Abstract

Hereditary cancers with cancer-predisposing mutations represent unique models of human oncogenesis, as a driving oncogenic event is present in germline. Currently, there are no satisfactory models to study these malignancies. We report the generation of IPSC from the somatic cells of a patient with hereditary c-met-mutated papillary renal cell carcinoma (PRCC). From these cells we have generated spontaneous aggregates organizing in structures which expressed kidney markers such as PODXL and Six2. These structures expressed PRCC markers both in vitro and in vivo in NSG mice. Gene-expression profiling showed striking molecular similarities with signatures found in a large cohort of PRCC tumor samples. This analysis, applied to primary cancers with and without c-met mutation, showed overexpression of the BHLHE40 and KDM4C only in the c-met-mutated PRCC tumors, as predicted by c-met-mutated embryoid bodies transcriptome. These data therefore represent the first proof of concept of “hereditary renal cancer in a dish” model using c-met-mutated iPSC-derived embryoid bodies, opening new perspectives for discovery of novel predictive progression markers and for drug-screening for future precision-medicine strategies.

Highlights

  • Hereditary cancers are due to oncogenic mutations or deletions and represent a major challenge in terms of diagnosis, prognosis, and prevention [1]

  • The propositus patient is a patient in whom the diagnosis of the c-met mutation was performed during a genetic study realized in her family, because of the occurrence of papillary renal cell carcinoma (PRCC) in her mother

  • In the “fusion embryoid bodies” type structures we detected strong expression of both PODXL and phalloidin throughout (Figure S5C,E). These results indicate that renal differentiation of met-IPSC could be assessed within 2 weeks with the formation of specific kidney embryoid bodies expressing PODXL glomerular marker

Read more

Summary

Introduction

Hereditary cancers are due to oncogenic mutations or deletions and represent a major challenge in terms of diagnosis, prognosis, and prevention [1]. The use of drug targeting strategies requires the generation of tissue-specific embryoid bodies, a technology which has been achieved in many tissues using either primary cells or pluripotent stem cells [7]. The feasibility and clinical use of iPSC-derived cancer embryoid bodies harboring a hereditary oncogenic mutation has not been shown so far. We show here the proof of concept of the feasibility of this strategy by generating iPSC lines bearing a c-met mutation. We developed from these patient-specific iPSC, embryoid bodies using a 3D in vitro culture system followed by extensive analysis by immunochemistry, transmission electron microscopy, and transcriptome studies

Methods
Results
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call