HIF-1α/PLOD2轴整合了细胞外基质组织和细胞代谢,导致肌肉骨骼修复失常。

IF 14.3 1区 医学 Q1 CELL & TISSUE ENGINEERING
Heeseog Kang, Amy L Strong, Yuxiao Sun, Lei Guo, Conan Juan, Alec C Bancroft, Ji Hae Choi, Chase A Pagani, Aysel A Fernandes, Michael Woodard, Juhoon Lee, Sowmya Ramesh, Aaron W James, David Hudson, Kevin N Dalby, Lin Xu, Robert J Tower, Benjamin Levi
{"title":"HIF-1α/PLOD2轴整合了细胞外基质组织和细胞代谢,导致肌肉骨骼修复失常。","authors":"Heeseog Kang, Amy L Strong, Yuxiao Sun, Lei Guo, Conan Juan, Alec C Bancroft, Ji Hae Choi, Chase A Pagani, Aysel A Fernandes, Michael Woodard, Juhoon Lee, Sowmya Ramesh, Aaron W James, David Hudson, Kevin N Dalby, Lin Xu, Robert J Tower, Benjamin Levi","doi":"10.1038/s41413-024-00320-0","DOIUrl":null,"url":null,"abstract":"<p><p>While hypoxic signaling has been shown to play a role in many cellular processes, its role in metabolism-linked extracellular matrix (ECM) organization and downstream processes of cell fate after musculoskeletal injury remains to be determined. Heterotopic ossification (HO) is a debilitating condition where abnormal bone formation occurs within extra-skeletal tissues. Hypoxia and hypoxia-inducible factor 1α (HIF-1α) activation have been shown to promote HO. However, the underlying molecular mechanisms by which the HIF-1α pathway in mesenchymal progenitor cells (MPCs) contributes to pathologic bone formation remain to be elucidated. Here, we used a proven mouse injury-induced HO model to investigate the role of HIF-1α on aberrant cell fate. Using single-cell RNA sequencing (scRNA-seq) and spatial transcriptomics analyses of the HO site, we found that collagen ECM organization is the most highly up-regulated biological process in MPCs. Zeugopod mesenchymal cell-specific deletion of Hif1α (Hoxa11-CreER<sup>T2</sup>; Hif1a<sup>fl/fl</sup>) significantly mitigated HO in vivo. ScRNA-seq analysis of these Hoxa11-CreER<sup>T2</sup>; Hif1a<sup>fl/fl</sup> mice identified the PLOD2/LOX pathway for collagen cross-linking as downstream of the HIF-1α regulation of HO. Importantly, our scRNA-seq data and mechanistic studies further uncovered that glucose metabolism in MPCs is most highly impacted by HIF-1α deletion. From a translational aspect, a pan-LOX inhibitor significantly decreased HO. A newly screened compound revealed that the inhibition of PLOD2 activity in MPCs significantly decreased osteogenic differentiation and glycolytic metabolism. This suggests that the HIF-1α/PLOD2/LOX axis linked to metabolism regulates HO-forming MPC fate. These results suggest that the HIF-1α/PLOD2/LOX pathway represents a promising strategy to mitigate HO formation.</p>","PeriodicalId":9134,"journal":{"name":"Bone Research","volume":null,"pages":null},"PeriodicalIF":14.3000,"publicationDate":"2024-03-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10933265/pdf/","citationCount":"0","resultStr":"{\"title\":\"The HIF-1α/PLOD2 axis integrates extracellular matrix organization and cell metabolism leading to aberrant musculoskeletal repair.\",\"authors\":\"Heeseog Kang, Amy L Strong, Yuxiao Sun, Lei Guo, Conan Juan, Alec C Bancroft, Ji Hae Choi, Chase A Pagani, Aysel A Fernandes, Michael Woodard, Juhoon Lee, Sowmya Ramesh, Aaron W James, David Hudson, Kevin N Dalby, Lin Xu, Robert J Tower, Benjamin Levi\",\"doi\":\"10.1038/s41413-024-00320-0\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"<p><p>While hypoxic signaling has been shown to play a role in many cellular processes, its role in metabolism-linked extracellular matrix (ECM) organization and downstream processes of cell fate after musculoskeletal injury remains to be determined. Heterotopic ossification (HO) is a debilitating condition where abnormal bone formation occurs within extra-skeletal tissues. Hypoxia and hypoxia-inducible factor 1α (HIF-1α) activation have been shown to promote HO. However, the underlying molecular mechanisms by which the HIF-1α pathway in mesenchymal progenitor cells (MPCs) contributes to pathologic bone formation remain to be elucidated. Here, we used a proven mouse injury-induced HO model to investigate the role of HIF-1α on aberrant cell fate. Using single-cell RNA sequencing (scRNA-seq) and spatial transcriptomics analyses of the HO site, we found that collagen ECM organization is the most highly up-regulated biological process in MPCs. Zeugopod mesenchymal cell-specific deletion of Hif1α (Hoxa11-CreER<sup>T2</sup>; Hif1a<sup>fl/fl</sup>) significantly mitigated HO in vivo. ScRNA-seq analysis of these Hoxa11-CreER<sup>T2</sup>; Hif1a<sup>fl/fl</sup> mice identified the PLOD2/LOX pathway for collagen cross-linking as downstream of the HIF-1α regulation of HO. Importantly, our scRNA-seq data and mechanistic studies further uncovered that glucose metabolism in MPCs is most highly impacted by HIF-1α deletion. From a translational aspect, a pan-LOX inhibitor significantly decreased HO. A newly screened compound revealed that the inhibition of PLOD2 activity in MPCs significantly decreased osteogenic differentiation and glycolytic metabolism. This suggests that the HIF-1α/PLOD2/LOX axis linked to metabolism regulates HO-forming MPC fate. These results suggest that the HIF-1α/PLOD2/LOX pathway represents a promising strategy to mitigate HO formation.</p>\",\"PeriodicalId\":9134,\"journal\":{\"name\":\"Bone Research\",\"volume\":null,\"pages\":null},\"PeriodicalIF\":14.3000,\"publicationDate\":\"2024-03-12\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10933265/pdf/\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"Bone Research\",\"FirstCategoryId\":\"3\",\"ListUrlMain\":\"https://doi.org/10.1038/s41413-024-00320-0\",\"RegionNum\":1,\"RegionCategory\":\"医学\",\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"\",\"PubModel\":\"\",\"JCR\":\"Q1\",\"JCRName\":\"CELL & TISSUE ENGINEERING\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"Bone Research","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.1038/s41413-024-00320-0","RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"CELL & TISSUE ENGINEERING","Score":null,"Total":0}
引用次数: 0

