Therapeutic efficacy of human hepatocyte transplantation in a SCID/uPA mouse model with inducible liver disease

PLoS One. 2010 Feb 18;5(2):e9209. doi: 10.1371/journal.pone.0009209.

Abstract

Background: Severe Combined Immune Deficient (SCID)/Urokinase-type Plasminogen Activator (uPA) mice undergo liver failure and are useful hosts for the propagation of transplanted human hepatocytes (HH) which must compete with recipient-derived hepatocytes for replacement of the diseased liver parenchyma. While partial replacement by HH has proven useful for studies with Hepatitis C virus, complete replacement of SCID/uPA mouse liver by HH has never been achieved and limits the broader application of these mice for other areas of biomedical research. The herpes simplex virus type-1 thymidine kinase (HSVtk)/ganciclovir (GCV) system is a powerful tool for cell-specific ablation in transgenic animals. The aim of this study was to selectively eliminate murine-derived parenchymal liver cells from humanized SCID/uPA mouse liver in order to achieve mice with completely humanized liver parenchyma. Thus, we reproduced the HSVtk (vTK)/GCV system of hepatic failure in SCID/uPA mice.

Methodology/principal findings: In vitro experiments demonstrated efficient killing of vTK expressing hepatoma cells after GCV treatment. For in vivo experiments, expression of vTK was targeted to the livers of FVB/N and SCID/uPA mice. Hepatic sensitivity to GCV was first established in FVB/N mice since these mice do not undergo liver failure inherent to SCID/uPA mice. Hepatic vTK expression was found to be an integral component of GCV-induced pathologic and biochemical alterations and caused death due to liver dysfunction in vTK transgenic FVB/N and non-transplanted SCID/uPA mice. In SCID/uPA mice with humanized liver, vTK/GCV caused death despite extensive replacement of the mouse liver parenchyma with HH (ranging from 32-87%). Surprisingly, vTK/GCV-dependent apoptosis and mitochondrial aberrations were also localized to bystander vTK-negative HH.

Conclusions/significance: Extensive replacement of mouse liver parenchyma by HH does not provide a secure therapeutic advantage against vTK/GCV-induced cytotoxicity targeted to residual mouse hepatocytes. Functional support by engrafted HH may be secured by strategies aimed at limiting this bystander effect.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Animals
  • Apoptosis / drug effects
  • Cell Line
  • Cell Line, Tumor
  • Cell Survival / drug effects
  • Cell Transplantation / methods*
  • Disease Models, Animal
  • Female
  • Ganciclovir / pharmacology
  • Ganciclovir / toxicity
  • Hepatocytes / cytology
  • Hepatocytes / transplantation*
  • Herpesvirus 1, Human / enzymology
  • Humans
  • Immunoblotting
  • Liver / drug effects
  • Liver / metabolism
  • Liver / ultrastructure
  • Liver Diseases / etiology
  • Liver Diseases / genetics
  • Liver Diseases / surgery*
  • Male
  • Mice
  • Mice, SCID
  • Mice, Transgenic
  • Microscopy, Electron
  • Reverse Transcriptase Polymerase Chain Reaction
  • Thymidine Kinase / genetics
  • Thymidine Kinase / metabolism
  • Transfection
  • Transplantation, Heterologous
  • Urokinase-Type Plasminogen Activator / genetics
  • Urokinase-Type Plasminogen Activator / metabolism*

Substances

  • Thymidine Kinase
  • Urokinase-Type Plasminogen Activator
  • Ganciclovir