
| | - Alexandra Milona
- Center for Molecular Medicine, UMC Utrecht, Utrecht, The Netherlands
| | - Harmjan R. Vos
- Center for Molecular Medicine, UMC Utrecht, Utrecht, The Netherlands
| | | | | |
Collapse
|
27
|
Ontsouka CE, Huang X, Aliyev E, Albrecht C. In vitro characterization and endocrine regulation of cholesterol and phospholipid transport in the mammary gland. Mol Cell Endocrinol 2017; 439:35-45. [PMID: 27760380 DOI: 10.1016/j.mce.2016.10.016] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Revised: 09/25/2016] [Accepted: 10/15/2016] [Indexed: 10/20/2022]
Abstract
Cell-based studies previously showed that the ATP-binding cassette transporter A1 (ABCA1) transfers cholesterol across mammary epithelial cells (MEC). Data for phospholipid transport are lacking, and it is unclear from which cellular source the transported cholesterol stems, whether this transport activates signaling pathways, and how lactogenic hormones regulate it. To clarify these aspects, lipid transport and expressional analyses were performed in bovine primary (bMEC) and/or immortalized (MAC-T) MEC cultures. Lipid efflux and ABCA1, ABCG1 and liver X receptorα mRNA levels were higher in MAC-T than bMEC. In MAC-T, the transported cholesterol originated mainly from the plasma membrane. ABCA1 dependent cholesterol efflux was higher than phosphatidylcholine efflux, was suppressed by probucol (ABCA1 inhibitor), AG490 (janus kinase-2 inhibitor), PD98059 (mitogen activated protein kinase kinase inhibitor) and pretreatment with β-cyclodextrin (lowering membrane cholesterol). Insulin was the only hormone significantly increasing cholesterol efflux. In conclusion, this study gives novel mechanistic and regulatory insights into the transport of cholesterol and phospholipids in MEC.
Collapse
Affiliation(s)
- Corneille Edgar Ontsouka
- Institute of Biochemistry and Molecular Medicine, Faculty of Medicine, University of Bern, Buehlstrasse 28, 3012, Bern, Switzerland; Swiss National Center of Competence in Research, NCCR TransCure, University of Bern, Bern, Switzerland
| | - Xiao Huang
- Institute of Biochemistry and Molecular Medicine, Faculty of Medicine, University of Bern, Buehlstrasse 28, 3012, Bern, Switzerland; Swiss National Center of Competence in Research, NCCR TransCure, University of Bern, Bern, Switzerland
| | - Eldar Aliyev
- Institute of Biochemistry and Molecular Medicine, Faculty of Medicine, University of Bern, Buehlstrasse 28, 3012, Bern, Switzerland
| | - Christiane Albrecht
- Institute of Biochemistry and Molecular Medicine, Faculty of Medicine, University of Bern, Buehlstrasse 28, 3012, Bern, Switzerland; Swiss National Center of Competence in Research, NCCR TransCure, University of Bern, Bern, Switzerland.
| |
Collapse
|
28
|
Ren K, Mo ZC, Liu X, Tang ZL, Jiang Y, Peng XS, Zhang QH, Shi JF, Yi GH. TGF-β Down-regulates Apolipoprotein M Expression through the TAK-1-JNK-c-Jun Pathway in HepG2 Cells. Lipids 2016; 52:109-117. [PMID: 28039587 DOI: 10.1007/s11745-016-4227-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Accepted: 12/14/2016] [Indexed: 12/22/2022]
Abstract
Apolipoprotein M (apoM) is a relatively novel apolipoprotein that plays pivotal roles in many dyslipidemia-associated diseases; however, its regulatory mechanisms are poorly understood. Many cytokines have been identified that down-regulate apoM expression in HepG2 cells, among which transforming growth factor-β (TGF-β) exerts the most potent effects. In addition, c-Jun, a member of the activated protein 1 (AP-1) family whose activity is modulated by c-Jun N-terminal kinase (JNK), decreases apoM expression at the transcriptional level by binding to the regulatory element in the proximal apoM promoter. In this study, we investigated the molecular mechanisms through which TGF-β decreases the apoM level in HepG2 cells. The results revealed that TGF-β inhibited apoM expression at both the mRNA and protein levels in a dose- and time-dependent manner and that it suppressed apoM secretion. These effects were attenuated by treatment of cells with either SP600125 (JNK inhibitor) or c-Jun siRNA. 5Z-7-oxozeaenol [(a TGF-β-activated kinase 1 (TAK-1) inhibitor)] also attenuated the TGF-β-mediated inhibition of apoM expression and suppressed the activation of JNK and c-Jun. These results have demonstrated that TGF-β suppresses apoM expression through the TAK-1-JNK-c-Jun pathway in HepG2 cells.
Collapse
Affiliation(s)
- Kun Ren
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, University of South China, 28 W Changsheng Road, Hengyang, 421001, Hunan, China
| | - Zhong-Cheng Mo
- Department of Histology and Embryology, University of South China, Hengyang, 421001, Hunan, China
| | - Xing Liu
- National Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, 100005, China
| | - Zhen-Li Tang
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, University of South China, 28 W Changsheng Road, Hengyang, 421001, Hunan, China
| | - Yue Jiang
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, University of South China, 28 W Changsheng Road, Hengyang, 421001, Hunan, China
| | - Xiao-Shan Peng
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, University of South China, 28 W Changsheng Road, Hengyang, 421001, Hunan, China
| | - Qing-Hai Zhang
- Clinical Research Institution, The First Affiliated Hospital, University of South China, Hengyang, 421001, Hunan, China
| | - Jin-Feng Shi
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, University of South China, 28 W Changsheng Road, Hengyang, 421001, Hunan, China.,Department of Histology and Embryology, University of South China, Hengyang, 421001, Hunan, China
| | - Guang-Hui Yi
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, University of South China, 28 W Changsheng Road, Hengyang, 421001, Hunan, China.
