Abstract

Chronic and persistent inflammation is a well-known carcinogenesis promoter. Hepatocellular carcinoma (HCC) is one of the most common inflammation-associated cancers; most HCCs arise in the setting of chronic inflammation and hepatic injury. Both NF-κB and STAT3 are important regulators of inflammation. Centrosomal P4.1-associated protein (CPAP), a centrosomal protein that participates primarily in centrosome functions, is overexpressed in HCC and can increase TNF-α-mediated NF-κB activation and IL-6-induced STAT3 activation. A transgenic (Tg) mouse model with hepatocyte-specific CPAP expression was established to investigate the physiological role of CPAP in hepatocarcinogenesis. Obvious inflammatory cell accumulation and fatty change were observed in the livers of CPAP Tg mice. The alanine aminotransferase (ALT) level and the expression levels of inflammatory genes, such as IL-6, IL-1β and TNF-α, were higher in CPAP Tg mice than in wild type (WT) mice. High-dose/short-term treatment with diethylnitrosamine (DEN) increased the ALT level, proinflammatory gene expression levels, and STAT3 and NF-κB activation in CPAP Tg mice; low-dose/long-term DEN treatment induced more severe liver tumor formation in CPAP Tg mice than in WT mice. CPAP can increase the expression of chemokine (C-C motif) ligand 16 (CCL-16), an important chemotactic cytokine, in human hepatocytes. CCL-16 expression is positively correlated with CPAP and TNF-α mRNA expression in the peritumoral part of HCC. In summary, these results suggest that CPAP may promote hepatocarcinogenesis through enhancing the inflammation pathway via increasing the expression of CCL-16.

Highlights

  • Hepatocellular carcinoma (HCC) is one of the most aggressive cancers and the second leading cause of cancer mortality worldwide [1]

  • Founder mice were generated by injecting the albumin-driven Centrosomal P4.1-associated protein (CPAP) construct into fertilized eggs of C57BL/6 mice (Supplementary Fig. S1A); three lines of liver-specific CPAP Tg mice were selected for further investigation

  • Our results showed that overexpressed CPAP increases the expression of proinflammatory cytokines, including TNF-α, IL-6, IL-1β, IL-8, and liver-enriched CCL-16, by promoting the activation of two important inflammatory transcriptional factors—STAT3 and NF-κB—to create and maintain a chronic inflammatory microenvironment that facilitates the development of HCC (Fig. 8)

Read more

Summary

Introduction

Hepatocellular carcinoma (HCC) is one of the most aggressive cancers and the second leading cause of cancer mortality worldwide [1]. 70–90% of HCC cases are associated with chronic inflammation which may result from hepatitis infection, alcoholic liver disease, or nonalcoholic fatty liver disease [2]. Various types of cells interact with chemokines to prevent the spread of pathogen infection; and these interactions maintain physiological homeostasis through a negative feedback mechanism [3, 4]. The uncontrolled action of regulatory mechanisms resulting from sustained inflammation (e.g., macrophage polarization toward the M2 phenotype, increased immunosuppressive cell infiltration, and prolonged or excessive inflammatory responses) will cause nonresolving inflammation and subsequently lead to cellular oncogenic transformation as well as genetic and epigenetic changes [4, 5]. Many studies have investigated whether the regulatory relationship between various proinflammatory cells (including immune cells) and inflammatory mediators within the tumor microenvironment is a potent target for cancer prevention and therapy [6,7,8]

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