期刊论文详细信息
PLoS Pathogens
Engineering HIV-Resistant Human CD4+ T Cells with CXCR4-Specific Zinc-Finger Nucleases
Carl H. June1  James L. Riley1  Anthony J. Secreto2  Gwenn A. Danet-Desnoyers2  Craig B. Wilen3  Frederic D. Bushman3  James A. Hoxie3  Robert W. Doms3  Scott A. Sherrill-Mix3  Andrea P. O. Jordan3  John C. Tilton3  Sean C. Patro3  Bruce A. Bunnell4  Pyone P. Aye4  Andrew A. Lackner4  Jeffrey C. Miller5  Michael C. Holmes5  Joshua Kahn5  Jianbin Wang5  Kenneth A. Kim5  Philip D. Gregory5  Gary Lee5  Edward J. Rebar5 
[1] Abramson Family Cancer Research Institute, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America;Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America;Department of Microbiology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America;Divisions of Regenerative Medicine and Comparative Pathology, Tulane National Primate Research Center, Tulane University School of Medicine, Covington, Louisiana, United States of America;Sangamo BioSciences, Richmond, California, United States of America
关键词: T cells;    Cell disruption;    HIV-1;    HIV;    Nucleases;    HIV infections;    Mouse models;    Zinc finger nucleases;   
DOI  :  10.1371/journal.ppat.1002020
学科分类:生物科学(综合)
来源: Public Library of Science
PDF
【 摘 要 】

HIV-1 entry requires the cell surface expression of CD4 and either the CCR5 or CXCR4 coreceptors on host cells. Individuals homozygous for the ccr5Δ32 polymorphism do not express CCR5 and are protected from infection by CCR5-tropic (R5) virus strains. As an approach to inactivating CCR5, we introduced CCR5-specific zinc-finger nucleases into human CD4+ T cells prior to adoptive transfer, but the need to protect cells from virus strains that use CXCR4 (X4) in place of or in addition to CCR5 (R5X4) remains. Here we describe engineering a pair of zinc finger nucleases that, when introduced into human T cells, efficiently disrupt cxcr4 by cleavage and error-prone non-homologous DNA end-joining. The resulting cells proliferated normally and were resistant to infection by X4-tropic HIV-1 strains. CXCR4 could also be inactivated in ccr5Δ32 CD4+ T cells, and we show that such cells were resistant to all strains of HIV-1 tested. Loss of CXCR4 also provided protection from X4 HIV-1 in a humanized mouse model, though this protection was lost over time due to the emergence of R5-tropic viral mutants. These data suggest that CXCR4-specific ZFNs may prove useful in establishing resistance to CXCR4-tropic HIV for autologous transplant in HIV-infected individuals.

【 授权许可】

CC BY   

【 预 览 】
附件列表
Files Size Format View
RO201902016855325ZK.pdf 1350KB PDF download
  文献评价指标  
  下载次数:9次 浏览次数:7次