| |
Collapse
|
29
|
Kamada Y, Kida S, Hirano KI, Yamaguchi S, Suzuki A, Hashimoto C, Kimura A, Sato M, Fujii H, Sobajima T, Yamamoto A, Ebisutani Y, Takamatsu S, Shinzaki S, Yoshida Y, Yamada M, Nagasaka H, Takehara T, Miyoshi E. Hepatic aberrant glycosylation by N-acetylglucosaminyltransferase V accelerates HDL assembly. Am J Physiol Gastrointest Liver Physiol 2016; 311:G859-G868. [PMID: 27659420 DOI: 10.1152/ajpgi.00231.2016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Accepted: 09/14/2016] [Indexed: 01/31/2023]
Abstract
Glycosylation is involved in various pathophysiological conditions. N-Acetylglucosaminyltransferase V (GnT-V), catalyzing β1-6 branching in asparagine-linked oligosaccharides, is one of the most important glycosyltransferases involved in cancer and the immune system. Recent findings indicate that aberrant N-glycan structure can modify lipid metabolism. In this study, we investigated the effects of aberrant glycosylation by GnT-V on high-density lipoprotein cholesterol (HDL) assembly. We used GnT-V transgenic (Tg) mice and GnT-V Hep3B cell (human hepatoma cell line) transfectants. The study also included 96 patients who underwent medical health check-ups. Total serum cholesterol levels, particularly HDL-cholesterol (HDL-C) levels, were significantly increased in Tg vs. wild-type (WT) mice. Hepatic expression of apolipoprotein AI (ApoAI) and ATP-binding cassette subfamily A member 1 (ABCA1), two important factors in HDL assembly, were higher in Tg mice compared with WT mice. ApoAI and ABCA1 were also significantly elevated in GnT-V transfectants compared with mock-transfected cells. Moreover, ApoAI protein in the cultured media of GnT-V transfectants was significantly increased. Finally, we found a strong correlation between serum GnT-V activity and HDL-C concentration in human subjects. Multivariate logistic analyses demonstrated that GnT-V activity was an independent and significant determinant for serum HDL-C levels even adjusted with age and gender differences. Further analyses represented that serum GnT-V activity had strong correlation especially with the large-size HDL particle concentration. These findings indicate that enhanced hepatic GnT-V activity accelerated HDL assembly and could be a novel mechanism for HDL synthesis.
Collapse
Affiliation(s)
- Yoshihiro Kamada
- Department of Molecular Biochemistry and Clinical Investigation, Osaka University Graduate School of Medicine, Suita, Osaka, Japan.,Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Sachiho Kida
- Department of Molecular Biochemistry and Clinical Investigation, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Ken-Ichi Hirano
- Laboratory of Cardiovascular Disease, Novel, Non-invasive and Nutritional Therapeutics, Osaka University, Suita, Osaka, Japan
| | - Satoshi Yamaguchi
- Laboratory of Cardiovascular Disease, Novel, Non-invasive and Nutritional Therapeutics, Osaka University, Suita, Osaka, Japan
| | - Akira Suzuki
- Laboratory of Cardiovascular Disease, Novel, Non-invasive and Nutritional Therapeutics, Osaka University, Suita, Osaka, Japan
| | - Chikako Hashimoto
- Laboratory of Cardiovascular Disease, Novel, Non-invasive and Nutritional Therapeutics, Osaka University, Suita, Osaka, Japan
| | - Akihiro Kimura
- Department of Molecular Biochemistry and Clinical Investigation, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Motoya Sato
- Department of Molecular Biochemistry and Clinical Investigation, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Hironobu Fujii
- Department of Molecular Biochemistry and Clinical Investigation, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Tomoaki Sobajima
- Department of Molecular Biochemistry and Clinical Investigation, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Akiko Yamamoto
- Department of Molecular Biochemistry and Clinical Investigation, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Yusuke Ebisutani
- Department of Molecular Biochemistry and Clinical Investigation, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Shinji Takamatsu
- Department of Molecular Biochemistry and Clinical Investigation, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Shinichiro Shinzaki
- Department of Molecular Biochemistry and Clinical Investigation, Osaka University Graduate School of Medicine, Suita, Osaka, Japan.,Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Yuichi Yoshida
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | | | - Hironori Nagasaka
- Department of Pediatrics, Takarazuka City Hospital, Takarazuka, Hyogo, Japan
| | - Tetsuo Takehara
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Eiji Miyoshi
- Department of Molecular Biochemistry and Clinical Investigation, Osaka University Graduate School of Medicine, Suita, Osaka, Japan;
| |
Collapse
|
30
|
Kamada Y, Ebisutani Y, Kida S, Mizutani K, Akita M, Yamamoto A, Fujii H, Sobajima T, Terao N, Takamatsu S, Yoshida Y, Takehara T, Miyoshi E. Ectopic expression of N-acetylglucosaminyltransferase V accelerates hepatic triglyceride synthesis. Hepatol Res 2016; 46:E118-29. [PMID: 26041473 DOI: 10.1111/hepr.12541] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Revised: 05/24/2015] [Accepted: 06/02/2015] [Indexed: 01/28/2023]
Abstract
AIM Glycosylation changes induce various types of biological phenomena in human diseases. N-Acetylglucosaminyltransferase V (GnT-V) is one of the most important glycosyltransferases involved in cancer biology. Recently, many researchers have challenged studies of lipid metabolism in cancer. To elucidate the relationships between cancer and lipid metabolism more precisely, we investigated the effects of GnT-V on lipid metabolism. In this study, we investigated the effects of aberrant glycosylation by GnT-V on hepatic triglyceride production. METHODS We compared lipid metabolism in GnT-V transgenic (Tg) mice with that of wild-type (WT) mice fed with normal chow or a choline-deficient amino acid-defined (CDAA) diet in vivo. HepG2 cells and GnT-V transfectants of Hep3B cells were used in an in vitro study. RESULTS Serum triglyceride levels and hepatic very low-density lipoprotein (VLDL) secretion in Tg mice were significantly elevated compared with that of WT mice. Hepatic lipogenic genes (Lxrα, Srebp1, Fas and Acc) and VLDL secretion-related gene (Mttp1) were significantly higher in Tg mice. Expression of these genes was also significantly higher in GnT-V transfectants than in mock cells. Knockdown of GnT-V decreased, while both epidermal growth factor and transforming growth factor-β1 stimulation increased LXRα gene expression in HepG2 cells. Finally, we found that the blockade of VLDL secretion by CDAA diet induced massive hepatic steatosis in Tg mice. CONCLUSION Our study demonstrates that enhancement of hepatic GnT-V activity accelerates triglyceride synthesis and VLDL secretion. Glycosylation modification by GnT-V regulation could be a novel target for a therapeutic approach to lipid metabolism.
