PUBLICATION

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

Authors
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.
ID
ZDB-PUB-191223-14
Date
2019
Source
Viruses   12(1): (Journal)
Registered Authors
Berman, Jason, Coombs, Andrew, Melong, Nicole, Veinotte, Chansey, Wertman, Jaime
Keywords
Kaposi’s sarcoma-associated herpesvirus (KSHV), ddPCR, human herpesvirus-8, hypoxia, primary effusion lymphoma (PEL), xenotransplantation, zebrafish
MeSH Terms
  • Animals
  • Cell Line, Tumor
  • Cell Proliferation
  • Disease Models, Animal
  • Disease Susceptibility*
  • Herpesviridae Infections/complications*
  • Herpesviridae Infections/virology*
  • Herpesvirus 8, Human/physiology*
  • Heterografts
  • Humans
  • Sarcoma, Kaposi/virology*
  • Zebrafish
PubMed
31861850 Full text @ Viruses
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 tumor 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 by 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.
Genes / Markers
Figures
Show all Figures
Expression
Phenotype
Mutations / Transgenics
Human Disease / Model
Sequence Targeting Reagents
Fish
Antibodies
Orthology
Engineered Foreign Genes
Mapping