Preprint Article Version 1 Preserved in Portico This version is not peer-reviewed

The Zebrafish Xenograft Platform – A Novel Tool for Modeling KSHV-Associated Diseases

Version 1 : Received: 7 November 2019 / Approved: 8 November 2019 / Online: 8 November 2019 (03:23:22 CET)

A peer-reviewed article of this Preprint also exists.

Pringle, E.S.; Wertman, J.; Melong, N.; Coombs, A.J.; Young, A.L.; O’Leary, D.; Veinotte, C.; Robinson, C.-A.; Ha, M.N.; Dellaire, G.; Druley, T.E.; McCormick, C.; Berman, J.N. The Zebrafish Xenograft Platform—A Novel Tool for Modeling KSHV-Associated Diseases. Viruses 2020, 12, 12. Pringle, E.S.; Wertman, J.; Melong, N.; Coombs, A.J.; Young, A.L.; O’Leary, D.; Veinotte, C.; Robinson, C.-A.; Ha, M.N.; Dellaire, G.; Druley, T.E.; McCormick, C.; Berman, J.N. The Zebrafish Xenograft Platform—A Novel Tool for Modeling KSHV-Associated Diseases. Viruses 2020, 12, 12.

Abstract

Kaposi’s sarcoma associated-herpesvirus (KSHV, also known as human herpesvirus-8) is a gammaherpesvirus that establishes life-long infection in human B lymphocytes. KSHV infection is typically asymptomatic but immunosuppression can predispose KSHV-infected individuals to primary effusion lymphoma (PEL); a malignancy driven by aberrant proliferation of latently infected B lymphocytes, and supported by pro-inflammatory cytokines and angiogenic factors produced by cells that succumb to lytic viral replication. Here, we report the development of the first in vivo model for a virally-induced lymphoma in zebrafish, whereby KSHV-infected PEL tumour cells engraft and proliferate in the yolk sac of zebrafish larvae. Using a PEL cell line engineered to produce the viral lytic switch protein RTA in the presence of doxycycline, we demonstrate drug-inducible reactivation from KSHV latency in vivo, which enabled real-time observation and evaluation of latent and lytic phases of KSHV infection. In addition, we developed a sensitive droplet digital PCR method to monitor latent and lytic viral gene expression and host cell gene expression in xenografts. The zebrafish yolk sac is not well-vascularized and using fluorogenic assays we confirmed that this site provides a hypoxic environment that may mimic the microenvironment of some human tumors. We found that PEL cell proliferation in xenografts was dependent on the host hypoxia-dependent translation initiation factor, eukaryotic initiation factor 4E2 (eIF4E2). This demonstrates that the zebrafish yolk sac is a functionally hypoxic environment and xenografted cells must switch to dedicated hypoxic gene expression machinery to survive and proliferate. The establishment of the PEL xenograft model enables future studies that exploit the innate advantages of the zebrafish as a model for genetic and pharmacologic screens.

Keywords

Kaposi’s sarcoma-associated herpesvirus (KSHV); human herpesvirus-8; zebrafish; ddPCR; xenotransplantation; primary effusion lymphoma (PEL); hypoxia

Subject

Biology and Life Sciences, Virology

Comments (0)

We encourage comments and feedback from a broad range of readers. See criteria for comments and our Diversity statement.

Leave a public comment
Send a private comment to the author(s)
* All users must log in before leaving a comment
Views 0
Downloads 0
Comments 0
Metrics 0


×
Alerts
Notify me about updates to this article or when a peer-reviewed version is published.
We use cookies on our website to ensure you get the best experience.
Read more about our cookies here.