Abstract

Abstract The stochasticity of transcription leads to phenotypic variation within a clonal population. This means that within a healthy tissue, a fraction of cells could assume an “edge” transcriptional state that is primed for a transition toward malignancy. We find such an edge transcriptional state amongst acinar cells in a publicly available PDAC single-cell RNA-seq dataset. These acinar edge cells (AE) cells have transcriptomes that are highly diverged from a typical normal acinar cell, and show transcriptional similarities to pancreatic ductal adenocarcinoma (PDAC). The AE state can be found in several single-cell datasets from healthy human pancreatic tissue. The AE state is not mutationally-driven as AE cells in healthy tissues do not possess any known oncogenic driver mutations, nor are they clonally derived. The transition from a non-edge acinar to an AE state recapitulates key aspects of pancreatic biology; genes associated with this transition are enriched for known markers of acinar de-differentiation and acinar-ductal metaplasia. Further, the AE state represents a partial reversion to a SOX9+PTF1A+ multi-potent embryonic pancreatic progenitor state, likely through the activation of KLF5, HES1 and RBPJ targets. Remarkably, the promoters of genes upregulated in the AE state are significantly hypomethylated in PDAC relative to normal pancreas, while the genes up-regulated in the AE state are up-regulated in PDAC samples in the Cancer Genome Atlas (TCGA) database. It is thus likely that transcriptomic changes in the edge state are fixed epigenetically during tumor initiation and progression. A gene signature derived from the AE cells shows strong association with survival among PDAC patients. Remarkably, we also find that the fraction of AE cells in the healthy pancreas increases with age, which points to a strong non-genetic and transcriptomic component to the known increase in PDAC risk with age. We also observed edge-like transcriptomic heterogeneity among alveolar, ciliated and club cells in the human lung, and amongst hepatocytes in liver tissues. This suggests that, across tissues, gene expression heterogeneity can lead to sub-populations of healthy cells across tissues existing in pre-malignant states. Citation Format: Vishaka Gopalan, Arashdeep Singh, Farid Rashidi, Li Wang, Eytan Ruppin, Efsun Arda, Sridhar Hannenhalli. A transcriptionally distinct subpopulation of healthy acinar cells exhibit features of pancreatic progenitors and pancreatic ductal adeno carcinoma [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2021; 2021 Apr 10-15 and May 17-21. Philadelphia (PA): AACR; Cancer Res 2021;81(13_Suppl):Abstract nr LB200.

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