Abstract

Cell-collagen interactions are crucial for cell migration and invasion during cancer development and progression. Heat shock protein 47 (HSP47) is an endoplasmic reticulum-resident molecular chaperone that facilitates collagen maturation and deposition. It has been previously shown that HSP47 expression in cancer cells is crucial for cancer invasiveness. However, exogenous collagen cannot rescue cell invasion in HSP47-silenced cancer cells, suggesting that other HSP47 targets contribute to cancer cell invasion. Here, we show that HSP47 expression is required for the stability and cell-surface expression of discoidin domain-containing receptor 2 (DDR2) in breast cancer tissues. HSP47 silencing reduced DDR2 protein stability, accompanied by suppressed cell migration and invasion. Co-immunoprecipitation results revealed that HSP47 binds to the DDR2 ectodomain. Using a photoconvertible technique and total internal reflection fluorescence microscopy, we further demonstrate that HSP47 expression significantly sustains the membrane localization of the DDR2 protein. These results suggest that binding of HSP47 to DDR2 increases DDR2 stability and regulates its membrane dynamics and thereby enhances cancer cell migration and invasion. Given that DDR2 has a crucial role in the epithelial-to-mesenchymal transition and cancer progression, targeting the HSP47-DDR2 interaction might be a potential strategy for inhibiting DDR2-dependent cancer progression.

Highlights

  • Cell– collagen interactions are crucial for cell migration and invasion during cancer development and progression

  • Using gene co-expression analysis, we showed that mRNA levels of Heat shock protein 47 (HSP47) and discoidin domain–containing receptor 2 (DDR2) were significantly correlated in human breast cancer tissues (Fig. 1A)

  • We found that DDR2 protein levels were reduced in HSP47-silenced MDA-MB-231 cells (Fig. 1, B and C), which is associated with reduced cell migration and invasion [7]

Read more

Summary

Introduction

Cell– collagen interactions are crucial for cell migration and invasion during cancer development and progression. HSP47 silencing reduced DDR2 protein stability, accompanied by suppressed cell migration and invasion. Using a photoconvertible technique and total internal reflection fluorescence microscopy, we further demonstrate that HSP47 expression significantly sustains the membrane localization of the DDR2 protein These results suggest that binding of HSP47 to DDR2 increases DDR2 stability and regulates its membrane dynamics and thereby enhances cancer cell migration and invasion. HSP47 is a heat shock protein that is produced when cells are exposed to stressful conditions [11,12,13] These results suggest that HSP47 has additional targets during cancer progression and in response to stressful conditions. Identification of HSP47 targets may advance our understanding how HSP47 expression promotes cell migration and provide new insights into function of HSP47 in cancer progression

Methods
Results
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