Collapse
Affiliation(s)
- Yoshihiro Kamada
- Departments of Molecular Biochemistry and Clinical Investigation, Suita, Japan.,Gastroenterology and Hepatology, Osaka University, Graduate School of Medicine, Suita, Japan
| | - Yusuke Ebisutani
- Departments of Molecular Biochemistry and Clinical Investigation, Suita, Japan
| | - Sachiho Kida
- Departments of Molecular Biochemistry and Clinical Investigation, Suita, Japan
| | - Kayo Mizutani
- Departments of Molecular Biochemistry and Clinical Investigation, Suita, Japan
| | - Maaya Akita
- Departments of Molecular Biochemistry and Clinical Investigation, Suita, Japan
| | - Akiko Yamamoto
- Departments of Molecular Biochemistry and Clinical Investigation, Suita, Japan
| | - Hironobu Fujii
- Departments of Molecular Biochemistry and Clinical Investigation, Suita, Japan
| | - Tomoaki Sobajima
- Departments of Molecular Biochemistry and Clinical Investigation, Suita, Japan
| | - Naoko Terao
- Departments of Molecular Biochemistry and Clinical Investigation, Suita, Japan
| | - Shinji Takamatsu
- Departments of Molecular Biochemistry and Clinical Investigation, Suita, Japan
| | - Yuichi Yoshida
- Gastroenterology and Hepatology, Osaka University, Graduate School of Medicine, Suita, Japan
| | - Tetsuo Takehara
- Gastroenterology and Hepatology, Osaka University, Graduate School of Medicine, Suita, Japan
| | - Eiji Miyoshi
- Departments of Molecular Biochemistry and Clinical Investigation, Suita, Japan
| |
Collapse
|
31
|
Fierro-Macías AE, Floriano-Sánchez E, Mena-Burciaga VM, Gutiérrez-Leonard H, Lara-Padilla E, Abarca-Rojano E, Fierro-Almanzán AE. [Association between IGF system and PAPP-A in coronary atherosclerosis]. ARCHIVOS DE CARDIOLOGIA DE MEXICO 2016; 86:148-56. [PMID: 26906607 DOI: 10.1016/j.acmx.2015.12.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Revised: 12/23/2015] [Accepted: 12/24/2015] [Indexed: 11/28/2022] Open
Abstract
Atherosclerosis is a condition that involves multiple pathophysiological mechanisms and whose knowledge has not been fully elucidated. Often, scientific advances on the atherogenic pathophysiology generate that molecules not previously considered in the scene of this disease, were attributed actions on the onset or progression of it. A representative example is the study of a new mechanism involved in the atherogenic process, consisting of the association between the insulin-like growth factor (IGF) system and pregnancy-associated plasma protein-A (PAPP-A). Insulin-like growth factor system is a family of peptides that include 3 peptide hormones, 4 transmembrane receptors and 6 binding proteins. Insulin-like growth factor-1 (IGF-1) is the main ligand of the IGF system involved in coronary atherosclerosis. IGF-1 exerts its effects via activation of the IGF-1R receptor on vascular smooth muscle cells or macrophages. In vascular smooth muscle cells promotes migration and prevents apoptosis which increases plaque stability while in macrophages reduces reverse cholesterol transport leading to the formation of foam cells. Regulation of IGF-1 endothelial bioavailability is carried out by IGFBP proteases, mainly by PAPP-A. In this review, we address the mechanisms between IGF system and PAPP-A in atherosclerosis with emphasis on molecular effects on vascular smooth muscle cells and macrophages.
Collapse
Affiliation(s)
- Alfonso Eduardo Fierro-Macías
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, México, DF, México.
| | - Esaú Floriano-Sánchez
- Laboratorio Multidisciplinario de Investigación, Escuela Militar de Graduados de Sanidad, Secretaría de la Defensa Nacional (SEDENA), México, DF, México
| | - Victoria Michelle Mena-Burciaga
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, México, DF, México
| | - Hugo Gutiérrez-Leonard
- Departamento de Hemodinamia, Hospital Central Militar, Secretaría de la Defensa Nacional (SEDENA), México, DF, México
| | - Eleazar Lara-Padilla
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, México, DF, México
| | - Edgar Abarca-Rojano
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, México, DF, México
| | - Alfonso Edmundo Fierro-Almanzán
- Departamento de Cirugía, Hospital General Regional N.(o) 66, Instituto Mexicano del Seguro Social (IMSS), Ciudad Juárez, Chihuahua, México
| |
Collapse
|
32
|
El Asmar Z, Terrand J, Jenty M, Host L, Mlih M, Zerr A, Justiniano H, Matz RL, Boudier C, Scholler E, Garnier JM, Bertaccini D, Thiersé D, Schaeffer C, Van Dorsselaer A, Herz J, Bruban V, Boucher P. Convergent Signaling Pathways Controlled by LRP1 (Receptor-related Protein 1) Cytoplasmic and Extracellular Domains Limit Cellular Cholesterol Accumulation. J Biol Chem 2016; 291:5116-27. [PMID: 26792864 DOI: 10.1074/jbc.m116.714485] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Indexed: 11/06/2022] Open
Abstract
The low density lipoprotein receptor-related protein 1 (LRP1) is a ubiquitously expressed cell surface receptor that protects from intracellular cholesterol accumulation. However, the underlying mechanisms are unknown. Here we show that the extracellular (α) chain of LRP1 mediates TGFβ-induced enhancement of Wnt5a, which limits intracellular cholesterol accumulation by inhibiting cholesterol biosynthesis and by promoting cholesterol export. Moreover, we demonstrate that the cytoplasmic (β) chain of LRP1 suffices to limit cholesterol accumulation in LRP1(-/-) cells. Through binding of Erk2 to the second of its carboxyl-terminal NPXY motifs, LRP1 β-chain positively regulates the expression of ATP binding cassette transporter A1 (ABCA1) and of neutral cholesterol ester hydrolase (NCEH1). These results highlight the unexpected functions of LRP1 and the canonical Wnt5a pathway and new therapeutic potential in cholesterol-associated disorders including cardiovascular diseases.
