抑制体外巨噬细胞样细胞中的 miR-33a-5p 可促进载脂蛋白 A 诱导的胆固醇外流。

IF 2.7 Q2 PATHOLOGY
Olanrewaju Oladosu, Emma Chin, Christian Barksdale, Rhonda R Powell, Terri Bruce, Alexis Stamatikos
{"title":"抑制体外巨噬细胞样细胞中的 miR-33a-5p 可促进载脂蛋白 A 诱导的胆固醇外流。","authors":"Olanrewaju Oladosu, Emma Chin, Christian Barksdale, Rhonda R Powell, Terri Bruce, Alexis Stamatikos","doi":"10.3390/pathophysiology31010009","DOIUrl":null,"url":null,"abstract":"<p><p>Atherosclerosis is caused by cholesterol accumulation within arteries. The intima is where atherosclerotic plaque accumulates and where lipid-laden foam cells reside. Intimal foam cells comprise of both monocyte-derived macrophages and macrophage-like cells (MLC) of vascular smooth muscle cell (VSMC) origin. Foam cells can remove cholesterol via apoAI-mediated cholesterol efflux and this process is regulated by the transporter ABCA1. The microRNA miR-33a-5p is thought to be atherogenic via silencing ABCA1 which promotes cholesterol retention and data has shown inhibiting miR-33a-5p in macrophages may be atheroprotective via enhancing apoAI-mediated cholesterol efflux. However, it is not entirely elucidated whether precisely inhibiting miR-33a-5p in MLC also increases ABCA1-dependent cholesterol efflux. Therefore, the purpose of this work is to test the hypothesis that inhibition of miR-33a-5p in cultured MLC enhances apoAI-mediated cholesterol efflux. In our study, we utilized the VSMC line MOVAS cells in our experiments, and cholesterol-loaded MOVAS cells to convert this cell line into MLC. Inhibition of miR-33a-5p was accomplished by transducing cells with a lentivirus that expresses an antagomiR directed at miR-33a-5p. Expression of miR-33a-5p was analyzed by qRT-PCR, ABCA1 protein expression was assessed via immunoblotting, and apoAI-mediated cholesterol efflux was measured using cholesterol efflux assays. In our results, we demonstrated that lentiviral vector-mediated knockdown of miR-33a-5p resulted in decreasing expression of this microRNA in cultured MLC. Moreover, reduction of miR-33a-5p in cultured MLC resulted in de-repression of ABCA1 expression, which caused ABCA1 protein upregulation in cultured MLC. Additionally, this increase in ABCA1 protein expression resulted in enhancing ABCA1-dependent cholesterol efflux through increasing apoAI-mediated cholesterol efflux in cultured MLC. From these findings, we conclude that inhibiting miR-33a-5p in MLC may protect against atherosclerosis by promoting ABCA1-dependent cholesterol efflux.</p>","PeriodicalId":19852,"journal":{"name":"Pathophysiology","volume":null,"pages":null},"PeriodicalIF":2.7000,"publicationDate":"2024-02-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10976131/pdf/","citationCount":"0","resultStr":"{\"title\":\"Inhibition of miR-33a-5p in Macrophage-like Cells In Vitro Promotes apoAI-Mediated Cholesterol Efflux.\",\"authors\":\"Olanrewaju Oladosu, Emma Chin, Christian Barksdale, Rhonda R Powell, Terri Bruce, Alexis Stamatikos\",\"doi\":\"10.3390/pathophysiology31010009\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"<p><p>Atherosclerosis is caused by cholesterol accumulation within arteries. The intima is where atherosclerotic plaque accumulates and where lipid-laden foam cells reside. Intimal foam cells comprise of both monocyte-derived macrophages and macrophage-like cells (MLC) of vascular smooth muscle cell (VSMC) origin. Foam cells can remove cholesterol via apoAI-mediated cholesterol efflux and this process is regulated by the transporter ABCA1. The microRNA miR-33a-5p is thought to be atherogenic via silencing ABCA1 which promotes cholesterol retention and data has shown inhibiting miR-33a-5p in macrophages may be atheroprotective via enhancing apoAI-mediated cholesterol efflux. However, it is not entirely elucidated whether precisely inhibiting miR-33a-5p in MLC also increases ABCA1-dependent cholesterol efflux. Therefore, the purpose of this work is to test the hypothesis that inhibition of miR-33a-5p in cultured MLC enhances apoAI-mediated cholesterol efflux. In our study, we utilized the VSMC line MOVAS cells in our experiments, and cholesterol-loaded MOVAS cells to convert this cell line into MLC. Inhibition of miR-33a-5p was accomplished by transducing cells with a lentivirus that expresses an antagomiR directed at miR-33a-5p. Expression of miR-33a-5p was analyzed by qRT-PCR, ABCA1 protein expression was assessed via immunoblotting, and apoAI-mediated cholesterol efflux was measured using cholesterol efflux assays. In our results, we demonstrated that lentiviral vector-mediated knockdown of miR-33a-5p resulted in decreasing expression of this microRNA in cultured MLC. Moreover, reduction of miR-33a-5p in cultured MLC resulted in de-repression of ABCA1 expression, which caused ABCA1 protein upregulation in cultured MLC. Additionally, this increase in ABCA1 protein expression resulted in enhancing ABCA1-dependent cholesterol efflux through increasing apoAI-mediated cholesterol efflux in cultured MLC. From these findings, we conclude that inhibiting miR-33a-5p in MLC may protect against atherosclerosis by promoting ABCA1-dependent cholesterol efflux.</p>\",\"PeriodicalId\":19852,\"journal\":{\"name\":\"Pathophysiology\",\"volume\":null,\"pages\":null},\"PeriodicalIF\":2.7000,\"publicationDate\":\"2024-02-28\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10976131/pdf/\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"Pathophysiology\",\"FirstCategoryId\":\"1085\",\"ListUrlMain\":\"https://doi.org/10.3390/pathophysiology31010009\",\"RegionNum\":0,\"RegionCategory\":null,\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"\",\"PubModel\":\"\",\"JCR\":\"Q2\",\"JCRName\":\"PATHOLOGY\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"Pathophysiology","FirstCategoryId":"1085","ListUrlMain":"https://doi.org/10.3390/pathophysiology31010009","RegionNum":0,"RegionCategory":null,"ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q2","JCRName":"PATHOLOGY","Score":null,"Total":0}
引用次数: 0

