在甲状腺眼病体外模型中,L型钙通道阻滞剂尼莫地平通过减轻TGF-β1诱导的钙反应发挥抗纤维化作用。

IF 4.1 1区 医学 Q1 OPHTHALMOLOGY Eye and Vision Pub Date : 2024-09-06 DOI:10.1186/s40662-024-00401-5
Qian Chen, Yuan Pan, Yunwei Hu, Guanyu Chen, Xiaoqing Chen, Yanyan Xie, Minzhen Wang, Zhuang Li, Jun Huang, Yuxun Shi, Haixiang Huang, Te Zhang, Mei Wang, Peng Zeng, Sha Wang, Rongxin Chen, Yongxin Zheng, Liuxueying Zhong, Huasheng Yang, Dan Liang
{"title":"在甲状腺眼病体外模型中,L型钙通道阻滞剂尼莫地平通过减轻TGF-β1诱导的钙反应发挥抗纤维化作用。","authors":"Qian Chen, Yuan Pan, Yunwei Hu, Guanyu Chen, Xiaoqing Chen, Yanyan Xie, Minzhen Wang, Zhuang Li, Jun Huang, Yuxun Shi, Haixiang Huang, Te Zhang, Mei Wang, Peng Zeng, Sha Wang, Rongxin Chen, Yongxin Zheng, Liuxueying Zhong, Huasheng Yang, Dan Liang","doi":"10.1186/s40662-024-00401-5","DOIUrl":null,"url":null,"abstract":"<p><strong>Background: </strong>Thyroid eye disease (TED) is a vision-threatening autoimmune disorder. Orbital tissue fibrosis leading to intractable complications remains a troublesome issue in TED management. Exploration of novel therapeutic targets and agents to ameliorate tissue fibrosis is crucial for TED. Recent work suggests that Ca<sup>2+</sup> signaling participates in tissue fibrosis. However, whether an alteration of Ca<sup>2+</sup> signaling has a role in fibrogenesis during TED remains unclear. In this study, we aimed to investigate the role of Ca<sup>2+</sup> signaling in the fibrogenesis process during TED and the potential therapeutic effects of a highly selective inhibitor of the L-type calcium channel (LTCC), nimodipine, through a TGF-β1 induced in vitro TED model.</p><p><strong>Methods: </strong>Primary culture of orbital fibroblasts (OFs) were established from orbital adipose connective tissues of patients with TED and healthy control donors. Real-time quantitative polymerase chain reaction (RT-qPCR) and RNA sequencing were used to assess the genes expression associated with LTCC in OFs. Flow cytometry, RT-qPCR, 5-ethynyl-2'-deoxyuridine (EdU) proliferation assay, wound healing assay and Western blot (WB) were used to assess the intracellular Ca<sup>2+</sup> response on TGF-β1 stimulation, and to evaluate the potential therapeutic effects of nimodipine in the TGF-β1 induced in vitro TED model. The roles of Ca<sup>2+</sup>/calmodulin-dependent protein kinase II (CaMKII) and signal transducer and activator of transcription 1 (STAT1) in fibrogenesis during TED were determined by immunohistochemistry, WB, flow cytometry and co-immunoprecipitation assay. Selective inhibitors were used to explore the downstream signaling pathways.</p><p><strong>Results: </strong>LTCC inhibitor nimodipine blocked the TGF-β1 induced intracellular Ca<sup>2+</sup> response and further reduced the expression of alpha-smooth muscle actin (α-SMA), collagen type I alpha 1 (Col1A1) and collagen type I alpha 2 (Col1A2) in OFs. Besides, nimodipine inhibited cell proliferation and migration of OFs. Moreover, our results provided evidence that activation of the CaMKII/STAT1 signaling pathway was involved in fibrogenesis during TED, and nimodipine inhibited the pro-fibrotic functions of OFs by down-regulating the CaMKII/STAT1 signaling pathway.</p><p><strong>Conclusions: </strong>TGF-β1 induces an LTCC-mediated Ca<sup>2+</sup> response, followed by activation of CaMKII/STAT1 signaling pathway, which promotes the pro-fibrotic functions of OFs and participates in fibrogenesis during TED. Nimodipine exerts potent anti-fibrotic benefits in vitro by suppressing the CaMKII/STAT1 signaling pathway. Our work deepens our understanding of the fibrogenesis process during TED and provides potential therapeutic targets and alternative candidate for TED.</p>","PeriodicalId":12194,"journal":{"name":"Eye and Vision","volume":null,"pages":null},"PeriodicalIF":4.1000,"publicationDate":"2024-09-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11378575/pdf/","citationCount":"0","resultStr":"{\"title\":\"An L-type calcium channel blocker nimodipine exerts anti-fibrotic effects by attenuating TGF-β1 induced calcium response in an in vitro model of thyroid eye disease.