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 2021 and some content may be unavailable. To unlock all content for 2021, please visit the archives.

Abstract: PO1212

Comparative Multiple-Species Analysis of Renal Disease Mutations in HNF1B

Session Information

Category: Genetic Diseases of the Kidneys

  • 1001 Genetic Diseases of the Kidneys: Cystic

Authors

  • Grand, Kelli, Universitat Zurich, Zurich, ZH, Switzerland
  • Naert, Thomas, Universitat Zurich, Zurich, ZH, Switzerland
  • Rizzo, Ludovica, Universitat Zurich, Zurich, ZH, Switzerland
  • Getwan, Maike, Universitat Zurich, Zurich, ZH, Switzerland
  • Pichler, Roman, Albert-Ludwigs-Universitat Freiburg Medizinische Fakultat, Freiburg, Baden-Württemberg, Germany
  • Lienkamp, Soeren S., Universitat Zurich, Zurich, ZH, Switzerland
Background

Hepatocyte nuclear factor 1-beta (HNF1B) is a transcription factor involved in various stages of nephrogenesis and maintenance of renal tubular functions. Mutations in HNF1B are the most common monogenic causes for developmental renal disease, yet the underlying pathways affected are not fully understood. By comparative analysis in Xenopus and directly reprogrammed mammalian cells (iRECs) we investigated a patient-specific mutation (R295C) associated with cystic-dysplastic kidneys.

Methods

We used HNF1B to form renal-like organoids from Xenopus explants. In parallel, we analyzed how HNF1B R295C affects nephrogenesis in iRECs. Transcriptional changes were comparatively analyzed in two different species. We confirmed HNF1B target candidates in vivo using CRISPR/Cas9 editing of Xenopus embryos.

Results

We show that HNF1B is not only an essential component in direct reprogramming but can also induce ectopic pronephric tissue in Xenopus ectodermal explants. Changes in the transcriptomic profile demonstrated alterations in specific transcriptional modules and identified novel direct and indirect targets of the transcription factor HNF1B, which are linked to signaling pathways associated with renal morphogenesis, cilia and organic anion transport.

Conclusion

In conclusion, the combined use of directly reprogrammed mammalian cells and Xenopus renal organoid experiments allow us to gain a unique perspective into evolutionary conserved mechanisms of renal development and HNF1B associated kidney disease.