scholarly journals P06.04 Transcriptome-wide network analysis predicts the role of lactate dehydrogenase C in breast cancer cell survival and immune dysfunction

2021 ◽  
Vol 9 (Suppl 1) ◽  
pp. A20.2-A21
Author(s):  
A Naik ◽  
J Decock

BackgroundCancer testis antigens (CTAs) are lucrative anti-cancer targets given their restricted expression patterns and known roles as mediators of cancer hallmarks, including cancer metabolism, proliferation, survival, and cell motility. Lactate dehydrogenase C (LDHC) is a CTA with upregulated expression in poor prognosis subtypes of breast cancer, however its tumorigenic role is less understood. We recently reported that silencing LDHC reduces breast cancer cell survival through a dysregulated DNA damage response, thus highlighting its potential as an anti-cancer target with limited off-target effects. This study aimed to explore the changes in the transcriptome of breast cancer cells and immune-related mediators upon in vitro LDHC targeting.Materials and MethodsWe silenced LDHC expression in breast cancer cell lines and investigated the downstream effects on the tumor cell transcriptome. Differentially expressed genes were subjected to regulatory network analyses. We further assessed the secretory profile of cytokines and immune checkpoint expression in LDHC-silenced cells and used the Tumor Immune Dysfunction and Exclusion (TIDE) algorithm to determine the effect of the interaction between LDHC expression and cytotoxic T lymphocyte (CTL) infiltration in the METABRIC breast cancer cohort.ResultsNetwork analysis to investigate the effects of silencing LDHC on the tumor cell transcriptome identified 47 up- and 55 down-regulated transcripts (2.0-fold change, adj p<0.05). Differentially expressed genes in the LDHC-silenced cells were particularly enriched in canonical pathways regulating cell cycle checkpoint control, BRCA1-mediated DNA damage response and NF-kb signaling in response to infection. Upstream regulator analyses revealed the altered expression profile was associated with mTOR (p=1.27e-5, z=2.242) and CASP3 (p=3.2e-4, z=2.250) pathways, which in the presence of LDHC are predicted to activate TP53, Myc, NF-KB complex, STAT1/3, PRKC, CDK2, FOXO3 and HIF-1a while inhibiting SMAD3, PTEN, ATM, IL18 and BCL2. Consequentially, the observed network-wide changes on LDHC silencing are predicted to negatively influence cellular growth and proliferation, cell migration and cell infiltration. The LDHC-associated network indicated a higher-level regulation by miR378a-3p (p=1.4e-7, z=-3.117), affecting the downstream mechanistic in LDHC-expressing cells. Interestingly, the miR378a causal network also indicated inhibition of the immune response in LDHC-positive cells. TIDE analysis indicated that high expression of LDHC in the METABRIC Her2 breast cancer cohort (TIDE score=1.97, p=0.049), and to a lesser extent in triple negative breast cancer (TIDE score=0.466, p=0.642), decreases the beneficial effect between CTLs and overall survival observed in LDHC Low tumors. Concurrently, LDHC-silenced cells displayed a pro-inflammatory gene expression and cytokine profile and down-regulated the expression of PD-L1 and Gal-9 immune checkpoints.ConclusionsOur findings provide an indication of potential CTL dysfunction in breast tumors with high LDHC expression and suggests that therapeutic targeting of LDHC may inhibit tumor growth while releasing the anti-tumor immune response in breast cancer.Disclosure InformationA. Naik: None. J. Decock: None.

2020 ◽  
Author(s):  
Laura Cheradame ◽  
Ida Chiara Guerrera ◽  
Julie Gaston ◽  
Alain Schmitt ◽  
Vincent Jung ◽  
...  

AbstractSTING (Stimulator of Interferon Genes) is a well-known endoplasmic reticulum-anchored adaptor of the innate immunity that triggers the expression of inflammatory cytokines in response to pathogen infection. In cancer cells, this pro-inflammatory pathway can be activated by genomic DNA damage potentiating antitumor immune responses. Here we report that STING promotes cancer cell survival and resistance to genotoxic treatment in a cell-autonomous manner. Mechanistically, we show that STING partly localizes at the inner nuclear membrane in various breast cancer cell lines and clinical tumor samples, and interacts with several proteins of the DNA damage response (DDR). STING overexpression enhances the amount of chromatin-bound DNA-dependent Protein Kinase (DNA-PK) complex, while STING silencing impairs DDR foci formation and DNA repair efficacy. Importantly, this function of STING is independent of its canonical pro-inflammatory pathway. This study highlights a previously unappreciated cell-autonomous tumor-promoting mechanism of STING that opposes its well-documented role in tumor immunosurveillance.Graphical abstract


2020 ◽  
Vol 16 ◽  
Author(s):  
Vibhavana Singh ◽  
Rakesh Reddy ◽  
Antarip Sinha ◽  
Venkatesh Marturi ◽  
Shravani Sripathi Panditharadyula ◽  
...  

: Diabetes and breast cancer are pathophysiologically similar and clinically established diseases that co-exist with a wider complex similar molecular signalling and having similar set of risk factors. Insulin plays a pivotal role for invasion and migration of breast cancer cells. Several ethnopharmacological evidences light the concomitant anti-diabetic and anti-cancer activity of medicinal plant and phytochemicals against breast tumor of patients with diabetes. This present article reviewed the findings on medicinal plants and phytochemicals with concomitant anti-diabetic and anti-cancer effects reported in scientific literature to facilitate the development of dual-acting therapies against diabetes and breast cancer. The schematic tabular form of published literatures on medicinal plants (63 plants belongs to 45 families) concluded the dynamics of phytochemicals against diabetes and breast tumor that could be explored further for the discovery of therapies for controlling of breast cancer cell invasion and migration in patient with diabetes.


Oncotarget ◽  
2017 ◽  
Vol 8 (60) ◽  
pp. 101461-101474 ◽  
Author(s):  
Yung-Lung Chang ◽  
Yu-Juei Hsu ◽  
Ying Chen ◽  
Yi-Wen Wang ◽  
Shih-Ming Huang

Oncogene ◽  
2001 ◽  
Vol 20 (50) ◽  
pp. 7413-7419 ◽  
Author(s):  
Yuvarani S Kuppumbatti ◽  
Brent Rexer ◽  
Shigeo Nakajo ◽  
Kazuyasu Nakaya ◽  
Rafael Mira-y-Lopez

Oncotarget ◽  
2019 ◽  
Vol 10 (42) ◽  
pp. 4307-4320 ◽  
Author(s):  
Anna Banach ◽  
Ya-Ping Jiang ◽  
Eric Roth ◽  
Cem Kuscu ◽  
Jian Cao ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document