Basic Study
Copyright ©The Author(s) 2018. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Hepatol. Feb 27, 2018; 10(2): 277-286
Published online Feb 27, 2018. doi: 10.4254/wjh.v10.i2.277
Homologous recombination mediates stable Fah gene integration and phenotypic correction in tyrosinaemia mouse-model
Norman Junge, Qinggong Yuan, Thu Huong Vu, Simon Krooss, Christien Bednarski, Asha Balakrishnan, Toni Cathomen, Michael P Manns, Ulrich Baumann, Amar Deep Sharma, Michael Ott
Norman Junge, Thu Huong Vu, Ulrich Baumann, Department of Pediatric Gastroenterology and Hepatology, Hannover Medical School, Hannover 30625, Germany
Qinggong Yuan, Asha Balakrishnan, Michael P Manns, Amar Deep Sharma, Michael Ott, Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover 30625, Germany
Qinggong Yuan, Simon Krooss, Asha Balakrishnan, Michael Ott, TWINCORE, Centre for Experimental and Clinical Infection Research, Hannover 30625, Germany
Christien Bednarski, Toni Cathomen, Medical Center, University of Freiburg, Institute for Cell and Gene Therapy, Freiburg 79108, Germany
Amar Deep Sharma, Research Group MicroRNA in Liver Regeneration, Cluster of Excellence REBIRTH, Hannover Medical School, Hannover 30625, Germany
Author contributions: Junge N was involved in conception and design of the research, performed the majority of the experiments, analysed the data and wrote the manuscript; Yuan QG performed mouse surgery and immunostaining and revised the work critically for important intellectual content; Huong Vu T had substantial contributions to the experiments, animal care and analysis and interpretation of data for the work; Krooss S, Bednarski C, Balakrishnan A and Cathomen T helped with the experiments and design of the research and revised the work critically for important intellectual content; Manns MP and Baumann U revised the work critically for important intellectual content; Sharma AD and Ott M were initiator and supervisor of the work, the developed initial concept and design of the research and conducted important preliminary studies.
Institutional animal care and use committee statement: All experiments were approved and performed according to guidelines and ethical regulations from Hannover Medical School and local government.
Conflict-of-interest statement: There are no conflicts of interest for any of the authors regarding this work.
Data sharing statement: Technical appendix, statistical code, and dataset available from the corresponding author (ott.michael@mh-hannover.de).
Open-Access: This article is an open-access article which was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/licenses/by-nc/4.0/
Correspondence to: Michael Ott, MD, Full Professor, TWINCORE, Centre for Experimental and Clinical Infection Research, Feodor-Lynen-Str 7, Hannover 30625, Germany. ott.michael@mh-hannover.de
Telephone: +49-511-220027120 Fax: +49-511-220027178
Received: December 7, 2017
Peer-review started: December 8, 2017
First decision: December 18, 2017
Revised: February 1, 2018
Accepted: February 23, 2018
Article in press: February 23, 2018
Published online: February 27, 2018
Abstract
AIM

To stably correct tyrosinaemia in proliferating livers of fumarylacetoacetate-hydrolase knockout (Fah-/-) mice by homologous-recombination-mediated targeted addition of the Fah gene.

METHODS

C57BL/6 Fah∆exon5 mice served as an animal model for human tyrosinaemia type 1 in our study. The vector was created by amplifying human Fah cDNA including the TTR promoter from a lentivirus plasmid as described. The Fah expression cassette was flanked by homologous arms (620 bp and 749 bp long) of the Rosa26 gene locus. Mice were injected with 2.1 × 108 VP of this vector (rAAV8-ROSA26.HAL-TTR.Fah-ROSA26.HAR) via the tail vein. Mice in the control group were injected with 2.1 × 108 VP of a similar vector but missing the homologous arms (rAAV8-TTR.Fah). Primary hepatocytes from Fah-/- recipient mice, treated with our vectors, were isolated and 1 × 106 hepatocytes were transplanted into secondary Fah-/- recipient mice by injection into the spleen. Upon either vector application or hepatocyte transplantation NTBC treatment was stopped in recipient mice.

RESULTS

Here, we report successful HR-mediated genome editing by integration of a Fah gene expression cassette into the “safe harbour locus” Rosa26 by recombinant AAV8. Both groups of mice showed long-term survival, weight gain and FAH positive clusters as determined by immunohistochemistry analysis of liver sections in the absence of NTBC treatment. In the group of C57BL/6 Fah∆exon5 mice, which have been transplanted with hepatocytes from a mouse injected with rAAV8-ROSA26.HAL-TTR.Fah-ROSA26.HAR 156 d before, 6 out of 6 mice showed long-term survival, weight gain and FAH positive clusters without need for NTBC treatment. In contrast only 1 out 5 mice, who received hepatocytes from rAAV8-TTR.Fah treated mice, survived and showed few and smaller FAH positive clusters. These results demonstrate that homologous recombination-mediated Fah gene transfer corrects the phenotype in a mouse model of human tyrosinaemia type 1 (Fah-/- mice) and is long lasting in a proliferating state of the liver as shown by withdrawal of NTBC treatment and serial transplantation of isolated hepatocytes from primary Fah-/- recipient mice into secondary Fah-/- recipient mice. This long term therapeutic efficacy is clearly superior to our control mice treated with episomal rAAV8 gene therapy approach.

CONCLUSION

HR-mediated rAAV8 gene therapy provides targeted transgene integration and phenotypic correction in Fah-/- mice with superior long-term efficacy compared to episomal rAAV8 therapy in proliferating livers.

Keywords: Gene therapy, AAV8, Liver based metabolic disease, Targeted integration, ROSA26, Paediatric liver disease

Core tip: Recombinant adeno-associated virus (rAAV) has been explored for gene delivery in various murine models of hereditary liver disease, but in young children transgene expression from AAV-epigenomes diminishes over time. We thus explored, whether homologous recombination-mediated targeted gene addition of the fumarylacetoacetate hydrolase (Fah) gene would stably correct tyrosinaemia in rapidly proliferating livers of Fah-/- mice. Here, we report successful homologous recombination-mediated genome editing of a Fah gene expression cassette at the Rosa26 locus by rAAV8. We demonstrate that this approach corrects the phenotype and is long lasting in a proliferating state of the liver, as shown by serial transplantation.