Astragalus mongholicus bunge and panax notoginseng formula (A&P) improves renal fibrosis in UUO mice via inhibiting the long non-coding RNA A330074K22Rik and downregulating ferroptosis signaling.

IF 3.3 2区 医学 Q1 INTEGRATIVE & COMPLEMENTARY MEDICINE
Xia Zhong, Yue Huang, Jian Jia, Jian Liu, Hongwei Su, Qiongdan Hu, Ruizhi Tan, Li Wang
{"title":"Astragalus mongholicus bunge and panax notoginseng formula (A&P) improves renal fibrosis in UUO mice via inhibiting the long non-coding RNA A330074K22Rik and downregulating ferroptosis signaling.","authors":"Xia Zhong, Yue Huang, Jian Jia, Jian Liu, Hongwei Su, Qiongdan Hu, Ruizhi Tan, Li Wang","doi":"10.1186/s12906-024-04557-4","DOIUrl":null,"url":null,"abstract":"<p><strong>Background: </strong>Chronic kidney disease (CKD) and its associated end-stage renal disease (ESRD) are significant health problems that pose a threat to human well-being. Renal fibrosis is a common feature and ultimate pathological outcome of various CKD leading to ESRD. The Astragalus mongholicus Bunge and Panax notoginseng formula (A&P) is a refined compound formulated by our research group, which has been clinically administered for over a decade and has demonstrated the ability to improve the inflammatory state of various acute or chronic kidney diseases. However, the underlying mechanism by which A&P ameliorates renal fibrosis remains unclear.</p><p><strong>Methods: </strong>We established a mouse model by surgically ligating the unilateral ureter to induce renal injury in vivo. And we utilized renal in situ electroporation of a plasmid with low LncRNA A33 expression to establish the unilateral ureteral obstruction(UUO)mouse model. In vitro, we stimulated primary tubular epithelial cells(pTEC) injury using TGF-β1, siRNA-A33, and pcDNA3.1-A33 plasmids were transfected into pTECs to respectively knockdown and overexpress LncRNA A33, and both in vitro and in vivo models were intervened with A&P.</p><p><strong>Results: </strong>The results demonstrated that A&P effectively alleviated renal fibrosis in mice. Subsequent findings indicated high expression of LncRNA A33 in the kidneys of UUO mice and TGF-β1-induced renal tubular cells. In situ, renal electroporation of a plasmid with reduced LncRNA A33 expression revealed that inhibiting LncRNA A33 significantly improved renal fibrosis in UUO mice. Moreover, A&P effectively suppressed LncRNA A33 expression both in vitro and in vivo. Subsequent downregulation of LncRNA A33 in renal tubular epithelial cells resulted in the downregulation of numerous fibrotic markers, a significant inhibition of LncRNA A33, and a notable reduction in downstream ferroptosis signaling. Cell experiments demonstrated that A&P improved renal fibrosis in UUO mice by inhibiting LncRNA A33 and downregulating ferroptosis signaling.</p><p><strong>Conclusion: </strong>Through the inhibition of LncRNA A33 and subsequent downregulation of ferroptosis signaling, A&P showed potential as a therapeutic approach for improving renal fibrosis in UUO mice, providing a potential treatment avenue for CKD.</p>","PeriodicalId":9128,"journal":{"name":"BMC Complementary Medicine and Therapies","volume":null,"pages":null},"PeriodicalIF":3.3000,"publicationDate":"2024-07-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11264518/pdf/","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"BMC Complementary Medicine and Therapies","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.1186/s12906-024-04557-4","RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"INTEGRATIVE & COMPLEMENTARY MEDICINE","Score":null,"Total":0}
引用次数: 0

Abstract

Background: Chronic kidney disease (CKD) and its associated end-stage renal disease (ESRD) are significant health problems that pose a threat to human well-being. Renal fibrosis is a common feature and ultimate pathological outcome of various CKD leading to ESRD. The Astragalus mongholicus Bunge and Panax notoginseng formula (A&P) is a refined compound formulated by our research group, which has been clinically administered for over a decade and has demonstrated the ability to improve the inflammatory state of various acute or chronic kidney diseases. However, the underlying mechanism by which A&P ameliorates renal fibrosis remains unclear.

