ASN's Mission

To create a world without kidney diseases, the ASN Alliance for Kidney Health elevates care by educating and informing, driving breakthroughs and innovation, and advocating for policies that create transformative changes in kidney medicine throughout the world.

learn more

Contact ASN

1401 H St, NW, Ste 900, Washington, DC 20005

email@asn-online.org

202-640-4660

The Latest on X

Kidney Week

Please note that you are viewing an archived section from 2019 and some content may be unavailable. To unlock all content for 2019, please visit the archives.

Abstract: FR-PO745

Heterozygous Inactivation of PKD1 in Miniature Pigs Induces Embryonic Cyst Formation and Progressive Autosomal Dominant Polycystic Kidney Disease (ADPKD)

Session Information

Category: Genetic Diseases of the Kidneys

  • 1001 Genetic Diseases of the Kidneys: Cystic

Authors

  • Daniel, Emily A., University of Kansas Medical Center, Kansas City, Kansas, United States
  • Raman, Archana, University of Pennsylvania, Philadelphia, Pennsylvania, United States
  • Ward, Christopher J., KUMC, Kansas City, Kansas, United States
  • Reif, Gail, University of Kansas Medical Center, Kansas City, Kansas, United States
  • Rogers, Christopher S., Exemplar Genetics, Coralville, Iowa, United States
  • Fields, Timothy A., University of Kansas Medical Center, Kansas City, Kansas, United States
  • Rowe, Peter S. N., University of Kansas Medical Center, Kansas City, Kansas, United States
  • Calvet, James P., University of Kansas Medical Center, Kansas City, Kansas, United States
  • Wallace, Darren P., University of Kansas Medical Center, Kansas City, Kansas, United States
Background

Murine models do not faithfully recapitulate important features of human ADPKD or its dominant inheritance. Gene editing and somatic cell nuclear transfer in large animals have provided superior models for human genetic disorders that have improved our understanding of disease mechanisms and the translational value of pre-clinical studies for novel therapies. Porcine anatomy, development, physiology, genetics and body size are like those of humans. We generated a new ADPKD model in the Yucatan miniature pig to monitor renal cyst formation in utero and postnatal PKD progression.

Methods

Gene targeting methods were used to insert a blasticidin cassette into intron 30 of PKD1, which resulted in a null allele, and somatic cell nuclear transfer to generate PKD1+/- cloned pigs. Breeding colonies were established to generate PKD1+/- and wildtype littermates. Kidneys were collected at embryonic days 30, 60, 90, newborn (~120 embryonic days) and 3, 6, 9 and 12 months postnatal to evaluate cyst development. Embryonic kidneys were imaged by micro-CT and larger kidneys were sectioned using a tissue slicer for measurement of cystic index. Thin tissue sections were stained with antibodies to AQP-2 to identify collecting ducts and PCNA, a marker of cell proliferation.

Results

Embryonic PKD1+/- pig kidneys had sporadic cysts and cystic dilations that appeared to form in clusters, consistent with the proposed mechanism of early cyst development in human ADPKD. The number of cysts and total cystic area progressively increased after birth; however, changes in kidney weight relative to body weight were not significantly different between PKD1+/- and wildtype pigs. Many, but not all, cysts stained positive for AQP-2, indicating the involvement of multiple segments of the nephron. There were increased PCNA-positive cells in cystic epithelia compared to non-cystic tissue in embryonic and adult PKD1+/- kidneys.

Conclusion

Our results demonstrate that inactivation of one PKD1 allele induces renal cyst formation in utero and progressive ADPKD in a new porcine model ADPKD.

Funding

  • NIDDK Support –