\",\"authors\":\"Qian Chen, Yuan Pan, Yunwei Hu, Guanyu Chen, Xiaoqing Chen, Yanyan Xie, Minzhen Wang, Zhuang Li, Jun Huang, Yuxun Shi, Haixiang Huang, Te Zhang, Mei Wang, Peng Zeng, Sha Wang, Rongxin Chen, Yongxin Zheng, Liuxueying Zhong, Huasheng Yang, Dan Liang\",\"doi\":\"10.1186/s40662-024-00401-5\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"<p><strong>Background: </strong>Thyroid eye disease (TED) is a vision-threatening autoimmune disorder. Orbital tissue fibrosis leading to intractable complications remains a troublesome issue in TED management. Exploration of novel therapeutic targets and agents to ameliorate tissue fibrosis is crucial for TED. Recent work suggests that Ca<sup>2+</sup> signaling participates in tissue fibrosis. However, whether an alteration of Ca<sup>2+</sup> signaling has a role in fibrogenesis during TED remains unclear. In this study, we aimed to investigate the role of Ca<sup>2+</sup> signaling in the fibrogenesis process during TED and the potential therapeutic effects of a highly selective inhibitor of the L-type calcium channel (LTCC), nimodipine, through a TGF-β1 induced in vitro TED model.</p><p><strong>Methods: </strong>Primary culture of orbital fibroblasts (OFs) were established from orbital adipose connective tissues of patients with TED and healthy control donors. Real-time quantitative polymerase chain reaction (RT-qPCR) and RNA sequencing were used to assess the genes expression associated with LTCC in OFs. Flow cytometry, RT-qPCR, 5-ethynyl-2'-deoxyuridine (EdU) proliferation assay, wound healing assay and Western blot (WB) were used to assess the intracellular Ca<sup>2+</sup> response on TGF-β1 stimulation, and to evaluate the potential therapeutic effects of nimodipine in the TGF-β1 induced in vitro TED model. The roles of Ca<sup>2+</sup>/calmodulin-dependent protein kinase II (CaMKII) and signal transducer and activator of transcription 1 (STAT1) in fibrogenesis during TED were determined by immunohistochemistry, WB, flow cytometry and co-immunoprecipitation assay. Selective inhibitors were used to explore the downstream signaling pathways.</p><p><strong>Results: </strong>LTCC inhibitor nimodipine blocked the TGF-β1 induced intracellular Ca<sup>2+</sup> response and further reduced the expression of alpha-smooth muscle actin (α-SMA), collagen type I alpha 1 (Col1A1) and collagen type I alpha 2 (Col1A2) in OFs. Besides, nimodipine inhibited cell proliferation and migration of OFs. Moreover, our results provided evidence that activation of the CaMKII/STAT1 signaling pathway was involved in fibrogenesis during TED, and nimodipine inhibited the pro-fibrotic functions of OFs by down-regulating the CaMKII/STAT1 signaling pathway.</p><p><strong>Conclusions: </strong>TGF-β1 induces an LTCC-mediated Ca<sup>2+</sup> response, followed by activation of CaMKII/STAT1 signaling pathway, which promotes the pro-fibrotic functions of OFs and participates in fibrogenesis during TED. Nimodipine exerts potent anti-fibrotic benefits in vitro by suppressing the CaMKII/STAT1 signaling pathway. Our work deepens our understanding of the fibrogenesis process during TED and provides potential therapeutic targets and alternative candidate for TED.</p>\",\"PeriodicalId\":12194,\"journal\":{\"name\":\"Eye and Vision\",\"volume\":null,\"pages\":null},\"PeriodicalIF\":4.1000,\"publicationDate\":\"2024-09-06\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11378575/pdf/\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"Eye and Vision\",\"FirstCategoryId\":\"3\",\"ListUrlMain\":\"https://doi.org/10.1186/s40662-024-00401-5\",\"RegionNum\":1,\"RegionCategory\":\"医学\",\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"\",\"PubModel\":\"\",\"JCR\":\"Q1\",\"JCRName\":\"OPHTHALMOLOGY\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"Eye and Vision","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.1186/s40662-024-00401-5","RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"OPHTHALMOLOGY","Score":null,"Total":0}
引用次数: 0