Collapse
Affiliation(s)
- Zeina El Asmar
- From the CNRS, UMR 7213, University of Strasbourg, 67401 Illkirch, France
| | - Jérome Terrand
- From the CNRS, UMR 7213, University of Strasbourg, 67401 Illkirch, France
| | - Marion Jenty
- From the CNRS, UMR 7213, University of Strasbourg, 67401 Illkirch, France
| | - Lionel Host
- From the CNRS, UMR 7213, University of Strasbourg, 67401 Illkirch, France
| | - Mohamed Mlih
- From the CNRS, UMR 7213, University of Strasbourg, 67401 Illkirch, France
| | - Aurélie Zerr
- From the CNRS, UMR 7213, University of Strasbourg, 67401 Illkirch, France
| | - Hélène Justiniano
- From the CNRS, UMR 7213, University of Strasbourg, 67401 Illkirch, France
| | - Rachel L Matz
- From the CNRS, UMR 7213, University of Strasbourg, 67401 Illkirch, France
| | - Christian Boudier
- From the CNRS, UMR 7213, University of Strasbourg, 67401 Illkirch, France
| | - Estelle Scholler
- From the CNRS, UMR 7213, University of Strasbourg, 67401 Illkirch, France
| | - Jean-Marie Garnier
- IGBMC (Institut de Génétique et de Biologie Moléculaire et Cellulaire), INSERM 964/CNRS UMR 7104, University of Strasbourg, 67401 Illkirch, France
| | - Diego Bertaccini
- CNRS, UMR 7178, University of Strasbourg, 67087 Strasbourg, France, and
| | - Danièle Thiersé
- CNRS, UMR 7178, University of Strasbourg, 67087 Strasbourg, France, and
| | | | | | - Joachim Herz
- Department of Molecular Genetics and Center for Translational Neurodegeneration Research, UT Southwestern Medical Center, Dallas, Texas 75390
| | - Véronique Bruban
- From the CNRS, UMR 7213, University of Strasbourg, 67401 Illkirch, France,
| | - Philippe Boucher
- From the CNRS, UMR 7213, University of Strasbourg, 67401 Illkirch, France,
| |
Collapse
|
33
|
Yao F, Lv YC, Zhang M, Xie W, Tan YL, Gong D, Cheng HP, Liu D, Li L, Liu XY, Zheng XL, Tang CK. Apelin-13 impedes foam cell formation by activating Class III PI3K/Beclin-1-mediated autophagic pathway. Biochem Biophys Res Commun 2015; 466:637-43. [DOI: 10.1016/j.bbrc.2015.09.045] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Accepted: 09/08/2015] [Indexed: 12/24/2022]
|
34
|
Li E, Wang T, Wang F, Wang T, Sun LQ, Li L, Niu SH, Zhang JY. FGF21 protects against ox-LDL induced apoptosis through suppressing CHOP expression in THP1 macrophage derived foam cells. BMC Cardiovasc Disord 2015. [PMID: 26223891 PMCID: PMC4518604 DOI: 10.1186/s12872-015-0077-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Background FGF21,as a member of the fibroblast growth factor superfamily, is an important endogenous regulator to systemic glucose and lipid metabolism. Elevated serum FGF21 levels have been reported in subjects with coronary heart disease and carotid artery plaques. The formation and apoptosis of foam cell, induced by ox-LDL and oxysterols, are key steps in the development of atherosclerosis. Methods In this study, THP1 derived macrophages were induced into foam cells by ox-LDL or sterols. The formation and apoptosis of foam cells treated with or without FGF21 were analyzed. Results We demonstrated that the accumulation of cholesterol was decreased after FGF21 treatment in THP1 macrophage derived foam cells. Consistently, the apoptosis of macrophage was alleviated dramatically with FGF21 treatment. ERK1/2 knockdown didn’t abrogate the effect of FGF21 on THP1 macrophage derived foam cells. However, FGF21 suppressed the induced expression of CHOP and DR5 in THP1 macrophage derived foam cells. Conclusion FGF21 protects against the formation and apoptosis of THP1 macrophages derived foam cells through suppressing the expression of CHOP.
Collapse
Affiliation(s)
- En Li
- Department of Cardiovascular internal medicine, The first Affiliated Hospital of Zhengzhou University, Zhengzhou, P. R. China. .,Department of Cardiovascular internal medicine, The second Affiliated Hospital of Zhengzhou University, Zhengzhou, P. R. China.
| | - Ting Wang
- Department of Gerontology, Shaanxi People's Hospital, Xi'an, P. R. China.
| | - Feng Wang
- CAS Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, P. R. China.
| | - Tao Wang
- Department of Gerontology, The second Affiliated Hospital of Zhengzhou University, Zhengzhou, P. R. China.
| | - Li-qiang Sun
- Department of Cardiovascular internal medicine, The second Affiliated Hospital of Zhengzhou University, Zhengzhou, P. R. China.
| | - Li Li
- Department of Cardiovascular internal medicine, The second Affiliated Hospital of Zhengzhou University, Zhengzhou, P. R. China.
| | - Shao-hui Niu
- Department of Cardiovascular internal medicine, The second Affiliated Hospital of Zhengzhou University, Zhengzhou, P. R. China.
| | - Jin-ying Zhang
- Department of Cardiovascular internal medicine, The first Affiliated Hospital of Zhengzhou University, Zhengzhou, P. R. China.
| |
Collapse
|
35
|
Abstract
Formation of foam cell macrophages, which sequester extracellular modified lipids, is a key event in atherosclerosis. How lipid loading affects macrophage phenotype is controversial, with evidence suggesting either pro- or anti-inflammatory consequences. To investigate this further, we compared the transcriptomes of foamy and non-foamy macrophages that accumulate in the subcutaneous granulomas of fed-fat ApoE null mice and normal chow fed wild-type mice in vivo. Consistent with previous studies, LXR/RXR pathway genes were significantly over-represented among the genes up-regulated in foam cell macrophages. Unexpectedly, the hepatic fibrosis pathway, associated with platelet derived growth factor and transforming growth factor-β action, was also over-represented. Several collagen polypeptides and proteoglycan core proteins as well as connective tissue growth factor and fibrosis-related FOS and JUN transcription factors were up-regulated in foam cell macrophages. Increased expression of several of these genes was confirmed at the protein level in foam cell macrophages from subcutaneous granulomas and in atherosclerotic plaques. Moreover, phosphorylation and nuclear translocation of SMAD2, which is downstream of several transforming growth factor-β family members, was also detected in foam cell macrophages. We conclude that foam cell formation in vivo leads to a pro-fibrotic macrophage phenotype, which could contribute to plaque stability, especially in early lesions that have few vascular smooth muscle cells.
Collapse
Affiliation(s)
- Anita C. Thomas
- Bristol Heart Institute, University of Bristol, Bristol, United Kingdom
- * E-mail:
| | - Wouter J. Eijgelaar
- Cardiovascular Research Institute Maastricht (CARIM), Maastricht, The Netherlands
| | - Mat J. A. P. Daemen
- Cardiovascular Research Institute Maastricht (CARIM), Maastricht, The Netherlands
- Academisch Medisch Centrum (AMC), Amsterdam, The Netherlands
| | - Andrew C. Newby
- Bristol Heart Institute, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
36
|
Abstract
The 9p21.3 locus was the first to yield to genome-wide association studies (GWAS) seeking common genetic variants predisposing to increased risk of coronary artery atherosclerotic disease (CAD). The 59 single nucleotide polymorphisms that show highest association with CAD are clustered in a region 100,000 to 150,000 base pairs 5' to the cyclin-dependent kinase inhibitors CDKN2B (coding for p15(ink4b)) and CDKN2A (coding for p16(ink4a) and p14(ARF)). This region also covers the 3' end of a long noncoding RNA transcribed antisense to CDKN2B (CDKN2BAS, aka ANRIL for antisense noncoding RNA at the ink4 locus) whose expression has been linked to chromatin remodeling at the locus. Despite intensive investigation over the past 7 years, the functional significance of the 9p21.3 locus remains elusive. Other variants at this locus have been associated with glaucoma, glioma, and type 2 diabetes mellitus, diseases that implicate tissue-resident macrophages. Here, we review the evidence that genetic variants at 9p21.3 disrupt tissue-specific enhancers and propose new insights to guide future studies.
