4.7 Article

Circulating macrophages as the mechanistic link between mosaic loss of Y-chromosome and cardiac disease

期刊

CELL AND BIOSCIENCE
卷 13, 期 1, 页码 -

出版社

BMC
DOI: 10.1186/s13578-023-01075-7

关键词

Mosaic loss of Y-chromosome (mLOY); Cardiac fibrosis; Hematopoietic macrophage; Profibrotic regulons; Proliferation of cardiac fibroblast; Age-related cardiac disease

向作者/读者索取更多资源

Research shows that mosaic loss of Y-chromosome in peripheral leukocytes is associated with male age-associated diseases. This study provides the first evidence linking mosaic loss of Y-chromosome to cardiac fibrosis and identifies the underlying mechanism, offering new insights into the relationship between mosaic loss of Y-chromosome and male age-related diseases.
BackgroundGenetics evidences have long linked mosaic loss of Y-chromosome (mLOY) in peripheral leukocytes with a wide range of male age-associated diseases. However, a lack of cellular and molecular mechanistic explanations for this link has limited further investigation into the relationship between mLOY and male age-related disease. Excitingly, Sano et al. have provided the first piece of evidence directly linking mLOY to cardiac fibrosis through mLOY enriched profibrotic transforming growth factor & beta;1 (TGF-& beta;1) regulons in hematopoietic macrophages along with suppressed interleukin-1 & beta; (IL-1 & beta;) proinflammatory regulons. The results of this novel finding can be extrapolated to other disease related to mLOY, such as cancer, cardiac disease, and age-related macular degeneration.ResultsSano et al. used a CRISPR-Cas9 gRNAs gene editing induced Y-chromosome ablation mouse model to assess results of a UK biobank prospective analysis implicating the Y-chromosome in male age-related disease. Using this in vivo model, Sano et al. showed that hematopoietic mLOY accelerated cardiac fibrosis and heart failure in male mice through profibrotic pathways. This process was linked to monocyte-macrophage differentiation during hematopoietic development. Mice confirmed to have mLOY in leukocytes, by loss of Y-chromosome genes Kdm5d, Uty, Eif2s3y, and Ddx3y, at similar percentages to the human population were shown to have accelerated rates of interstitial and perivascular fibrosis and abnormal echocardiograms. These mice also recovered poorly from the transverse aortic constriction (TAC) model of heart failure and developed left ventricular dysfunction at higher rates. This was attributed to aberrant proliferation of cardiac MEF-SK4 + fibroblasts promoted by mLOY macrophages enriched in profibrotic regulons and lacking in proinflammatory regulons. These pro-fibrotic macrophages localized to heart and eventually resulted in cardiac fibrosis via enhanced TGF-& beta;1 and suppressed IL-1 & beta; signaling. Furthermore, treatment of mLOY mice with TGF & beta;1 neutralizing antibody was able to improve their cardiac function. This study by Sano et al. was able to provide a causative link between the known association between mLOY and male cardiac disease morbidity and mortality for the first time, and thereby provide a new target for improving human health.ConclusionsUsing a CRISPR-Cas9 induced Y-chromosome ablation mouse model, Sano et al. has proven mosaic loss of Y-chromosome in peripheral myeloid cells to have a causative effect on male mobility and mortality due to male age-related cardiac disease. They traced the mechanism of this effect to hyper-expression of the profibrotic TGF-& beta;1 and reduced pro-inflammatory IL-1 & beta; signaling, attenuation of which could provide another potential strategy in improving outcomes against age-related diseases in men.

作者

我是这篇论文的作者
点击您的名字以认领此论文并将其添加到您的个人资料中。

评论

主要评分

4.7
评分不足

次要评分

新颖性
-
重要性
-
科学严谨性
-
评价这篇论文

推荐

暂无数据
暂无数据