来自受 HTLV-1 感染细胞的胞外囊泡可调节靶细胞和病毒传播。

IF 2.7 3区 医学 Q3 VIROLOGY
Daniel O Pinto, Sarah Al Sharif, Gifty Mensah, Maria Cowen, Pooja Khatkar, James Erickson, Heather Branscome, Thomas Lattanze, Catherine DeMarino, Farhang Alem, Ruben Magni, Weidong Zhou, Sandrine Alais, Hélène Dutartre, Nazira El-Hage, Renaud Mahieux, Lance A Liotta, Fatah Kashanchi
{"title":"来自受 HTLV-1 感染细胞的胞外囊泡可调节靶细胞和病毒传播。","authors":"Daniel O Pinto, Sarah Al Sharif, Gifty Mensah, Maria Cowen, Pooja Khatkar, James Erickson, Heather Branscome, Thomas Lattanze, Catherine DeMarino, Farhang Alem, Ruben Magni, Weidong Zhou, Sandrine Alais, Hélène Dutartre, Nazira El-Hage, Renaud Mahieux, Lance A Liotta, Fatah Kashanchi","doi":"10.1186/s12977-021-00550-8","DOIUrl":null,"url":null,"abstract":"<p><strong>Background: </strong>The Human T-cell Lymphotropic Virus Type-1 (HTLV-1) is a blood-borne pathogen and etiological agent of Adult T-cell Leukemia/Lymphoma (ATLL) and HTLV-1 Associated Myelopathy/Tropical Spastic Paraparesis (HAM/TSP). HTLV-1 has currently infected up to 10 million globally with highly endemic areas in Japan, Africa, the Caribbean and South America. We have previously shown that Extracellular Vesicles (EVs) enhance HTLV-1 transmission by promoting cell-cell contact.</p><p><strong>Results: </strong>Here, we separated EVs into subpopulations using differential ultracentrifugation (DUC) at speeds of 2 k (2000×g), 10 k (10,000×g), and 100 k (100,000×g) from infected cell supernatants. Proteomic analysis revealed that EVs contain the highest viral/host protein abundance in the 2 k subpopulation (2 k > 10 k > 100 k). The 2 k and 10 k populations contained viral proteins (i.e., p19 and Tax), and autophagy proteins (i.e., LC3 and p62) suggesting presence of autophagosomes as well as core histones. Interestingly, the use of 2 k EVs in an angiogenesis assay (mesenchymal stem cells + endothelial cells) caused deterioration of vascular-like-tubules. Cells commonly associated with the neurovascular unit (i.e., astrocytes, neurons, and macrophages) in the blood-brain barrier (BBB) showed that HTLV-1 EVs may induce expression of cytokines involved in migration (i.e., IL-8; 100 k > 2 k > 10 k) from astrocytes and monocyte-derived macrophages (i.e., IL-8; 2 k > 10 k). Finally, we found that EVs were able to promote cell-cell contact and viral transmission in monocytic cell-derived dendritic cell. The EVs from both 2 k and 10 k increased HTLV-1 spread in a humanized mouse model, as evidenced by an increase in proviral DNA and RNA in the Blood, Lymph Node, and Spleen.</p><p><strong>Conclusions: </strong>Altogether, these data suggest that various EV subpopulations induce cytokine expression, tissue damage, and viral spread.</p>","PeriodicalId":21123,"journal":{"name":"Retrovirology","volume":null,"pages":null},"PeriodicalIF":2.7000,"publicationDate":"2021-02-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7901226/pdf/","citationCount":"0","resultStr":"{\"title\":\"Extracellular vesicles from HTLV-1 infected cells modulate target cells and viral spread.\",\"authors\":\"Daniel O Pinto, Sarah Al Sharif, Gifty Mensah, Maria Cowen, Pooja Khatkar, James Erickson, Heather Branscome, Thomas Lattanze, Catherine DeMarino, Farhang Alem, Ruben Magni, Weidong Zhou, Sandrine Alais, Hélène Dutartre, Nazira El-Hage, Renaud Mahieux, Lance A Liotta, Fatah Kashanchi\",\"doi\":\"10.1186/s12977-021-00550-8\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"<p><strong>Background: </strong>The Human T-cell Lymphotropic Virus Type-1 (HTLV-1) is a blood-borne pathogen and etiological agent of Adult T-cell Leukemia/Lymphoma (ATLL) and HTLV-1 Associated Myelopathy/Tropical Spastic Paraparesis (HAM/TSP). HTLV-1 has currently infected up to 10 million globally with highly endemic areas in Japan, Africa, the Caribbean and South America. We have previously shown that Extracellular Vesicles (EVs) enhance HTLV-1 transmission by promoting cell-cell contact.</p><p><strong>Results: </strong>Here, we separated EVs into subpopulations using differential ultracentrifugation (DUC) at speeds of 2 k (2000×g), 10 k (10,000×g), and 100 k (100,000×g) from infected cell supernatants. Proteomic analysis revealed that EVs contain the highest viral/host protein abundance in the 2 k subpopulation (2 k > 10 k > 100 k). The 2 k and 10 k populations contained viral proteins (i.e., p19 and Tax), and autophagy proteins (i.e., LC3 and p62) suggesting presence of autophagosomes as well as core histones. Interestingly, the use of 2 k EVs in an angiogenesis assay (mesenchymal stem cells + endothelial cells) caused deterioration of vascular-like-tubules. Cells commonly associated with the neurovascular unit (i.e., astrocytes, neurons, and macrophages) in the blood-brain barrier (BBB) showed that HTLV-1 EVs may induce expression of cytokines involved in migration (i.e., IL-8; 100 k > 2 k > 10 k) from astrocytes and monocyte-derived macrophages (i.e., IL-8; 2 k > 10 k). Finally, we found that EVs were able to promote cell-cell contact and viral transmission in monocytic cell-derived dendritic cell. The EVs from both 2 k and 10 k increased HTLV-1 spread in a humanized mouse model, as evidenced by an increase in proviral DNA and RNA in the Blood, Lymph Node, and Spleen.</p><p><strong>Conclusions: </strong>Altogether, these data suggest that various EV subpopulations induce cytokine expression, tissue damage, and viral spread.</p>\",\"PeriodicalId\":21123,\"journal\":{\"name\":\"Retrovirology\",\"volume\":null,\"pages\":null},\"PeriodicalIF\":2.7000,\"publicationDate\":\"2021-02-23\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7901226/pdf/\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"Retrovirology\",\"FirstCategoryId\":\"3\",\"ListUrlMain\":\"https://doi.org/10.1186/s12977-021-00550-8\",\"RegionNum\":3,\"RegionCategory\":\"医学\",\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"\",\"PubModel\":\"\",\"JCR\":\"Q3\",\"JCRName\":\"VIROLOGY\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"Retrovirology","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.1186/s12977-021-00550-8","RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q3","JCRName":"VIROLOGY","Score":null,"Total":0}
引用次数: 0

