Abstract

BackgroundThe initiation and progression of hepatocellular carcinoma (HCC) are largely dependent on its local microenvironment. Adipocytes are an important component of hepatic microenvironment in nonalcoholic fatty liver disease (NAFLD), which is a significant risk factor for HCC. Given the global prevalence of NAFLD, a better understanding of the interplay between HCC cells and adipocytes is urgently needed. Exosomes, released by malignant cells, represent a novel way of cell-cell interaction and have been shown to play an important role in cancer cell communication with their microenvironment. Here, we explore the role of HCC-derived exosomes in the cellular and molecular conversion of adipocytes into tumor-promoting cells.MethodsExosomes were isolated from HCC cell line HepG2 and added to adipocytes. Transcriptomic alterations of exosome-stimulated adipocytes were analyzed using gene expression profiling, and secretion of inflammation-associated cytokines was detected by RT-PCR and ELISA. In vivo mouse xenograft model was used to evaluate the growth-promoting and angiogenesis-enhancing effects of exosome-treated adipocytes. Protein content of tumor exosomes was analyzed by mass spectrometry. Activated phospho-kinases involved in exosome-treated adipocytes were detected by phospho-kinase antibody array and Western blot.ResultsOur results demonstrated that HCC cell HepG2-derived exosomes could be actively internalized by adipocytes and caused significant transcriptomic alterations and in particular induced an inflammatory phenotype in adipocytes. The tumor exosome-treated adipocytes, named exo-adipocytes, promoted tumor growth, enhanced angiogenesis, and recruited more macrophages in mouse xenograft model. In vitro, conditioned medium from exo-adipocytes promoted HepG2 cell migration and increased tube formation of human umbilical vein endothelial cells (HUVECs). Mechanistically, we found HepG2 exosomes activated several phopho-kinases and NF-κB signaling pathway in exo-adipocytes. Additionally, a total of 1428 proteins were identified in HepG2 exosomes by mass spectrometry.ConclusionsOur results provide new insights into the concept that tumor cell-derived exosomes can educate surrounding adipocytes to create a favorable microenvironment for tumor progression.

Highlights

  • The initiation and progression of hepatocellular carcinoma (HCC) are largely dependent on its local microenvironment

  • Our results demonstrated that HCC cell line HepG2-derived exosomes could be actively internalized by adipocytes differentiated from mesenchymal stem cells (MSCs) and caused significant transcriptomic alterations, and in particular, induced an inflammatory phenotype in adipocytes

  • Exo-adipocyte-conditioned medium is chemotaxic and promotes HepG2 migration The increased number of F4/80 macrophages in tumors receiving exo-adipocytes prompted us to investigate the effect of exo-adipocyte-conditioned medium on THP-1 cells in vitro as THP-1 cells are one of the exo-adipocyte-conditioned medium enhanced tube formation of human umbilical vein endothelial cells (HUVECs) To confirm our in vivo findings that exo-adipocytes promote angiogenesis in tumors, we examined the effect of exo-adipocyte-conditioned medium on HUVECs

Read more

Summary

Introduction

The initiation and progression of hepatocellular carcinoma (HCC) are largely dependent on its local microenvironment. Adipocytes are an important component of hepatic microenvironment in nonalcoholic fatty liver disease (NAFLD), which is a significant risk factor for HCC. Given the global prevalence of NAFLD, a better understanding of the interplay between HCC cells and adipocytes is urgently needed. Hepatocellular carcinoma (HCC) represents the fifth most common cancer worldwide and the third leading cause of cancer-related mortality [1, 2]. Both diagnostic and therapeutic strategies for HCC have improved over the past decades, the 5-year survival rate only is around 10%, and HCC continues to be a global health issue, especially in Asian countries [3, 4]. The interplay between the HCC cells and adjacent adipocytes remains poorly understood so far

Methods
Results
Discussion
Conclusion
Full Text
Paper version not known

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

Disclaimer: All third-party content on this website/platform is and will remain the property of their respective owners and is provided on "as is" basis without any warranties, express or implied. Use of third-party content does not indicate any affiliation, sponsorship with or endorsement by them. Any references to third-party content is to identify the corresponding services and shall be considered fair use under The CopyrightLaw.