4.8 Article

Positive selection in dNTPase SAMHD1 throughout mammalian evolution

出版社

NATL ACAD SCIENCES
DOI: 10.1073/pnas.1908755116

关键词

SAMHD1; HIV-1; restriction; evolution; mammals

资金

  1. UK Medical Research Council [MC_U117573805, G0900950]
  2. UK Biotechnology and Biological Sciences Research Council [BB/P007562/1]
  3. European Union Framework Program 7 HIVINNOV Grant [305137]
  4. Wellcome Trust [108014/Z/15/Z, 108012/Z/15/Z]
  5. Francis Crick Institute (Cancer Research UK) [FC001042, FC001162, FC001178]
  6. Francis Crick Institute (UK Medical Research Council) [FC001042, FC001162, FC001178]
  7. Francis Crick Institute (Wellcome Trust) [FC001042, FC001162, FC001178]
  8. University College London CoMPLEX
  9. Wellcome Trust [108014/Z/15/Z] Funding Source: Wellcome Trust
  10. BBSRC [BB/P007562/1] Funding Source: UKRI
  11. MRC [MC_U117592729, MC_U117512710, MC_U117573805, MC_PC_13056, G0900950] Funding Source: UKRI

向作者/读者索取更多资源

The vertebrate protein SAMHD1 is highly unusual in having roles in cellular metabolic regulation, antiviral restriction, and regulation of innate immunity. Its deoxynucleoside triphosphohydrolase activity regulates cellular dNTP concentration, reducing levels below those required by lentiviruses and other viruses to replicate. To counter this threat, some primate lentiviruses encode accessory proteins that bind SAMHD1 and induce its degradation; in turn, positive diversifying selection has been observed in regions bound by these lentiviral proteins, suggesting that primate SAMHD1 has coevolved to evade these countermeasures. Moreover, deleterious polymorphisms in human SAMHD1 are associated with autoimmune disease linked to uncontrolled DNA synthesis of endogenous retroelements. Little is known about how evolutionary pressures affect these different SAMHD1 functions. Here, we examine the deeper history of these interactions by testing whether evolutionary signatures in SAMHD1 extend to other mammalian groups and exploring the molecular basis of this coevolution. Using codon-based likelihood models, we find positive selection in SAMHD1 within each mammal lineage for which sequence data are available. We observe positive selection at sites clustered around T592, a residue that is phosphorylated to regulate SAMHD1 activity. We verify experimentally that mutations within this cluster affect catalytic rate and lentiviral restriction, suggesting that virus-host coevolution has required adaptations of enzymatic function. Thus, persistent positive selection may have involved the adaptation of SAMHD1 regulation to balance antiviral, metabolic, and innate immunity functions.

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