Repository logo
 

A minimally invasive, lentiviral based method for the rapid and sustained genetic manipulation of renal tubules.


Change log

Authors

Espana-Agusti, Judit 
Tuveson, David A 
Adams, David J 
Matakidou, Athena 

Abstract

The accelerated discovery of disease-related genes emerging from genomic studies has strained the capacity of traditional genetically engineered mouse models (GEMMs) to provide in-vivo validation. Direct, somatic, genetic engineering approaches allow for accelerated and flexible genetic manipulation and represent an attractive alternative to GEMMs. In this study we investigated the feasibility, safety and efficiency of a minimally invasive, lentiviral based approach for the sustained in-vivo modification of renal tubular epithelial cells. Using ultrasound guidance, reporter vectors were directly injected into the mouse renal parenchyma. We observed transgene expression confined to the renal cortex (specifically proximal and distal tubules) and sustained beyond 2 months post injection. Furthermore, we demonstrate the ability of this methodology to induce long-term, in-vivo knockdown of candidate genes either through somatic recombination of floxed alleles or by direct delivery of specific shRNA sequences. This study demonstrates that ultrasound-guided injection of lentiviral vectors provides a safe and efficient method for the genetic manipulation of renal tubules, representing a quick and versatile alternative to GEMMs for the functional characterisation of disease-related genes.

Description

Keywords

Animals, Animals, Genetically Modified, Epithelial Cells, Gene Transfer Techniques, Genetic Vectors, HEK293 Cells, Humans, Kidney Tubules, Lentivirus, Mice, Mice, Inbred C57BL, NIH 3T3 Cells, RNA Interference, RNA, Small Interfering, Tuberous Sclerosis Complex 1 Protein, Tumor Suppressor Proteins, Ultrasonography

Journal Title

Sci Rep

Conference Name

Journal ISSN

2045-2322
2045-2322

Volume Title

5

Publisher

Springer Science and Business Media LLC
Sponsorship
The authors wish to thank the core facilities (Biological Research Unit, Histopathology, Flow Cytometry and Microscopy) of the CRUK Cambridge Institute for advice and technical assistance. This work was funded by a CRUK Clinician Scientist Fellowship award (C37839/A12177).