在小鼠肥厚性心肌病(HCM)模型中抑制miR-199a-3p可减轻纤维化重塑

Irina Zalivina , Temo Barwari , Xiaoke Yin , Sarah R. Langley , Javier Barallobre-Barreiro , Hiroko Wakimoto , Anna Zampetaki , Manuel Mayr , Metin Avkiran , Seda Eminaga
{"title":"在小鼠肥厚性心肌病(HCM)模型中抑制miR-199a-3p可减轻纤维化重塑","authors":"Irina Zalivina ,&nbsp;Temo Barwari ,&nbsp;Xiaoke Yin ,&nbsp;Sarah R. Langley ,&nbsp;Javier Barallobre-Barreiro ,&nbsp;Hiroko Wakimoto ,&nbsp;Anna Zampetaki ,&nbsp;Manuel Mayr ,&nbsp;Metin Avkiran ,&nbsp;Seda Eminaga","doi":"10.1016/j.jmccpl.2023.100056","DOIUrl":null,"url":null,"abstract":"<div><h3>Background</h3><p>Hypertrophic cardiomyopathy (HCM) is an autosomal dominant genetic disorder, characterized by cardiomyocyte hypertrophy, cardiomyocyte disarray and fibrosis, which has a prevalence of ∼1: 200–500 and predisposes individuals to heart failure and sudden death. The mechanisms through which diverse HCM-causing mutations cause cardiac dysfunction remain mostly unknown and their identification may reveal new therapeutic avenues. MicroRNAs (miRNAs) have emerged as critical regulators of gene expression and disease phenotype in various pathologies. We explored whether miRNAs could play a role in HCM pathogenesis and offer potential therapeutic targets.</p></div><div><h3>Methods and results</h3><p>Using high-throughput miRNA expression profiling and qPCR analysis in two distinct mouse models of HCM, we found that miR-199a-3p expression levels are upregulated in mutant mice compared to age- and treatment-matched wild-type mice. We also found that miR-199a-3p expression is enriched in cardiac non-myocytes compared to cardiomyocytes. When we expressed miR-199a-3p mimic in cultured murine primary cardiac fibroblasts and analyzed the conditioned media by proteomics, we found that several extracellular matrix (ECM) proteins (<em>e.g.</em>, TSP2, FBLN3, COL11A1, LYOX) were differentially secreted (data are available <em>via</em> ProteomeXchange with identifier <span>PXD042904</span><svg><path></path></svg>). We confirmed our proteomics findings by qPCR analysis of selected mRNAs and demonstrated that miR-199a-3p mimic expression in cardiac fibroblasts drives upregulation of ECM gene expression, including <em>Tsp2</em>, <em>Fbln3</em>, <em>Pcoc1</em>, <em>Col1a1</em> and <em>Col3a1</em>. To examine the role of miR-199a-3p <em>in vivo</em>, we inhibited its function using lock-nucleic acid (LNA)-based inhibitors (antimiR-199a-3p) in an HCM mouse model. Our results revealed that progression of cardiac fibrosis is attenuated when miR-199a-3p function is inhibited in mild-to-moderate HCM. Finally, guided by computational target prediction algorithms, we identified mRNAs <em>Cd151</em> and <em>Itga3</em> as direct targets of miR-199a-3p and have shown that miR-199a-3p mimic expression negatively regulates AKT activation in cardiac fibroblasts.</p></div><div><h3>Conclusions</h3><p>Altogether, our results suggest that miR-199a-3p may contribute to cardiac fibrosis in HCM through its actions in cardiac fibroblasts. Thus, inhibition of miR-199a-3p in mild-to-moderate HCM may offer therapeutic benefit in combination with complementary approaches that target the primary defect in cardiac myocytes.</p></div>","PeriodicalId":73835,"journal":{"name":"Journal of molecular and cellular cardiology plus","volume":null,"pages":null},"PeriodicalIF":0.0000,"publicationDate":"2023-11-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S2772976123000260/pdfft?md5=d7ef72bd8c44be43fadde9936ac62981&pid=1-s2.0-S2772976123000260-main.pdf","citationCount":"1","resultStr":"{\"title\":\"Inhibition of miR-199a-3p in a murine hypertrophic cardiomyopathy (HCM) model attenuates fibrotic remodeling\",\"authors\":\"Irina Zalivina ,&nbsp;Temo Barwari ,&nbsp;Xiaoke Yin ,&nbsp;Sarah R. Langley ,&nbsp;Javier Barallobre-Barreiro ,&nbsp;Hiroko Wakimoto ,&nbsp;Anna Zampetaki ,&nbsp;Manuel Mayr ,&nbsp;Metin Avkiran ,&nbsp;Seda Eminaga\",\"doi\":\"10.1016/j.jmccpl.2023.100056\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"<div><h3>Background</h3><p>Hypertrophic cardiomyopathy (HCM) is an autosomal dominant genetic disorder, characterized by cardiomyocyte hypertrophy, cardiomyocyte disarray and fibrosis, which has a prevalence of ∼1: 200–500 and predisposes individuals to heart failure and sudden death. The mechanisms through which diverse HCM-causing mutations cause cardiac dysfunction remain mostly unknown and their identification may reveal new therapeutic avenues. MicroRNAs (miRNAs) have emerged as critical regulators of gene expression and disease phenotype in various pathologies. We explored whether miRNAs could play a role in HCM pathogenesis and offer potential therapeutic targets.</p></div><div><h3>Methods and results</h3><p>Using high-throughput miRNA expression profiling and qPCR analysis in two distinct mouse models of HCM, we found that miR-199a-3p expression levels are upregulated in mutant mice compared to age- and treatment-matched wild-type mice. We also found that miR-199a-3p expression is enriched in cardiac non-myocytes compared to cardiomyocytes. When we expressed miR-199a-3p mimic in cultured murine primary cardiac fibroblasts and analyzed the conditioned media by proteomics, we found that several extracellular matrix (ECM) proteins (<em>e.g.</em>, TSP2, FBLN3, COL11A1, LYOX) were differentially secreted (data are available <em>via</em> ProteomeXchange with identifier <span>PXD042904</span><svg><path></path></svg>). We confirmed our proteomics findings by qPCR analysis of selected mRNAs and demonstrated that miR-199a-3p mimic expression in cardiac fibroblasts drives upregulation of ECM gene expression, including <em>Tsp2</em>, <em>Fbln3</em>, <em>Pcoc1</em>, <em>Col1a1</em> and <em>Col3a1</em>. To examine the role of miR-199a-3p <em>in vivo</em>, we inhibited its function using lock-nucleic acid (LNA)-based inhibitors (antimiR-199a-3p) in an HCM mouse model. Our results revealed that progression of cardiac fibrosis is attenuated when miR-199a-3p function is inhibited in mild-to-moderate HCM. Finally, guided by computational target prediction algorithms, we identified mRNAs <em>Cd151</em> and <em>Itga3</em> as direct targets of miR-199a-3p and have shown that miR-199a-3p mimic expression negatively regulates AKT activation in cardiac fibroblasts.</p></div><div><h3>Conclusions</h3><p>Altogether, our results suggest that miR-199a-3p may contribute to cardiac fibrosis in HCM through its actions in cardiac fibroblasts. Thus, inhibition of miR-199a-3p in mild-to-moderate HCM may offer therapeutic benefit in combination with complementary approaches that target the primary defect in cardiac myocytes.</p></div>\",\"PeriodicalId\":73835,\"journal\":{\"name\":\"Journal of molecular and cellular cardiology plus\",\"volume\":null,\"pages\":null},\"PeriodicalIF\":0.0000,\"publicationDate\":\"2023-11-22\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"https://www.sciencedirect.com/science/article/pii/S2772976123000260/pdfft?md5=d7ef72bd8c44be43fadde9936ac62981&pid=1-s2.0-S2772976123000260-main.pdf\",\"citationCount\":\"1\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"Journal of molecular and cellular cardiology plus\",\"FirstCategoryId\":\"1085\",\"ListUrlMain\":\"https://www.sciencedirect.com/science/article/pii/S2772976123000260\",\"RegionNum\":0,\"RegionCategory\":null,\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"\",\"PubModel\":\"\",\"JCR\":\"\",\"JCRName\":\"\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"Journal of molecular and cellular cardiology plus","FirstCategoryId":"1085","ListUrlMain":"https://www.sciencedirect.com/science/article/pii/S2772976123000260","RegionNum":0,"RegionCategory":null,"ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"","JCRName":"","Score":null,"Total":0}
引用次数: 1

摘要

肥厚性心肌病(HCM)是一种常染色体显性遗传病,以心肌细胞肥大、心肌细胞紊乱和纤维化为特征,其患病率约为1:20 00 - 500,易导致个体心力衰竭和猝死。各种hcm引起的突变引起心功能障碍的机制仍然是未知的,它们的鉴定可能会揭示新的治疗途径。MicroRNAs (miRNAs)已成为各种病理中基因表达和疾病表型的关键调节因子。我们探索mirna是否在HCM发病机制中发挥作用并提供潜在的治疗靶点。方法和结果通过对两种不同HCM小鼠模型的高通量miRNA表达谱和qPCR分析,我们发现与年龄匹配和治疗匹配的野生型小鼠相比,miR-199a-3p在突变小鼠中的表达水平上调。我们还发现,与心肌细胞相比,miR-199a-3p在心脏非肌细胞中的表达更丰富。当我们在培养的小鼠原代心脏成纤维细胞中表达miR-199a-3p模拟物并通过蛋白质组学分析条件培养基时,我们发现几种细胞外基质(ECM)蛋白(例如,TSP2, FBLN3, COL11A1, LYOX)分泌差异(数据可通过ProteomeXchange获得,标识号为PXD042904)。我们通过对所选mrna的qPCR分析证实了我们的蛋白质组学发现,并证明miR-199a-3p在心脏成纤维细胞中的模拟表达驱动ECM基因表达上调,包括Tsp2、Fbln3、Pcoc1、Col1a1和Col3a1。为了研究miR-199a-3p在体内的作用,我们在HCM小鼠模型中使用基于锁定核酸(LNA)的抑制剂(anti - miR-199a-3p)抑制其功能。我们的研究结果显示,当miR-199a-3p功能在轻度至中度HCM中被抑制时,心脏纤维化的进展会减弱。最后,在计算靶标预测算法的指导下,我们发现mrna Cd151和Itga3是miR-199a-3p的直接靶标,并表明miR-199a-3p模拟表达负调控AKT在心脏成纤维细胞中的激活。综上所述,我们的研究结果表明miR-199a-3p可能通过其在心脏成纤维细胞中的作用促进HCM的心脏纤维化。因此,在轻度至中度HCM中抑制miR-199a-3p可能与针对心肌细胞原发性缺陷的互补方法相结合,提供治疗益处。
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-199a-3p in a murine hypertrophic cardiomyopathy (HCM) model attenuates fibrotic remodeling

Inhibition of miR-199a-3p in a murine hypertrophic cardiomyopathy (HCM) model attenuates fibrotic remodeling

Background

Hypertrophic cardiomyopathy (HCM) is an autosomal dominant genetic disorder, characterized by cardiomyocyte hypertrophy, cardiomyocyte disarray and fibrosis, which has a prevalence of ∼1: 200–500 and predisposes individuals to heart failure and sudden death. The mechanisms through which diverse HCM-causing mutations cause cardiac dysfunction remain mostly unknown and their identification may reveal new therapeutic avenues. MicroRNAs (miRNAs) have emerged as critical regulators of gene expression and disease phenotype in various pathologies. We explored whether miRNAs could play a role in HCM pathogenesis and offer potential therapeutic targets.

Methods and results

Using high-throughput miRNA expression profiling and qPCR analysis in two distinct mouse models of HCM, we found that miR-199a-3p expression levels are upregulated in mutant mice compared to age- and treatment-matched wild-type mice. We also found that miR-199a-3p expression is enriched in cardiac non-myocytes compared to cardiomyocytes. When we expressed miR-199a-3p mimic in cultured murine primary cardiac fibroblasts and analyzed the conditioned media by proteomics, we found that several extracellular matrix (ECM) proteins (e.g., TSP2, FBLN3, COL11A1, LYOX) were differentially secreted (data are available via ProteomeXchange with identifier PXD042904). We confirmed our proteomics findings by qPCR analysis of selected mRNAs and demonstrated that miR-199a-3p mimic expression in cardiac fibroblasts drives upregulation of ECM gene expression, including Tsp2, Fbln3, Pcoc1, Col1a1 and Col3a1. To examine the role of miR-199a-3p in vivo, we inhibited its function using lock-nucleic acid (LNA)-based inhibitors (antimiR-199a-3p) in an HCM mouse model. Our results revealed that progression of cardiac fibrosis is attenuated when miR-199a-3p function is inhibited in mild-to-moderate HCM. Finally, guided by computational target prediction algorithms, we identified mRNAs Cd151 and Itga3 as direct targets of miR-199a-3p and have shown that miR-199a-3p mimic expression negatively regulates AKT activation in cardiac fibroblasts.

Conclusions

Altogether, our results suggest that miR-199a-3p may contribute to cardiac fibrosis in HCM through its actions in cardiac fibroblasts. Thus, inhibition of miR-199a-3p in mild-to-moderate HCM may offer therapeutic benefit in combination with complementary approaches that target the primary defect in cardiac myocytes.

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
Journal of molecular and cellular cardiology plus
Journal of molecular and cellular cardiology plus Cardiology and Cardiovascular Medicine
自引率
0.00%
发文量
0
审稿时长
31 days
×
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