Pancreatic stellate cell-derived citrate switches pancreatic cancer cell metabolism towards a more mitochondrial phenotype with minimal effects on bioenergetics and cytosolic Ca2+ homeostasis

Pancreatology ◽  
2019 ◽  
Vol 19 ◽  
pp. S84
Author(s):  
Jason I.E. Bruce ◽  
Ahlam Sultan ◽  
Andrew James ◽  
Daniel A. Richardson ◽  
Howbeer Muhamad-Ali ◽  
...  
2015 ◽  
Author(s):  
Fernando F. Blanco ◽  
Mahsa Zarei ◽  
Jonathan R. Brody ◽  
Laszlo G. Boros ◽  
Jordan M. Winter

2016 ◽  
Vol 61 ◽  
pp. S82
Author(s):  
T. Tataranni ◽  
F. Agriesti ◽  
V. Ruggieri ◽  
C. Mazzoccoli ◽  
I. Laurenzana ◽  
...  

2022 ◽  
Author(s):  
Daniel R Plaugher ◽  
Boris Aguilar ◽  
David Murrugarra

Pancreatic Ductal Adenocarcinoma (PDAC) is widely known for its poor prognosis because it is often diagnosed when the cancer is in a later stage. We built a model to analyze the microenvironment of pancreatic cancer in order to better understand the interplay between pancreatic cancer, stellate cells, and their signaling cytokines. Specifically, we have used our model to study the impact of inducing four common mutations: KRAS, TP53, SMAD4, and CDKN2A. After implementing the various mutation combinations, we used our stochastic simulator to derive aggressiveness scores based on simulated attractor probabilities and long-term trajectory approximations. These aggression scores were then corroborated with clinical data. Moreover, we found sets of control targets that are effective among common mutations. These control sets contain nodes within both the pancreatic cancer cell and the pancreatic stellate cell, including PIP3, RAF, PIK3 and BAX in pancreatic cancer cell as well as ERK and PIK3 pancreatic stellate cell. Many of these nodes were found to be differentially expressed among pancreatic cancer patients in the TCGA database. Furthermore, literature suggests that many of these nodes can be targeted by drugs currently in circulation. The results herein help provide a proof of concept in the path towards personalized medicine through a means of mathematical systems biology. All data and code used for running simulations, statistical analysis, and plotting is available on a GitHub repository at https://github.com/drplaugher/PCC_Mutations .


2020 ◽  
Vol 158 (6) ◽  
pp. S-221
Author(s):  
Chamini Perera ◽  
Zhihong Xu ◽  
Alpha Raj Mekapogu ◽  
S.M. Zahid Hosen ◽  
Srinivasa Pothula ◽  
...  

Author(s):  
Silpa Gampala ◽  
Fenil Shah ◽  
Xiaoyu Lu ◽  
Hye-ran Moon ◽  
Olivia Babb ◽  
...  

Abstract Background Pancreatic cancer is a complex disease with a desmoplastic stroma, extreme hypoxia, and inherent resistance to therapy. Understanding the signaling and adaptive response of such an aggressive cancer is key to making advances in therapeutic efficacy. Redox factor-1 (Ref-1), a redox signaling protein, regulates the conversion of several transcription factors (TFs), including HIF-1α, STAT3 and NFκB from an oxidized to reduced state leading to enhancement of their DNA binding. In our previously published work, knockdown of Ref-1 under normoxia resulted in altered gene expression patterns on pathways including EIF2, protein kinase A, and mTOR. In this study, single cell RNA sequencing (scRNA-seq) and proteomics were used to explore the effects of Ref-1 on metabolic pathways under hypoxia. Methods scRNA-seq comparing pancreatic cancer cells expressing less than 20% of the Ref-1 protein was analyzed using left truncated mixture Gaussian model and validated using proteomics and qRT-PCR. The identified Ref-1’s role in mitochondrial function was confirmed using mitochondrial function assays, qRT-PCR, western blotting and NADP assay. Further, the effect of Ref-1 redox function inhibition against pancreatic cancer metabolism was assayed using 3D co-culture in vitro and xenograft studies in vivo. Results Distinct transcriptional variation in central metabolism, cell cycle, apoptosis, immune response, and genes downstream of a series of signaling pathways and transcriptional regulatory factors were identified in Ref-1 knockdown vs Scrambled control from the scRNA-seq data. Mitochondrial DEG subsets downregulated with Ref-1 knockdown were significantly reduced following Ref-1 redox inhibition and more dramatically in combination with Devimistat in vitro. Mitochondrial function assays demonstrated that Ref-1 knockdown and Ref-1 redox signaling inhibition decreased utilization of TCA cycle substrates and slowed the growth of pancreatic cancer co-culture spheroids. In Ref-1 knockdown cells, a higher flux rate of NADP + consuming reactions was observed suggesting the less availability of NADP + and a higher level of oxidative stress in these cells. In vivo xenograft studies demonstrated that tumor reduction was potent with Ref-1 redox inhibitor similar to Devimistat. Conclusion Ref-1 redox signaling inhibition conclusively alters cancer cell metabolism by causing TCA cycle dysfunction while also reducing the pancreatic tumor growth in vitro as well as in vivo.


Sign in / Sign up

Export Citation Format

Share Document