Skip to main content
Top
Published in: Journal of Experimental & Clinical Cancer Research 1/2021

Open Access 01-12-2021 | Pancreatic Cancer | Research

Ref-1 redox activity alters cancer cell metabolism in pancreatic cancer: exploiting this novel finding as a potential target

Authors: Silpa Gampala, Fenil Shah, Xiaoyu Lu, Hye-ran Moon, Olivia Babb, Nikkitha Umesh Ganesh, George Sandusky, Emily Hulsey, Lee Armstrong, Amber L. Mosely, Bumsoo Han, Mircea Ivan, Jing-Ruey Joanna Yeh, Mark R. Kelley, Chi Zhang, Melissa L. Fishel

Published in: Journal of Experimental & Clinical Cancer Research | Issue 1/2021

Login to get access

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.
Appendix
Available only for authorised users
Literature
15.
go back to reference Fung H, Demple B. A vital role for Ape1/Ref1 protein in repairing spontaneous DNA damage in human cells. Mol Cell. 2005;17:463–70.CrossRef Fung H, Demple B. A vital role for Ape1/Ref1 protein in repairing spontaneous DNA damage in human cells. Mol Cell. 2005;17:463–70.CrossRef
16.
go back to reference Izumi T, et al. Two essential but distinct functions of the mammalian abasic endonuclease. Proc Natl Acad Sci U S A. 2005;102:5739–43.CrossRef Izumi T, et al. Two essential but distinct functions of the mammalian abasic endonuclease. Proc Natl Acad Sci U S A. 2005;102:5739–43.CrossRef
20.
go back to reference Lando D, Pongratz I, Poellinger L, Whitelaw ML. A redox mechanism controls differential DNA binding activities of hypoxia-inducible factor (HIF) 1alpha and the HIF-like factor. J Biol Chem. 2000;275:4618–27.CrossRef Lando D, Pongratz I, Poellinger L, Whitelaw ML. A redox mechanism controls differential DNA binding activities of hypoxia-inducible factor (HIF) 1alpha and the HIF-like factor. J Biol Chem. 2000;275:4618–27.CrossRef
22.
go back to reference Kelley MR, Georgiadis MM, Fishel ML. APE1/Ref-1role in redox signaling: translational applications of targeting the redox function of the DNA repair/redox protein APE1/Ref-1. Curr Mol Pharmacol. 2012;5:36–53.CrossRef Kelley MR, Georgiadis MM, Fishel ML. APE1/Ref-1role in redox signaling: translational applications of targeting the redox function of the DNA repair/redox protein APE1/Ref-1. Curr Mol Pharmacol. 2012;5:36–53.CrossRef
32.
go back to reference Jiang Y, Zhou S, Sandusky GE, Kelley MR, Fishel ML. Reduced expression of DNA repair and redox signaling protein APE1/Ref-1 impairs human pancreatic cancer cell survival, proliferation, and cell cycle progression. Cancer Invest. 2010;28:885–95.CrossRef Jiang Y, Zhou S, Sandusky GE, Kelley MR, Fishel ML. Reduced expression of DNA repair and redox signaling protein APE1/Ref-1 impairs human pancreatic cancer cell survival, proliferation, and cell cycle progression. Cancer Invest. 2010;28:885–95.CrossRef
34.
go back to reference McIlwain DW, Fishel ML, Boos A, Kelley MR, Jerde TJ. APE1/Ref-1 redox-specific inhibition decreases survivin protein levels and induces cell cycle arrest in prostate cancer cells. Oncotarget. 2018;9:10962–77.CrossRef McIlwain DW, Fishel ML, Boos A, Kelley MR, Jerde TJ. APE1/Ref-1 redox-specific inhibition decreases survivin protein levels and induces cell cycle arrest in prostate cancer cells. Oncotarget. 2018;9:10962–77.CrossRef
45.
go back to reference Wan C, et al. Fast and Efficient Boolean Matrix Factorization by Geometric Segmentation. Proceedings of the AAAI Conference on Artificial Intelligence. 2019. p. 6086–6093. Wan C, et al. Fast and Efficient Boolean Matrix Factorization by Geometric Segmentation. Proceedings of the AAAI Conference on Artificial Intelligence. 2019. p. 6086–6093.
49.
go back to reference Fan Z, et al. Cleaving the oxidative repair protein Ape1 enhances cell death mediated by granzyme A. Nat Immunol. 2003;4:145–53.CrossRef Fan Z, et al. Cleaving the oxidative repair protein Ape1 enhances cell death mediated by granzyme A. Nat Immunol. 2003;4:145–53.CrossRef
50.
go back to reference Wang D, Luo M, Kelley MR. Human apurinic endonuclease 1 (APE1) expression and prognostic significance in osteosarcoma: enhanced sensitivity of osteosarcoma to DNA damaging agents using silencing RNA APE1 expression inhibition. Mol Cancer Ther. 2004;3:679–86.PubMed Wang D, Luo M, Kelley MR. Human apurinic endonuclease 1 (APE1) expression and prognostic significance in osteosarcoma: enhanced sensitivity of osteosarcoma to DNA damaging agents using silencing RNA APE1 expression inhibition. Mol Cancer Ther. 2004;3:679–86.PubMed
54.
go back to reference Sempere LF, Gunn JR, Korc M. A novel 3-dimensional culture system uncovers growth stimulatory actions by TGFbeta in pancreatic cancer cells. Cancer Biol Ther. 2011;12:198–207.CrossRef Sempere LF, Gunn JR, Korc M. A novel 3-dimensional culture system uncovers growth stimulatory actions by TGFbeta in pancreatic cancer cells. Cancer Biol Ther. 2011;12:198–207.CrossRef
60.
go back to reference Fitzmaurice GM, Laird NM, Ware JH. Statistic in Medicine. In; Applied longitudinal analysis. Hoboken: Wiley Online Library; 2004. Fitzmaurice GM, Laird NM, Ware JH. Statistic in Medicine. In; Applied longitudinal analysis. Hoboken: Wiley Online Library; 2004.
65.
go back to reference Wan C, et al. Association for the advancement of artificial intelligence. 2019. Wan C, et al. Association for the advancement of artificial intelligence. 2019.
89.
go back to reference Wang X, et al. APE1/Ref-1 regulates 5-FU resistance in colon cancer cells through its redox and endonuclease activity. Int J Clin Exp Med. 2019;12:3870–8. Wang X, et al. APE1/Ref-1 regulates 5-FU resistance in colon cancer cells through its redox and endonuclease activity. Int J Clin Exp Med. 2019;12:3870–8.
Metadata
Title
Ref-1 redox activity alters cancer cell metabolism in pancreatic cancer: exploiting this novel finding as a potential target
Authors
Silpa Gampala
Fenil Shah
Xiaoyu Lu
Hye-ran Moon
Olivia Babb
Nikkitha Umesh Ganesh
George Sandusky
Emily Hulsey
Lee Armstrong
Amber L. Mosely
Bumsoo Han
Mircea Ivan
Jing-Ruey Joanna Yeh
Mark R. Kelley
Chi Zhang
Melissa L. Fishel
Publication date
01-12-2021
Publisher
BioMed Central
Published in
Journal of Experimental & Clinical Cancer Research / Issue 1/2021
Electronic ISSN: 1756-9966
DOI
https://doi.org/10.1186/s13046-021-02046-x

Other articles of this Issue 1/2021

Journal of Experimental & Clinical Cancer Research 1/2021 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine