Your browser doesn't support javascript.
loading
Reactivation of Epstein-Barr virus by a dual-responsive fluorescent EBNA1-targeting agent with Zn2+-chelating function.
Jiang, Lijun; Lung, Hong Lok; Huang, Tao; Lan, Rongfeng; Zha, Shuai; Chan, Lai Sheung; Thor, Waygen; Tsoi, Tik-Hung; Chau, Ho-Fai; Boreström, Cecilia; Cobb, Steven L; Tsao, Sai Wah; Bian, Zhao-Xiang; Law, Ga-Lai; Wong, Wing-Tak; Tai, William Chi-Shing; Chau, Wai Yin; Du, Yujun; Tang, Lucas Hao Xi; Chiang, Alan Kwok Shing; Middeldorp, Jaap M; Lo, Kwok-Wai; Mak, Nai Ki; Long, Nicholas J; Wong, Ka-Leung.
Affiliation
  • Jiang L; Department of Chemistry, Hong Kong Baptist University, Kowloon Tong, Kowloon, Hong Kong.
  • Lung HL; Department of Chemistry, Imperial College London, London W12 0BZ, United Kingdom.
  • Huang T; Department of Biology, Hong Kong Baptist University, Kowloon Tong, Kowloon, Hong Kong.
  • Lan R; School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Kowloon, Hong Kong.
  • Zha S; Department of Cell Biology & Medical Genetics, Shenzhen University Health Science Center, 518071 Shenzhen, China.
  • Chan LS; Department of Chemistry, Hong Kong Baptist University, Kowloon Tong, Kowloon, Hong Kong.
  • Thor W; Department of Biology, Hong Kong Baptist University, Kowloon Tong, Kowloon, Hong Kong.
  • Tsoi TH; Department of Chemistry, Hong Kong Baptist University, Kowloon Tong, Kowloon, Hong Kong.
  • Chau HF; Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Hong Kong.
  • Boreström C; Department of Chemistry, Hong Kong Baptist University, Kowloon Tong, Kowloon, Hong Kong.
  • Cobb SL; Laboratory for Clinical Chemistry, Sahlgrenska University Hospital, SE-41345 Göteborg, Sweden.
  • Tsao SW; Department of Chemistry, Durham University, Durham DH1 3LE, United Kingdom.
  • Bian ZX; Department of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong.
  • Law GL; School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Kowloon, Hong Kong.
  • Wong WT; Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Hong Kong.
  • Tai WC; Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Hong Kong.
  • Chau WY; Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Hong Kong.
  • Du Y; Department of Biology, Hong Kong Baptist University, Kowloon Tong, Kowloon, Hong Kong.
  • Tang LHX; Department of Biology, Hong Kong Baptist University, Kowloon Tong, Kowloon, Hong Kong.
  • Chiang AKS; Department of Biology, Hong Kong Baptist University, Kowloon Tong, Kowloon, Hong Kong.
  • Middeldorp JM; Department of Paediatrics and Adolescent Medicine, The University of Hong Kong, Pokfulam, Hong Kong.
  • Lo KW; Department of Pathology, VU University Medical Center, 1081HV Amsterdam, The Netherlands.
  • Mak NK; Department of Anatomical and Cellular Pathology, State Key Laboratory of Oncology in South China, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, NT, Hong Kong.
  • Long NJ; Department of Biology, Hong Kong Baptist University, Kowloon Tong, Kowloon, Hong Kong.
  • Wong KL; Department of Chemistry, Imperial College London, London W12 0BZ, United Kingdom.
Proc Natl Acad Sci U S A ; 116(52): 26614-26624, 2019 Dec 26.
Article in En | MEDLINE | ID: mdl-31822610
ABSTRACT
Epstein-Barr nuclear antigen 1 (EBNA1) plays a vital role in the maintenance of the viral genome and is the only viral protein expressed in nearly all forms of Epstein-Barr virus (EBV) latency and EBV-associated diseases, including numerous cancer types. To our knowledge, no specific agent against EBV genes or proteins has been established to target EBV lytic reactivation. Here we report an EBNA1- and Zn2+-responsive probe (ZRL5P4) which alone could reactivate the EBV lytic cycle through specific disruption of EBNA1. We have utilized the Zn2+ chelator to further interfere with the higher order of EBNA1 self-association. The bioprobe ZRL5P4 can respond independently to its interactions with Zn2+ and EBNA1 with different fluorescence changes. It can selectively enter the nuclei of EBV-positive cells and disrupt the oligomerization and oriP-enhanced transactivation of EBNA1. ZRL5P4 can also specifically enhance Dicer1 and PML expression, molecular events which had been reported to occur after the depletion of EBNA1 expression. Importantly, we found that treatment with ZRL5P4 alone could reactivate EBV lytic induction by expressing the early and late EBV lytic genes/proteins. Lytic induction is likely mediated by disruption of EBNA1 oligomerization and the subsequent change of Dicer1 expression. Our probe ZRL5P4 is an EBV protein-specific agent that potently reactivates EBV from latency, leading to the shrinkage of EBV-positive tumors, and our study also suggests the association of EBNA1 oligomerization with the maintenance of EBV latency.
Key words

Full text: 1 Collection: 01-internacional Database: MEDLINE Language: En Journal: Proc Natl Acad Sci U S A Year: 2019 Type: Article Affiliation country: Hong Kong

Full text: 1 Collection: 01-internacional Database: MEDLINE Language: En Journal: Proc Natl Acad Sci U S A Year: 2019 Type: Article Affiliation country: Hong Kong