261 results on '"abca1"'
Search Results
2. Gypenoside XVII inhibits ox-LDL-induced macrophage inflammatory responses and promotes cholesterol efflux through activating the miR-182-5p/HDAC9 signaling pathway
- Author
-
Deng, Wen-Yi, Zhou, Cheng-Long, and Zeng, Meng-Ya
- Published
- 2024
- Full Text
- View/download PDF
3. Novel bioactive lipids enhanced HDL-mediated cholesterol efflux from macrophages through the ABCA1 receptor pathway
- Author
-
Khattib, Ali, Shmet, Manar, Ashkar, Rasha, Hayek, Tony, and Khatib, Soliman
- Published
- 2024
- Full Text
- View/download PDF
4. Hepatic steatosis aggravates atherosclerosis via small extracellular vesicle-mediated inhibition of cellular cholesterol efflux.
- Author
-
Chen, Xu, Chen, Shen, Pang, Juan, Huang, Rong, You, Yiran, Zhang, Haoyang, Xiao, Jinghe, Xue, Hongliang, and Ling, Wenhua
- Subjects
- *
HIGH cholesterol diet , *EFFLUX (Microbiology) , *FATTY liver , *NON-alcoholic fatty liver disease , *ATHEROSCLEROSIS , *FOAM cells , *EXTRACELLULAR vesicles , *ATP-binding cassette transporters , *P-glycoprotein - Abstract
While it is recognized that non-alcoholic fatty liver disease (NAFLD) is associated with cardiovascular disease (CVD), how NAFLD affects the development and progression of CVD remains unclear and debatable. Hence, we aimed to determine the role of steatotic hepatocyte-derived small extracellular vesicles (sEVs) in foam cell formation and atherosclerosis progression. sEVs from steatotic hepatocytes were isolated and characterized. MicroRNA (miRNA) deep sequencing was utilized to identify functional miRNA in sEVs. Lastly, we conducted a cross-sectional study on patients with NAFLD to validate these findings. Treatment of sEVs from steatotic hepatocytes promoted macrophage-derived foam cell formation and atherosclerosis progression via inhibition of ABCA1-mediated cholesterol efflux. Macrophage-specific deletion of Abca1 in ApoE -/- mice abolished the role of steatotic hepatocyte-derived sEVs in atherosclerosis progression. In addition, hepatocyte-specific deletion of Rab27a , which is the key GTPase regulating sEV release, significantly ameliorated high-fat, high-cholesterol diet-induced atherosclerosis progression in ApoE -/- mice. The miRNA deep sequencing results showed that miR-30a-3p was enriched in sEVs from steatotic hepatocytes. miR-30a-3p directly targeted the 3' untranslated region of ABCA1 to inhibit ABCA1 expression and cholesterol efflux. Treatment with antagomiR-30a-3p significantly attenuated atherosclerosis progression in high-fat, high-cholesterol diet-fed ApoE -/- mice. Moreover, serum sEVs from patients with NAFLD and sEV-miR-30a-3p expression were associated with decreased cholesterol efflux levels in foam cells. Steatotic hepatocyte-derived sEVs promote foam cell formation and facilitate atherogenesis via the miR-30a-3p/ABCA1 axis. Reducing sEV secretion by steatotic hepatocytes or targeting miR-30a-3p may be potential therapeutic approaches to slow the progression of NAFLD-driven atherosclerosis. The presence of hepatic steatosis is strongly correlated with the risk of cardiovascular disease and cardiovascular events, yet the molecular mechanisms linking steatosis to progression of atherosclerosis are unclear. Herein, we identified small extracellular vesicles from steatotic hepatocytes as a trigger that accelerated the progression of atherosclerosis. Steatotic hepatocyte-derived small extracellular vesicles promoted foam cell formation via the miR-30a-3p/ABCA1 axis. Our findings not only provide mechanistic insight into non-alcoholic fatty liver disease-driven atherosclerosis but also provide potential therapeutic targets for patients with atherosclerosis. [Display omitted] • Extracellular vesicles from steatotic hepatocytes promote foam cell formation in atherosclerosis. • Steatotic hepatocyte-derived extracellular vesicles inhibit ABCA1-mediated cholesterol efflux via miR-30a-3p. • Inhibition of extracellular vesicle release from hepatocytes ameliorates atherosclerosis progression in ApoE -/- mice. • Treatment of antagomiR-30a-3p attenuates high-fat, high-cholesterol diet-induced atherosclerosis progression in ApoE -/- mice. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
5. 25-Hydroxycholesterol attenuates tumor necrosis factor alpha-induced blood-brain barrier breakdown in vitro.
- Author
-
Loiola, Rodrigo Azevedo, Nguyen, Cindy, Dib, Shiraz, Saint-Pol, Julien, Dehouck, Lucie, Sevin, Emmanuel, Naudot, Marie, Landry, Christophe, Pahnke, Jens, Pot, Caroline, and Gosselet, Fabien
- Subjects
- *
CHOLESTEROL metabolism , *TUMOR necrosis factors , *TIGHT junctions , *BLOOD-brain barrier , *CELL metabolism , *ENCEPHALITIS - Abstract
Intracellular cholesterol metabolism is regulated by the SREBP-2 and LXR signaling pathways. The effects of inflammation on these molecular mechanisms remain poorly studied, especially at the blood-brain barrier (BBB) level. Tumor necrosis factor α (TNFα) is a proinflammatory cytokine associated with BBB dysfunction. Therefore, the aim of our study was to investigate the effects of TNFα on BBB cholesterol metabolism, focusing on its underlying signaling pathways. Using a human in vitro BBB model composed of human brain-like endothelial cells (hBLECs) and brain pericytes (HBPs), we observed that TNFα increases BBB permeability by degrading the tight junction protein CLAUDIN-5 and activating stress signaling pathways in both cell types. TNFα also promotes cholesterol release and decreases cholesterol accumulation and APOE secretion. In hBLECs, the expression of SREBP-2 targets (LDLR and HMGCR) is increased, while ABCA1 expression is decreased. In HBPs, only LDLR and ABCA1 expression is increased. TNFα treatment also induces 25-hydroxycholesterol (25-HC) production, a cholesterol metabolite involved in the immune response and intracellular cholesterol metabolism. 25-HC pretreatment attenuates TNFα-induced BBB leakage and partially alleviates the effects of TNFα on ABCA1, LDLR, and HMGCR expression. Overall, our results suggest that TNFα favors cholesterol efflux via an LXR/ABCA1-independent mechanism at the BBB, while it activates the SREBP-2 pathway. Treatment with 25-HC partially reversed the effect of TNFα on the LXR/SREBP-2 pathways. Our study provides novel perspectives for better understanding cerebrovascular signaling events linked to BBB dysfunction and cholesterol metabolism in neuroinflammatory diseases. • TNFα increases BBB permeability via CLAUDIN-5 degradation • TNFα deeply alters cholesterol metabolism of BBB cells • TNFα promotes 25-hydroxycholesterol production • 25-HC partially abolishes TNFα effect on BBB cells [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
6. ABCA1 deficiency causes tissue-specific dysregulation of the SREBP2 pathway in mice.
- Author
-
Yamauchi, Yoshio, Abe-Dohmae, Sumiko, Iwamoto, Noriyuki, Sato, Ryuichiro, and Yokoyama, Shinji
- Subjects
- *
HIGH density lipoproteins , *KNOCKOUT mice , *ATHEROSCLEROSIS , *CHOLESTEROL , *MICE - Abstract
ABCA1 plays an essential role in the formation of high-density lipoprotein (HDL), and its mutations cause Tangier disease (TD), a familial HDL deficiency. In addition to the disappearance of HDL, TD patients exhibit cholesterol deposition in peripheral tissues through a mechanism poorly understood, which may contribute to the development of premature atherosclerosis. We and others previously showed that ABCA1 deficiency causes hyperactivation of the SREBP2 pathway in vitro. Here, we show using Abca1 knockout mice that ABCA1 deficiency leads to tissue-specific dysregulation of SREBP2 activity in a nutritional status-dependent manner, which may underlie the pathophysiology of TD. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
7. Inhibition of the cholesterol transporter ABCA1 by probucol decreases capacitation and tyrosine phosphorylation of dog spermatozoa, and is dose dependent.
- Author
-
Schäfer-Somi, S., Claaßen, S., and Lechner, D.
- Subjects
- *
CHOLESTEROL , *TYROSINE , *ACROSOME reaction , *DOGS , *PHOSPHORYLATION , *DOG breeds , *SPERMATOZOA , *FROZEN semen - Abstract
The ATP binding cassette (ABC) transporter molecule ABCA1 participates in the cholesterol transport within and through cell membranes. We recently demonstrated that in dog spermatozoa, capacitation could be decreased with probucol (PRO), an ABCA1 specific antagonist. In this study, a dose-effect relationship of PRO on dog sperm capacitation, tyrosine phosphorylation and cholesterol efflux from the sperm plasma membrane was investigated. A total of 16 ejaculates from dogs of different breeds, aged 2–4 years were used. Sperm motility and membrane integrity in the main fraction was determined by CASA. Samples were stained with a boron dipyrromethene difluoride (BODIPY) fluorophore (P9672, Sigma- Aldrich, A) diluted in DMSO at a final concentration of 0.4 μM. All samples were divided into 5 aliquots, with 0, 100, 250, 500 and 1000 μM of PRO. After incubation at 37 °C for 2 h, PI was added and flow cytometry performed. All aliquots were examined for capacitation and acrosome reaction by using the CTC assay and tyrosine phosphorylation (TP). Membrane integrity was measured in all aliquots to investigate the effect of PRO on cell membranes. Membrane integrity did not differ between controls (0 μM), and 100, 250 and 500 μM PRO, but decreased with 1000 μM PRO (p < 0.05). Increasing PRO concentration decreased the percentage alive cells with cholesterol efflux per PRO group (0 μM: 77.8 ± 10.6%, 100 μM: 63.7 ± 11.7%, 250 μM: 52.1 ± 12.9%, 500 μM: 37.7 ± 11.6%, 1000 μM: 33.1 ± 14.4%; p < 0.05), decreased head and entire tail phosphorylated cells (0 μM: 34.6%, 1000 μM: 5.1% p < 0.05); and decreased the percentage capacitated cells (maximum with PRO 500 μM: capacitated vs. control: 54.2 ± 17% vs 25 ± 7.7%, p < 0.05). Conclusion: PRO decreased the cholesterol efflux, and decreased tyrosine phosphorylation and capacitation in a dose-dependent manner. This suggests a strong involvement of the ABCA1 transporter in different functional aspects of sperm capacitation in dogs. • Probucol is an ABCA1 specific antagonist and effective in dog spermatozoa membranes. • Increasing probucol decreased the percentage alive cells with cholesterol efflux. • Increasing probucol decreased head and entire tail phosphorylated cells. • Increasing probucol decreased the percentage capacitated cells. • The ABCA1 transporter is involved in sperm capacitation in dogs. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
8. Ilexgenin A inhibits lipid accumulation in macrophages and reduces the progression of atherosclerosis through PTPN2/ERK1/2/ABCA1 signalling pathway.
- Author
-
Zhou, Qinyi, Wang, Yang, Cheng, Yaqiong, Zhou, Jing, Liu, Wang, Ma, Xiaofeng, Tang, Shilin, Tang, Shangshu, and Tang, Chaoke
- Subjects
- *
FOAM cells , *PHOSPHOPROTEIN phosphatases , *CELLULAR signal transduction , *FATTY liver , *PATHOLOGICAL physiology , *ATP-binding cassette transporters , *PROTEIN-tyrosine phosphatase - Abstract
Macrophage lipid accumulation indicates a pathological change in atherosclerosis. Ilexgenin A (IA), a pentacyclic triterpenoid compound, plays a role in preventing inflammation, bacterial infection, and fatty liver and induces a potential anti-atherogenic effect. However, the anti-atherosclerotic mechanism remains unclear. The present study investigated the effects of IA on lipid accumulation in macrophage-derived foam cells and atherogenesis in apoE−/− mice. Our results indicated that the expression of adenosine triphosphate-binding cassette transporter A1 (ABCA1) was up-regulated by IA, promoting cholesterol efflux and reducing lipid accumulation in macrophages, which may be regulated by the protein tyrosine phosphatase non-receptor type 2 (PTPN2)/ERK1/2 signalling pathway. IA attenuated the progression of atherosclerosis in high-fat diet-fed apoE−/− mice. PTPN2 knockdown with siRNA or treatment with an ERK1/2 agonist (Ro 67–7476) impeded the effects of IA on ABCA1 upregulation and cholesterol efflux in macrophages. These results suggest that IA inhibits macrophage lipid accumulation and alleviates atherosclerosis progression via the PTPN2/ERK1/2 signalling pathway. • Ilexgenin A increases ABCA1 expression and promotes cholesterol efflux to apoA-I. • IA attenuated the progression of atherosclerosis in high-fat diet-fed apoE−/− mice. • IA inhibits macrophage lipid accumulation and alleviates atherosclerosis progression via the PTPN2/ERK1/2 signalling pathway. • IA is a potential therapeutic agent against atherosclerosis. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
9. A high-fat/high-cholesterol diet, but not high-cholesterol alone, increases free cholesterol and apoE-rich HDL serum levels in rats and upregulates hepatic ABCA1 expression.