Collapse
|
37
|
Chou JL, Huang RL, Shay J, Chen LY, Lin SJ, Yan PS, Chao WT, Lai YH, Lai YL, Chao TK, Lee CI, Tai CK, Wu SF, Nephew KP, Huang THM, Lai HC, Chan MWY. Hypermethylation of the TGF-β target, ABCA1 is associated with poor prognosis in ovarian cancer patients. Clin Epigenetics 2015; 7:1. [PMID: 25628764 PMCID: PMC4307187 DOI: 10.1186/s13148-014-0036-2] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Accepted: 12/15/2014] [Indexed: 12/28/2022] Open
Abstract
Background The dysregulation of transforming growth factor-β (TGF-β) signaling plays a crucial role in ovarian carcinogenesis and in maintaining cancer stem cell properties. Classified as a member of the ATP-binding cassette (ABC) family, ABCA1 was previously identified by methylated DNA immunoprecipitation microarray (mDIP-Chip) to be methylated in ovarian cancer cell lines, A2780 and CP70. By microarray, it was also found to be upregulated in immortalized ovarian surface epithelial (IOSE) cells following TGF-β treatment. Thus, we hypothesized that ABCA1 may be involved in ovarian cancer and its initiation. Results We first compared the expression level of ABCA1 in IOSE cells and a panel of ovarian cancer cell lines and found that ABCA1 was expressed in HeyC2, SKOV3, MCP3, and MCP2 ovarian cancer cell lines but downregulated in A2780 and CP70 ovarian cancer cell lines. The reduced expression of ABCA1 in A2780 and CP70 cells was associated with promoter hypermethylation, as demonstrated by bisulfite pyro-sequencing. We also found that knockdown of ABCA1 increased the cholesterol level and promoted cell growth in vitro and in vivo. Further analysis of ABCA1 methylation in 76 ovarian cancer patient samples demonstrated that patients with higher ABCA1 methylation are associated with high stage (P = 0.0131) and grade (P = 0.0137). Kaplan-Meier analysis also found that patients with higher levels of methylation of ABCA1 have shorter overall survival (P = 0.019). Furthermore, tissue microarray using 55 ovarian cancer patient samples revealed that patients with a lower level of ABCA1 expression are associated with shorter progress-free survival (P = 0.038). Conclusions ABCA1 may be a tumor suppressor and is hypermethylated in a subset of ovarian cancer patients. Hypermethylation of ABCA1 is associated with poor prognosis in these patients. Electronic supplementary material The online version of this article (doi:10.1186/s13148-014-0036-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jian-Liang Chou
- Department of Life Science, National Chung Cheng University, 168 University Road, Min-Hsiung, Chia-Yi 621, Taiwan ; Institute of Molecular Biology, National Chung Cheng University, Min-Hsiung, Chia-Yi, Taiwan ; Division of Gastroenterology, Chang Gung Memorial Hospital, Chia-Yi, Taiwan
| | - Rui-Lan Huang
- Department of Obstetrics and Gynecology, Shuang Ho Hospital, Taipei Medical University, No 291, Zhongzheng Rd., Zhonghe District, New Taipei City, 23561 Taiwan
| | - Jacqueline Shay
- Department of Life Science, National Chung Cheng University, 168 University Road, Min-Hsiung, Chia-Yi 621, Taiwan ; Institute of Molecular Biology, National Chung Cheng University, Min-Hsiung, Chia-Yi, Taiwan
| | - Lin-Yu Chen
- Department of Life Science, National Chung Cheng University, 168 University Road, Min-Hsiung, Chia-Yi 621, Taiwan
| | - Sheng-Jie Lin
- Department of Life Science, National Chung Cheng University, 168 University Road, Min-Hsiung, Chia-Yi 621, Taiwan ; Institute of Molecular Biology, National Chung Cheng University, Min-Hsiung, Chia-Yi, Taiwan
| | - Pearlly S Yan
- Division of Hematology, Department of Internal Medicine, Comprehensive Cancer Center, The Ohio State University, Columbus, OH USA
| | - Wei-Ting Chao
- Department of Life Science, Tunghai University, Taichung, Taiwan
| | - Yi-Hui Lai
- Department of Life Science, National Chung Cheng University, 168 University Road, Min-Hsiung, Chia-Yi 621, Taiwan ; Institute of Molecular Biology, National Chung Cheng University, Min-Hsiung, Chia-Yi, Taiwan
| | - Yen-Ling Lai
- Department of Life Science, National Chung Cheng University, 168 University Road, Min-Hsiung, Chia-Yi 621, Taiwan ; Institute of Molecular Biology, National Chung Cheng University, Min-Hsiung, Chia-Yi, Taiwan
| | - Tai-Kuang Chao
- Department of Pathology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Cheng-I Lee
- Department of Life Science, National Chung Cheng University, 168 University Road, Min-Hsiung, Chia-Yi 621, Taiwan ; Institute of Molecular Biology, National Chung Cheng University, Min-Hsiung, Chia-Yi, Taiwan
| | - Chien-Kuo Tai
- Department of Life Science, National Chung Cheng University, 168 University Road, Min-Hsiung, Chia-Yi 621, Taiwan ; Institute of Molecular Biology, National Chung Cheng University, Min-Hsiung, Chia-Yi, Taiwan
| | - Shu-Fen Wu
- Department of Life Science, National Chung Cheng University, 168 University Road, Min-Hsiung, Chia-Yi 621, Taiwan ; Institute of Molecular Biology, National Chung Cheng University, Min-Hsiung, Chia-Yi, Taiwan
| | - Kenneth P Nephew
- Medical Sciences, Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Bloomington, IN USA
| | - Tim H-M Huang
- Department of Molecular Medicine, Institute of Biotechnology, University of Texas Health Science Center, San Antonio, TX USA
| | - Hung-Cheng Lai
- Department of Obstetrics and Gynecology, Shuang Ho Hospital, Taipei Medical University, No 291, Zhongzheng Rd., Zhonghe District, New Taipei City, 23561 Taiwan ; Department of Obstetrics and Gynecology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan ; Department of Clinical Pharmacology, Xiangya Hospital; Institute of Clinical Pharmacology, Central South University and Hunan Key Laboratory of Pharmacogenetics, Changsha, China
| | - Michael W Y Chan
- Department of Life Science, National Chung Cheng University, 168 University Road, Min-Hsiung, Chia-Yi 621, Taiwan ; Institute of Molecular Biology, National Chung Cheng University, Min-Hsiung, Chia-Yi, Taiwan
| |
Collapse
|
38
|
Kardassis D, Gafencu A, Zannis VI, Davalos A. Regulation of HDL genes: transcriptional, posttranscriptional, and posttranslational. Handb Exp Pharmacol 2015; 224:113-179. [PMID: 25522987 DOI: 10.1007/978-3-319-09665-0_3] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
HDL regulation is exerted at multiple levels including regulation at the level of transcription initiation by transcription factors and signal transduction cascades; regulation at the posttranscriptional level by microRNAs and other noncoding RNAs which bind to the coding or noncoding regions of HDL genes regulating mRNA stability and translation; as well as regulation at the posttranslational level by protein modifications, intracellular trafficking, and degradation. The above mechanisms have drastic effects on several HDL-mediated processes including HDL biogenesis, remodeling, cholesterol efflux and uptake, as well as atheroprotective functions on the cells of the arterial wall. The emphasis is on mechanisms that operate in physiologically relevant tissues such as the liver (which accounts for 80% of the total HDL-C levels in the plasma), the macrophages, the adrenals, and the endothelium. Transcription factors that have a significant impact on HDL regulation such as hormone nuclear receptors and hepatocyte nuclear factors are extensively discussed both in terms of gene promoter recognition and regulation but also in terms of their impact on plasma HDL levels as was revealed by knockout studies. Understanding the different modes of regulation of this complex lipoprotein may provide useful insights for the development of novel HDL-raising therapies that could be used to fight against atherosclerosis which is the underlying cause of coronary heart disease.
