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: PO1355

Vascularized Kidney Organoids on Chip for Efficacy and Toxicity Testing of Somatic Genome Editing

Session Information

Category: Genetic Diseases of the Kidneys

  • 1002 Genetic Diseases of the Kidneys: Non-Cystic

Authors

  • Gupta, Navin R., Massachusetts General Hospital, Boston, Massachusetts, United States
  • Wolf, Kayla J., Harvard University Wyss Institute for Biologically Inspired Engineering, Boston, Massachusetts, United States
  • Maremanda, Krishna Prahlad, Brigham and Women's Hospital, Boston, Massachusetts, United States
  • Kroll, Katharina T., Harvard University Wyss Institute for Biologically Inspired Engineering, Boston, Massachusetts, United States
  • Zhang, Chengcheng, Massachusetts General Hospital, Boston, Massachusetts, United States
  • Sabbisetti, Venkata, Brigham and Women's Hospital, Boston, Massachusetts, United States
  • Lewis, Jennifer A., Harvard University Wyss Institute for Biologically Inspired Engineering, Boston, Massachusetts, United States
  • Morizane, Ryuji, Massachusetts General Hospital, Boston, Massachusetts, United States
Background

Somatic genome editing has therapeutic potential to cure inheritable disease. Clinical translation requires safety and efficacy analyses. DNA editing may be widely discrepant across species due to genomic differences, necessitating human tissue-based platforms. AAV-based delivery of DNA editing elements is under clinical investigation. If delivered systemically, the kidney may be particularly susceptible to genome editing owing to high blood flow. Kidney organoids have been generated from human stem cells through the co-induction of nephron, stromal, and endothelial progenitor cells that mature to form multicompartment human kidney tissue. We married organoid and organ-on-chip technologies to facilitate the maturation of nephron epithelia and the development of perfusable vascular networks to simulate systemic delivery of genome editing elements.

Methods

Using human recombinant growth factors and defined small molecules, kidney organoids were generated from male and female, embryonic and induced, stem cell lines. To test the efficacy of an AAV2-based delivery system, the tropism of varied capsids, 2/8/9, for kidney compartments was assessed under static and perfused conditions. The AAV receptor expression was evaluated by single nuclear RNA-seq.

Results

The greatest infectivity was with capsid protein 2, whose receptor of heparin sulfate proteoglycan is expressed in proximal & distal tubules and podocytes of kidney organoids by single cellular transcriptomics. Biomarker analysis demonstrated a statistically significant increase in tubular injury markers, KIM-1 and MCP-1, after infection with AAV2/2 as compared to other AAVs. Following treatment with AAV2/2CMVeGFP, the majority of LTL+ and CDH1+ tubular epithelia were GFP+, while PODXL+ podocytes were poorly infected. The optimal AAV serotype, MOI, and duration of infection under static conditions were applied to vascularized kidney organoids. Initial on-chip testing supports enhanced infectivity by live-cell monitoring and wholemount immunostaining, including AAV infection in GFP+PODXL+ podocytes.

Conclusion

We propose vascularized kidney organoids may simulate the systemic delivery of AAVs across kidney compartments, as a pre-clinical testing platform of the efficacy and safety of somatic cell genome editing.

Funding

  • Other NIH Support