Melatonin induces RAW264.7 cell apoptosis via the BMAL1/ROS/MAPK-p38 pathway to improve postmenopausal osteoporosis.

IF 4.7 2区 医学 Q2 CELL & TISSUE ENGINEERING Bone & Joint Research Pub Date : 2023-11-01 DOI:10.1302/2046-3758.1211.BJR-2022-0425.R3
Xiaochuan Wang, Wen Jiang, Kexin Pan, Lin Tao, Yue Zhu
{"title":"Melatonin induces RAW264.7 cell apoptosis via the BMAL1/ROS/MAPK-p38 pathway to improve postmenopausal osteoporosis.","authors":"Xiaochuan Wang,&nbsp;Wen Jiang,&nbsp;Kexin Pan,&nbsp;Lin Tao,&nbsp;Yue Zhu","doi":"10.1302/2046-3758.1211.BJR-2022-0425.R3","DOIUrl":null,"url":null,"abstract":"<p><strong>Aims: </strong>Currently, the effect of drug treatment for osteoporosis is relatively poor, and the side effects are numerous and serious. Melatonin is a potential drug to improve bone mass in postmenopausal women. Unfortunately, the mechanism by which melatonin improves bone metabolism remains unclear. The aim of this study was to further investigate the potential mechanism of melatonin in the treatment of osteoporosis.</p><p><strong>Methods: </strong>The effects of melatonin on mitochondrial apoptosis protein, bmal1 gene, and related pathway proteins of RAW264.7 (mouse mononuclear macrophage leukaemia cells) were analyzed by western blot. Cell Counting Kit-8 was used to evaluate the effect of melatonin on cell viability. Flow cytometry was used to evaluate the effect of melatonin on the apoptosis of RAW264.7 cells and mitochondrial membrane potential. A reactive oxygen species (ROS) detection kit was used to evaluate the level of ROS in osteoclast precursors. We used bmal1-small interfering RNAs (siRNAs) to downregulate the <i>Bmal1</i> gene. We established a postmenopausal mouse model and verified the effect of melatonin on the bone mass of postmenopausal osteoporosis in mice via micro-CT. <i>Bmal1</i> lentiviral activation particles were used to establish an in vitro model of overexpression of the bmal1 gene.</p><p><strong>Results: </strong>Melatonin promoted apoptosis of RAW264.7 cells and increased the expression of BMAL1 to inhibit the activation of ROS and phosphorylation of mitogen-activated protein kinase (MAPK)-p38. Silencing the bmal1 gene weakened the above effects of melatonin. After that, we used dehydrocorydaline (DHC) to enhance the activation of MAPK-p38, and the effects of melatonin on reducing ROS levels and promoting apoptosis of RAW264.7 cells were also blocked. Then, we constructed a mouse model of postmenopausal osteoporosis and administered melatonin. The results showed that melatonin improves bone loss in ovariectomized mice. Finally, we established a model of overexpression of the bmal1 gene, and these results suggest that the bmal1 gene can regulate ROS activity and change the level of the MAPK-p38 signalling pathway.</p><p><strong>Conclusion: </strong>Our study confirmed that melatonin promotes the apoptosis of RAW264.7 cells through BMAL1/ROS/MAPK-p38, and revealed the therapeutic effect and mechanism of melatonin in postmenopausal osteoporosis. This finding enriches BMAL1 as a potential target for the treatment of osteoporosis and the pathogenesis of postmenopausal osteoporosis.</p>","PeriodicalId":9074,"journal":{"name":"Bone & Joint Research","volume":null,"pages":null},"PeriodicalIF":4.7000,"publicationDate":"2023-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10618049/pdf/","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Bone & Joint Research","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.1302/2046-3758.1211.BJR-2022-0425.R3","RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q2","JCRName":"CELL & TISSUE ENGINEERING","Score":null,"Total":0}
引用次数: 0

Abstract

Aims: Currently, the effect of drug treatment for osteoporosis is relatively poor, and the side effects are numerous and serious. Melatonin is a potential drug to improve bone mass in postmenopausal women. Unfortunately, the mechanism by which melatonin improves bone metabolism remains unclear. The aim of this study was to further investigate the potential mechanism of melatonin in the treatment of osteoporosis.

Methods: The effects of melatonin on mitochondrial apoptosis protein, bmal1 gene, and related pathway proteins of RAW264.7 (mouse mononuclear macrophage leukaemia cells) were analyzed by western blot. Cell Counting Kit-8 was used to evaluate the effect of melatonin on cell viability. Flow cytometry was used to evaluate the effect of melatonin on the apoptosis of RAW264.7 cells and mitochondrial membrane potential. A reactive oxygen species (ROS) detection kit was used to evaluate the level of ROS in osteoclast precursors. We used bmal1-small interfering RNAs (siRNAs) to downregulate the Bmal1 gene. We established a postmenopausal mouse model and verified the effect of melatonin on the bone mass of postmenopausal osteoporosis in mice via micro-CT. Bmal1 lentiviral activation particles were used to establish an in vitro model of overexpression of the bmal1 gene.

