Response by Owens and Deaton to Letter Regarding Article, "Dichotomous Roles of Smooth Muscle Cell-Derived MCP1 (Monocyte Chemoattractant Protein 1) in Development of Atherosclerosis".

Arteriosclerosis, thrombosis, and vascular biology(2023)

引用 0|浏览9
暂无评分
摘要
HomeArteriosclerosis, Thrombosis, and Vascular BiologyVol. 43, No. 1Response by Owens and Deaton to Letter Regarding Article, “Dichotomous Roles of Smooth Muscle Cell–Derived MCP1 (Monocyte Chemoattractant Protein 1) in Development of Atherosclerosis” Free AccessLetterPDF/EPUBAboutView PDFView EPUBSections ToolsAdd to favoritesDownload citationsTrack citationsPermissions ShareShare onFacebookTwitterLinked InMendeleyReddit Jump toFree AccessLetterPDF/EPUBResponse by Owens and Deaton to Letter Regarding Article, “Dichotomous Roles of Smooth Muscle Cell–Derived MCP1 (Monocyte Chemoattractant Protein 1) in Development of Atherosclerosis” Gary K. Owens and Rebecca A. Deaton Gary K. OwensGary K. Owens https://orcid.org/0000-0002-7119-9657 University of Virginia School of Medicine, Robert M. Berne Cardiovascular Research Center, Charlottesville, VA. Search for more papers by this author and Rebecca A. DeatonRebecca A. Deaton https://orcid.org/0000-0001-7569-1924 University of Virginia School of Medicine, Robert M. Berne Cardiovascular Research Center, Charlottesville, VA. Search for more papers by this author Originally published21 Dec 2022https://doi.org/10.1161/ATVBAHA.122.318638Arteriosclerosis, Thrombosis, and Vascular Biology. 2023;43:e64In Response:We thank Wang et al for their kind remarks and further insights regarding our Owsiany et al1 2022 Arteriosclerosis, Thrombosis, and Vascular Biology article. We fully agree with virtually all their comments including there being multiple possible cellular sources of MCP1 (monocyte chemoattractant protein 1) and secondary responses to its knockout in smooth muscle cells (SMC) including activating other cytokines. As is the case with any gene knockout study, the phenotype observed is a function not only of the initial gene knockout but also any downstream adaptive and maladaptive responses. Our observation that SMC heterozygous, but not homozygous MCP1 knockout mice, showed a paradoxical increase in plaque size and macrophage content is consistent with this idea since the homozygous SMC MCP1 knockout mice are more likely to undergo compensatory changes. Moreover, these results unveiled an unexpected beneficial role of SMC-derived MCPI in a Western diet–fed Apoe−/− mouse model. We present evidence, but certainly not proof, that this phenotype may be the result of systemic monocytosis secondary to loss of SMC-derived MCP1 causing increased release of monocytes from hematopoietic stem cell niches. However, as indicated by Wang et al, there are numerous other possibilities that remain to be tested. One interesting possibility is that initial production of MCP1 by SMC is beneficial because it promotes recruitment of monocytes-macrophages to early-stage atherosclerotic lesions as a means to remove oxidized lipids and apoptotic cells. However, as suggested by Randolph,2 the process may only be effective for a short time before the macrophages are overwhelmed, become engorged with lipids, fail to egress to the lymphatics, and give rise to foam cells and contribute to a chronic inflammatory state.We also agree with their comment that there is stage-specific regulation due to the different cellular sources of MCP1 at different stages of atherosclerosis. In support of this idea, we observed that mice with MCP1 knockout in the subset of SMC that have transitioned through a Lgals3 (Galectin3)+ state exhibit a phenotype virtually opposite to that of mice with MCP1 knockout in all SMC. SMC-Lgals3 dual recombinase MCP1 knockout mice had lesions with an increased ACTA2 (smooth muscle alpha-actin)+ 2 fibrous cap and decreased investment of Lgals3-transitioned SMCs, consistent with increased plaque stability. That is, MCP1 expression by this subset of SMC appears to be detrimental.Finally, we wish to caution readers to be careful in directly comparing results of cell-specific conditional gene knockout to global conventional gene knockout studies given the many undefined variables between such models including the timing of gene knockout and complex differences in the phenotypic state of lesion cells at any given time point. Our SMC-specific conditional MCP1 knockout mouse models provide valuable mechanistic insights regarding the contributions of SMC-derived MCP1 to lesion pathogenesis. However, these models are not intended or likely to predict overall effects of systemic MCP1 inhibitors.Article InformationDisclosures None.References1. Owsiany KM, Deaton RA, Soohoo KG, Tram NA, Owens GK. Dichotomous roles of smooth muscle cell-derived MCP1 (monocyte chemoattractant protein 1) in development of atherosclerosis.Arterioscler Thromb Vasc Biol. 2022; 42:942–956. doi: 10.1161/ATVBAHA.122.317882LinkGoogle Scholar2. Randolph GJ. Emigration of monocyte-derived cells to lymph nodes during resolution of inflammation and its failure in atherosclerosis.Curr.Opin.Lipidol. 2008 Oct; 19(5):462–468. doi: 10.1097/MOL.0b013e32830d5f09CrossrefMedlineGoogle Scholar Previous Back to top Next FiguresReferencesRelatedDetails January 2023Vol 43, Issue 1 Advertisement Article InformationMetrics © 2022 American Heart Association, Inc.https://doi.org/10.1161/ATVBAHA.122.318638PMID: 36542726 Originally publishedDecember 21, 2022 PDF download Advertisement
更多
查看译文
关键词
atherosclerosis”,monocyte chemoattractant protein,smooth muscle cell–derived,mcp1
AI 理解论文
溯源树
样例
生成溯源树,研究论文发展脉络
Chat Paper
正在生成论文摘要