Abstract

Pancreatic ductal adenocarcinoma (PDAC) is one of the most aggressive malignancies with limited survival rate. Roles for peptidylarginine deiminases (PADs) have been studied in relation to a range of cancers with roles in epigenetic regulation (including histone modification and microRNA regulation), cancer invasion, and extracellular vesicle (EV) release. Hitherto though, knowledge on PADs in PDAC is limited. In the current study, two PDAC cell lines (Panc-1 and MiaPaCa-2) were treated with pan-PAD inhibitor Cl-amidine as well as PAD2, PAD3, and PAD4 isozyme-specific inhibitors. Effects were assessed on changes in EV signatures, including EV microRNA cargo (miR-21, miR-126, and miR-221), on changes in cellular protein expression relevant for pancreatic cancer progression and invasion (moesin), for mitochondrial housekeeping (prohibitin, PHB), and gene regulation (deiminated histone H3, citH3). The two pancreatic cancer cell lines were found to predominantly express PAD2 and PAD3, which were furthermore expressed at higher levels in Panc-1, compared with MiaPaCa-2 cells. PAD2 isozyme-specific inhibitor had the strongest effects on reducing Panc-1 cell invasion capability, which was accompanied by an increase in moesin expression, which in pancreatic cancer is found to be reduced and associated with pancreatic cancer aggressiveness. Some reduction, but not significant, was also found on PHB levels while effects on histone H3 deimination were variable. EV signatures were modulated in response to PAD inhibitor treatment, with the strongest effects observed for PAD2 inhibitor, followed by PAD3 inhibitor, showing significant reduction in pro-oncogenic EV microRNA cargo (miR-21, miR-221) and increase in anti-oncogenic microRNA cargo (miR-126). While PAD2 inhibitor, followed by PAD3 inhibitor, had most effects on reducing cancer cell invasion, elevating moesin expression, and modulating EV signatures, PAD4 inhibitor had negligible effects and pan-PAD inhibitor Cl-amidine was also less effective. Compared with MiaPaCa-2 cells, stronger modulatory effects for the PAD inhibitors were observed in Panc-1 cells, which importantly also showed strong response to PAD3 inhibitor, correlating with previous observations that Panc-1 cells display neuronal/stem-like properties. Our findings report novel PAD isozyme regulatory roles in PDAC, highlighting roles for PAD isozyme-specific treatment, depending on cancer type and cancer subtypes, including in PDAC.

Highlights

  • Pancreatic ductal adenocarcinoma (PDAC) is a major cause of cancer-associated deaths in Western countries and is the eighth main source of cancer-related deaths globally [1].Despite advances in diagnostic technology, PDAC is usually diagnosed late and at an incurable stage, while early diagnosis is directly linked to improved survival [2]

  • peptidylarginine deiminases (PADs) Isozymes Are Differently Expressed in PDAC Cells

  • We found that extracellular vesicle (EV) signatures were differently modulated in response to the PAD isozymespecific inhibitors, including some effects observed on EV subpopulations and EV miR

Read more

Summary

Introduction

Pancreatic ductal adenocarcinoma (PDAC) is a major cause of cancer-associated deaths in Western countries and is the eighth main source of cancer-related deaths globally [1].Despite advances in diagnostic technology, PDAC is usually diagnosed late and at an incurable stage, while early diagnosis is directly linked to improved survival [2]. Peptidylarginine deiminases (PADs) are a group of calcium-dependent enzymes that cause post-translational deimination/citrullination in target proteins, leading to changes in their structure and function, affecting protein–protein interactions, generation of neoepitopes, and modulating gene regulation [6,7,8]. This post-translational modification may aid protein moonlighting, a phylogenetically conserved mechanism that allows proteins to carry out numerous functions within one polypeptide chain, in relation to both physiological and pathophysiological functions [9,10].

Objectives
Methods
Discussion
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