Collapse
Affiliation(s)
- Dimitris Kardassis
- Department of Biochemistry, University of Crete Medical School and Institute of Molecular Biology and Biotechnology, Foundation of Research and Technology of Hellas, Heraklion, Crete, 71110, Greece,
| | | | | | | |
Collapse
|
39
|
SHANG WEI, YU XUEJING, WANG HONGLIAN, CHEN TIELIN, FANG YING, YANG XIANGGUI, ZHOU PUHUI, NIE FANG, ZHOU QIN, ZHOU JIANZHONG. Fibroblast growth factor 21 enhances cholesterol efflux in THP-1 macrophage-derived foam cells. Mol Med Rep 2014; 11:503-8. [DOI: 10.3892/mmr.2014.2731] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2013] [Accepted: 04/25/2014] [Indexed: 11/06/2022] Open
|
40
|
Wang Y, Wu JF, Tang YY, Zhang M, Li Y, Chen K, Zeng MY, Yao F, Xie W, Zheng XL, Zeng GF, Tang CK. Urotensin II increases foam cell formation by repressing ABCA1 expression through the ERK/NF-κB pathway in THP-1 macrophages. Biochem Biophys Res Commun 2014; 452:998-1003. [DOI: 10.1016/j.bbrc.2014.09.030] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Accepted: 09/08/2014] [Indexed: 12/21/2022]
|
41
|
Lin XL, He XL, Zeng JF, Zhang H, Zhao Y, Tan JK, Wang Z. FGF21 Increases Cholesterol Efflux by Upregulating ABCA1 Through the ERK1/2–PPARγ–LXRα Pathway in THP1 Macrophage-Derived Foam Cells. DNA Cell Biol 2014; 33:514-21. [PMID: 24735204 DOI: 10.1089/dna.2013.2290] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Affiliation(s)
- Xiao-Long Lin
- Key Laboratory for Arteriosclerology of Hunan Province, Institute of Cardiovascular Disease, University of South China, Hengyang, China
- Pathology Department, The Third People's Hospital of Huizhou, Guangdong Huizhou, China
| | - Xing-Lan He
- Key Laboratory for Arteriosclerology of Hunan Province, Institute of Cardiovascular Disease, University of South China, Hengyang, China
| | - Jun-Fa Zeng
- The Second Affiliated Hospital of the University of South China, Hengyang, China
| | - Hai Zhang
- Key Laboratory for Arteriosclerology of Hunan Province, Institute of Cardiovascular Disease, University of South China, Hengyang, China
| | - Yue Zhao
- The First Affiliated Hospital of the University of South China, Hengyang, China
| | - Jian-Kai Tan
- The First Affiliated Hospital of the University of South China, Hengyang, China
| | - Zuo Wang
- Key Laboratory for Arteriosclerology of Hunan Province, Institute of Cardiovascular Disease, University of South China, Hengyang, China
| |
Collapse
|
42
|
MicroRNA-19b promotes macrophage cholesterol accumulation and aortic atherosclerosis by targeting ATP-binding cassette transporter A1. Atherosclerosis 2014; 236:215-26. [PMID: 25084135 DOI: 10.1016/j.atherosclerosis.2014.07.005] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2014] [Revised: 06/19/2014] [Accepted: 07/07/2014] [Indexed: 12/20/2022]
Abstract
RATIONALE Macrophage accumulation of cholesterol leads to foam cell formation which is a major pathological event of atherosclerosis. Recent studies have shown that microRNA (miR)-19b might play an important role in cholesterol metabolism and atherosclerotic diseases. Here, we have identified miR-19b binding to the 3'UTR of ATP-binding cassette transporter A1 (ABCA1) transporters, and further determined the potential roles of this novel interaction in atherogenesis. OBJECTIVE To investigate the molecular mechanisms involved in a miR-19b promotion of macrophage cholesterol accumulation and the development of aortic atherosclerosis. METHODS AND RESULTS We performed bioinformatics analysis using online websites, and found that miR-19b was highly conserved during evolution and directly bound to ABCA1 mRNA with very low binding free energy. Luciferase reporter assay confirmed that miR-19b bound to 3110-3116 sites within ABCA1 3'UTR. MiR-19b directly regulated the expression levels of endogenous ABCA1 in foam cells derived from human THP-1 macrophages and mouse peritoneal macrophages (MPMs) as determined by qRT-PCR and western blot. Cholesterol transport assays revealed that miR-19b dramatically suppressed apolipoprotein AI-mediated ABCA1-dependent cholesterol efflux, resulting in the increased levels of total cholesterol (TC), free cholesterol (FC) and cholesterol ester (CE) as revealed by HPLC. The excretion of (3)H-cholesterol originating from cholesterol-laden MPMs into feces was decreased in mice overexpressing miR-19b. Finally, we evaluated the proatherosclerotic role of miR-19b in apolipoprotein E deficient (apoE(-/-)) mice. Treatment with miR-19b precursor reduced plasma high-density lipoprotein (HDL) levels, but increased plasma low-density lipoprotein (LDL) levels. Consistently, miR-19b precursor treatment increased aortic plaque size and lipid content, but reduced collagen content and ABCA1 expression. In contrast, treatment with the inhibitory miR-19b antisense oligonucleotides (ASO) prevented or reversed these effects. CONCLUSION MiR-19b promotes macrophage cholesterol accumulation, foam cell formation and aortic atherosclerotic development by targeting ABCA1.
Collapse
|
43
|
Yin K, Agrawal DK. High-density lipoprotein: a novel target for antirestenosis therapy. Clin Transl Sci 2014; 7:500-11. [PMID: 25043950 DOI: 10.1111/cts.12186] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Restenosis is an integral pathological process central to the recurrent vessel narrowing after interventional procedures. Although the mechanisms for restenosis are diverse in different pathological conditions, endothelial dysfunction, inflammation, vascular smooth muscle cell (SMC) proliferation, and myofibroblasts transition have been thought to play crucial role in the development of restenosis. Indeed, there is an inverse relationship between high-density lipoprotein (HDL) levels and risk for coronary heart disease (CHD). However, relatively studies on the direct assessment of HDL effect on restenosis are limited. In addition to involvement in the cholesterol reverse transport, many vascular protective effects of HDL, including protection of endothelium, antiinflammation, antithrombus actions, inhibition of SMC proliferation, and regulation by adventitial effects may contribute to the inhibition of restenosis, though the exact relationships between HDL and restenosis remain to be elucidated. This review summarizes the vascular protective effects of HDL, emphasizing the potential role of HDL in intimal hyperplasia and vascular remodeling, which may provide novel prophylactic and therapeutic strategies for antirestenosis.