摘要

动脉粥样硬化是由胆固醇在动脉内积聚造成的。动脉内膜是动脉粥样硬化斑块的聚集地,也是脂质泡沫细胞的栖息地。内膜泡沫细胞由来源于单核细胞的巨噬细胞和来源于血管平滑肌细胞(VSMC)的巨噬细胞样细胞(MLC)组成。泡沫细胞可通过载脂蛋白AI介导的胆固醇外流清除胆固醇,这一过程受转运体ABCA1的调节。有数据显示,抑制巨噬细胞中的 miR-33a-5p 可通过增强 apoAI 介导的胆固醇外流起到保护动脉粥样硬化的作用。然而,精确抑制 MLC 中的 miR-33a-5p 是否也会增加 ABCA1 依赖的胆固醇外流,目前还没有完全阐明。因此,本研究的目的是检验在培养的 MLC 中抑制 miR-33a-5p 是否会增强 apoAI 介导的胆固醇外流的假设。在我们的研究中,我们利用 VSMC 细胞系 MOVAS 细胞进行实验,并利用胆固醇负载的 MOVAS 细胞将该细胞系转化为 MLC。抑制 miR-33a-5p 的方法是用表达针对 miR-33a-5p 的抗噬菌体的慢病毒转导细胞。miR-33a-5p 的表达通过 qRT-PCR 进行分析,ABCA1 蛋白的表达通过免疫印迹进行评估,apoAI 介导的胆固醇外流通过胆固醇外流测定进行测量。我们的研究结果表明,慢病毒载体介导的 miR-33a-5p 基因敲除导致培养的 MLC 中该 microRNA 的表达下降。此外,培养的 MLC 中 miR-33a-5p 的减少导致 ABCA1 表达的去抑制,从而引起培养的 MLC 中 ABCA1 蛋白的上调。此外,ABCA1 蛋白表达的增加导致培养 MLC 中载脂蛋白介导的胆固醇外流增加,从而增强了 ABCA1 依赖性胆固醇外流。根据这些发现,我们得出结论:抑制 MLC 中的 miR-33a-5p 可促进 ABCA1 依赖性胆固醇外流,从而预防动脉粥样硬化。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
Inhibition of miR-33a-5p in Macrophage-like Cells In Vitro Promotes apoAI-Mediated Cholesterol Efflux.

Atherosclerosis is caused by cholesterol accumulation within arteries. The intima is where atherosclerotic plaque accumulates and where lipid-laden foam cells reside. Intimal foam cells comprise of both monocyte-derived macrophages and macrophage-like cells (MLC) of vascular smooth muscle cell (VSMC) origin. Foam cells can remove cholesterol via apoAI-mediated cholesterol efflux and this process is regulated by the transporter ABCA1. The microRNA miR-33a-5p is thought to be atherogenic via silencing ABCA1 which promotes cholesterol retention and data has shown inhibiting miR-33a-5p in macrophages may be atheroprotective via enhancing apoAI-mediated cholesterol efflux. However, it is not entirely elucidated whether precisely inhibiting miR-33a-5p in MLC also increases ABCA1-dependent cholesterol efflux. Therefore, the purpose of this work is to test the hypothesis that inhibition of miR-33a-5p in cultured MLC enhances apoAI-mediated cholesterol efflux. In our study, we utilized the VSMC line MOVAS cells in our experiments, and cholesterol-loaded MOVAS cells to convert this cell line into MLC. Inhibition of miR-33a-5p was accomplished by transducing cells with a lentivirus that expresses an antagomiR directed at miR-33a-5p. Expression of miR-33a-5p was analyzed by qRT-PCR, ABCA1 protein expression was assessed via immunoblotting, and apoAI-mediated cholesterol efflux was measured using cholesterol efflux assays. In our results, we demonstrated that lentiviral vector-mediated knockdown of miR-33a-5p resulted in decreasing expression of this microRNA in cultured MLC. Moreover, reduction of miR-33a-5p in cultured MLC resulted in de-repression of ABCA1 expression, which caused ABCA1 protein upregulation in cultured MLC. Additionally, this increase in ABCA1 protein expression resulted in enhancing ABCA1-dependent cholesterol efflux through increasing apoAI-mediated cholesterol efflux in cultured MLC. From these findings, we conclude that inhibiting miR-33a-5p in MLC may protect against atherosclerosis by promoting ABCA1-dependent cholesterol efflux.

求助全文
通过发布文献求助,成功后即可免费获取论文全文。 去求助
来源期刊
Pathophysiology
Pathophysiology Medicine-Pathology and Forensic Medicine
CiteScore
3.10
自引率
0.00%
发文量
48
期刊介绍: Pathophysiology is an international journal which publishes papers in English which address the etiology, development, and elimination of pathological processes. Contributions on the basic mechanisms underlying these processes, model systems and interdisciplinary approaches are strongly encouraged.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
确定
请完成安全验证×
copy
已复制链接
快去分享给好友吧!
我知道了
右上角分享
点击右上角分享
0
联系我们:info@booksci.cn Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。 Copyright © 2023 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术官方微信