- Author
-
Shinohata, Ryoko, Shibakura, Misako, Arao, Yujiro, Watanabe, Shogo, Hirohata, Satoshi, and Usui, Shinichi
- Subjects
- *
HDL cholesterol , *HIGH density lipoproteins , *ATP-binding cassette transporters , *DIETARY fats , *ANIMAL nutrition , *APOLIPOPROTEIN E - Abstract
A high-fat/high-cholesterol (HFC) diet, but not a high-cholesterol (HC) diet, is known to induce elevated serum apolipoprotein E (apoE)-rich high-density lipoprotein (HDL) levels in animal models. However, the exact mechanisms by which the combination of dietary fat and cholesterol induces apoE-rich HDL production is not well understood. Here, we investigated the effects of dietary fat and cholesterol on serum lipoprotein profiles and hepatic mRNA expression that are associated with HDL production, cholesterol transport, and bile acid metabolism. Male Sprague-Dawley rats were fed HFC, HC, high-fat, or control diets and then evaluated. The HFC diet induced significant increases in hepatic free-cholesterol accumulation (1.4-fold, p < 0.01) and serum apoE-rich HDL cholesterol (8.7-fold, p < 0.001) levels compared with the HC diet. The apoE-rich HDL induced by the HFC diet was remarkably rich in free cholesterol. Liver gene-expression was mostly similar between the HC and HFC diet groups. However, there was a significant increase of ATP-binding cassette transporter A1 (ABCA1) levels in the HFC group compared to the HC group for both mRNA (1.9-fold, p < 0.001) and protein (6.6-fold, p < 0.01). These results suggest that an increase in apoE-rich HDL induced by dietary fat and cholesterol may be involved in cholesterol efflux from the liver through increased ABCA1-mediated free-cholesterol efflux. • An HFC diet increases hepatic free-cholesterol and serum apoE-rich HDL cholesterol. • HFC-induced apoE-rich HDL is remarkably rich in free cholesterol. • HFC significantly increases ABCA1 mRNA and protein levels. • HFC-induced apoE-rich HDL acts via increased ABCA1-mediated free-cholesterol efflux. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
10. The role of HDL- and non-HDL-related parameters in cell-cholesterol efflux capacity.
- Author
-
Asztalos, Bela F., Hauser, Thomas H., Goldfine, Allison B., Welty, Francine K., Horvath, Katalin V., and Schaefer, Ernst J.
- Subjects
- *
CORONARY disease , *LIPOPROTEINS , *LDL cholesterol , *HDL cholesterol - Abstract
The regulation of cell-cholesterol efflux is not completely understood. Our aim was to assess the role of HDL- and non-HDL-related parameters in ATP-binding cassette transporter-A1 (ABCA1) and scavenger receptor class B-type-I (SRBI) cell-cholesterol efflux capacity (CEC) in coronary heart disease (CHD) cases and controls. Lipids and apoA-I-containing HDL particles (by 2D gel-electrophoresis and immunodetection) were measured in 534 statin-treated CHD patients and in 1076 age-, gender-, and BMI-matched controls. ABCA1-CEC and SRBI-CEC were measured in apoB-depleted serum of 100 cases and 100 controls. Cases had significantly higher concentrations of preβ-1 particles (88%) and ABCA1-CEC (34%) compared to controls. ABCA1-CEC was positively correlated with the concentrations of preβ-1 particles, triglycerides, small-dense (sd) LDL-C, and LDL-C in both cases and controls. Moreover, both the concentration and the functionality of preβ-1 particles (ABCA1-CEC/mg preβ-1) were positively associated with the concentrations of sdLDL-C and triglycerides. Cases had 27% lower levels of large HDL particles but similar SRBI-CEC compared to controls. SRBI-CEC was correlated positively with HDL-C, apoA-I, and large-HDL particle levels. However, the functionality of large-HDL particles (SRBI-CEC/mg large particles) was significantly and positively correlated with the preβ-1/α-1 ratio, sdLDL-C, and triglycerides. CHD patients have significantly higher concentration, but less functional preβ-1 particles in term of cholesterol efflux capacity compared to controls. Triglyceride-rich lipoproteins have significant influence on either the concentration or the functionality or both of HDL particles and consequently HDL-CEC. [Display omitted] • Coronary heart disease (CHD) patients have significantly higher ABCA1-cholesterol efflux capacity (CEC) compared to controls. • CHD patients have significantly higher concentration, but less functional preβ-1 particles, compared to controls. • TG-rich lipoproteins significantly influence the concentration and functionality of HDL particles and consequently HDL-CEC. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
11. The ABCA1-efferocytosis axis: A new strategy to protect against atherosclerosis.
- Author
-
Chen, Wujun, Li, Lu, Wang, Jie, Zhang, Renshuai, Zhang, Tingting, Wu, Yudong, Wang, Shuai, and Xing, Dongming
- Subjects
- *
ATHEROSCLEROSIS , *HDL cholesterol , *CORONARY disease , *MYOCARDIAL infarction , *PHAGOCYTOSIS - Abstract
• Efferocytosis involves the clearance of apoptotic cells by phagocytes to suppress atherosclerosis. • ABCA1 promotes not only cholesterol efflux but also efferocytosis. • ABCA1 promotes efferocytosis by regulating "find-me" ligands and "eat-me" ligands expression. • ABCA1 has an efferocytosis pathway similar to that of TG2, which is an "eat-me" ligand. • ABCA1 can form several regulatory feedback axes with ANXA1, MEGF10, GULP1, TNFα, IL-6. Atherosclerosis, a disease process characterized by lipid accumulation and inflammation, is the main cause of coronary heart disease (CHD) and myocardial infarction (MI). Efferocytosis involves the clearance of apoptotic cells by phagocytes. Successful engulfment triggers the release of anti-inflammatory cytokines to suppress atherosclerosis. ABCA1 is a key mediator of cholesterol efflux to apoA-I for the generation of HDL-C in reverse cholesterol transport (RCT). Intriguingly, ABCA1 promotes not only cholesterol efflux but also efferocytosis. ABCA1 promotes efferocytosis by regulating the release of "find-me" ligands, including LPC, and the exposure, release, and expression of "eat-me" ligands, including PtdSer, ANXA1, ANXA5, MEGF10, and GULP1. ABCA1 has a pathway similar to TG2, which is an "eat-me" ligand. ABCA1 has the highest known homology to ABCA7, which controls efferocytosis as the engulfment and processing ligand. In addition, ABCA1 can form several regulatory feedback axes with ANXA1, MEGF10, GULP1, TNFα, and IL-6. Therefore, ABCA1 is the central factor that links cholesterol efflux and apoptotic cell clearance. Several drugs have been studied or approved for apoptotic cell clearance, such as CD47 antibody and PD1-/PD-L1 antibody. In this article, we review the role and mechanism of action of ABCA1 in efferocytosis and focus on new insights into the ABCA1-efferocytosis axis and its potential as a novel therapeutic target in atherosclerosis. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
12. TIGAR mitigates atherosclerosis by promoting cholesterol efflux from macrophages.
- Author
-
Zhao, Zhen-Wang, Zhang, Min, Zou, Jin, Wan, Xiang-Jun, Zhou, Li, Wu, Yao, Liu, Shang-Ming, Liao, Ling-Xiao, Li, Heng, Qin, Yu-Sheng, Yu, Xiao-Hua, and Tang, Chao-Ke
- Subjects
- *
ATP-binding cassette transporters , *ATHEROSCLEROSIS , *CHOLESTEROL , *CHOLESTEROL metabolism , *MACROPHAGES , *BLOOD lipids - Abstract
TP53-induced glycolysis and apoptosis regulator (TIGAR) is now characterized as a fructose-2,6-bisphosphatase to reduce glycolysis and protect against oxidative stress. Recent studies have demonstrated that TIGAR is associated with cardiovascular disease. However, little is known about its role in atherosclerogenesis. In this study, we aimed to investigate the effect of TIGAR on atherosclerosis and explore the underlying molecular mechanism. The Gene Expression Omnibus (GEO) datasets were used to analyze the differential expression of relative proteins. THP-1-derived macrophages were used as an in vitro model and apolipoprotein E-deficient (Apoe −/−) mice were used as an in vivo model. [3H] labeled cholesterol was used to assess the capacity of cholesterol efflux and reverse cholesterol transport (RCT). Both qPCR and Western blot were used to evaluate the mRNA and protein expression, respectively. Lentiviral vectors were used to disturb the expression of TIGAR in vitro and in vivo. Oil Red O, hematoxylin-eosin, and Masson staining were performed to evaluate atherosclerotic plaques in Apoe −/− mice fed a Western diet. Conventional assay kits were used to measure the levels of reactive oxygen species (ROS), plasma lipid profiles and 27-hydroxycholesterol (27-HC). Our results showed that TIGAR is increased upon the formation of macrophage foam cells and atherosclerosis. TIGAR knockdown markedly promoted lipid accumulation in macrophages. Silencing of TIGAR impaired cholesterol efflux and down-regulated the expression of ATP-binding cassette transporter A1 (ABCA1) and ABCG1 by interfering with liver X receptor α (LXRα) expression and activity, but did not influence cholesterol uptake by macrophages. Additionally, this inhibitory effect of TIGAR deficiency on cholesterol metabolism was mediated through the ROS/CYP27A1 pathway. In vivo experiments revealed that TIGAR deficiency decreased the levels of ABCA1 and ABCG1 in plaques and aorta and impaired the capacity of RCT, thereby leading to the progression of atherosclerosis in Apoe −/− mice. TIGAR mitigates the development of atherosclerosis by up-regulating ABCA1 and ABCG1 expression via the ROS/CYP27A1/LXRα pathway. [Display omitted] • TIGAR is involved in macrophage foam cells formation and atherosclerosis development. • TIGAR promotes ABCA1-and ABCG1-dependent cholesterol efflux, but does not influence cholesterol uptake in macrophages. • TIGAR increases ABCA1 and ABCG1 expression by reducing ROS and activating CYP27A1/LXRα pathway. • TIGAR facilitatesreverse cholesterol transport (RCT) and mitigates the development of atherosclerosis in Apoe −/− mice. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
13. Recent advances in the regulation of ABCA1 and ABCG1 by lncRNAs.
- Author
-
Zhang, Shun, Li, Lu, Wang, Jie, Zhang, Tingting, Ye, Ting, Wang, Shuai, Xing, Dongming, and Chen, Wujun
- Subjects
- *
ATHEROSCLEROSIS , *CORONARY disease , *CHOLESTEROL , *SIRTUINS , *LINCRNA , *EFFLUX (Microbiology) - Abstract
• ABCA1 and ABCG1 promote cholesterol efflux to suppress foam cell generation. • lncRNAs regulate cholesterol efflux to influence atherosclerosis development. • ABCA1 and ABCG1 are regulated by different lncRNAs. • Some lncRNAs play dual roles in ABCA1 expression and atherosclerosis. • Some lncRNAs form a feedback loop with target gene. Coronary heart disease (CHD) with atherosclerosis is the leading cause of death worldwide. ABCA1 and ABCG1 promote cholesterol efflux to suppress foam cell generation and reduce atherosclerosis development. Long noncoding RNAs (lncRNAs) are emerging as a unique group of RNA transcripts that longer than 200 nucleotides and have no protein-coding potential. Many studies have found that lncRNAs regulate cholesterol efflux to influence atherosclerosis development. ABCA1 is regulated by different lncRNAs, including MeXis, GAS5, TUG1, MEG3, MALAT1, Lnc-HC, RP5-833A20.1, LOXL1-AS1, CHROME, DAPK1-IT1, SIRT1 AS lncRNA, DYNLRB2-2, DANCR, LeXis, LOC286367, and LncOR13C9. ABCG1 is also regulated by different lncRNAs, including TUG1, GAS5, RP5-833A20.1, DYNLRB2-2, ENST00000602558.1, and AC096664.3. Thus, various lncRNAs are associated with the roles of ABCA1 and ABCG1 on cholesterol efflux in atherosclerosis regulation. However, some lncRNAs play dual roles in ABCA1 expression and atherosclerosis, and the functions of some lncRNAs in atherosclerosis have not been investigated in vivo. In this article, we review the roles of lncRNAs in atherosclerosis and focus on new insights into lncRNAs associated with the roles of ABCA1 and ABCG1 on cholesterol efflux and the potential of these lncRNAs as novel therapeutic targets in atherosclerosis. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
14. The effect of transgender hormonal treatment on high density lipoprotein cholesterol efflux capacity.
- Author
-
van Velzen, Daan M., Adorni, Maria Pia, Zimetti, Francesca, Strazzella, Arianna, Simsek, Suat, Sirtori, Cesare R., Heijer, Martin den, and Ruscica, Massimiliano
- Subjects
- *
ATP-binding cassette transporters , *TRANS women , *TRANSGENDER people , *CHOLESTEROL , *TRANS men - Abstract
A decrease in high-density lipoprotein (HDL)-cholesterol concentrations during transgender hormone therapy has been shown. However, the ability of HDL to remove cholesterol from arterial wall macrophages, termed cholesterol efflux capacity (CEC), has proven to be a better predictor of cardiovascular disease (CVD) largely independently of HDL-concentrations. In addition, the serum capacity to load macrophages with cholesterol (cholesterol loading capacity, CLC) represents an index of pro-atherogenic potential. As transgender individuals are exposed to lifelong exogenous hormone therapy (HT), it becomes of interest to study whether HDL-CEC and serum CLC are affected by HT. HDL-CEC and serum CLC have been evaluated in 15 trans men treated with testosterone and in 15 trans women treated with estradiol and cyproterone acetate at baseline and after 12 months of HT. Total HDL-CEC from macrophages and its major contributors, the ATP-binding cassette transporters (ABC) A1 and ABCG1 HDL-CEC and HDL-CEC by aqueous diffusion were determined by a radioisotopic assay. CLC was evaluated in human THP-1 macrophages. In trans women, total HDL-CEC decreased by 10.8% (95%CI: −14.3;-7.3; p < 0.001), ABCA1 HDL-CEC by 23.8% (−34.7; −12.9; p < 0.001) and aqueous diffusion HDL-CEC by 4.8% (−8.4;-1.1; p < 0.01). In trans men, only aqueous diffusion HDL-CEC decreased significantly, −9.8% (−15.7;-3.9; p < 0.01). ABCG1 HDL-CEC did not change in either group. Serum CLC and HDL subclass distribution were not modified by HT in both groups. Total HDL-CEC decreased during HT in trans women, with a specific reduction in ABCA1 CEC. This finding might contribute to a higher CVD risk. [Display omitted] • In trans women, total, ABCA1 and aqueous diffusion high density lipoprotein cholesterol efflux capacity (HDL-CEC) decreased after hormone therapy. • In trans women, changes in HDL-CEC are not associated with plasma HDL-C levels. • Results in trans women may explain the increased cardiovascular risk in feminizing hormone therapy. • In trans men only aqueous diffusion HDL-CEC significantly decreased after therapy. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
15. Impact of bone marrow ATP-binding cassette transporter A1 deficiency on atherogenesis is independent of the presence of the low-density lipoprotein receptor.