摘要

虽然缺氧信号已被证明在许多细胞过程中发挥作用,但它在肌肉骨骼损伤后与新陈代谢相关的细胞外基质(ECM)组织和细胞命运的下游过程中的作用仍有待确定。异位骨化(HO)是一种使人衰弱的病症,即骨骼外组织内出现异常骨形成。缺氧和缺氧诱导因子 1α (HIF-1α)激活已被证明可促进异位骨化。然而,间充质祖细胞(MPCs)中的HIF-1α通路导致病理性骨形成的潜在分子机制仍有待阐明。在这里,我们使用一种成熟的小鼠损伤诱导 HO 模型来研究 HIF-1α 对异常细胞命运的作用。利用单细胞 RNA 测序(scRNA-seq)和 HO 位点的空间转录组学分析,我们发现胶原 ECM 组织是间充质干细胞中上调率最高的生物过程。Zeugopod间充质细胞特异性缺失Hif1α(Hoxa11-CreERT2;Hif1afl/fl)可显著减轻体内的HO。对这些 Hoxa11-CreERT2; Hif1afl/fl 小鼠进行的 ScRNA 序列分析发现,胶原交联的 PLOD2/LOX 通路是 HIF-1α 调节 HO 的下游通路。重要的是,我们的scRNA-seq数据和机理研究进一步发现,HIF-1α缺失对MPCs的葡萄糖代谢影响最大。从转化方面来看,泛LOX抑制剂能显著降低HO。一种新筛选出的化合物显示,抑制 MPCs 中 PLOD2 的活性可显著降低成骨分化和糖代谢。这表明,与新陈代谢相关的HIF-1α/PLOD2/LOX轴调节HO形成MPC的命运。这些结果表明,HIF-1α/PLOD2/LOX途径是缓解HO形成的一种有前途的策略。
本文章由计算机程序翻译,如有差异,请以英文原文为准。

The HIF-1α/PLOD2 axis integrates extracellular matrix organization and cell metabolism leading to aberrant musculoskeletal repair.

The HIF-1α/PLOD2 axis integrates extracellular matrix organization and cell metabolism leading to aberrant musculoskeletal repair.

While hypoxic signaling has been shown to play a role in many cellular processes, its role in metabolism-linked extracellular matrix (ECM) organization and downstream processes of cell fate after musculoskeletal injury remains to be determined. Heterotopic ossification (HO) is a debilitating condition where abnormal bone formation occurs within extra-skeletal tissues. Hypoxia and hypoxia-inducible factor 1α (HIF-1α) activation have been shown to promote HO. However, the underlying molecular mechanisms by which the HIF-1α pathway in mesenchymal progenitor cells (MPCs) contributes to pathologic bone formation remain to be elucidated. Here, we used a proven mouse injury-induced HO model to investigate the role of HIF-1α on aberrant cell fate. Using single-cell RNA sequencing (scRNA-seq) and spatial transcriptomics analyses of the HO site, we found that collagen ECM organization is the most highly up-regulated biological process in MPCs. Zeugopod mesenchymal cell-specific deletion of Hif1α (Hoxa11-CreERT2; Hif1afl/fl) significantly mitigated HO in vivo. ScRNA-seq analysis of these Hoxa11-CreERT2; Hif1afl/fl mice identified the PLOD2/LOX pathway for collagen cross-linking as downstream of the HIF-1α regulation of HO. Importantly, our scRNA-seq data and mechanistic studies further uncovered that glucose metabolism in MPCs is most highly impacted by HIF-1α deletion. From a translational aspect, a pan-LOX inhibitor significantly decreased HO. A newly screened compound revealed that the inhibition of PLOD2 activity in MPCs significantly decreased osteogenic differentiation and glycolytic metabolism. This suggests that the HIF-1α/PLOD2/LOX axis linked to metabolism regulates HO-forming MPC fate. These results suggest that the HIF-1α/PLOD2/LOX pathway represents a promising strategy to mitigate HO formation.

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
Bone Research
Bone Research CELL & TISSUE ENGINEERING-
CiteScore
20.00
自引率
4.70%
发文量
289
审稿时长
20 weeks
期刊介绍: Established in 2013, Bone Research is a newly-founded English-language periodical that centers on the basic and clinical facets of bone biology, pathophysiology, and regeneration. It is dedicated to championing key findings emerging from both basic investigations and clinical research concerning bone-related topics. The journal's objective is to globally disseminate research in bone-related physiology, pathology, diseases, and treatment, contributing to the advancement of knowledge in this field.
×
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