Collapse
Affiliation(s)
- Kai Yin
- Center for Clinical & Translational Science, Creighton University School of Medicine, Omaha, Nebraska, USA
| | | |
Collapse
|
44
|
Abstract
Low plasma levels of HDL-cholesterol (HDL-C) represent a strong and independent risk factor for cardiovascular disease. HDL particles display a wide spectrum of atheroprotective activities, which include effluxing cellular cholesterol, diminishing cellular death, decreasing vascular constriction, reducing inflammatory response, protecting from pathological oxidation, combating bacterial infection, lessening platelet activation, regulating gene expression by virtue of microRNAs, and improving glucose metabolism. It remains presently indeterminate as to whether some biological activities of HDL are more relevant for the protection of the endothelium from atherogenesis when compared with others. The multitude of such activities raises the question of a proper assay to assess HDL functionality ex vivo. Together with clear understanding of molecular mechanisms underlying atheroprotective properties of HDL, such assay will provide a basis to resolve the ultimate question of the HDL field to allow the development of efficient HDL-targeting therapies.
Collapse
Affiliation(s)
- Anatol Kontush
- National Institute for Health and Medical Research (INSERM), UMR-ICAN 1166, University of Pierre and Marie Curie - Paris 6, Pitié - Salpétrière University Hospital, ICAN, 75651 Paris Cedex 13, France
| |
Collapse
|
45
|
Wu JF, Wang Y, Zhang M, Tang YY, Wang B, He PP, Lv YC, Ouyang XP, Yao F, Tan YL, Tang SL, Tang DP, Cayabyab FS, Zheng XL, Zhang DW, Zeng GF, Tang CK. Growth differentiation factor-15 induces expression of ATP-binding cassette transporter A1 through PI3-K/PKCζ/SP1 pathway in THP-1 macrophages. Biochem Biophys Res Commun 2014; 444:325-31. [DOI: 10.1016/j.bbrc.2014.01.048] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2013] [Accepted: 01/14/2014] [Indexed: 11/26/2022]
|
46
|
Cerda A, Issa MH, Genvigir FDV, Rohde CB, Cavalli SA, Bertolami MC, Faludi AA, Hirata MH, Hirata RDC. Atorvastatin and hormone therapy influence expression of ABCA1, APOA1 and SCARB1 in mononuclear cells from hypercholesterolemic postmenopausal women. J Steroid Biochem Mol Biol 2013; 138:403-9. [PMID: 24007717 DOI: 10.1016/j.jsbmb.2013.08.017] [Citation(s) in RCA: 7] [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: 04/12/2013] [Revised: 08/22/2013] [Accepted: 08/26/2013] [Indexed: 11/15/2022]
Abstract
BACKGROUND Reverse cholesterol transport (RCT) has been inversely related to atherosclerosis and cardiovascular risk. The influence of menopause in the RCT process is poorly understood and the effects of cholesterol-lowering interventions, including statins and hormone therapy (HT), on genes controlling the RCT in postmenopausal women are also unknown. METHODS The effects on serum lipids and expression profile of genes involved in RCT - APOA1, ABCA1, ABCG1, SCARB1 and LXRA - were evaluated by TaqMan(®) quantitative PCR in peripheral blood mononuclear cells (PBMC) from 87 postmenopausal hypercholesterolemic women treated with atorvastatin (AT, n=17), estrogen or estrogen plus progestin (HT, n=34) and estrogen or estrogen plus progestin associated with atorvastatin (HT+AT, n=36). RESULTS Atorvastatin and HT treatments reduced the mRNA levels of APOA1 and SCARB1, respectively, whereas ABCA1 expression was reduced after all treatments. Although the expression of LXRA, an important transcription factor controlling the expression of genes involved in RCT, was not modified after any treatment, it was correlated with ABCA1, APOA1 and SCARB1 RNAm values before and after treatments, however no correlation with ABCG1 was observed. In a linear regression analysis, HT was related to an increase in apoAI levels after treatment when compared to atorvastatin and, moreover, higher SCARB1 and ABCA1 basal expression were also associated with decreased apoAI levels after treatments. CONCLUSION ABCA1 mRNA levels are decreased by atorvastatin and HT, however these treatments have a differential effect on APOA1 and SCARB1 expression in PBMC from postmenopausal women. Basal ABCA1 and SCARB1 expression profile could be helpful markers in predicting the effect of atorvastatin and HT on RCT, according to the changes in apoAI levels in this sample population.
Collapse
Affiliation(s)
- Alvaro Cerda
- School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo, Brazil.
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Hu YW, Ma X, Huang JL, Mao XR, Yang JY, Zhao JY, Li SF, Qiu YR, Yang J, Zheng L, Wang Q. Dihydrocapsaicin Attenuates Plaque Formation through a PPARγ/LXRα Pathway in apoE(-/-) Mice Fed a High-Fat/High-Cholesterol Diet. PLoS One 2013; 8:e66876. [PMID: 23840542 PMCID: PMC3694162 DOI: 10.1371/journal.pone.0066876] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2012] [Accepted: 05/10/2013] [Indexed: 12/26/2022] Open
Abstract
Aims Atherosclerosis is a chronic inflammatory disease and represents the major cause of cardiovascular morbidity and mortality. There is evidence that dihydrocapsaicin (DHC) can exert multiple pharmacological and physiological effects. Here, we explored the effect of DHC in atherosclerotic plaque progression in apoE−/− mice fed a high-fat/high-cholesterol diet. Methods and Results apoE−/− mice were randomly divided into two groups and fed a high-fat/high-cholesterol diet with or without DHC for 12 weeks. We demonstrated that cellular cholesterol content was significantly decreased while apoA1-mediated cholesterol efflux was significantly increased following treatment with DHC in THP-1 macrophage-derived foam cells. We also observed that plasma levels of TG, LDL-C, VLDL-C, IL-1β, IL-6, TNF-α and CRP were markedly decreased while plasma levels of apoA1 and HDL-C were significantly increased, and consistent with this, atherosclerotic lesion development was significantly inhibited by DHC treatment of apoE−/− mice fed a high-fat/high-cholesterol diet. Moreover, treatment with both LXRα siRNA and PPARγ siRNA made the up-regulation of DHC on ABCA1, ABCG1, ABCG5, SR-B1, NPC1, CD36, LDLR, HMGCR, apoA1 and apoE expression notably abolished while made the down-regulation of DHC on SRA1 expression markedly compensated. And treatment with PPARγ siRNA made the DHC-induced up-regulation of LXRα expression notably abolished while treatment with LXRα siRNA had no effect on DHC-induced PPARγ expression. Conclusion These observations provide direct evidence that DHC can significantly decrease atherosclerotic plaque formation involving in a PPARγ/LXRα pathway and thus DHC may represent a promising candidate for a therapeutic agent for the treatment or prevention of atherosclerosis.