- Author
-
Ouweneel, Amber B., Zhao, Ying, Calpe-Berdiel, Laura, Lammers, Bart, Hoekstra, Menno, Van Berkel, Theo J.C., and Van Eck, Miranda
- Subjects
- *
ATP-binding cassette transporters , *LIPOPROTEIN receptors , *BONE marrow , *ATHEROSCLEROSIS , *BONE marrow transplantation , *LYMPHOCYTE count - Abstract
There is extensive evidence from bone marrow transplantation studies that hematopoietic ATP binding cassette A1 (Abca1) is atheroprotective in low-density lipoprotein receptor (Ldlr) deficient mice. In contrast, studies using lysosyme M promoter-driven deletion of Abca1 in Ldlr deficient mice failed to show similar effects. It was hypothesized that the discrepancy between these studies might be due to the presence of Ldlr in bone marrow-derived cells in the transplantation model. In this study, we aim to determine the contribution of Ldlr to the atheroprotective effect of hematopoietic Abca1 in the murine bone marrow transplantation model. Wild-type, Ldlr −/−, Abca1 −/−, and Abca1 −/− Ldlr −/− bone marrow was transplanted into hypercholesterolemic Ldlr −/− mice. Bone marrow Lldr deficiency did not influence the effects of Abca1 on macrophage cholesterol efflux, foam cell formation, monocytosis or plasma cholesterol. Ldlr deficiency did reduce circulating and peritoneal lymphocyte counts, albeit only in animals lacking Abca1 in bone marrow-derived cells. Importantly, the effects of Abca1 deficiency on atherosclerosis susceptibility were unaltered by the presence or absence of Ldlr. Bone marrow Ldlr deficiency did lead to marginally but consistently decreased atherosclerosis, regardless of Abca1 deficiency. Thus, Ldlr expression on bone marrow-derived cells does, to a minimal extent, influence atherosclerotic lesion development, albeit independent of Abca1. This study provides novel insight into the relative impact of Ldlr and Abca1 in bone marrow-derived cells on macrophage foam cell formation and atherosclerosis development in vivo. We have shown that Ldlr and Abca1 differentially and independently influence atherosclerosis development in a murine bone marrow transplantation model of atherosclerosis. Image 1 • Hematopoietic Ldlr and Abca1 independently influence atherosclerosis development. • Ldlr deletion does not alter Abca1 deficiency-induced effects on macrophage cholesterol handling. • Bone marrow Abca1 deletion-driven lymphocytosis is reduced by the additional deletion of Ldlr. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
16. Breast cancer resistance protein (Bcrp/Abcg2) is selectively modulated by lipopolysaccharide (LPS) in the mouse yolk sac.
- Author
-
Martinelli, L.M., Reginatto, M.W., Fontes, K.N., Andrade, C.B.V., Monteiro, V.R.S., Gomes, H.R., Almeida, F.R.C.L., Bloise, F.F., Matthews, S.G., Ortiga-Carvalho, T.M., and Bloise, E.
- Subjects
- *
YOLK sac , *TOXIC substance exposure , *BREAST cancer , *ATP-binding cassette transporters , *GESTATIONAL age - Abstract
• ABC transporters modulate transfer of xenobiotics, toxins, nutrients and cytokines. • Bcrp is localized in the yolk sac endodermal epithelium and in the mesothelium. • Lipopolysaccharide affect yolk Bcrp/ Abcg2 expression in a gestational-age dependent-manner. • These changes may alter fetal exposure to xenobiotics and toxic substances. Bacterial infection alters placental ABC transporters expression. These transporters provide fetal protection against circulating xenobiotics and environmental toxins present in maternal blood. We hypothesized that lipopolysaccharide (LPS-bacterial mimic) alters the yolk sac morphology and expression of key ABC transporters in a gestational-age dependent manner. Yolk sac samples from C57BL/6 mice were obtained at gestational ages (GD) 15.5 and GD18.5, 4 or 24 h after LPS exposure (150ug/kg; n = 8/group). Samples underwent morphometrical, qPCR and immunohistochemistry analysis. The volumetric proportions of the histological components of the yolk sac did not change in response to LPS. LPS increased Abcg2 expression at GD15.5, after 4 h of treatment (p < 0.05). No changes in Abca1, Abcb1a/b, Abcg1, Glut1, Snat1, Il-1β, Ccl2 and Mif were observed. Il-6 and Cxcl1 were undetectable in the yolk sac throughout pregnancy. Abca1, breast cancer resistance protein (Bcrp, encoded by Abcg2) and P-glycoprotein (P-gp/ Abcb1a/b) were localized in the endodermal (uterine-facing) epithelium and to a lesser extent in the mesothelium (amnion-facing), whereas Abca1 was also localized to the endothelium of the yolk sac blood vessels. LPS increased the labeling area and intensity of Bcrp in the yolk sac's mesothelial cells at GD15.5 (4 h), whereas at GD18.5, the area of Bcrp labeling in the mesothelium (4 and 24 h) was decreased (p < 0.05). Bacterial infection has the potential to change yolk sac barrier function by affecting Bcrp and Abcg2 expression in a gestational-age dependent-manner. These changes may alter fetal exposure to xenobiotics and toxic substances present in the maternal circulation and in the uterine cavity. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
17. Preβ1-high-density lipoprotein metabolism is delayed in patients with chronic kidney disease not on hemodialysis.
- Author
-
Yamatani, Kotoko, Hirayama, Satoshi, Seino, Utako, Hirayama, Akiko, Hori, Atsushi, Suzuki, Koya, Idei, Mayumi, Kitahara, Masaki, and Miida, Takashi
- Subjects
CARRIER proteins ,CHRONIC kidney failure ,COMPARATIVE studies ,CONTINUOUS ambulatory peritoneal dialysis ,HIGH density lipoproteins ,IMMUNOASSAY ,KIDNEYS ,MEMBRANE proteins ,TRANSFERASES - Abstract
Preβ1-high-density lipoprotein (HDL) is a lipid-poor cholesterol acceptor that is converted to lipid-rich HDL by lecithin–cholesterol acyltransferase (LCAT). In patients receiving hemodialysis, preβ1-HDL metabolism is hampered even if HDL cholesterol is normal. Hemodialysis may affect preβ1-HDL metabolism by releasing lipases from the vascular wall due to heparin. We investigated whether preβ1-HDL metabolism is delayed in patients with chronic kidney disease (CKD) who are not receiving hemodialysis. We examined 44 patients with Stage 3 or higher CKD and 22 healthy volunteers (Control group). The patients with CKD were divided into those without renal replacement therapy (CKD group, n = 22) and those undergoing continuous ambulatory peritoneal dialysis (CAPD group, n = 22). Plasma preβ1-HDL concentrations were determined by immunoassay. During incubation at 37°C, we used 5,5-dithio-bis (2-nitrobenzoic acid) (DTNB) to inhibit LCAT activity and defined the conversion halftime of preβ1-HDL (CHT preβ1) as the time required for the difference in preβ1-HDL concentration in the presence and absence of 5,5-DTNB to reach half the baseline concentration. The absolute and relative preβ1-HDL concentrations were higher, and CHT preβ1 was longer in the CKD and CAPD groups than in the Control group. Preβ1-HDL concentration was significantly correlated with CHT preβ1 but not with LCAT activity in patients with CKD and CAPD. Preβ1-HDL metabolism is delayed in patients with CKD who are not on hemodialysis. This preβ1-HDL metabolic delay may progress as renal function declines. • We measured preβ1-high-density lipoprotein (HDL) levels in patients with chronic kidney disease not on hemodialysis. • Preβ1-HDL concentrations increased gradually as chronic kidney disease reached more advanced stages. • High preβ1-HDL was associated with delayed preβ1-HDL metabolism. • Impaired HDL metabolism is likely to progress as renal function declines. • Preβ1-HDL concentration could be a clinical marker for HDL dysfunction. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
18. Molecular mechanism for nobiletin to enhance ABCA1/G1 expression in mouse macrophages.
- Author
-
Tsuboi, Tomoe, Lu, Rui, Yonezawa, Takayuki, Watanabe, Akio, Woo, Je-Tae, Abe-Dohmae, Sumiko, and Yokoyama, Shinji
- Subjects
- *
MICE , *PEROXISOME proliferator-activated receptors , *MESSENGER RNA - Abstract
Nobiletin (NOB), a functional ingredient found in citrus peel, is said to act against diabetes, obesity, and atherosclerosis. It has been reported to activate AMPK pathway, as well as increase SREBP1c, PPARα and PPARγ expression. However, no molecular mechanism has been elucidated to be able to integrate these sporadic findings with some controversies to lead to concrete outcomes. In this study, regulation of HDL biogenesis by NOB was investigated modulating ABCA1 and ABCG1 expression. Regulation of ABCA1/G1 by NOB was investigated in mouse macrophages J774.1. NOB increased mRNA and protein levels of ABCA1/G1, and cell cholesterol release by these factors. It also increased mRNA of PPARγ and LXRα but not PPARα. The increase in ABCA1/G1 mRNA levels by NOB was suppressed by antagonists of PPARγ and LXRα. The increase in PPAR γ mRNA levels by NOB was suppressed by an LXRα antagonist, and the increase in LXRα mRNA levels was suppressed by a PPARγ antagonist. NOB increased CD3 6 mRNA and this was suppressed by an LXRα antagonist. The increase in ABCA1 mRNA by a PPARγ agonist was also suppressed by an LXRα antagonist. NOB did not influence LPL1 mRNA expression levels. NOB stimulated AMPK phosphorylation, and the increase in ABCA1/G1 , LXRα and PPARγ mRNA levels and ABCA1/G1 protein levels by NOB was reversed by an AMPK inhibitor. AMPK siRNA suppressed ABCA1 expression. NOB activates AMPK and subsequently LXRα to promote the expression of ABCA1 and ABCG1, and an LXRα - PPARγ loop pathway amplifies these signals. Image 1 • Nobiletin activates AMPK to enhance LXRα transcription and increase transcription of ABCA1 and ABCG1 for HDL biogenesis. • Nobiletin-activated pathway for ABCA1/G1 expression is amplified by LXRα-PPARγ loop. • LXRα-PPARα loop is not involved in amplification of nobiletin-activated increase of ABCA1/G1 expression. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
19. Cav3.1 T-type calcium channel blocker NNC 55-0396 reduces atherosclerosis by increasing cholesterol efflux.
- Author
-
Tsai, Min-Chien, Cho, Rou-Ling, Lin, Chin-Sheng, Jheng, Yu-Sin, Lien, Chih-Feng, Chen, Chien-Chang, and Tzeng, Bing-Hsiean
- Subjects
- *
ATP-binding cassette transporters , *CALCIUM antagonists , *HIGH cholesterol diet , *CHOLESTEROL , *LOW density lipoprotein receptors , *LIPID metabolism - Abstract
Ca v 3.1 T-type calcium channel blocker reduces atherosclerosis by increasing cholesterol efflux and inhibition of lipid accumulation through p38/JNK-LXRα-ABCA1/ABCG1 signaling pathways. [Display omitted] Calcium channel blockers (CCBs) are commonly used as antihypertensive agents. While certain L-type CCBs exhibit antiatherogenic effects, the impact of Ca v 3.1 T-type CCBs on antiatherogenesis and lipid metabolism remains unexplored. NNC 55–0396 (NNC) is a highly selective blocker of T-type calcium channels (Ca v 3.1 channels). We investigated the effects of NNC on relevant molecules and molecular mechanisms in human THP-1 macrophages. Cholesterol efflux, an indicator of reverse cholesterol transport (RCT) efficiency, was assessed using [3H]-labeled cholesterol. In vivo, high cholesterol diet (HCD)-fed LDL receptor knockout (Ldlr -/-) mice, an atherosclerosis-prone model, underwent histochemical staining to analyze plaque burden. Treatment of THP-1 macrophages with NNC facilitated cholesterol efflux and reduced intracellular cholesterol accumulation. Pharmacological and genetic interventions demonstrated that NNC treatment or Ca v 3.1 knockdown significantly enhanced the protein expression of scavenger receptor B1 (SR-B1), ATP-binding cassette transporter A1 (ABCA1), ATP-binding cassette transporter G1 (ABCG1), and liver X receptor alpha (LXRα) transcription factor. Mechanistic analysis revealed that NNC activates p38 and c-Jun N-terminal kinase (JNK) phosphorylation, leading to increased expression of ABCA1, ABCG1, and LXRα-without involving the microRNA pathway. LXRα is required for NNC–induced ABCA1 and ABCG1 expression. Administering NNC diminished atherosclerotic lesion area and lipid deposition in HCD-fed Ldlr -/- mice. NNC's anti-atherosclerotic effects, achieved through enhanced cholesterol efflux and inhibition of lipid accumulation, suggest a promising therapeutic approach for hypertensive patients with atherosclerosis. This research highlights the potential of Ca v 3.1 T-type CCBs in addressing cardiovascular complications associated with hypertension. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
20. A pan-PPAR agonist E17241 ameliorates hyperglycemia and diabetic dyslipidemia in KKAy mice via up-regulating ABCA1 in islet, liver, and white adipose tissue.
