4.8 Article

In-frame deletion of SMC5 related with the phenotype of primordial dwarfism, chromosomal instability and insulin resistance

期刊

出版社

JOHN WILEY & SONS LTD
DOI: 10.1002/ctm2.1007

关键词

insulin resistance; primordial dwarfism; SMC5 mutation

向作者/读者索取更多资源

The SMC5/6 complex is found to play a crucial role in maintaining genome stability, but its relationship with human diseases has not been well described. Through whole-exome sequencing and other experimental methods, a homozygous in-frame deletion of Arg372 in SMC5 was identified in an adult patient with microcephalic primordial dwarfism, chromosomal instability, and insulin resistance. Animal models, including smc5 knockout zebrafish and Smc5(K371del) knock-in mouse, were created to investigate the phenotypic characteristics and potential mechanisms. The findings suggest a link between growth disorders and metabolic homeostasis imbalance through defective cell survival and differentiation.
BackgroundSMC5/6 complex plays a vital role in maintaining genome stability, yet the relationship with human diseases has not been described. MethodsSMC5 variation was identified through whole-exome sequencing (WES) and verified by Sanger sequencing. Immunoprecipitation, cytogenetic analysis, fluorescence activated cell sorting (FACS) and electron microscopy were used to elucidate the cellular consequences of patient's cells. smc5 knockout (KO) zebrafish and Smc5(K371del) knock-in mouse models were generated by CRISPR-Cas9. RNA-seq, quantitative real-time PCR (qPCR), western blot, microquantitative computed tomography (microCT) and histology were used to explore phenotypic characteristics and potential mechanisms of the animal models. The effects of Smc5 knockdown on mitotic clonal expansion (MCE) during adipogenesis were investigated through Oil Red O staining, proliferation and apoptosis assays in vitro. ResultsWe identified a homozygous in-frame deletion of Arg372 in SMC5, one of the core subunits of the SMC5/6 complex, from an adult patient with microcephalic primordial dwarfism, chromosomal instability and insulin resistance. SMC5 mutation disrupted its interaction with its interacting protein NSMCE2, leading to defects in DNA repair and chromosomal instability in patient fibroblasts. Smc5 KO zebrafish showed microcephaly, short length and disturbed glucose metabolism. Smc5 depletion triggers a p53-related apoptosis, as concomitant deletion of the p53 rescued growth defects phenotype in zebrafish. An smc5(K371del) knock-in mouse model exhibited high mortality, severe growth restriction and fat loss. In 3T3-L1 cells, the knockdown of smc5 results in impaired MCE, a crucial step in adipogenesis. This finding implies that defective cell survival and differentiation is an important mechanism linking growth disorders and metabolic homeostasis imbalance.

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

4.8
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