Identification of Copper Transporter 1 as a Receptor for Feline Endogenous Retrovirus ERV-DC14

IF 4 2区 医学 Q2 VIROLOGY
Sandrine Tury, D. Giovannini, S. Ivanova, Jawida Touhami, V. Courgnaud, J. Battini
{"title":"Identification of Copper Transporter 1 as a Receptor for Feline Endogenous Retrovirus ERV-DC14","authors":"Sandrine Tury, D. Giovannini, S. Ivanova, Jawida Touhami, V. Courgnaud, J. Battini","doi":"10.1128/jvi.00229-22","DOIUrl":null,"url":null,"abstract":"Receptor usage is an important determinant of diseases induced by pathogenic retroviruses. In the case of feline leukemia viruses, three subgroups (A, B, and C) based on their ability to recognize different cell host receptors, respectively, the thiamine transporter THTR1, the phosphate transporter PiT1, and the heme exporter FLVCR1, are associated with distinct feline diseases. ABSTRACT Vertebrates harbor hundreds of endogenous retroviral (ERV) sequences in their genomes, which are considered signs of past infections that occurred during evolution. On rare occasions, ERV genes like env are maintained and coopted by hosts for physiological functions, but they also participate in recombination events with exogenous retroviruses to generate rearranged viruses with novel tropisms. In domestic cats, feline leukemia virus type D (FeLV-D) has been described as a recombinant virus between the infectious FeLV-A and likely the ERV-DC14 env gene that resulted in an extended tropism due to the usage of a new uncharacterized retroviral receptor. Here, we report the identification of SLC31A1 encoding the copper transporter 1 (CTR1) as a susceptibility gene for ERV-DC14 infection. Expression of human CTR1 into nonpermissive cells was sufficient to confer sensitivity to ERV-DC14 pseudotype infection and to increase the binding of an ERV-DC14 Env ligand. Moreover, inactivation of CTR1 by genome editing or cell surface downmodulation of CTR1 by a high dose of copper dramatically decreased ERV-DC14 infection and binding, while magnesium treatment had no effect. We also investigated the role of CTR1 in the nonpermissivity of feline and hamster cells. While feline CTR1 was fully functional for ERV-DC14, we found that binding was strongly reduced upon treatment with conditioned medium of feline cells, suggesting that the observed resistance to infection was a consequence of CTR1 saturation. In contrast, hamster CTR1 was inactive due to the presence of a N-linked glycosylation site at position 27, which is absent in the human ortholog. These results provide evidence that CTR1 is a receptor for ERV-DC14. Along with chimpanzee endogenous retrovirus type 2, ERV-DC14 is the second family of endogenous retrovirus known to have used CTR1 during past infections of vertebrates. IMPORTANCE Receptor usage is an important determinant of diseases induced by pathogenic retroviruses. In the case of feline leukemia viruses, three subgroups (A, B, and C) based on their ability to recognize different cell host receptors, respectively, the thiamine transporter THTR1, the phosphate transporter PiT1, and the heme exporter FLVCR1, are associated with distinct feline diseases. FeLV-A is horizontally transmitted and found in all naturally infected cats, while FeLV-B and FeLV-C have emerged from FeLV-A, respectively, by recombination with endogenous retroviral env sequences or by mutations in the FeLV-A env gene, both leading to a switch in receptor usage and in subsequent in vivo tropism. Here, we set up a genetic screen to identify the retroviral receptor of ERV-DC14, a feline endogenous provirus whose env gene has been captured by infectious FeLV-A to give rise to FeLV-D in a process similar to FeLV-B. Our results reveal that the copper transporter CTR1 was such a receptor and provide new insights into the acquisition of an expanded tropism by FeLV-D.","PeriodicalId":17583,"journal":{"name":"Journal of Virology","volume":null,"pages":null},"PeriodicalIF":4.0000,"publicationDate":"2022-06-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":"3","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Journal of Virology","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.1128/jvi.00229-22","RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q2","JCRName":"VIROLOGY","Score":null,"Total":0}
引用次数: 3