- Author
-
Sheng, Ren, Li, Yining, Wu, Yexiang, Liu, Chang, Wang, Weizhi, Han, Xiaowan, Li, Yinghong, Lei, Lijuan, Jiang, Xinhai, Zhang, Yuyan, Zhang, Yuhao, Li, Shunwang, Hong, Bin, Liu, Chao, Xu, Yanni, and Si, Shuyi
- Subjects
- *
ATP-binding cassette transporters , *WHITE adipose tissue , *TYPE 2 diabetes , *PEROXISOME proliferator-activated receptors , *ISLANDS , *DYSLIPIDEMIA - Abstract
Type 2 diabetes mellitus (T2DM) is a common chronic metabolic disease. Peroxisome proliferator-activated receptors (PPARs) play crucial roles in regulating glucolipid metabolism. Previous studies showed that E17241 could ameliorate atherosclerosis and lower fasting blood glucose levels in ApoE −/− mice. In this work, we investigated the role of E17241 in glycolipid metabolism in diabetic KKAy mice. We confirmed that E17241 is a powerful pan-PPAR agonist with a potent agonistic activity on PPARγ, a high activity on PPARα, and a moderate activity on PPARδ. E17241 also significantly increased the protein expression of ATP-binding cassette transporter 1 (ABCA1), a crucial downstream target gene for PPARs. E17241 clearly lowered plasma glucose levels, improved OGTT and ITT, decreased islet cholesterol content, improved β-cell function, and promoted insulin secretion in KKAy mice. Moreover, E17241 could significantly lower plasma total cholesterol and triglyceride levels, reduce liver lipid deposition, and improve the adipocyte hypertrophy and the inflammatory response in epididymal white adipose tissue. Further mechanistic studies indicated that E17241 boosts cholesterol efflux and insulin secretion in an ABCA1 dependent manner. RNA-seq and qRT-PCR analysis demonstrated that E17241 induced different expression of PPAR target genes in liver and adipose tissue differently from the PPARγ agonist rosiglitazone. In addition, E17241 treatment was also demonstrated to have an exhilarating cardiorenal benefits. Our results demonstrate that E17241 regulates glucolipid metabolism in KKAy diabetic mice while having cardiorenal benefits without inducing weight gain. It is a promising drug candidate for the treatment of T2DM. [Display omitted] • E17241, a unique pan-PPAR agonist and an ABCA1 upregulator, effectively ameliorates diabetic dyslipidemia in KKAy mice. • E17241 significantly lowers plasma fasting glucose levels and improves OGTT and ITT in KKAy mice. • E17241 improves islet function by promoting islet cholesterol efflux in an ABCA1-dependent way. • E17241 decreases liver lipid accumulation and eWAT mass by regulating PPAR target genes differently from rosiglitazone. • E17241 has cardiorenal benefits in KKAy mice. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
21. High dose rosuvastatin increases ABCA1 transporter in human atherosclerotic plaques in a cholesterol-independent fashion.
- Author
-
Santovito, Donato, Marcantonio, Pamela, Mastroiacovo, Daniela, Natarelli, Lucia, Mandolini, Claudia, De Nardis, Velia, Paganelli, Camilla, De Cesare, Domenico, Affaitati, Giannapia, Giamberardino, Maria Adele, Stellin, Luisa, Pinelli, Mauro, Weber, Christian, De Blasis, Giovanni, Occhiuzzi, Umberto, Bucci, Marco, Desideri, Giovambattista, and Cipollone, Francesco
- Subjects
- *
ATHEROSCLEROTIC plaque , *INTERNAL carotid artery - Abstract
ATP-binding cassette A1 (ABCA1) and G1 (ABCG1) mediate cholesterol efflux from lipid-laden macrophages, thus promoting anti-atherosclerotic outcomes. The mechanism(s) linking treatment with statins and ABCA1/ABCG1 in human atherosclerosis are not fully understood and require further investigation. Therefore, we studied whether short-term treatment with low- or high-dose rosuvastatin may affect ABCA1 and ABCG1 expression in human atherosclerotic plaques. Seventy patients with severe stenosis of the internal carotid artery were randomized to receive low (10 mg/day) or high (40 mg/day) dose rosuvastatin for 12 weeks before elective endarterectomy. As controls, we analyzed a reference group of 10 plaques from subjects with hypercholesterolemia but not receiving statin treatment and an additional set of 11 plaques collected from normocholesterolemic patients. On atherosclerotic plaques, ABCA1 and ABCG1 expression was evaluated at RNA level by qPCR and at protein level by immunoblotting and immunohistochemistry. Both rosuvastatin doses were associated with lower plaque ABCA1 mRNA levels and with a trend toward reduction for ABCG1. However, ABCA1 protein was paradoxically higher in patients treated with high-dose rosuvastatin and was associated with lower levels of miR-33b-5p, a microRNA known as a regulator of ABCA1. Multivariate analyses showed that the effect is cholesterol-independent. Finally, no effects were found for ABCG1 protein. High-dose rosuvastatin increases macrophage ABCA1 protein levels in human atherosclerotic plaque despite mRNA reduction in a mechanism unrelated to plasma cholesterol reduction and potentially involving miR-33b-5p. This pathway may reflect an additional feature contributing to the anti-atherosclerotic effect for high-dose rosuvastatin. Trial registration : ISRCTN16590640 • In atherosclerotic plaques, ABCA1 and ABCG1 mRNA are reduced by rosuvastatin treatment. • Despite mRNA reduction, high-dose rosuvastatin increased protein levels of ABCA1. • A similar trend was found for ABCG1 although not reaching statistical significance. • This effect doesn't appear to be related to reduction of plasma cholesterol. • This work highlights a potential additive atheroprotective mechanism of rosuvastatin. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
22. Associations between dietary vitamin intake, ABCA1 gene promoter DNA methylation, and lipid profiles in a Japanese population.
- Author
-
Fujii, Ryosuke, Yamada, Hiroya, Munetsuna, Eiji, Yamazaki, Mirai, Ando, Yoshitaka, Mizuno, Genki, Tsuboi, Yoshiki, Ohashi, Koji, Ishikawa, Hiroaki, Hagiwara, Chiharu, Maeda, Keisuke, Hashimoto, Shuji, and Suzuki, Koji
- Subjects
VITAMIN C metabolism ,VITAMIN A metabolism ,FOLIC acid metabolism ,VITAMIN D metabolism ,CARDIOVASCULAR diseases risk factors ,HIGH density lipoproteins ,INGESTION ,QUESTIONNAIRES ,VITAMIN E ,VITAMINS ,CROSS-sectional method ,BETA carotene ,DNA methylation ,SEQUENCE analysis ,MEMBRANE transport proteins - Abstract
Background Higher intake of fruits and vegetables is associated with reduced risk of specific types of cancer and of cardiovascular disease (CVD), but the protective role of the vitamins contained in fruits and vegetables on CVD is controversial. This discrepancy can raise the question of the effects of antioxidants in vitamins on CVD. Recently, we reported that higher vegetable intake was significantly associated with the decreased DNA methylation level of ATP-binding cassette transporter A1 (ABCA1), a gene associated with HDL-cholesterol metabolism. Objective We investigated whether ABCA1 DNA methylation mediates an effect of dietary vitamin intake on lipid profiles, an important risk factor for CVD, in a Japanese population. Methods A total of 225 individuals (108 men and 117 women) with no clinical history and no drug use for dyslipidemia participated in this cross-sectional study. We used the pyrosequencing method to measure the ABCA1 DNA methylation levels at 8 CpG sites, and we used mean DNA methylation level in statistical analysis. Dietary vitamin intake was assessed with the FFQ and adjusted for the residual method. Results In women, higher dietary vitamin intake [vitamin A, β-carotene, folic acid, vitamin C (VC), vitamin D, and vitamin E] was significantly associated with lower mean ABCA1 DNA methylation levels (P = 0.004, 0.03, 0.005, 0.001, 0.03, and 0.04, respectively). In addition, in women, we found a significant inverse association between mean ABCA1 DNA methylation and HDL cholesterol (P = 0.04) but not for other lipid indexes. Mediation analysis showed a significant indirect effect of VC intake on HDL cholesterol through ABCA1 DNA methylation level in women (P = 0.04). Conclusions Although this study does not prove causality, the results suggest that ABCA1 DNA methylation mediates the protective effect of VC on HDL cholesterol in women, which could offer a novel biological mechanism in CVD prevention. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
23. Astaxanthin exerts protective effects similar to bexarotene in Alzheimer's disease by modulating amyloid-beta and cholesterol homeostasis in blood-brain barrier endothelial cells.
- Author
-
Fanaee-Danesh, Elham, Gali, Chaitanya Chakravarthi, Tadic, Jelena, Zandl-Lang, Martina, Carmen Kober, Alexandra, Agujetas, Vicente Roca, de Dios, Cristina, Tam-Amersdorfer, Carmen, Stracke, Anika, Albrecher, Nicole Maria, Manavalan, Anil Paul Chirackal, Reiter, Marielies, Sun, Yidan, Colell, Anna, Madeo, Frank, Malle, Ernst, and Panzenboeck, Ute
- Subjects
- *
BLOOD-brain barrier , *ASTAXANTHIN , *ALZHEIMER'S disease , *ENDOTHELIAL cells , *RETINOID X receptors , *AMYLOID beta-protein precursor , *CHOLESTEROL - Abstract
The pathogenesis of Alzheimer's disease (AD) is characterized by overproduction, impaired clearance, and deposition of amyloid-β peptides (Aβ) and connected to cholesterol homeostasis. Since the blood-brain barrier (BBB) is involved in these processes, we investigated effects of the retinoid X receptor agonist, bexarotene (Bex), and the peroxisome proliferator-activated receptor α agonist and antioxidant, astaxanthin (Asx), on pathways of cellular cholesterol metabolism, amyloid precursor protein processing/Aβ production and transfer at the BBB in vitro using primary porcine brain capillary endothelial cells (pBCEC), and in 3xTg AD mice. Asx/Bex downregulated transcription/activity of amyloidogenic BACE1 and reduced Aβ oligomers and ~80 kDa intracellular 6E10-reactive APP/Aβ species, while upregulating non-amyloidogenic ADAM10 and soluble (s)APPα production in pBCEC. Asx/Bex enhanced Aβ clearance to the apical/plasma compartment of the in vitro BBB model. Asx/Bex increased expression levels of ABCA1 , LRP1 , and/or APOA-I. Asx/Bex promoted cholesterol efflux, partly via PPARα/RXR activation, while cholesterol biosynthesis/esterification was suppressed. Silencing of LRP-1 or inhibition of ABCA1 by probucol reversed Asx/Bex-mediated effects on levels of APP/Aβ species in pBCEC. Murine (m)BCEC isolated from 3xTg AD mice treated with Bex revealed elevated expression of APOE and ABCA1. Asx/Bex reduced BACE1 and increased LRP-1 expression in mBCEC from 3xTg AD mice when compared to vehicle-treated or non-Tg treated mice. In parallel, Asx/Bex reduced levels of Aβ oligomers in mBCEC and Aβ species in brain soluble and insoluble fractions of 3xTg AD mice. Our results suggest that both agonists exert beneficial effects at the BBB by balancing cholesterol homeostasis and enhancing clearance of Aβ from cerebrovascular endothelial cells. • Asx exerts beneficial effects similar to Bex in brain capillary endothelial cells. • Non-amyloidogenic APP processing is enhanced via BACE1 and ADAM10 modulation. • ABCA1 is upregulated and cellular cholesterol efflux is stimulated involving nuclear receptor-dependent mechanisms. • LRP-1 is up regulated along with augmented Aβ uptake/transcytosis. • Silencing of LRP-1 or inhibition of ABCA1 reverses effects of Asx and Bex on APP processing/Aβ species. • Asx and Bex considerably reduce cerebral and cerebrovascular Aβ load in vivo in 3xTg AD mice. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
24. Intracellular cholesterol stimulates ENaC by interacting with phosphatidylinositol‑4,5‑bisphosphate and mediates cyclosporine A-induced hypertension.