Results: Melatonin promoted apoptosis of RAW264.7 cells and increased the expression of BMAL1 to inhibit the activation of ROS and phosphorylation of mitogen-activated protein kinase (MAPK)-p38. Silencing the bmal1 gene weakened the above effects of melatonin. After that, we used dehydrocorydaline (DHC) to enhance the activation of MAPK-p38, and the effects of melatonin on reducing ROS levels and promoting apoptosis of RAW264.7 cells were also blocked. Then, we constructed a mouse model of postmenopausal osteoporosis and administered melatonin. The results showed that melatonin improves bone loss in ovariectomized mice. Finally, we established a model of overexpression of the bmal1 gene, and these results suggest that the bmal1 gene can regulate ROS activity and change the level of the MAPK-p38 signalling pathway.

Conclusion: Our study confirmed that melatonin promotes the apoptosis of RAW264.7 cells through BMAL1/ROS/MAPK-p38, and revealed the therapeutic effect and mechanism of melatonin in postmenopausal osteoporosis. This finding enriches BMAL1 as a potential target for the treatment of osteoporosis and the pathogenesis of postmenopausal osteoporosis.

Abstract Image

Abstract Image

Abstract Image

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
褪黑素通过BMAL1/ROS/MAPK-p38途径诱导RAW264.7细胞凋亡,改善绝经后骨质疏松症。
目的:目前,药物治疗骨质疏松症的效果相对较差,副作用多且严重。褪黑素是一种潜在的药物,可以改善绝经后妇女的骨量。不幸的是,褪黑激素改善骨代谢的机制尚不清楚。本研究的目的是进一步探讨褪黑素治疗骨质疏松症的潜在机制。方法:采用蛋白质印迹法分析褪黑素对小鼠单核巨噬细胞白血病细胞RAW264.7线粒体凋亡蛋白、bmal1基因及相关通路蛋白的影响。细胞计数试剂盒-8用于评估褪黑素对细胞活力的影响。流式细胞术评价褪黑素对RAW264.7细胞凋亡和线粒体膜电位的影响。活性氧(ROS)检测试剂盒用于评估破骨细胞前体中ROS的水平。我们使用bmal1小干扰RNA(siRNA)下调bmal1基因。我们建立了绝经后小鼠模型,并通过显微CT验证了褪黑素对小鼠绝经后骨质疏松症骨量的影响。Bmal1慢病毒激活颗粒用于建立Bmal1基因过表达的体外模型。结果:褪黑素促进RAW264.7细胞凋亡,增加BMAL1的表达,抑制ROS的活化和丝裂原活化蛋白激酶(MAPK)-p38的磷酸化。bmal1基因的沉默削弱了褪黑激素的上述作用。之后,我们使用脱氢corydaline(DHC)来增强MAPK-p38的激活,褪黑素对降低ROS水平和促进RAW264.7细胞凋亡的作用也被阻断。然后,我们构建了绝经后骨质疏松症小鼠模型,并给予褪黑素。结果表明,褪黑素可改善去卵巢小鼠的骨丢失。最后,我们建立了bmal1基因过表达的模型,这些结果表明bmal1可以调节ROS活性并改变MAPK-p38信号通路的水平。结论:我们的研究证实了褪黑素通过BMAL1/ROS/MAPK-p38促进RAW264.7细胞凋亡,并揭示了褪黑素对绝经后骨质疏松症的治疗作用和机制。这一发现丰富了BMAL1作为治疗骨质疏松症和绝经后骨质疏松症发病机制的潜在靶点。
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 去求助
来源期刊
Bone & Joint Research
Bone & Joint Research CELL & TISSUE ENGINEERING-ORTHOPEDICS
CiteScore
7.40
自引率
23.90%
发文量
156
审稿时长
12 weeks
期刊介绍: The gold open access journal for the musculoskeletal sciences. Included in PubMed and available in PubMed Central.
期刊最新文献
Identification of age-related genes in rotator cuff tendon. The interactions of macrophages, lymphocytes, and mesenchymal stem cells during bone regeneration. Mechanical influence of facet tropism in patients with chronic discogenic pain disorder. Sonodynamic effect based on vancomycin-loaded microbubbles or meropenem-loaded microbubbles enhances elimination of different biofilms and bactericidal efficacy. Guanylate cyclase promotes osseointegration by inhibiting oxidative stress and inflammation in aged rats with iron overload.
×
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