Methods: We established a mouse model by surgically ligating the unilateral ureter to induce renal injury in vivo. And we utilized renal in situ electroporation of a plasmid with low LncRNA A33 expression to establish the unilateral ureteral obstruction(UUO)mouse model. In vitro, we stimulated primary tubular epithelial cells(pTEC) injury using TGF-β1, siRNA-A33, and pcDNA3.1-A33 plasmids were transfected into pTECs to respectively knockdown and overexpress LncRNA A33, and both in vitro and in vivo models were intervened with A&P.

Results: The results demonstrated that A&P effectively alleviated renal fibrosis in mice. Subsequent findings indicated high expression of LncRNA A33 in the kidneys of UUO mice and TGF-β1-induced renal tubular cells. In situ, renal electroporation of a plasmid with reduced LncRNA A33 expression revealed that inhibiting LncRNA A33 significantly improved renal fibrosis in UUO mice. Moreover, A&P effectively suppressed LncRNA A33 expression both in vitro and in vivo. Subsequent downregulation of LncRNA A33 in renal tubular epithelial cells resulted in the downregulation of numerous fibrotic markers, a significant inhibition of LncRNA A33, and a notable reduction in downstream ferroptosis signaling. Cell experiments demonstrated that A&P improved renal fibrosis in UUO mice by inhibiting LncRNA A33 and downregulating ferroptosis signaling.

Conclusion: Through the inhibition of LncRNA A33 and subsequent downregulation of ferroptosis signaling, A&P showed potential as a therapeutic approach for improving renal fibrosis in UUO mice, providing a potential treatment avenue for CKD.

Abstract Image

黄芪党参三七方(A&P)通过抑制长非编码 RNA A330074K22Rik 和下调铁蛋白沉积信号转导改善 UUO 小鼠的肾纤维化。
背景:慢性肾脏病(CKD)及其相关的终末期肾病(ESRD)是威胁人类福祉的重大健康问题。肾脏纤维化是各种 CKD 导致 ESRD 的共同特征和最终病理结果。黄芪三七方(A&P)是我们研究小组精制的复方制剂,经过十多年的临床应用,已证明能够改善各种急性或慢性肾脏疾病的炎症状态。然而,A&P改善肾脏纤维化的内在机制仍不清楚:方法:我们通过手术结扎单侧输尿管建立了小鼠模型,诱导体内肾损伤。方法:我们通过手术结扎单侧输尿管建立了小鼠模型,并利用肾脏原位电穿孔技术将低 LncRNA A33 表达的质粒植入小鼠体内,建立了单侧输尿管梗阻(UUO)小鼠模型。在体外,我们使用TGF-β1刺激原发性肾小管上皮细胞(pTEC)损伤,将siRNA-A33和pcDNA3.1-A33质粒转染至pTEC,分别敲除和过表达LncRNA A33,并对体外和体内模型进行A&P干预:结果:A&P能有效缓解小鼠肾脏纤维化。随后的研究结果表明,LncRNA A33在UUO小鼠肾脏和TGF-β1诱导的肾小管细胞中高表达。在肾脏原位电穿孔抑制 LncRNA A33 表达的质粒后发现,抑制 LncRNA A33 能显著改善 UUO 小鼠的肾脏纤维化。此外,A&P 能有效抑制 LncRNA A33 在体外和体内的表达。随后在肾小管上皮细胞中下调 LncRNA A33 导致许多纤维化标志物下调、LncRNA A33 被显著抑制,以及下游铁变态反应信号明显减少。细胞实验表明,A&P 通过抑制 LncRNA A33 和下调铁突变信号转导改善了 UUO 小鼠的肾纤维化:结论:通过抑制 LncRNA A33 和随后下调铁突变信号,A&P 显示出作为一种治疗方法改善 UUO 小鼠肾纤维化的潜力,为 CKD 提供了一种潜在的治疗途径。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
求助全文
约1分钟内获得全文 求助全文
来源期刊
BMC Complementary Medicine and Therapies
BMC Complementary Medicine and Therapies INTEGRATIVE & COMPLEMENTARY MEDICINE-
CiteScore
6.10
自引率
2.60%
发文量
300
审稿时长
19 weeks
×
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