- Author
-
Zhai, Yu-Jia, Wu, Ming-Ming, Linck, Valerie A., Zou, Li, Yue, Qiang, Wei, Shi-Peng, Song, Chang, Zhang, Shuai, Williams, Clintoria R., Song, Bin-Lin, Zhang, Zhi-Ren, and Ma, He-Ping
- Subjects
- *
SYSTOLIC blood pressure , *ATP-binding cassette transporters , *CHOLESTEROL , *APOLIPOPROTEIN E , *BLOOD pressure , *SODIUM channels - Abstract
We have previously shown that blockade of ATP-binding cassette transporter A1 (ABCA1) with cyclosporine A (CsA) stimulates the epithelial sodium channel (ENaC) in cultured distal nephron cells. Here we show that CsA elevated systolic blood pressure in both wild-type and apolipoprotein E (ApoE) knockout (KO) mice to a similar level. The elevated systolic blood pressure was completely reversed by inhibition of cholesterol (Cho) synthesis with lovastatin. Inside-out patch-clamp data show that intracellular Cho stimulated ENaC in cultured distal nephron cells by interacting with phosphatidylinositol‑4,5‑bisphosphate (PIP 2), an ENaC activator. Confocal microscopy data show that both α‑ENaC and PIP 2 were localized in microvilli via a Cho-dependent mechanism. Deletion of membrane Cho reduced the levels of γ‑ENaC in the apical membrane. Reduced ABCA1 expression and elevated intracellular Cho were observed in old mice, compared to young mice. In parallel, cell-attached patch-clamp data from the split-open cortical collecting ducts (CCD) show that ENaC activity was significantly increased in old mice. These data suggest that elevation of intracellular Cho due to blockade of ABCA1 stimulates ENaC, which may contribute to CsA-induced hypertension. This study also implies that reduced ABCA1 expression may mediate age-related hypertension by increasing ENaC activity via elevation of intracellular Cho. • Increased intracellular cholesterol due to reduction of ABCA1 expression stimulates ENaC in aging mice • Increased intracellular cholesterol enhances ENaC activity by promoting the interaction between ENaC and PIP 2 • Pharmacological blockade of ABCA1 with cyclosporine A elevates the blood pressure via a Cholesterol-dependent mechanism. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
25. Cadmium induced redistribution of cholesterol by upregulating ABCA1 and downregulating OSBP.
- Author
-
Wang, Yanwei, Ji, Xingqi, Dai, Shuya, Liu, Haitao, Yan, Dandan, Zhou, Yang, Gu, Jie, and Shi, Haifeng
- Subjects
- *
PHYSIOLOGICAL effects of cadmium , *LIPID metabolism , *CHOLESTEROL , *ATP-binding cassette transporters , *OXYSTEROLS - Abstract
Abstract Human exposure to cadmium (Cd) could lead to alterations in lipid metabolism. However, the underlying mechanism is still unclear. In the present study, the data revealed that Cd exposure induced cholesterol redistribution both in vivo from mouse liver tissue into the serum, and in vitro from the HepG2 cells to the cultured medium, which were associated with modulating the expressions of cholesterol efflux proteins, including upregulating cholesterol exporter ATP-binding cassette transporter A1 (ABCA1) and downregulating oxysterol-binding protein (OSBP). Further investigation in HepG2 cells revealed that Cd upregulated ABCA1 expression with increased stability by inhibiting lysosomal pathway, and downregulated OSBP expression by increasing ubiquitination. Cd-induced cholesterol redistribution was completely inhibited by knockdown of ABCA1 expression using siRNA, and was significantly reduced by overexpression of OSBP. Taken together, these results suggested that Cd induced cholesterol redistribution by upregulating ABCA1 and downregulating OSBP. Graphical abstract Cadmium (Cd) induced cholesterol exporter ATP-binding cassette transporter A1 (ABCA1) accumulation through inhibiting lysosomal pathways by disrupting its acidic environment of lysosomes and autolysosomes and autophagic flux. And Cd also increased oxysterol-binding protein (OSBP) degradation by increasing ubiquitination. Cd induced ABCA1 accumulation and OSBP degradation facilitated cholesterol excretion. Unlabelled Image Highlights • Cd changed the cholesterol levels in vivo and in vitro. • Cd upregulated the expressions of ATP-binding cassette transporter A1 (ABCA1). • Cd downregulated oxysterol-binding protein (OSBP). • Cd-regulated ABCA1 and OSBP expression promoted cholesterol redistribution. • Cd-regulated ABCA1 and OSBP expression through lysosomal pathway and ubiquitination. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
26. Selective peroxisome proliferator-activated receptor-α modulator K-877 regulates the expression of ATP-binding cassette transporter A1 in pancreatic beta cells.
- Author
-
Dong, Tao, Lyu, Jingya, Imachi, Hitomi, Kobayashi, Toshihiro, Fukunaga, Kensaku, Sato, Seisuke, Ibata, Tomohiro, Yoshimoto, Takuo, Yonezaki, Kazuko, Murao, Koji, Iwama, Hisakazu, and Zhang, Guoxing
- Subjects
- *
PEROXISOME proliferator-activated receptors , *GENE expression , *CHOLESTEROL , *INSULIN , *ATP-binding cassette transporters - Abstract
Abstract ATP-binding cassette transporter A1 (ABCA1) protein is a pivotal regulator of cholesterol and phospholipid efflux from cells to high-density lipoprotein (HDL) particles. Pancreatic ABCA1 functions in beta cell cholesterol homeostasis and affects insulin secretion. We investigated the effect of pemafibrate (K-877), a novel selective PPARα modulator (SPPARMα), on pancreatic ABCA1 expression. In vivo experiment , mice were divided into four treatment groups, namely, normal food plus placebo, high fat diet (HFD) plus placebo, normal food plus K-877 (0.3 mg/kg/day), or HFD plus K-877 (0.3 mg/kg/day), and treated for eight weeks. The results in vitro experiment indicate that K-877 treatment increased levels of ABCA1 mRNA, as well as protein, subsequently reduced the cellular cholesterol content in INS-1 cells. PPARα specific antagonist GW6471 attenuate K-877 induced ABCA1 expression in INS-1 cells. ABCA1 promoter activity increased with K-877 treatment at concentration 1 μM and 10 μM. Glucose-stimulated insulin secretion was ameliorated by K-877 treatment in INS-1 cells and isolated mouse islets. Although the expression of ABCA1 was reduced in mice with HFD treatment, both ABCA1 protein and mRNA levels were increased in mice with K-877 treatment. K-877 treatment improved glucose intolerance induced by HFD in mice. These findings raise the possibility that K-877 may affect insulin secretion by controlling ABCA1 expression in pancreatic beta cells. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
27. miR-143 and miR-145 promote hypoxia-induced proliferation and migration of pulmonary arterial smooth muscle cells through regulating ABCA1 expression.
- Author
-
Yue, Yuxia, Zhang, Zhiyong, Zhang, Lei, Chen, Songhu, Guo, Yilin, and Hong, Yan
- Subjects
- *
HYPOXEMIA , *MUSCLE cells , *MICRORNA , *PULMONARY artery , *CELL migration - Abstract
Abstract Background Excessive proliferation and migration of pulmonary artery smooth muscle cells (PASMCs) play an important role in the occurrence and development of pulmonary arterial hypertension (PAH). miR-143/145 was reported to be up-regulated in the animal models and PAH patients, and deletion of miR-143/145 cluster prevented the development of hypoxia-induced pulmonary hypertension, but its underlying mechanism has not been elucidated. Methods qRT-PCR and Western blot were performed to detect the expressions of miR-143/145 and ATP-binding cassette transporter A1 (ABCA1) in PAH patients and PASMCs under hypoxic conditions. Cell proliferation and migration were assessed by Cell Counting Kit-8 and wound-healing assay, respectively. Luciferase reporter assay and RNA immunoprecipitation were conducted to confirm the interaction between miR-143/145 and ABCA1. Hypoxia-induced PAH rat model was established to confirm the functions of miR-143/145 in the pathogenesis of PAH and its underlying mechanism in vivo. Results miR-143 and miR-145 were up-regulated and ABCA1 was down-regulated in PAH patients and PASMCs under hypoxic conditions for different time, as well as hypoxia-induced PAH rats. miR-143/145 inhibition and ABCA1 overexpression suppressed hypoxia-induced proliferation and migration in PASMCs. ABCA1 was identified as a direct target of miR-143/145 in PASMCs. Moreover, ABCA1 partially reversed miR-143/145-mediated promotion of hypoxia-induced cell proliferation and migration in PASMCs. Furthermore, in vivo experiments confirmed that inhibition of miR-143/145 prevents hypoxia-induced pulmonary hypertension and pulmonary vascular remodeling by targeting ABCA1. Conclusion miR-143/145 promoted hypoxia-induced proliferation and migration of PASMCs by targeting ABCA1, contributing to better understanding of the mechanism underlying the pathogenesis of PAH. Highlights • miR-143 and miR-145 were significantly up-regulated in PAH patients and hypoxia-treated PASMCs. • Inhibition of miR-143/145 suppressed hypoxia-induced proliferation and migration in PASMCs. • ABCA1 was a direct target of miR-143/145 in PASMCs. • ABCA1 overexpression impeded hypoxia-induced proliferation and migration in PASMCs. • ABCA1 partially reversed miR-143/145-mediated promotion of cell proliferation and migration in PASMCs exposed to hypoxia. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
28. New roles of reactive astrocytes in the brain; an organizer of cerebral ischemia.
- Author
-
Koizumi, Schuichi, Hirayama, Yuri, and Morizawa, Yosuke M.
- Subjects
- *
CEREBRAL ischemia , *ASTROCYTES , *NEUROGLIA , *NEUROPROTECTIVE agents , *BRAIN physiology , *SYNAPSES - Abstract
The brain consists of neurons and much higher number of glial cells. They communicate each other, by which they control brain functions. The brain is highly vulnerable to several insults such as ischemia, but has a self-protective and self-repairing mechanisms against these. Ischemic tolerance or preconditioning is an endogenous neuroprotective phenomenon, where a mild non-lethal ischemic episode can induce resistance to a subsequent severe ischemic injury in the brain. Because of its neuroprotective effects against cerebral ischemia or stroke, ischemic tolerance has been widely studied. However, almost all studies have been performed from the viewpoint of neurons. Glial cells are structurally in close association with synapses. Recent studies have uncovered the active roles of astrocytes in modulating synaptic connectivity, such as synapse formation, elimination and maturation, during development or pathology. However, glia-mediated ischemic tolerance and/or neuronal repairing have received only limited attention. We and others have demonstrated that glial cells, especially astrocytes, play a pivotal role in regulation of induction of ischemic tolerance as well as repairing/remodeling of neuronal networks by phagocytosis. Here, we review our current understanding of (1) glial-mediated ischemic tolerance and (2) glia-mediated repairing/remodeling of the penumbra neuronal networks, and highlight their mechanisms as well as their potential benefits, problems, and therapeutic application. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
29. Impairment of trophoblast survival and differentiation by LXR ligands is prevented by cholesterol but not ABCA1 silencing.
- Author
-
Harmon, C. Miles, McGonigal, Stacy, and Larkin, Jacob C.
- Abstract
Introduction: The Liver X Receptors (LXRs) drive the transcriptional response to excess intracellular cholesterol. Oxysterols, the products of cholesterol oxidation, are activating ligands for LXR that can accumulate under conditions of oxidative stress and disrupt cholesterol homeostasis. While activation of LXR inhibits trophoblast differentiation, the impact of LXR on trophoblast physiology or cholesterol homeostasis is incompletely understood. We sought to determine if the effects of LXR activation can be ameliorated through modification of cholesterol bioavailability or inhibition of LXR-driven cholesterol efflux in trophoblasts.Methods: We measured the effect of oxysterol exposure on BeWo cells and primary human trophoblasts (PHT cells) cultured in lipoprotein-deficient medium. We also measured the effect of the synthetic, LXR-specific ligand T0901317 on PHT cell differentiation and survival. Finally, we silenced the ATP-binding cassette transporter A1 (ABCA1), a transcriptional target of LXR that drives cholesterol efflux, to determine if inhibition of cholesterol efflux could block the effects of T0901317.Results: Oxysterols inhibited BeWo survival and PHT cell differentiation, and these effects were blocked by cholesterol supplementation. T0901317 also inhibited PHT cell differentiation, and this effect was similarly blocked by cholesterol. Unlike cholesterol however, ABCA1 silencing did not modify the effect of T0901317 on PHT cell differentiation.Discussion: Oxysterols and LXR inhibit trophoblast survival and differentiation exclusively in conditions of cholesterol scarcity. These findings underscore the importance of cholesterol homeostasis in the maintenance of placental function and suggest that pathways regulating cholesterol homeostasis may represent therapeutic targets to mitigate harmful sequelae of placental injury. [ABSTRACT FROM AUTHOR]- Published
- 2018
- Full Text
- View/download PDF
30. Heat shock protein 70 accelerates atherosclerosis by downregulating the expression of ABCA1 and ABCG1 through the JNK/Elk-1 pathway.
- Author
-
Zhao, Zhen-Wang, Zhang, Min, Chen, Ling-Yan, Gong, Duo, Xia, Xiao-Dan, Yu, Xiao-Hua, Wang, Si-Qi, Ou, Xiang, Dai, Xiao-Yan, Zheng, Xi-Long, Zhang, Da-Wei, and Tang, Chao-Ke
- Subjects
- *
ATHEROSCLEROSIS , *HSP70 heat-shock proteins , *C-Jun N-terminal kinases , *IMMUNOPRECIPITATION , *CHOLESTEROL - Abstract
Background and aims Recent studies have suggested that heat shock protein 70 (HSP70) may play critical roles in cardiovascular disease. However, the effects of HSP70 on the development of atherosclerosis in apoE −/− mice remain largely unknown. This study was to investigate the role and potential mechanism of HSP70 in atherosclerosis. Methods HSP70 was overexpressed in apoE −/− mice and THP-1-derived macrophages with lentiviral vectors. Oil Red O, hematoxylin-eosin, and Masson staining were performed to evaluate atherosclerotic plaque in apoE −/− mice fed the Western type diet. Moreover, immunostaining was employed to detect the expression of relative proteins in aortic sinus. Reporter gene and chromatin immunoprecipitation were performed to analyze the effect of Elk-1 on the promoter activity of ABCA1 and ABCG1; [ 3 H] labeled cholesterol was used to assess the capacity of cholesterol efflux and reverse cholesterol transport (RCT). Results Our results showed that HSP70 increased lipid accumulation in arteries and promoted the formation of atherosclerotic lesion. The capacity of cholesterol efflux was reduced in peritoneal macrophages isolated from HSP70-overexpressed apoE −/− mice. The levels of ABCA1 and ABCG1 expression were also reduced in the peritoneal macrophages and the aorta from apoE −/− mice in response to HSP70. The c-Jun N-terminal kinase (JNK) and ETS transcription factor (Elk-1) played a critical role in HSP70-induced downregulation ABCA1 and ABCG1. Further, HSP70 reduced RCT from macrophages to plasma, liver, and feces in apoE −/− mice. Conclusions HSP70 promotes the progression of atherosclerosis in apoE −/− mice by suppressing the expression of ABCA1 and ABCG1 through the JNK/Elk-1 pathway. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
31. SIRT1 activator E1231 protects from experimental atherosclerosis and lowers plasma cholesterol and triglycerides by enhancing ABCA1 expression.