摘要

背景:甲状腺眼病(TED)是一种威胁视力的自身免疫性疾病:甲状腺眼病(TED)是一种威胁视力的自身免疫性疾病。眼眶组织纤维化导致难以治愈的并发症,仍然是治疗 TED 的一个棘手问题。探索新的治疗靶点和药物以改善组织纤维化对 TED 至关重要。最近的研究表明,Ca2+ 信号转导参与了组织纤维化。然而,Ca2+ 信号的改变是否在 TED 期间的纤维化中发挥作用仍不清楚。在本研究中,我们旨在通过TGF-β1诱导的体外TED模型,研究Ca2+信号在TED期间纤维化过程中的作用,以及L型钙通道(LTCC)高选择性抑制剂尼莫地平的潜在治疗效果:方法:从 TED 患者和健康对照供体的眼眶脂肪结缔组织中建立眼眶成纤维细胞(OFS)的原代培养。采用实时定量聚合酶链反应(RT-qPCR)和 RNA 测序评估眼眶成纤维细胞中与 LTCC 相关的基因表达。流式细胞术、RT-qPCR、5-乙炔基-2'-脱氧尿苷(EdU)增殖试验、伤口愈合试验和 Western 印迹(WB)用于评估细胞内 Ca2+ 对 TGF-β1 刺激的反应,并评估尼莫地平在 TGF-β1 诱导的体外 TED 模型中的潜在治疗效果。通过免疫组化、WB、流式细胞术和共免疫沉淀实验确定了TED过程中Ca2+/钙调蛋白依赖性蛋白激酶II(CaMKII)和信号转导及激活转录1(STAT1)在纤维化过程中的作用。使用选择性抑制剂探索下游信号通路:结果:LTCC抑制剂尼莫地平阻断了TGF-β1诱导的细胞内Ca2+反应,并进一步降低了OFs中α-平滑肌肌动蛋白(α-SMA)、Ⅰ型胶原α1(Col1A1)和Ⅰ型胶原α2(Col1A2)的表达。此外,尼莫地平还能抑制 OFs 的细胞增殖和迁移。尼莫地平通过下调 CaMKII/STAT1 信号通路抑制了 OFs 的促纤维化功能:结论:TGF-β1诱导LTCC介导的Ca2+反应,继而激活CaMKII/STAT1信号通路,促进OFs的促纤维化功能,并参与TED过程中的纤维生成。尼莫地平通过抑制 CaMKII/STAT1 信号通路,在体外发挥了强大的抗纤维化作用。我们的研究加深了对TED过程中纤维生成过程的理解,并为TED提供了潜在的治疗靶点和替代候选药物。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
查看原文
分享 分享
微信好友 朋友圈 QQ好友 复制链接
本刊更多论文
An L-type calcium channel blocker nimodipine exerts anti-fibrotic effects by attenuating TGF-β1 induced calcium response in an in vitro model of thyroid eye disease.

