4.7 Article

Editing TINF2 as a potential therapeutic approach to restore telomere length in dyskeratosis congenita

期刊

BLOOD
卷 140, 期 6, 页码 608-618

出版社

AMER SOC HEMATOLOGY
DOI: 10.1182/blood.2021013750

关键词

-

资金

  1. American Cancer Society [133396RSG-19-029-01-DMC]
  2. Pew Charitable Trusts
  3. Alexander and Margaret Stewart Trust
  4. Siebel Stem Cell Institute
  5. National Institutes of Health (NIH) [R01-CA196884]
  6. Chen Zuckerberg Biohub
  7. D.O.D. [W81XWH-19-1-0586]
  8. Hellman Graduate Fellowship
  9. NIH [R01 HL131744]

向作者/读者索取更多资源

Mutations in the TINF2 gene cause telomere shortening and BM failure syndrome. This study introduces TIN2-DC mutations in hESCs and HSPCs to understand the disease mechanism and develop therapeutic strategies. The study demonstrates that editing the mutant TINF2 allele restores telomere length and replicative potential in stem cells. The findings provide a versatile platform for evaluating potential therapeutic approaches for BM failure.
Mutations in the TINF2 gene, encoding the shelterin protein TIN2, cause telomere shortening and the inherited bone marrow (BM) failure syndrome dyskeratosis congenita (DC). A lack of suitable model systems limits the mechanistic understanding of telomere shortening in the stem cells and thus hinders the development of treatment options for BM failure. Here, we endogenously introduced TIN2-DC mutations in human embryonic stem cells (hESCs) and human hematopoietic stem and progenitor cells (HSPCs) to dissect the disease mechanism and identify a gene-editing strategy that rescued the disease phenotypes. The hESCs with the T284R disease mutation exhibited the short telomere phenotype observed in DC patients. Yet, telomeres in mutant hESCs did not trigger DNA damage responses at telomeres or show exacerbated telomere shortening when differentiated into telomerase-negative cells. Disruption of the mutant TINF2 allele by introducing a frameshift mutation in exon 2 restored telomere length in stem cells and the replicative potential of differentiated cells. Similarly, we introduced TIN2-DC disease variants in human HSPCs to assess the changes in telomere length and proliferative capacity. Lastly, we showed that editing at exon 2 of TINF2 that restored telomere length in hESCs could be generated in TINF2-DC patient HSPCs. Our study demonstrates a simple genetic intervention that rescues the TIN2-DC disease phenotype in stem cells and provides a versatile platform to assess the efficacy of potential therapeutic approaches in vivo.

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