- Author
-
Feng, Tingting, Liu, Peng, Wang, Xiao, Luo, Jinque, Zuo, Xuan, Jiang, Xinhai, Liu, Chang, Li, Yongzhen, Li, Ni, Chen, Minghua, Zhu, Ningyu, Han, Xiaowan, Liu, Chao, Xu, Yanni, and Si, Shuyi
- Subjects
- *
SIRTUINS , *DEACETYLASES , *LOW density lipoproteins , *SURFACE plasmon resonance , *CHOLESTEROL - Abstract
Background and aims Sirtuin 1 (SIRT1) is a nicotinamide adenine dinucleotide-dependent protein deacetylase. Recent studies have demonstrated that enhancing SIRT1 expression or activity may modulate cholesterol and lipid metabolism. However, pharmacological and molecular regulators for SIRT1 are scarce. Here, we aimed to find novel small molecule modulators of SIRT1 to regulate cholesterol and lipid metabolism. Methods A high-throughput screening assay was established to identify SIRT1 activators. Surface plasmon resonance and immunoprecipitation were performed to confirm the interaction of E1231 with SIRT1. Cholesterol assay was performed to demonstrate the in vitro effect of E1231. The in vivo effect of E1231 was evaluated in experimental models. Results E1231, a piperazine 1,4-diamide compound, was identified as a SIRT1 activator with EC 50 value of 0.83 μM. E1231 interacted with recombinant human SIRT1 protein and deacetylated liver X receptor-alpha (LXRα). E1231 increased ATP-binding cassette transporter A1 (ABCA1) expression in RAW 264.7 cells dependent on SIRT1 and LXRα. E1231 promoted cholesterol efflux and inhibited lipid accumulation in RAW 264.7 cells via SIRT1 and ABCA1. In the golden hamster hyperlipidemia model, E1231 treatment decreased total cholesterol and triglyceride levels in both serum and the liver, while increased cholesterol content in feces. Moreover, E1231 increased ABCA1 and SIRT1 protein expression in the liver. In ApoE −/− mice, E1231 treatment reduced atherosclerotic plaque development compared with untreated ApoE −/− mice. Conclusions We identified a novel SIRT1 activator E1231 and elucidated its beneficial effects on lipid and cholesterol metabolism. Our study suggests that E1231 might be developed as a novel drug for treating atherosclerosis. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
32. The E3 ubiquitin ligase, HECTD1, is involved in ABCA1-mediated cholesterol export from macrophages.
- Author
-
Aleidi, Shereen M., Yang, Alryel, Sharpe, Laura J., Rao, Geetha, Cochran, Blake J., Rye, Kerry-Anne, Kockx, Maaike, Brown, Andrew J., and Gelissen, Ingrid C.
- Subjects
- *
UBIQUITIN ligases , *CHOLESTEROL , *MACROPHAGES , *CELL growth , *LIGASES - Abstract
The ABC lipid transporters, ABCA1 and ABCG1, are essential for maintaining lipid homeostasis in cells such as macrophages by exporting excess cholesterol to extracellular acceptors. These transporters are highly regulated at the post-translational level, including protein ubiquitination. Our aim was to investigate the role of the E3 ubiquitin ligase HECTD1, recently identified as associated with ABCG1, on ABCG1 and ABCA1 protein levels and cholesterol export function. Here, we show that HECTD1 protein is widely expressed in a range of human and murine primary cells and cell lines, including macrophages, neuronal cells and insulin secreting β-cells. siRNA knockdown of HECTD1 unexpectedly decreased overexpressed ABCG1 protein levels and cell growth, but increased native ABCA1 protein in CHO-K1 cells. Knockdown of HECTD1 in unloaded THP-1 macrophages did not affect ABCG1 but significantly increased ABCA1 protein levels, in wild-type as well as THP-1 cells that do not express ABCG1. Cholesterol export from macrophages to apoA-I over time was increased after knockdown of HECTD1, however these effects were not sustained in cholesterol-loaded cells. In conclusion, we have identified a new candidate, the E3 ubiquitin ligase HECTD1, that may be involved in the regulation of ABCA1-mediated cholesterol export from unloaded macrophages to apoA-I. The exact mechanism by which this ligase affects this pathway remains to be elucidated. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
33. Curcumin enhances LXRα in an AMP-activated protein kinase-dependent manner in human macrophages.
- Author
-
Sáenz, Javier, Alba, Gonzalo, Reyes-Quiroz, María E., Geniz, Isabel, Jiménez, Juan, Sobrino, Francisco, and Santa-María, Consuelo
- Subjects
- *
CURCUMIN , *NUCLEAR receptors (Biochemistry) , *PROTEIN expression , *PROTEIN genetics , *MACROPHAGES - Abstract
Liver X receptor alpha (LXRα) is a nuclear receptor involved in cholesterol homeostasis. Curcumin, a traditional Chinese derivative from the rhizomes of Curcuma longa and a well-known AMP-activated protein kinase (AMPK) activator, possess hypocholesterolemic activity, however, the possible link between AMPK and cholesterol is unknown. In this study, we have investigated whether curcumin regulates metabolic changes in cholesterol metabolism via LXRα in THP-1 human macrophages, the cells implicated in atheroma plaques formation. Results showed that curcumin induced AMPK phosphorylation, increased LXRα mRNA and protein expression. Curcumin up-regulated mRNA expression of genes involved in cholesterol transport and metabolism as ATP-binding cassette (ABC) transporters ABCA1 and ABCG1, and the sterol response element binding protein 1c (SREBP1c). On the other hand, this increased LXRα mRNA and protein expression was reverted when AMPK was inhibited by its chemical inhibitor, compound C. Transfection with AMPK α1 and α2 siRNA decreased the LXRα mRNA expression and its target genes. Curcumin treatment inhibited cell migration and was also able to promote reverse cholesterol transport in THP-1 cells. This enhanced reverse cholesterol transport might be related to the up-regulating of ABCA1 and ABCG1 mRNA expression by activating AMPK-LXRα signaling in THP-1 cells. This study describes a possible mechanism for understanding the hypocholesterolemic effects of curcumin and expand knowledge about the LXRα regulation by AMPK. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
34. Tanshindiol C inhibits oxidized low-density lipoprotein induced macrophage foam cell formation via a peroxiredoxin 1 dependent pathway.
- Author
-
Yang, Yuyu, Li, Xueyan, Peng, Liying, An, Lin, Sun, Ningyuan, Hu, Xuewen, Zhou, Ping, Xu, Yong, Li, Ping, and Chen, Jun
- Subjects
- *
LOW density lipoproteins , *MACROPHAGES , *PEROXIREDOXINS , *OXIDATION , *ATHEROSCLEROSIS - Abstract
NF-E2-related factor 2 (Nrf2) has been shown to be protective in atherosclerosis. The loss of Nrf2 in macrophages enhances foam cell formation and promotes early atherogenesis. Tanshindiol C (Tan C) is isolated from the root of Salvia miltiorrhiza Bge., a traditional Chinese medicine that has been used for the treatment of several cardiovascular diseases for many years. This study was aimed to test the potential role of Tan C against macrophage foam cell formation and to explore the underlying mechanism. Firstly, we observed that Tan C markedly suppressed oxidized low-density lipoprotein (oxLDL) induced macrophage foam cell formation. Then, we found that Tan C was an activator of both Nrf2 and Sirtuin 1 (Sirt1) in macrophages. Nrf2 and Sirt1 synergistically activated the transcription of anti-oxidant peroxiredoxin 1 (Prdx1) after Tan C treatment. More important, we demonstrated that silencing of Prdx1 promoted oxLDL-induced macrophage foam cell formation. Prdx1 upregulated adenosine triphosphate-binding cassette (ABC) transporter A1 (ABCA1) expression and decreased intracellular lipid accumulation. Furthermore, Tan C ameliorated oxLDL induced macrophage foam cell formation in a Prdx1-dependent manner. These observations suggest that Tan C protects macrophages from oxLDL induced foam cell formation via activation of Prdx1/ABCA1 signaling and that Prdx1 may be a novel target for therapeutic intervention of atherosclerosis. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
35. Global gene expression analysis reveals a subtle effect of DEHP in human granulosa cell line HGrC1.
- Author
-
Samardzija Nenadov, Dragana, Tesic, Biljana, Tomanic, Tamara, Opacic, Marija, Stanic, Bojana, Pogrmic-Majkic, Kristina, and Andric, Nebojsa
- Subjects
- *
GRANULOSA cells , *GENE expression , *CELL lines , *ENDOCRINE disruptors , *PROGESTERONE , *PROGESTERONE receptors - Abstract
Di(2-ethylhexyl) phthalate (DEHP) is an endocrine disruptor that exerts anti-steroidogenic effects in human granulosa cells; however, the extent of this effect depends on the concentration of DEHP and granulosa cell models used for exposure. The objective of this study was to identify the effects of low- and high-dose DEHP exposure in human granulosa cells. We exposed human granulosa cell line HGrC1 to 3 nM and 25 μM DEHP for 48 h. The whole genome transcriptome was analyzed using the DNBSEQ sequencing platform and bioinformatics tools. The results revealed that 3 nM DEHP did not affect global gene expression, whereas 25 µM DEHP affected the expression of only nine genes in HGrC1 cells: ABCA1 , SREBF1 , MYLIP , TUBB3 , CENPT , NUPR1 , ASS1 , PCK2 , and CTSD. We confirmed the downregulation of ABCA1 mRNA and SREBP-1 protein (encoded by the SREBF1 gene), both involved in cholesterol homeostasis. Despite these changes, progesterone production remained unaffected in low- and high-dose DEHP-exposed HGrC1 cells. The high concentration of DEHP decreased the levels of ABC1A mRNA and SREBP-1 protein and prevented the upregulation of STAR, a protein involved in progesterone synthesis, in forskolin-stimulated HGrC1 cells; however, the observed changes were not sufficient to alter progesterone production in forskolin-stimulated HGrC1 cells. Overall, this study suggests that acute exposure to low concentration of DEHP does not compromise the function of HGrC1 cells, whereas high concentration causes only subtle effects. The identified nine novel targets of high-dose DEHP require further investigation to determine their role and importance in DEHP-exposed human granulosa cells. • 3 nM DEHP does not affect global gene expression in human granulosa cell line HGrC1. • 25 µM DEHP affects mRNA expression of nine genes in HGrC1 cells. • 25 µM DEHP decreases ABCA1 and SREBF1, two genes involved in cholesterol homeostasis. • 25 µM DEHP decreases ABCA1, SREBF1, and STAR in forskolin-stimulated HGrC1 cells. • 3 nM and 25 µM DEHP do not change progesterone levels in HGrC1 cells. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
36. Bisphenol A decreases progesterone synthesis by disrupting cholesterol homeostasis in rat granulosa cells.
- Author
-
Samardzija, Dragana, Pogrmic-Majkic, Kristina, Fa, Svetlana, Stanic, Bojana, Jasnic, Jovana, and Andric, Nebojsa
- Subjects
- *
BISPHENOL A , *PROGESTERONE , *CHOLESTEROL , *HOMEOSTASIS , *GRANULOSA cells - Abstract
Bisphenol A (BPA) is an endocrine disruptor used in a variety of consumer products. Exposure to BPA leads to alterations in steroidogenesis of ovarian granulosa cells. Here, we analyzed the mechanism by which BPA alters progesterone biosynthesis in immature rat granulosa cells. BPA increased expression of steroidogenic acute regulatory protein (StAR), cholesterol side-chain cleavage enzyme and 3β-hydroxysteroid dehydrogenase in granulosa cells; however, BPA prevented the basal and the FSH-induced progesterone production. BPA caused sequestration of cholesterol to the perinuclear area, as evident by the Filipin staining. BPA decreased mRNA expression of ATP binding cassette transporter-A1 ( Abca1 ) and increased level of sterol regulatory element binding protein 1. Addition of exogenous cell-permeable cholesterol restored the effect of BPA on Abca1 and Star mRNA expression and partially reversed BPA's effect on progesterone production. These results indicate that exposure to BPA disrupts cholesterol homeostasis leading to decreased progesterone production in immature rat granulosa cells. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
37. LXR activation increases the expression of GnRH AND αMSH in the rat hypothalamus in vivo.
- Author
-
Kruse, María Sol, Suarez, Lucas Gabriel, and Coirini, Héctor
- Subjects
- *
GONADOTROPIN releasing hormone , *MSH (Hormone) , *TRANSCRIPTION factors , *HYPOTHALAMUS physiology , *LIPID metabolism , *PHYSIOLOGICAL effects of carbohydrates , *LABORATORY rats - Abstract
Liver X receptors (LXR) are important transcription factors involved in the regulation of carbohydrate and lipid metabolism. Recently, we described LXR receptors expression in the hypothalamus but their function in this brain area remains unknown. Here, we evaluated the function of LXR on the expression of factors produced in the hypothalamus in vitro and in vivo by Western blotting and immunocytochemistry. More precisely we studied the expression of GnRH and GHRH, αMSH and NPY in male Sprague-Dawley rats. The effects of two synthetic LXR agonists, T0901317 and GW3965, were first tested in vitro . Hypothalamic explants were treated with either T0901317 or GW3965 (10 μM) for 2, 4, 6 and 8 h. As a positive control the cholesterol ABCA1 and glucose GLUT2 transporters were used. No changes were observed in the expression of the factors evaluated in vitro . The effects of the LXR agonists were then tested in vivo . Rats were injected ICV into the third ventricle with either T0901317 or GW3965 (2.5 μg/5 μL ICV) and after 3.5 h or 24 h the hypothalami were dissected out and rapidly frozen for analysis. αMSH and GnRH expression was significantly increased after 3.5 h of T0901317 treatment. Anterior/posterior hypothalamic ratio increases for αMSH expression and decreases for GnRH expression after 24 h of LXR activation. Altogether these results show that LXR activation affects the expression of GnRH and αMSH, suggesting that LXR in the hypothalamus is capable of modulating hypothalamic responses related to appetite, sexual behavior and reproductive functions. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
38. Discovery of molecular mechanism of a clinical herbal formula upregulating serum HDL-c levels in treatment of metabolic syndrome by in vivo and computational studies.