Background: Thyroid eye disease (TED) is a vision-threatening autoimmune disorder. Orbital tissue fibrosis leading to intractable complications remains a troublesome issue in TED management. Exploration of novel therapeutic targets and agents to ameliorate tissue fibrosis is crucial for TED. Recent work suggests that Ca2+ signaling participates in tissue fibrosis. However, whether an alteration of Ca2+ signaling has a role in fibrogenesis during TED remains unclear. In this study, we aimed to investigate the role of Ca2+ signaling in the fibrogenesis process during TED and the potential therapeutic effects of a highly selective inhibitor of the L-type calcium channel (LTCC), nimodipine, through a TGF-β1 induced in vitro TED model.

Methods: Primary culture of orbital fibroblasts (OFs) were established from orbital adipose connective tissues of patients with TED and healthy control donors. Real-time quantitative polymerase chain reaction (RT-qPCR) and RNA sequencing were used to assess the genes expression associated with LTCC in OFs. Flow cytometry, RT-qPCR, 5-ethynyl-2'-deoxyuridine (EdU) proliferation assay, wound healing assay and Western blot (WB) were used to assess the intracellular Ca2+ response on TGF-β1 stimulation, and to evaluate the potential therapeutic effects of nimodipine in the TGF-β1 induced in vitro TED model. The roles of Ca2+/calmodulin-dependent protein kinase II (CaMKII) and signal transducer and activator of transcription 1 (STAT1) in fibrogenesis during TED were determined by immunohistochemistry, WB, flow cytometry and co-immunoprecipitation assay. Selective inhibitors were used to explore the downstream signaling pathways.

Results: LTCC inhibitor nimodipine blocked the TGF-β1 induced intracellular Ca2+ response and further reduced the expression of alpha-smooth muscle actin (α-SMA), collagen type I alpha 1 (Col1A1) and collagen type I alpha 2 (Col1A2) in OFs. Besides, nimodipine inhibited cell proliferation and migration of OFs. Moreover, our results provided evidence that activation of the CaMKII/STAT1 signaling pathway was involved in fibrogenesis during TED, and nimodipine inhibited the pro-fibrotic functions of OFs by down-regulating the CaMKII/STAT1 signaling pathway.

Conclusions: TGF-β1 induces an LTCC-mediated Ca2+ response, followed by activation of CaMKII/STAT1 signaling pathway, which promotes the pro-fibrotic functions of OFs and participates in fibrogenesis during TED. Nimodipine exerts potent anti-fibrotic benefits in vitro by suppressing the CaMKII/STAT1 signaling pathway. Our work deepens our understanding of the fibrogenesis process during TED and provides potential therapeutic targets and alternative candidate for TED.

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
Eye and Vision
Eye and Vision OPHTHALMOLOGY-
CiteScore
8.60
自引率
2.40%
发文量
89
审稿时长
15 weeks
期刊介绍: Eye and Vision is an open access, peer-reviewed journal for ophthalmologists and visual science specialists. It welcomes research articles, reviews, methodologies, commentaries, case reports, perspectives and short reports encompassing all aspects of eye and vision. Topics of interest include but are not limited to: current developments of theoretical, experimental and clinical investigations in ophthalmology, optometry and vision science which focus on novel and high-impact findings on central issues pertaining to biology, pathophysiology and etiology of eye diseases as well as advances in diagnostic techniques, surgical treatment, instrument updates, the latest drug findings, results of clinical trials and research findings. It aims to provide ophthalmologists and visual science specialists with the latest developments in theoretical, experimental and clinical investigations in eye and vision.
期刊最新文献
An L-type calcium channel blocker nimodipine exerts anti-fibrotic effects by attenuating TGF-β1 induced calcium response in an in vitro model of thyroid eye disease. Efficacy of orthokeratology lens with the modified small treatment zone on myopia progression and visual quality: a randomized clinical trial. Nominal and achieved stromal ablation depth after myopic transepithelial photorefractive keratectomy: implications for residual stromal thickness calculation. Quantitative analysis of peripapillary capillary volume using dense B-scan OCT angiography in normal and diabetic retina. Correction: Prognostic potentials of AI in ophthalmology: systemic disease forecasting via retinal imaging.
×
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
现在去查看 取消
×
提示
确定
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
已复制链接
已复制链接
快去分享给好友吧!
我知道了
×
扫码分享
扫码分享
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 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 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1