Collapse
Affiliation(s)
- Yan-Wei Hu
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Xin Ma
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jin-Lan Huang
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Xin-Ru Mao
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jun-Yao Yang
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jia-Yi Zhao
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Shu-Fen Li
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yu-Rong Qiu
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jia Yang
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Lei Zheng
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- * E-mail: (LZ); (QW)
| | - Qian Wang
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- * E-mail: (LZ); (QW)
| |
Collapse
|
48
|
Yoon HS, Ju JH, Lee JE, Park HJ, Lee JM, Shin HK, Holzapfel W, Park KY, Do MS. The probiotic Lactobacillus rhamnosus BFE5264 and Lactobacillus plantarum NR74 promote cholesterol efflux and suppress inflammation in THP-1 cells. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2013; 93:781-787. [PMID: 22806829 DOI: 10.1002/jsfa.5797] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2012] [Revised: 05/15/2012] [Accepted: 06/06/2012] [Indexed: 06/01/2023]
Abstract
BACKGROUND The balance between the rate of cholesterol uptake/accumulation and the rate of cholesterol efflux is reflected in the amount of lipid accumulation in macrophages. Based upon the fact that liver X receptors (LXRs) play a role in cholesterol efflux, we studied the effects of probiotics on cholesterol efflux and anti-inflammatory action in macrophages. We confirmed changes in LXR expression by treatment of LXR-transfected CHO-K1 cells with lactic acid bacteria (LAB), and co-cultured THP-1 cells with LAB to investigate changes in cholesterol efflux and inflammation. RESULTS The experiment with CHO-K1 cells showed upregulation of LXR-β by LAB. Treatment of THP-1 cells with LAB promoted LXR expression in THP-1, which eventually led to significant upregulation of ABCA1 and ABCG1 expression. The treatment with live LAB also significantly promoted cholesterol efflux. LAB suppressed expression of interleukin (IL)-1β and tumor necrosis factor (TNF)-α, which resulted from activation of LXR. CONCLUSION Our study shows that Lactobacillus rhamnosus BFE5264 and Lactobacillus plantarum NR74 activated LXR and induced cholesterol efflux by promoting expression of ABCA1 and ABCG1. Both strains also suppressed proinflammatory cytokines including IL-1β and TNF-α. This study could account for the observation that LAB may block foam cell formation by cholesterol efflux and immune modulation.
Collapse
Affiliation(s)
- Hong-sup Yoon
- School of Life Science, Handong Global University, Pohang, Gyeongbuk 791-708, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Dubé E, Ethier-Chiasson M, Lafond J. Modulation of cholesterol transport by insulin-treated gestational diabetes mellitus in human full-term placenta. Biol Reprod 2013; 88:16. [PMID: 23221398 DOI: 10.1095/biolreprod.112.105619] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Gestational diabetes mellitus (GDM) is a common complication of pregnancy that is characterized by glucose intolerance, leads to dyslipidemia, and is aggravated by obesity. Cholesterol is taken up by the placenta as part of lipoproteins through the scavenger receptor class B type I receptor (SRBI), low-density lipoprotein receptor (LDLR), and very low density lipoprotein receptor (VLDLR), and its efflux is then mediated by ABCA1 and ABCG1. PCSK9 is involved in the degradation of LDLR and VLDLR. The goal of this study was to evaluate the impact of GDM and prepregnancy body mass index (BMI) on cholesterol transport through the modulation of the expression of several key players. Human full-term placenta, maternal, and venous cord blood samples were obtained at delivery from normal-weight women without GDM (n = 10), normal-weight women with GDM (n = 6), and overweight/obese women with GDM (n = 6). Lipids (total cholesterol, high-density lipoprotein, low-density lipoprotein, triglycerides, free fatty acids, apolipoprotein A1, apolipoprotein B100) levels were evaluated in blood samples. Messenger RNA and protein expression levels (LDLR, VLDLR, SRBI, ABCA1, ABCG1, proprotein convertase subtilisin/kexin type 9, liver x receptors, peroxisome proliferator-activated receptors) were assessed in human full-term placenta, respectively, by real-time RT-PCR and Western blots. Lipoprotein lipase activity was evaluated using a commercial kit on tissue homogenates. Overall, our study demonstrates that GDM affects the maternal and neonatal lipid profiles as well as different key players of placental cholesterol transfer from the maternal to the fetal circulation, depending on the maternal BMI. These changes could affect the fetal metabolism and predispose the fetus to future metabolic diseases.
Collapse
Affiliation(s)
- Evemie Dubé
- Laboratoire de Physiologie Materno-Fœtale, Département des Sciences Biologiques, Université du Québec à Montréal, Montréal, Québec, Canada
| | | | | |
Collapse
|
50
|
Liu XY, Lu Q, Ouyang XP, Tang SL, Zhao GJ, Lv YC, He PP, Kuang HJ, Tang YY, Fu Y, Zhang DW, Tang CK. Apelin-13 increases expression of ATP-binding cassette transporter A1 via activating protein kinase C α signaling in THP-1 macrophage-derived foam cells. Atherosclerosis 2012; 226:398-407. [PMID: 23290264 DOI: 10.1016/j.atherosclerosis.2012.12.002] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2012] [Revised: 12/05/2012] [Accepted: 12/05/2012] [Indexed: 01/22/2023]
Abstract
Apelin has an antiatherogenic function through activating protein kinase C (PKC) to initiate a series of cellular signaling pathways. PKC phosphorylates and stabilizes ATP-binding cassette transporter A1 (ABCA1) through inhibiting its degradation mediated by calpain. Thus, in the present study, we investigated whether apelin-13 affects expression of ABCA1 through PKC signaling. The results showed that apelin-13 dramatically increased cholesterol efflux from THP-1 macrophage-derived foam cells and reduced cellular cholesterol levels. ABCA1 protein but not mRNA levels were dramatically increased by apelin-13, and calpain-induced degradation of ABCA1 and calpain activity were suppressed with treatment of apelin-13. However, the effects of apelin-13 on ABCA1 protein expression, cellular cholesterol efflux and calpain activity were abolished by depletion of PKCα, suggesting the potential important role of PKCα. In addition, apelin-13 was shown to phosphorylate serine residues in ABCA1 through the PKCα pathway. Thus, apelin-13 appears to activate PKCα, phosphorylate ABCA1 and inhibit calpain-mediated proteolysis, thereby promoting cholesterol efflux and reducing foam cell formation. Our study herein described a possible mechanism for understanding the antiatherogenic effects of apelin on attenuating the progression of atherosclerosis.
Collapse
Affiliation(s)
- Xiao-Yan Liu
- Institute of Cardiovascular Research, Key Laboratory for Atherosclerology of Hunan Province, Life Science Research Center, University of South China, Hengyang, Hunan 421001, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|