- Author
-
Chen, Meimei, Yang, Fafu, Kang, Jie, Gan, Huijuan, Lai, Xinmei, and Gao, Yuxing
- Subjects
- *
HERBAL medicine , *HIGH density lipoproteins , *METABOLIC syndrome treatment , *BLOOD serum analysis , *CARDIOVASCULAR disease treatment - Abstract
Decreased HDL cholesterol (HDL-c) is considered as an independent risk factor of cardiovascular disease in metabolic syndrome (Mets). Wendan decoction (WDD), a famous clinical traditional Chinese medicine formula in Mets in China, which can obviously up-regulate serum HDL-c levels in Mets. However, till now, the molecular mechanism of up-regulation still remained unclear. In this study, an integrated approach that combined serum ABCA1 in vivo assay, QSAR modeling and molecular docking was developed to explore the molecular mechanism and chemical substance basis of WDD upregulating HDL-c levels. Compared with Mets model group, serum ABCA1 and HDL-c levels intervened by two different doses of WDD for two weeks were significantly up-regulated. Then, kohonen and LDA were applied to develop QSAR models for ABCA1 up-regulators based flavonoids. The derived QSAR model produced the overall accuracy of 100%, a very powerful tool for screening ABCA1 up-regulators. The QSAR model prediction revealed 67 flavonoids in WDD were ABCA1 up-regulators. Finally, they were subjected to the molecular docking to understand their roles in up-regulating ABCA1 expression, which led to discovery of 23 ABCA1 up-regulators targeting LXR beta. Overall, QSAR modeling and docking studies well accounted for the observed in vivo activities of ABCA1 affected by WDD. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
39. AGE-albumin enhances ABCA1 degradation by ubiquitin-proteasome and lysosomal pathways in macrophages.
- Author
-
Iborra, Rodrigo Tallada, Machado-Lima, Adriana, Okuda, Ligia Shimabukuro, Pinto, Paula Ramos, Nakandakare, Edna Regina, Machado, Ubiratan Fabres, Correa-Giannella, Maria Lucia, Pickford, Russell, Woods, Tom, Brimble, Margaret A., Rye, Kerry-Anne, Lu, Rui, Yokoyama, Shinji, and Passarelli, Marisa
- Abstract
Background and Aims: Advanced glycation end products (AGEs) induce cellular oxidative/endoplasmic reticulum stress and inflammation. We investigated its underlying mechanisms for atherogenesis focusing on regulation of ABCA1 protein decay in macrophages.Methods: The ABCA1 decay rate was evaluated in macrophages after treatment with LXR agonist and by incubation with control (C) or AGE-albumin concomitant or not with cycloheximide, MG-132, ammonium chloride and calpain inhibitors were utilized to inhibit, respectively, proteasome, lysosome and ABCA1 proteolysis at cell surface. ABCA1 was determined by immunoblot and the protein decay rate calculated along time by the slope of the linear regression. Ubiquitination level was determined in ABCA1 immunoprecipitated from whole cell lysate or bulk cell membrane. AGE effect was also analyzed in THP-1 cells transfected with siRNA-RAGE. Carboxymethyllysine (CML) and pyrraline (PYR) were determined by LC/MS. One-way ANOVA and Student t test were utilized to compare results.Results: CML and PYR-albumin were higher in AGE-albumin as compared to C. AGE-albumin reduced ABCA1 in J774 and THP-1 macrophages (20-30%) and induced a higher ABCA1 ubiquitination and a faster protein decay rate that was dependent on the presence of AGE during the kinetics of measurement in the presence of cycloheximide. Proteasomal inhibition restored and lysosomal inhibition partially recovered ABCA1 in cells treated with AGE-albumin. Calpain inhibition was not able to rescue ABCA1. RAGE knockdown prevented the reduction in ABCA1 elicited by AGE.Conclusions: AGE-albumin diminishes ABCA1 by accelerating its degradation through the proteasomal and lysosomal systems. This may increase lipid accumulation in macrophages by diminishing cholesterol efflux via RAGE signaling contributing to atherosclerosis in diabetes mellitus. [ABSTRACT FROM AUTHOR]- Published
- 2018
- Full Text
- View/download PDF
40. Increased prevalence of clinical and subclinical atherosclerosis in patients with damaging mutations in ABCA1 or APOA1.
- Author
-
Abdel-Razek, Omar, Sadananda, Singh N., Li, Xuan, Cermakova, Lubomira, Frohlich, Jiri, and Brunham, Liam R.
- Subjects
ATHEROSCLEROSIS ,CARRIER proteins ,CHOLESTEROL ,COMPARATIVE studies ,HIGH density lipoproteins ,GENETIC mutation ,DISEASE prevalence ,DESCRIPTIVE statistics ,SEQUENCE analysis - Abstract
Background A low level of high-density lipoprotein cholesterol (HDL-C) is a common clinical scenario and poses challenges for management. Many patients with low HDL-C harbor a damaging mutation in ABCA1 or APOA1 , but the clinical implications of genetic testing for these mutations are unclear. Objective The purpose of this study was to investigate the prevalence of clinical or subclinical atherosclerosis among patients with low HDL-C due to a mutation in ABCA1 or APOA1 , compared with patients with low HDL-C without such a mutation. Methods We performed targeted next-generation sequencing to identify mutations in ABCA1 and APOA1 in 72 patients with HDL-C levels below the 10 th percentile. We examined the prevalence of clinical atherosclerosis and subclinical atherosclerosis in these patients. We also measured cholesterol efflux capacity (CEC) in plasma. Results We identified a known disease-causing or likely pathogenic variant in the ABCA1 or APOA1 genes in 22% of patients with low HDL-C. Eighty-three percent of patients with a damaging mutation in ABCA1 or APOA1 had evidence of atherosclerosis compared with 38.6% with low HDL-C without such a mutation ( P = .04). Patients with damaging mutations in ABCA1 or APOA1 had lower CEC compared with patients without a mutation (25.9% vs 30.1%). Conclusion The presence of a damaging mutation in ABCA1 or APOA1 confers an increased risk of atherosclerosis relative to patients without such a mutation at a comparable level of HDL cholesterol, possibly because of a reduction in CEC. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
41. ABCA1 affects placental function via trophoblast and macrophage.
- Author
-
Chengmao, Xie, Li, Lin, Yan, Long, Jie, Yang, Xiaoju, Wang, Xiaohui, Cai, and Huimin, Guan
- Subjects
- *
ATP-binding cassette transporters , *PLACENTA physiology , *TROPHOBLAST , *MACROPHAGES , *IMMUNOFLUORESCENCE - Abstract
Aims To study the potential impact of ABCA1 on the function of the placenta. Main methods Trophoblasts and macrophages were isolated from the placenta with enzymatic digestion; Immunofluorescence assay was used to detect the location of ABCA1 in cells; RT-PCR and Western-blot were used to detect the expression of ABCA1; The cholesterol efflux assays of primary trophoblasts was detected by Amplex Red cholesterol assay kit (Invitrogen);Inflammatory factor secretion from primary macrophages was detected by Elisa. Key findings ABCA1 was mainly located on trophoblast membranes. Decreased ABCA1 expression in trophoblasts reduced the cholesterol efflux of trophoblasts (P < 0.01). while increased ABCA1 expression in trophoblasts reduced the cholesterol efflux of trophoblasts (P < 0.05). ABCA1 was uniformly expressed on the cell membrane, cytoplasm, and nucleus of macrophages. Decreased ABCA1 expression in macrophages, increased inflammatory factors but reduced IL-10 (P < 0.01). While increased ABCA1 expression in macrophages, reduced inflammatory factors but increased IL-10 (P < 0.01). Significance ABCA1 may be a potential target for the prevention of gestation diseases. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
42. Doxorubicin enhances oxysterol levels resulting in a LXR-mediated upregulation of cardiac cholesterol transporters.
- Author
-
Monzel, Judith V., Budde, Thomas, Meyer zu Schwabedissen, Henriette E., Schwebe, Matthias, Bien-Möller, Sandra, Lütjohann, Dieter, Kroemer, Heyo K., Jedlitschky, Gabriele, and Grube, Markus
- Subjects
- *
DOXORUBICIN , *OXYSTEROLS , *CHOLESTEROL metabolism , *HEART cells , *TRANSFORMING growth factors - Abstract
The anthracycline-mediated cardiotoxicity is still not completely understood. To examine the impact of cholesterol metabolism and transport in this context, cholesterol and oxysterol levels as well as the expression of the cholesterol transporters ABCA1 and ABCG1 were analyzed in doxorubicin-treated HL-1 murine cardiomyocytes as well as in mouse model for acute doxorubicin-induced cardiotoxicity. Doxorubicin-treated HL-1 cells exhibited enhanced cholesterol (153 ± 20% of control), oxysterol (24 S-hydroxycholesterol: 206 ± 29% of control) and cholesterol precursor levels (lathosterol: 122 ± 12% of control; desmosterol: 188 ± 10% of control) indicating enhanced cholesterol synthesis. Moreover, abca1 and abcg1 were upregulated on mRNA, protein and functional level caused by a doxorubicin-mediated activation of the nuclear receptor LXR. In addition, the oxysterols not only induced the abca1 and abcg1 in HL-1 cells but also enhanced the expression of endothelin-1 and transforming growth factor-β, which have already been identified as important factors in doxorubicin-induced cardiotoxicity. These in vitro findings were verified in a murine model for acute doxorubicin-induced cardiotoxicity, demonstrating elevated cardiac (2.1 ± 0.2 vs. 3.6 ± 1.0 ng/mg) and systemic cholesterol levels (105.0 ± 8.4 vs. 130.0 ± 4.3 mg/dl), respectively, as well as enhanced oxysterol levels such as cardiac 24 S-hydroxycholesterol (2.1 ± 0.2 vs. 3.6 ± 1.0 ng/mg). In line with these findings cardiac mRNA expression of abca1 (303% of control) and abcg1 (161% of control) was induced. Taken together, our data demonstrate enhanced cholesterol and oxysterol levels by doxorubicin, resulting in a LXR-dependent upregulation of abca1 and abcg1. In this context, the cytotoxic effects of oxysterols and their impact on cardiac gene expression should be considered as an important factor in doxorubicin-induced cardiotoxicity. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
43. ABCA1 and HDL3 are required to modulate smooth muscle cells phenotypic switch after cholesterol loading.
- Author
-
Castiglioni, Silvia, Monti, Matteo, Arnaboldi, Lorenzo, Canavesi, Monica, Ainis Buscherini, Giuditta, Calabresi, Laura, Corsini, Alberto, and Bellosta, Stefano
- Subjects
- *
SMOOTH muscle physiology , *HIGH-density lipoprotein receptors , *PHENOTYPES , *PHYSIOLOGICAL effects of cholesterol , *CALPONIN , *DOWNREGULATION - Abstract
Background and aims Cholesterol-loaded smooth muscle cells (SMCs) modify their phenotypic behavior becoming foam cells. To characterize the role of ABCA1 and HDL 3 in this process, we evaluated HDL 3 effects on cholesterol-induced phenotypic changes in SMCs expressing or not ABCA1. Methods SMCs, isolated from the aortae of wild-type (WT) and Abca1 knock-out (KO) mice, were cholesterol-loaded using a “water-soluble cholesterol’’. Results Cholesterol loading downregulates the expression of Acta 2 and calponin (SMC markers), and increases the expression of Mac- 2, CD11b and MHCII (inflammation-related genes and surface antigens) and Abca1, Abcg 1. HDL 3 normalizes SMC marker expression and reduces the expression of inflammation-related genes/proteins in WT cells, an effect not observed with free apoA-I. The effect of HDL 3 is almost lost in Abca1 KO cells, as well as when Abca1 is silenced in WT SMC. HDL 3 does not differently affect cholesterol downloading in WT or KO cells and stimulates phospholipids removal in WT cells. Similarly, the expression of myocardin and its modulators, such as miR-143/145, is reduced by cholesterol loading in WT and Abca1 KO SMCs; HDL 3 normalizes their levels in WT cells but not in KO cells. On the contrary, cholesterol loading induces Klf 4 expression while HDL 3 restores Klf 4 to basal levels in WT cells, but again this effect is not observed in KO cells. Conclusions Our results indicate that HDL 3 , by interacting with ABCA1, modulates the miR143/145-myocardin axis and prevents the cholesterol-induced gene expression modification in SMCs regardless of its cholesterol unloading capacity. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
44. The impact of APOA5, APOB, APOC3 and ABCA1 gene polymorphisms on ischemic stroke: Evidence from a meta-analysis.
- Author
-
Au, Anthony, Griffiths, Lyn R., Irene, Looi, Kooi, Cheah Wee, and Wei, Loo Keat
- Subjects
- *
GENETIC polymorphisms , *CORONARY disease , *ATHEROSCLEROSIS , *META-analysis , *GENETIC models - Abstract
Background and aims Genetic studies have been reported on the association between APOA5 , APOB , APOC3 and ABCA1 gene polymorphisms and ischemic stroke, but results remain controversial. Hence, this meta-analysis aimed to infer the causal relationships of APOA5 (rs662799, rs3135506), APOB (rs693, rs1042031, rs1801701), APOC3 (rs4520, rs5128, rs2854116, rs2854117) and ABCA1 rs2230806 with ischemic stroke risk. Methods A systematic review was performed for all the articles retrieved from multiple databases, up until March 2017. Data were extracted from all eligible studies, and meta-analysis was carried out using RevMan 5.3 and R package 3.2.1. The strength of association between each studied polymorphism and ischemic stroke risk was measured as odds ratios (ORs) and 95% confidence intervals (CIs), under fixed- and random-effect models. Results A total of 79 studies reporting on the association between the studied polymorphisms and ischemic stroke risk were identified. The pooled data indicated that all genetic models of APOA5 rs662799 (ORs = 1.23–1.43), allelic and over-dominant models of APOA5 rs3135506 (ORs = 1.77–1.97), APOB rs1801701 (ORs = 1.72–2.13) and APOB rs1042031 (ORs = 1.66–1.88) as well as dominant model of ABCA1 rs2230806 (OR = 1.31) were significantly associated with higher risk of ischemic stroke. However, no significant associations were observed between ischemic stroke and the other five polymorphisms, namely ApoB (rs693) and APOC3 (rs4520, rs5128, rs2854116 and rs2854117), under any genetic model. Conclusions The present meta-analysis confirmed a significant association of APOA5 rs662799 CC, APOA5 rs3135506 CG, APOB rs1801701 GA, APOB rs1042031 GA and ABCA1 rs2230806 GG with increased risk of ischemic stroke. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
