Abstract

Cancer-associated fibroblasts (CAFs) play critical roles in cancer progression by regulating tumor cell proliferation, angiogenesis, and metastasis. Recent studies demonstrated that CAFs induce inhibitory immune cell infiltration and chemotherapy resistance in gastric cancer by activating the NF-κB signaling pathway to secrete IL6, IL8, and other inflammatory factors. Inhibition of the NF-κB signaling pathway in CAFs might be a potential therapeutic strategy in gastric cancer. However, how the NF-κB pathway is activated in CAFs remains unclear. We showed that mesenchymal stem cells (MSCs) differentiated into CAFs, induced by the exosomes derived from gastric cancer cells. During the process of differentiation from MSCs into CAFs, we showed that nuclear PKM2 expression was continuously upregulated and associated with NF-κB P65 acetylation, contributing to P65 nuclear retention in CAFs and constant transcription of IL-6, IL-8, and other inflammatory factors, thus promoting gastric cancer cell proliferation. We showed that NF-κB P65 acetylation was induced by P300. We showed that nuclear PKM2 was derived from exosomes of gastric cancer cell lines and the positive feedback loop induced by PKM2-P65 combination. It is also proved that P300 inhibitors can inhibit tumor proliferation in an AGS subcutaneous xenograft tumor model. Our study showed that gastric cancer cells influence the continuous activation of the NF-κB signaling pathway in CAFs by secreting gastric cancer exosomes containing PKM2, thus inducing abnormal metabolism and inflammation activation. This study provides a new therapeutic target for CAF normalization or deactivation strategies.

Highlights

  • Gastric cancer is the third leading cause of cancer-related death worldwide

  • In the in vitro model of Cancer-associated fibroblasts (CAFs) differentiated from mesenchymal stem cells (MSCs) by gastric cancer-derived exosomes, we found that the main protein components of gastric cancer-derived exosomes are metabolism-related proteins, especially glycolytic enzymes, M2 pyruvate kinase (PKM2), lactate dehydrogenase A (LDHA), and phosphofructokinase P (PFKP), while PKM2 was frequently upregulated at the mRNA and protein levels in CAFs differentiated from MSCs

  • We presented that PKM2 can directly bind to nuclear factor (NF)-κB P65 in CAFs compared to the MSC controls, using the Co-IP assay (Fig. 5C, D)

Read more

Summary

Introduction

Gastric cancer is the third leading cause of cancer-related death worldwide. The overall survival time of patients withOfficial journal of the Cell Death Differentiation AssociationGu et al Cell Death and Disease (2021)12:291metastasis[3]. Gastric cancer is the third leading cause of cancer-related death worldwide. Official journal of the Cell Death Differentiation Association. Gu et al Cell Death and Disease (2021)12:291. CAFs play a prominent role in enhancing gastric cancer cell proliferation, angiogenesis, migration, and invasion and in promoting inhibitory immune cell infiltration, thereby leading to chemotherapy resistance in gastric cancer[3]. There are two main practices: direct CAF depletion via surface markers and normalization of CAFs4. Direct CAF depletion relies on targeting unique CAF markers. It is difficult to practice direct CAF depletion. Normalization of CAFs, via suppressing the tumor-promoting effective molecules and CAF-activated signaling pathways, seems to be more practical. It is important to investigate the mechanism of abnormal activation of CAFs to screen an appropriate therapeutic target

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