Abstract

Receptor usage is an important determinant of diseases induced by pathogenic retroviruses. In the case of feline leukemia viruses, three subgroups (A, B, and C) based on their ability to recognize different cell host receptors, respectively, the thiamine transporter THTR1, the phosphate transporter PiT1, and the heme exporter FLVCR1, are associated with distinct feline diseases. ABSTRACT Vertebrates harbor hundreds of endogenous retroviral (ERV) sequences in their genomes, which are considered signs of past infections that occurred during evolution. On rare occasions, ERV genes like env are maintained and coopted by hosts for physiological functions, but they also participate in recombination events with exogenous retroviruses to generate rearranged viruses with novel tropisms. In domestic cats, feline leukemia virus type D (FeLV-D) has been described as a recombinant virus between the infectious FeLV-A and likely the ERV-DC14 env gene that resulted in an extended tropism due to the usage of a new uncharacterized retroviral receptor. Here, we report the identification of SLC31A1 encoding the copper transporter 1 (CTR1) as a susceptibility gene for ERV-DC14 infection. Expression of human CTR1 into nonpermissive cells was sufficient to confer sensitivity to ERV-DC14 pseudotype infection and to increase the binding of an ERV-DC14 Env ligand. Moreover, inactivation of CTR1 by genome editing or cell surface downmodulation of CTR1 by a high dose of copper dramatically decreased ERV-DC14 infection and binding, while magnesium treatment had no effect. We also investigated the role of CTR1 in the nonpermissivity of feline and hamster cells. While feline CTR1 was fully functional for ERV-DC14, we found that binding was strongly reduced upon treatment with conditioned medium of feline cells, suggesting that the observed resistance to infection was a consequence of CTR1 saturation. In contrast, hamster CTR1 was inactive due to the presence of a N-linked glycosylation site at position 27, which is absent in the human ortholog. These results provide evidence that CTR1 is a receptor for ERV-DC14. Along with chimpanzee endogenous retrovirus type 2, ERV-DC14 is the second family of endogenous retrovirus known to have used CTR1 during past infections of vertebrates. IMPORTANCE Receptor usage is an important determinant of diseases induced by pathogenic retroviruses. In the case of feline leukemia viruses, three subgroups (A, B, and C) based on their ability to recognize different cell host receptors, respectively, the thiamine transporter THTR1, the phosphate transporter PiT1, and the heme exporter FLVCR1, are associated with distinct feline diseases. FeLV-A is horizontally transmitted and found in all naturally infected cats, while FeLV-B and FeLV-C have emerged from FeLV-A, respectively, by recombination with endogenous retroviral env sequences or by mutations in the FeLV-A env gene, both leading to a switch in receptor usage and in subsequent in vivo tropism. Here, we set up a genetic screen to identify the retroviral receptor of ERV-DC14, a feline endogenous provirus whose env gene has been captured by infectious FeLV-A to give rise to FeLV-D in a process similar to FeLV-B. Our results reveal that the copper transporter CTR1 was such a receptor and provide new insights into the acquisition of an expanded tropism by FeLV-D.
猫内源性逆转录病毒ERV-DC14受体铜转运蛋白1的鉴定
受体的使用是致病逆转录病毒引起疾病的重要决定因素。在猫白血病病毒中,三个亚群(A、B和C)基于它们识别不同细胞宿主受体的能力,分别是硫胺素转运体THTR1、磷酸盐转运体PiT1和血红素输出体FLVCR1,它们与不同的猫疾病相关。脊椎动物的基因组中含有数百个内源性逆转录病毒(ERV)序列,这些序列被认为是进化过程中发生的过去感染的迹象。在极少数情况下,像env这样的ERV基因被宿主维持和吸收用于生理功能,但它们也参与与外源性逆转录病毒的重组事件,产生具有新倾向的重排病毒。在家猫中,猫白血病病毒D型(FeLV-D)被描述为一种介于传染性FeLV-A和可能的ERV-DC14 env基因之间的重组病毒,由于使用了一种新的未表征的逆转录病毒受体,导致了扩展的向性。在这里,我们报道了SLC31A1编码铜转运蛋白1 (CTR1)作为ERV-DC14感染的易感基因的鉴定。人CTR1在非受纳细胞中的表达足以赋予对ERV-DC14假型感染的敏感性,并增加ERV-DC14 Env配体的结合。此外,通过基因组编辑使CTR1失活或通过高剂量铜下调CTR1细胞表面可显著降低ERV-DC14感染和结合,而镁处理没有效果。我们还研究了CTR1在猫和仓鼠细胞不允许性中的作用。虽然猫的CTR1对ERV-DC14具有完全的功能,但我们发现,在猫细胞的条件培养基中处理后,这种结合明显减少,这表明观察到的对感染的抵抗是CTR1饱和的结果。相比之下,仓鼠CTR1是无活性的,因为在27号位置存在一个n -连接的糖基化位点,这在人类同源基因中是不存在的。这些结果为CTR1是ERV-DC14的受体提供了证据。除了黑猩猩内源性2型逆转录病毒外,ERV-DC14是已知在过去脊椎动物感染中使用CTR1的第二个内源性逆转录病毒家族。受体的使用是致病逆转录病毒引起疾病的重要决定因素。在猫白血病病毒中,三个亚群(A、B和C)基于它们识别不同细胞宿主受体的能力,分别是硫胺素转运体THTR1、磷酸盐转运体PiT1和血红素输出体FLVCR1,它们与不同的猫疾病相关。FeLV-A是水平传播的,在所有自然感染的猫中都有发现,而FeLV-B和FeLV-C分别来自FeLV-A,通过与内源性逆转录病毒env序列的重组或FeLV-A env基因的突变,两者都导致受体使用的转换和随后的体内向性。在这里,我们建立了一个基因筛选来鉴定ERV-DC14的逆转录病毒受体,ERV-DC14是一种猫内源性病毒,其env基因被传染性FeLV-A捕获,以类似于FeLV-B的过程产生FeLV-D。我们的研究结果揭示了铜转运体CTR1就是这样一个受体,并为FeLV-D获得扩展向性提供了新的见解。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
求助全文
约1分钟内获得全文 求助全文
来源期刊
Journal of Virology
Journal of Virology 医学-病毒学
CiteScore
10.10
自引率
7.40%
发文量
906
审稿时长
1 months
期刊介绍: Journal of Virology (JVI) explores the nature of the viruses of animals, archaea, bacteria, fungi, plants, and protozoa. We welcome papers on virion structure and assembly, viral genome replication and regulation of gene expression, genetic diversity and evolution, virus-cell interactions, cellular responses to infection, transformation and oncogenesis, gene delivery, viral pathogenesis and immunity, and vaccines and antiviral agents.
文献相关原料
公司名称 产品信息 采购帮参考价格
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
确定
请完成安全验证×
copy
已复制链接
快去分享给好友吧!
我知道了
右上角分享
点击右上角分享
0
联系我们:info@booksci.cn Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。 Copyright © 2023 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术官方微信