45. Puerarin promotes ABCA1-mediated cholesterol efflux and decreases cellular lipid accumulation in THP-1 macrophages.
- Author
-
Li, Cong-hui, Gong, Duo, Chen, Ling-yan, Zhang, Min, Xia, Xiao-dan, Cheng, Hai-peng, Huang, Chong, Zhao, Zhen-wang, Zheng, Xi-Long, Tang, Xiao-er, and Tang, Chao-ke
- Subjects
- *
PUERARIA , *CHOLESTEROL in the body , *LIPOPROTEINS , *TRIGLYCERIDES , *MACROPHAGES , *THERAPEUTICS - Abstract
It was reported that puerarin decreases the total cholesterol, low-density lipoprotein cholesterol (LDL-C), triglyceride (TG) and increases high-density lipoprotein cholesterol (HDL-C) level, but the underlying mechanism is unclear. This study was designed to determine whether puerarin decreased lipid accumulation via up-regulation of ABCA1-mediated cholesterol efflux in THP-1 macrophage-derived foam cells. Our results showed that puerarin significantly promoted the expression of ATP-binding cassette transporter A1 (ABCA1) mRNA and protein via the AMP-activated protein kinase (AMPK)-peroxisome proliferator-activated receptor gamma (PPARγ)-liver X receptor-alpha (LXR-α) pathway and decreased cellular lipid accumulation in human THP-1 macrophage-derived foam cells. The miR-7 directly targeted 3′ untranslated region of STK11 (Serine/Threonine Kinase 11), which activated the AMPK pathway. Transfection with miR-7 mimic significantly reduced STK11 expression in puerarin-treated macrophages, decreased the phosphorylation of AMPK, down-regulated the expression of the PPARγ-LXR-α-ABCA1 expression. Additionally, treatment with miR-7 decreased cholesterol efflux and increased cholesterol levels in THP-1 macrophage-derived foam cells. Our study demonstrates that puerarin promotes ABCA1-mediated cholesterol efflux and decreases intracellular cholesterol levels through the pathway involving miR-7, STK11, and the AMPK-PPARγ-LXR-α-ABCA1 cascade. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
46. Hsp27 promotes ABCA1 expression and cholesterol efflux through the PI3K/PKCζ/Sp1 pathway in THP-1 macrophages.
- Author
-
Kuang, Hai-Jun, Zhao, Guo-Jun, Chen, Wu-Jun, Zhang, Min, Zeng, Gao-Feng, Zheng, Xi-Long, and Tang, Chao-Ke
- Subjects
- *
HEAT shock proteins , *ATHEROSCLEROSIS treatment , *PHOSPHOINOSITIDES , *PROTEIN expression , *MACROPHAGES , *ATP-binding cassette transporters , *BIOMARKERS , *THERAPEUTICS - Abstract
Heat shock protein 27 (Hsp27) is a putative biomarker and therapeutic target in atherosclerosis. This study was to explore the potential mechanisms underlying Hsp27 effects on ATP-binding cassette transporter A1 (ABCA1) expression and cellular cholesterol efflux. THP-1 macrophage-derived foam cells were infected with adenovirus to express wild-type Hsp27, hyper-phosphorylated Hsp27 mimic (3D Hsp27), antisense Hsp27 or hypo-phosphorylated Hsp27 mimic (3A Hsp27). Wild-type and 3D Hsp27 were found to up-regulate ABCA1 mRNA and protein expression and increase cholesterol efflux from cells. Expression of antisense or 3A Hsp27 suppressed the expression of ABCA1 and cholesterol efflux. Furthermore, over-expression of wild-type and 3D Hsp27 significantly increased the levels of phosphorylated specificity protein 1 (Sp1), protein kinase C ζ (PKCζ) and phosphatidylinositol 3-kinase (PI3K). In addition, the up-regulation of ABCA1 expression and cholesterol efflux induced by 3D Hsp27 was suppressed by inhibition of Sp1, PKCζ and PI3K with specific kinase inhibitors. Taken together, our results revealed that Hsp27 may up-regulate the expression of ABCA1 and promotes cholesterol efflux through activation of the PI3K/PKCζ/Sp1 signal pathway in THP-1 macrophage-derived foam cells. Our findings may partly explain the mechanisms underlying the anti-atherogenic effect of Hsp27. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
47. Structure-function relationships in reconstituted HDL: Focus on antioxidative activity and cholesterol efflux capacity.
- Author
-
Cukier, Alexandre M.O., Therond, Patrice, Didichenko, Svetlana A., Guillas, Isabelle, Chapman, M. John, Wright, Samuel D., and Kontush, Anatol
- Subjects
- *
LIPOPROTEINS , *APOLIPOPROTEIN A , *PHOSPHOLIPIDS , *HYDROPEROXIDES , *ETHYLENEDIAMINETETRAACETIC acid , *BUTYLATED hydroxytoluene - Abstract
Aims High-density lipoprotein (HDL) contains multiple components that endow it with biological activities. Apolipoprotein A-I (apoA-I) and surface phospholipids contribute to these activities; however, structure-function relationships in HDL particles remain incompletely characterised. Methods Reconstituted HDLs (rHDLs) were prepared from apoA-I and soy phosphatidylcholine (PC) at molar ratios of 1:50, 1:100 and 1:150. Oxidative status of apoA-I was varied using controlled oxidation of Met112 residue. HDL-mediated inactivation of PC hydroperoxides (PCOOH) derived from mildly pre-oxidized low-density lipoprotein (LDL) was evaluated by HPLC with chemiluminescent detection in HDL + LDL mixtures and re-isolated LDL. Cellular cholesterol efflux was characterised in RAW264.7 macrophages. Results rHDL inactivated LDL-derived PCOOH in a dose- and time-dependent manner. The capacity of rHDL to both inactivate PCOOH and efflux cholesterol via ATP-binding cassette transporter A1 (ABCA1) increased with increasing apoA-I/PC ratio proportionally to the apoA-I content in rHDL. Controlled oxidation of apoA-I Met112 gradually decreased PCOOH-inactivating capacity of rHDL but increased ABCA1-mediated cellular cholesterol efflux. Conclusions Increasing apoA-I content in rHDL enhanced its antioxidative activity towards oxidized LDL and cholesterol efflux capacity via ABCA1, whereas oxidation of apoA-I Met112 decreased the antioxidative activity but increased the cholesterol efflux. These findings provide important considerations in the design of future HDL therapeutics. Non-standard abbreviations and acronyms: AAPH, 2,2′-azobis(-amidinopropane) dihydrochloride; ABCA1, ATP-binding cassette transporter A1; apoA-I, apolipoprotein A-I; BHT, butylated hydroxytoluene; CV, cardiovascular; EDTA, ethylenediaminetetraacetic acid; HDL-C, high-density lipoprotein cholesterol; LOOH, lipid hydroperoxides; Met(O), methionine sulfoxide; Met112, methionine 112 residue; Met86, methionine 86 residue; oxLDL, oxidized low-density lipoprotein; PBS, phosphate-buffered saline; PC, phosphatidylcholine; PL, phospholipid; PCOOH, phosphatidylcholine hydroperoxide; PLOOH, phospholipid hydroperoxide. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
48. Lysophosphatidylcholine export by human ABCA7.
- Author
-
Tomioka, Maiko, Toda, Yoshinobu, Mañucat, Noralyn B., Akatsu, Hiroyasu, Fukumoto, Manabu, Kono, Nozomu, Arai, Hiroyuki, Kioka, Noriyuki, and Ueda, Kazumitsu
- Subjects
- *
LYSOPHOSPHATIDYLCHOLINE acyltransferase , *ADENOSINE triphosphate , *ALZHEIMER'S disease , *IMMUNOHISTOCHEMISTRY , *SPHINGOMYELIN - Abstract
The ATP-binding cassette transporter A7 (ABCA7), which is highly expressed in the brain, is associated with the pathogenesis of Alzheimer's disease (AD). However, the physiological function of ABCA7 and its transport substrates remain unclear. Immunohistochemical analyses of human brain sections from AD and non-AD subjects revealed that ABCA7 is expressed in neuron and microglia cells in the cerebral cortex. The transport substrates and acceptors were identified in BHK/ABCA7 cells and compared with those of ABCA1. Like ABCA1, ABCA7 exported choline phospholipids in the presence of apoA-I and apoE; however, unlike ABCA1, cholesterol efflux was marginal. Lipid efflux by ABCA7 was saturated by 5 μg/ml apoA-I and was not dependent on apoE isoforms, whereas efflux by ABCA1 was dependent on apoA-I up to 20 μg/ml and apoE isoforms. Liquid chromatography–tandem mass spectrometry analyses revealed that the two proteins had different preferences for phospholipid export: ABCA7 preferred phosphatidylcholine (PC) ≥ lysoPC > sphingomyelin (SM) = phosphatidylethanolamine (PE), whereas ABCA1 preferred PC > > SM > PE = lysoPC. The major difference in the pattern of lipid peaks between ABCA7 and ABCA1 was the high lysoPC/PC ratio of ABCA7. These results suggest that lysoPC is one of the major transport substrates for ABCA7 and that lysoPC export may be a physiologically important function of ABCA7 in the brain. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
49. The lipid moiety 7-ketocholesteryl-9-carboxynonanoate mediates binding interaction of oxLDL to LOX-1 and upregulates ABCA1 expression through PPARγ.
- Author
-
Li, Jingda, Xiu, Zhilong, Wang, Renjun, Yu, Chengjie, Chi, Yan, Qin, Jianzhong, Fu, Changzhen, Matsuura, Eiji, and Liu, Qingping
- Subjects
- *
LECTINS , *LOW density lipoproteins , *PEROXISOME proliferator-activated receptors , *IMMUNOCYTOCHEMISTRY , *CELLULAR signal transduction - Abstract
Aim Lectin-like oxidized low-density lipoprotein receptor-1 (LOX-1), a specific membrane receptor for oxidized low-density lipoprotein (oxLDL), plays a crucial role in atherosclerosis progression. The aim of this study was to elucidate the role of 7-ketocholesteryl-9-carboxynonanoate (oxLig-1), a lipid component of oxLDL, in the binding of oxLDL to LOX-1 and to determine whether oxLig-1 binding to LOX-1 is involved in the upregulation of ABCA1 expression. Main methods OxLig-1 binding to LOX-1 was analysed by AutoDock 4.2.6 and confirmed by fluorescence immunocytochemistry and enzyme-linked immunosorbent assays (ELISAs). LOX-1, LXRα and ABCA1 protein expression induced by oxLig-1 or methylated oxLig-1 was measured by western blotting. In addition, PPARγ activation was investigated using a dual-luciferase reporter system. Furthermore, the signalling cascade involved in oxLig-1-induced ABCA1 expression was assessed using inhibitors for PPARγ and LXRα and specific siRNAs against LOX-1, PPARγ and LXRα. Key findings Docking, fluorescence immunocytochemistry and ELISA analyses showed that oxLig-1 bound LOX-1 and that the ω-carboxyl group was critical for this binding. Moreover, oxLig-1, but not methylated oxLig-1, increased LOX-1, LXRα, and ABCA1 expression. Luciferase reporter assays indicated that oxLig-1 activated PPARγ in the presence of LOX-1. Additionally, the inhibitor and siRNA experiments showed that oxLig-1 triggered the PPARγ-LXRα signalling pathway, leading to upregulation of ABCA1, and that this process required the participation of LOX-1. Significance Our observations indicate that oxLig-1 is a critical epitope of oxLDL that mediates the binding of oxLDL to LOX-1 and initiates PPARγ signal transduction to upregulate the expression of ABCA1. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
50. Unveiling the role of G-quadruplex structure in promoter region: Regulation of ABCA1 expression in macrophages possibly via NONO protein recruitment.
- Author
-
Xiao, Chao-Da, Jia, Meng-Hao, Zhong, Ming-Qing, Xu, Yan, Yu, Zu-Tao, He, Zhi-Yong, Lu, Xu, Zhang, Yan, Zhou, Xue, Fu, Lin-Yun, and Shen, Xiang-Chun
- Subjects
- *
GENE expression , *PROMOTERS (Genetics) , *GENETIC regulation , *MORINDA citrifolia , *MACROPHAGES , *QUADRUPLEX nucleic acids - Abstract
ABCA1 has been found to be critical for cholesterol efflux in macrophages. Understanding the mechanism regulating ABCA1 expression is important for the prevention and treatment of atherosclerosis. In the present study, a G-quadruplex (G4) structure was identified in the ABCA1 promoter region. This G4 was shown to be essential for ABCA1 transcription. Stabilizing the G4 by ligands surprisingly upregulated ABCA1 expression in macrophages. Knocking out the G4 remarkably reduced ABCA1 expression, and abolished the increase of ABCA1 expression induced by the G4 ligand. By pull-down assays, the protein NONO was identified as an ABCA1 G4 binder. Overexpression or repression of NONO significantly induced upregulation and downregulation of ABCA1 expression, respectively. ChIP and EMSA experiments showed that the G4 ligand promoted the binding between the ABCA1 G4 and NONO, which led to more recruitment of NONO to the promoter region and enhanced ABCA1 transcription. Finally, the G4 ligand was shown to significantly reduce the accumulation of cholesterol in macrophages. This study showed a new insight into the regulation of gene expression by G4, and provided a new molecular mechanism regulating ABCA1 expression in macrophages. Furthermore, the study showed a possible novel application of the G4 ligand: preventing and treating atherosclerosis. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.