Abstract

Simple SummaryTumours are now known to develop more quickly when the tumour cell mass is located in a tissue that shows signs of chronic inflammation. Under such conditions, inflammatory cells from the surrounding tumour microenvironment provide survival signals to which cancer cells respond. We have previously found that some colorectal tumours overexpress the protein RAC1B that sustains tumour cell survival. Here we used a colon mucosa-like in vitro cell model and found that the presence of cancer-associated fibroblasts and pro-inflammatory macrophages stimulated colorectal cells to increase their RAC1B levels. Under these conditions, the secreted survival signals were analysed, and interleukin-6 identified as the main trigger for the increase in RAC1B levels. The results contribute to understand the tumour-promoting effect of inflammation at the molecular level.An inflammatory microenvironment is a tumour-promoting condition that provides survival signals to which cancer cells respond with gene expression changes. One example is the alternative splicing variant Rat Sarcoma Viral Oncogene Homolog (Ras)-Related C3 Botulinum Toxin Substrate 1 (RAC1)B, which we previously identified in a subset of V-Raf Murine Sarcoma Viral Oncogene Homolog B (BRAF)-mutated colorectal tumours. RAC1B was also increased in samples from inflammatory bowel disease patients or in an acute colitis mouse model. Here, we used an epithelial-like layer of polarized Caco-2 or T84 colorectal cancer (CRC) cells in co-culture with fibroblasts, monocytes or macrophages and analysed the effect on RAC1B expression in the CRC cells by RT-PCR, Western blot and confocal fluorescence microscopy. We found that the presence of cancer-associated fibroblasts and M1 macrophages induced the most significant increase in RAC1B levels in the polarized CRC cells, accompanied by a progressive loss of epithelial organization. Under these conditions, we identified interleukin (IL)-6 as the main trigger for the increase in RAC1B levels, associated with Signal Transducer and Activator of Transcription (STAT)3 activation. IL-6 neutralization by a specific antibody abrogated both RAC1B overexpression and STAT3 phosphorylation in polarized CRC cells. Our data identify that pro-inflammatory extracellular signals from stromal cells can trigger the overexpression of tumour-related RAC1B in polarized CRC cells. The results will help to understand the tumour-promoting effect of inflammation and identify novel therapeutic strategies.

Highlights

  • The mucosa of our gastrointestinal tract forms a physiological barrier that separates the surrounding tissues from the gut lumen, containing ingested food as well as microorganisms and environmental toxins [1]

  • Our studies further suggested that the overexpression of RAC1B is a subsequent genetic event in tumour-initiated colorectal cells, allowing them to escape from oncogene-induced senescence [31] and continue malignant progression

  • RAC1B was detected at the basolateral membranes

Read more

Summary

Introduction

The mucosa of our gastrointestinal tract forms a physiological barrier that separates the surrounding tissues from the gut lumen, containing ingested food as well as microorganisms and environmental toxins [1]. Highly polarized columnar cells form a tight-junction belt that seals the intercellular spaces and restricts any uptake of molecules or particles to highly selective transport mechanisms at the apical surface. This polarized cell layer is supported in the underlying submucosal tissue by a high immune cell surveillance. Activating mutations in the Kirsten Rat Sarcoma Viral Oncogene Homolog (KRAS) or V-Raf

Methods
Results
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