Abstract

Simple SummaryProtein canopy homolog 2 (CNPY2) controls the outgrowth of neurites through positive regulation of their outgrowth in neuroblastoma and pheochromocytoma due to stabilizing the myosin regulatory light chain (MRLC). Given the important role in endoplasmic reticulum (ER) stress and proteolysis, we focused on investigation of CNPY2 expression in mouse and human hepatocarcinogenesis and its clinical relevance in virus-associated liver cancer. CNPY2 elevation and coordinated accumulation of numerous cytoskeletal proteins and those involved in ER and mitochondrial stresses was found in mouse altered foci and tumors by proteome analysis of microdissected lesions. In Huh7 and HepG2 human liver cancer cells, CNPY2 increase was significantly associated with cell cycle progression, activated cell proliferation and invasion. CNPY2 expression was high in HCV-associated HCC patients, being positively associated with survival. To the best of our knowledge, this is the first report to demonstrate that CNPY2 may become a useful prognostic biomarker in HCV-associated HCC. In the present study, the role of a novel protein involved in neurite development and endoplasmic reticulum (ER) stress, canopy homolog 2 (CNPY2), was investigated in mouse and human hepatocarcinogenesis. Firstly, a sensitive quantitative and qualitative detection of protein expression using QSTAR Elite LC-Ms/Ms was performed for the analysis of lysates of microdissected hepatocellular altered foci (AF), adenomas (HCAs), carcinomas (HCCs) and peri-tumoral livers from C57Bl/6J mice treated with diethylnitrosamine (DEN) and then maintained for 27 or 38 weeks on basal diet. Significant overexpression of 18.5 kDa CNPY2 processed form was demonstrated in AF, HCAs and HCCs, while low expression was observed in the livers of DEN-treated and control mice. Furthermore, CNPY2 elevation in AF and tumors was coordinated with accumulation of numerous cytoskeletal proteins, including cytokeratins 8 and 18, actin, non-muscle myosin and septin 9 and those involved in ER and mitochondrial stresses such as calreticulin, prohibitins 1 and 2 and YME1-like-1. Knockdown of CNPY2 in Huh7 and HepG2 human liver cancer cells resulted in significant suppression of cell survival and invasive potential, inhibition of cyclin D1, induction of p21Waf1/Cip1 and suppression of the apoptosis inhibitor Bcl2. In contrast, transfection of a mouse CNPY2 (mCNPY2-Ds-Red) vector plasmid in Huh7 and HepG2 cancer cells, with subsequent accumulation of CNPY2 in the ER, resulted in significant increase in cancer cells survival. Clinicopathological analysis in 90 HCV-positive HCC patients, revealed significant association of CNPY2 overexpression with poor overall (p = 0.041) survival. Furthermore, CNPY2 increase was associated with vessel invasion (p = 0.038), poor histological differentiation (p = 0.035) and advanced clinical stage (p = 0.016). In conclusion, CNPY2 is a promising molecular target elevated early in hepatocarcinogenesis and prognostic marker for human HCV-associated HCC. CNPY2 is involved in the processes of ER stress, cell cycle progression, proliferation, survival and invasion of liver tumor cells.

Highlights

  • Hepatocellular carcinoma (HCC), the fifth most common cancer and the third leading cause of death from cancer worldwide [1], commonly emerges on a background of chronic liver disease with continuous rounds of necrosis and regeneration, inflammation and oxidative stress [2,3]

  • Formalin-fixed and paraffin embedded (FFPE) serial liver sections with altered foci (AF), hepatocellular adenomas (HCAs) and HCCs were prepared using liver samples from our previous experiment, in which development of liver preneoplastic and neoplastic lesions was observed after DEN administration to 6-week-old C57Bl/6J mice [6]

  • For the identification of proteins participating in the onset and progression of mice hepatocarcinogenesis, we compared the proteomes of microdissected AF (27-week time-point), HCAs and HCCs (38-week time-point), peri-tumoral liver tissue, and normal liver tissue of untreated mice

Read more

Summary

Introduction

Hepatocellular carcinoma (HCC), the fifth most common cancer and the third leading cause of death from cancer worldwide [1], commonly emerges on a background of chronic liver disease with continuous rounds of necrosis and regeneration, inflammation and oxidative stress [2,3]. It was previously reported to control the outgrowth of neurites, through positive regulation of their outgrowth in neuroblastoma and pheochromocytoma PC12 cells due to stabilizing the myosin regulatory light chain (MRLC) [8], and has been suggested as a novel modulator of cell motility [9] In neurite, this effect is likely to be related to the prevention of MRLC-interacting protein (MIR)-mediated ubiquitination of myosin and its subsequent proteasomal degradation [8]. Co-localization of human CNPY2 and monkey ezrin-radixin-moesin (ERM)-like MIR proteins have been reported to occur in intracellular spaces, such as with COS-7 cells and in primary neurons [8] These processes could induce dynamic interactions between the actin cytoskeleton and specific proteins, crucial for changes in cell shape and motility. In vitro functional and clinicopathological analyses were carried out to investigate CNPY2 role in human liver cancer and its association with HCV+ HCC patients survival and clinicopathological variables

Results
Immunohistochemical Assessment of CNPY2 and Related Proteins in Mice Livers
Discussion
Institutional Review Board Approval
Liver Tissue from In Vivo Experiment
Laser Microdissection
Immunohistochemistry and Scoring
Patients and Tissue Samples
Generation of the Mouse CNPY2 Containing Vector
Invasion Assay
Real-Time Quantitative PCR
Protein Extraction and Western Blot Analysis
4.10. Statistical Analysis
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