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 2022 and some content may be unavailable. To unlock all content for 2022, please visit the archives.

Abstract: FR-PO400

Chimeric Nephrons in Neonatal Mice for Repeated-Dose Toxicity Evaluation

Session Information

Category: Development‚ Stem Cells‚ and Regenerative Medicine

  • 500 Development‚ Stem Cells‚ and Regenerative Medicine

Authors

  • Matsui, Kenji, Tokyo Jikeikai Ika Daigaku, Minato-ku, Tokyo, Japan
  • Yamanaka, Shuichiro, Tokyo Jikeikai Ika Daigaku, Minato-ku, Tokyo, Japan
  • Matsumoto, Naoto, Tokyo Jikeikai Ika Daigaku, Minato-ku, Tokyo, Japan
  • Saito, Yatsumu, Tokyo Jikeikai Ika Daigaku, Minato-ku, Tokyo, Japan
  • Matsumoto, Kei, Tokyo Jikeikai Ika Daigaku, Minato-ku, Tokyo, Japan
  • Kobayashi, Eiji, Tokyo Jikeikai Ika Daigaku, Minato-ku, Tokyo, Japan
  • Yokoo, Takashi, Tokyo Jikeikai Ika Daigaku, Minato-ku, Tokyo, Japan

Group or Team Name

  • Division of Nephrology and Hypertension, Department of Internal Medicine, The Jikei University School of Medicine
Background

The detailed molecular mechanisms of drug-induced nephrotoxicity (DIN) have not been established to date.
It is difficult to examine multiple target molecules comprehensively in vivo because mice genetic modification requires time. Cultured kidney cells that lose polarity do not accurately reproduce toxicity. Renal organoids are not suitable to validate the recovery process and chronic toxicity because they cannot be cultured for a long time.
In this study, we have generated a new evaluation model using chimeric mice.

Methods

1. Renal progenitor cells (RPCs) extracted from EGFP-labeled mouse fetuses were incubated to form spheres. The spheres were then transplanted under the renal capsules of adult C57BL/6 (B6) and NOG mice.
2. RPCs cell suspension was injected into neonatal B6 renal capsules. The mice were treated with repeated-dose cisplatin and were harvested 2 months after the injection for an evaluation with immunostaining and scRNA-seq.

Results

1. The RPCs spheres in adult B6 showed a marked T-cell infiltration, suggesting an EGFP proteins rejection. Those transplanted into NOG formed nephrons, but their tubules became markedly dilated after 2 months.
2. The RPCs injected into the neonatal B6 were integrated into the host nephrogenic region without rejection. Subsequently, they formed mature chimeric nephrons from the glomeruli to the distal tubules that were retained for 4 months. Their function was confirmed by the presence of systemically administered dextran in their lumens. The repeated cisplatin administration caused them to express injury markers equivalent to the host nephrons.

Conclusion

We have developed a method to generate chimeric nephrons from the RPCs in neonatal mice that reproduced repeated-dose toxicity. The advantages of this model are as follows: 1, chimeric mice were generated easily; 2, chimeric nephrons showed a similar maturity and drug response to the host; 3, they survived for a longer time than the spheres in adults with connection to the host excretory tract; 4, immunocompetent neonates were tolerant to EGFP antigens, thus, traceable chimeric nephrons were formed; 5, scRNA-seq could be used to compare the nephron behavior of different origins.
Furthermore, this model provided the potential to easily change tubular phenotypes by gene-editing RPCs and may be applied to elucidate the DIN mechanism.

Funding

  • Government Support – Non-U.S.