摘要

背景:人类 T 细胞淋巴细胞病毒 1 型(HTLV-1)是一种血液传播病原体,也是成人 T 细胞白血病/淋巴瘤(ATLL)和 HTLV-1 相关脊髓病/热带痉挛性瘫痪(HAM/TSP)的病原体。目前,全球感染 HTLV-1 的人数已高达 1000 万,日本、非洲、加勒比海和南美洲是 HTLV-1 的高流行区。我们之前已经证明,细胞外小泡(EVs)可通过促进细胞间接触来增强 HTLV-1 的传播:结果:在此,我们采用差速超速离心(DUC)技术,以 2 k(2000×g)、10 k(10,000×g)和 100 k(100,000×g)的速度从感染细胞上清液中分离出了亚群的 EVs。蛋白质组分析表明,在 2 k 亚群(2 k > 10 k > 100 k)中,EV 含有最高的病毒/宿主蛋白质丰度。2 k 和 10 k 亚群含有病毒蛋白(即 p19 和 Tax)和自噬蛋白(即 LC3 和 p62),表明存在自噬体和核心组蛋白。有趣的是,在血管生成试验(间充质干细胞+内皮细胞)中使用 2 k EVs 会导致血管样管退化。血脑屏障(BBB)中通常与神经血管单元相关的细胞(即星形胶质细胞、神经元和巨噬细胞)显示,HTLV-1 EVs 可诱导星形胶质细胞和单核细胞衍生巨噬细胞表达参与迁移的细胞因子(即 IL-8;100 k > 2 k > 10 k)(即 IL-8;2 k > 10 k)。最后,我们发现 EVs 能够促进单核细胞衍生树突状细胞的细胞-细胞接触和病毒传播。在人源化小鼠模型中,2 k 和 10 k 的 EVs 都能增加 HTLV-1 的传播,血液、淋巴结和脾脏中的前病毒 DNA 和 RNA 的增加就是证明:总之,这些数据表明,各种 EV 亚群可诱导细胞因子表达、组织损伤和病毒传播。
本文章由计算机程序翻译,如有差异,请以英文原文为准。

Extracellular vesicles from HTLV-1 infected cells modulate target cells and viral spread.

Extracellular vesicles from HTLV-1 infected cells modulate target cells and viral spread.

Extracellular vesicles from HTLV-1 infected cells modulate target cells and viral spread.

Extracellular vesicles from HTLV-1 infected cells modulate target cells and viral spread.

Background: The Human T-cell Lymphotropic Virus Type-1 (HTLV-1) is a blood-borne pathogen and etiological agent of Adult T-cell Leukemia/Lymphoma (ATLL) and HTLV-1 Associated Myelopathy/Tropical Spastic Paraparesis (HAM/TSP). HTLV-1 has currently infected up to 10 million globally with highly endemic areas in Japan, Africa, the Caribbean and South America. We have previously shown that Extracellular Vesicles (EVs) enhance HTLV-1 transmission by promoting cell-cell contact.

Results: Here, we separated EVs into subpopulations using differential ultracentrifugation (DUC) at speeds of 2 k (2000×g), 10 k (10,000×g), and 100 k (100,000×g) from infected cell supernatants. Proteomic analysis revealed that EVs contain the highest viral/host protein abundance in the 2 k subpopulation (2 k > 10 k > 100 k). The 2 k and 10 k populations contained viral proteins (i.e., p19 and Tax), and autophagy proteins (i.e., LC3 and p62) suggesting presence of autophagosomes as well as core histones. Interestingly, the use of 2 k EVs in an angiogenesis assay (mesenchymal stem cells + endothelial cells) caused deterioration of vascular-like-tubules. Cells commonly associated with the neurovascular unit (i.e., astrocytes, neurons, and macrophages) in the blood-brain barrier (BBB) showed that HTLV-1 EVs may induce expression of cytokines involved in migration (i.e., IL-8; 100 k > 2 k > 10 k) from astrocytes and monocyte-derived macrophages (i.e., IL-8; 2 k > 10 k). Finally, we found that EVs were able to promote cell-cell contact and viral transmission in monocytic cell-derived dendritic cell. The EVs from both 2 k and 10 k increased HTLV-1 spread in a humanized mouse model, as evidenced by an increase in proviral DNA and RNA in the Blood, Lymph Node, and Spleen.

Conclusions: Altogether, these data suggest that various EV subpopulations induce cytokine expression, tissue damage, and viral spread.

求助全文
通过发布文献求助,成功后即可免费获取论文全文。 去求助
来源期刊
Retrovirology
Retrovirology 医学-病毒学
CiteScore
5.80
自引率
3.00%
发文量
24
审稿时长
>0 weeks
期刊介绍: Retrovirology is an open access, online journal that publishes stringently peer-reviewed, high-impact articles on host-pathogen interactions, fundamental mechanisms of replication, immune defenses, animal models, and clinical science relating to retroviruses. Retroviruses are pleiotropically found in animals. Well-described examples include avian, murine and primate retroviruses. Two human retroviruses are especially important pathogens. These are the human immunodeficiency virus, HIV, and the human T-cell leukemia virus, HTLV. HIV causes AIDS while HTLV-1 is the etiological agent for adult T-cell leukemia and HTLV-1-associated myelopathy/tropical spastic paraparesis. Retrovirology aims to cover comprehensively all aspects of human and animal retrovirus research.
×
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