Skip to main content
Top
Published in: Journal of Hematology & Oncology 1/2019

Open Access 01-12-2019 | Metastasis | Research

CD73 promotes hepatocellular carcinoma progression and metastasis via activating PI3K/AKT signaling by inducing Rap1-mediated membrane localization of P110β and predicts poor prognosis

Authors: Xiao-Lu Ma, Min-Na Shen, Bo Hu, Bei-Li Wang, Wen-Jing Yang, Li-Hua Lv, Hao Wang, Yan Zhou, An-Li Jin, Yun-Fan Sun, Chuan-Yan Zhang, Shuang-Jian Qiu, Bai-Shen Pan, Jian Zhou, Jia Fan, Xin-Rong Yang, Wei Guo

Published in: Journal of Hematology & Oncology | Issue 1/2019

Login to get access

Abstract

Background

Hepatocellular carcinoma (HCC) is one of the most prevalent malignancies worldwide because of rapid progression and high incidence of metastasis or recurrence. Accumulating evidence shows that CD73-expressing tumor cell is implicated in development of several types of cancer. However, the role of CD73 in HCC cell has not been systematically investigated and its underlying mechanism remains elusive.

Methods

CD73 expression in HCC cell was determined by RT-PCR, Western blot, and immunohistochemistry staining. Clinical significance of CD73 was evaluated by Cox regression analysis. Cell counting kit-8 and colony formation assays were used for proliferation evaluation. Transwell assays were used for motility evaluations. Co-immunoprecipitation, cytosolic and plasma membrane fractionation separation, and ELISA were applied for evaluating membrane localization of P110β and its catalytic activity. NOD/SCID/γc(null) (NOG) mice model was used to investigate the in vivo functions of CD73.

Results

In the present study, we demonstrate that CD73 was crucial for epithelial-mesenchymal transition (EMT), progression and metastasis in HCC. CD73 expression is increased in HCC cells and correlated with aggressive clinicopathological characteristics. Clinically, CD73 is identified as an independent poor prognostic indicator for both time to recurrence and overall survival. CD73 knockdown dramatically inhibits HCC cells proliferation, migration, invasion, and EMT in vitro and hinders tumor growth and metastasis in vivo. Opposite results could be observed when CD73 is overexpressed. Mechanistically, adenosine produced by CD73 binds to adenosine A2A receptor (A2AR) and activates Rap1, which recruits P110β to the plasma membrane and triggers PIP3 production, thereby promoting AKT phosphorylation in HCC cells. Notably, a combination of anti-CD73 and anti-A2AR achieves synergistic depression effects on HCC growth and metastasis than single agent alone.

Conclusions

CD73 promotes progression and metastasis through activating PI3K/AKT signaling, indicating a novel prognostic biomarker for HCC. Our data demonstrate the importance of CD73 in HCC in addition to its immunosuppressive functions and revealed that co-targeting CD73 and A2AR strategy may be a promising novel therapeutic strategy for future HCC management.
Appendix
Available only for authorised users
Literature
1.
go back to reference Llovet JM, Montal R, Sia D, Finn RS. Molecular therapies and precision medicine for hepatocellular carcinoma. Nat Rev Clin Oncol. 2018;15:599–616.CrossRef Llovet JM, Montal R, Sia D, Finn RS. Molecular therapies and precision medicine for hepatocellular carcinoma. Nat Rev Clin Oncol. 2018;15:599–616.CrossRef
2.
go back to reference Reig M, Da FL, Faivre S. New trials and results in systemic treatment of HCC. J Hepatol. 2018;69:525–33.CrossRef Reig M, Da FL, Faivre S. New trials and results in systemic treatment of HCC. J Hepatol. 2018;69:525–33.CrossRef
3.
go back to reference Dutta R, Mahato RI. Recent advances in hepatocellular carcinoma therapy. Pharmacol Ther. 2017;173:106–17.CrossRef Dutta R, Mahato RI. Recent advances in hepatocellular carcinoma therapy. Pharmacol Ther. 2017;173:106–17.CrossRef
4.
go back to reference Galle PR, Tovoli F, Foerster F, Worns MA, Cucchetti A, Bolondi L. The treatment of intermediate stage tumours beyond TACE: from surgery to systemic therapy. J Hepatol. 2017;67:173–83.CrossRef Galle PR, Tovoli F, Foerster F, Worns MA, Cucchetti A, Bolondi L. The treatment of intermediate stage tumours beyond TACE: from surgery to systemic therapy. J Hepatol. 2017;67:173–83.CrossRef
5.
go back to reference Huo X, Han S, Wu G, Latchoumanin O, Zhou G, Hebbard L, et al. Dysregulated long noncoding RNAs (lncRNAs) in hepatocellular carcinoma: implications for tumorigenesis, disease progression, and liver cancer stem cells. Mol Cancer. 2017;16:165.CrossRef Huo X, Han S, Wu G, Latchoumanin O, Zhou G, Hebbard L, et al. Dysregulated long noncoding RNAs (lncRNAs) in hepatocellular carcinoma: implications for tumorigenesis, disease progression, and liver cancer stem cells. Mol Cancer. 2017;16:165.CrossRef
6.
go back to reference Liu FF, Liu YN, Chen Z. Tim-3 expression and its role in hepatocellular carcinoma. J Hematol Oncol. 2018;11:126.CrossRef Liu FF, Liu YN, Chen Z. Tim-3 expression and its role in hepatocellular carcinoma. J Hematol Oncol. 2018;11:126.CrossRef
7.
go back to reference Zhou SL, Hu ZQ, Zhou ZJ, Dai Z, Wang Z, Cao Y, et al. miR-28-5p-IL-34-macrophage feedback loop modulates hepatocellular carcinoma metastasis. Hepatology. 2016;63:1560–75.CrossRef Zhou SL, Hu ZQ, Zhou ZJ, Dai Z, Wang Z, Cao Y, et al. miR-28-5p-IL-34-macrophage feedback loop modulates hepatocellular carcinoma metastasis. Hepatology. 2016;63:1560–75.CrossRef
8.
go back to reference Antonioli L, Pacher P, Vizi ES, Hasko G. CD39 and CD73 in immunity and inflammation. Trends Mol Med. 2013;19:355–67.CrossRef Antonioli L, Pacher P, Vizi ES, Hasko G. CD39 and CD73 in immunity and inflammation. Trends Mol Med. 2013;19:355–67.CrossRef
9.
go back to reference Allard D, Allard B, Gaudreau PO, Chrobak P, Stagg J. CD73-adenosine: a next-generation target in immuno-oncology. Immunotherapy. 2016;8:145–63.CrossRef Allard D, Allard B, Gaudreau PO, Chrobak P, Stagg J. CD73-adenosine: a next-generation target in immuno-oncology. Immunotherapy. 2016;8:145–63.CrossRef
10.
go back to reference Allard B, Longhi MS, Robson SC, Stagg J. The ectonucleotidases CD39 and CD73: novel checkpoint inhibitor targets. Immunol Rev. 2017;276:121–44.CrossRef Allard B, Longhi MS, Robson SC, Stagg J. The ectonucleotidases CD39 and CD73: novel checkpoint inhibitor targets. Immunol Rev. 2017;276:121–44.CrossRef
11.
go back to reference Gourdin N, Bossennec M, Rodriguez C, Vigano S, Machon C, Jandus C, et al. Autocrine adenosine regulates tumor polyfunctional CD73(+)CD4(+) effector T cells devoid of immune checkpoints. Cancer Res. 2018;78:3604–18.CrossRef Gourdin N, Bossennec M, Rodriguez C, Vigano S, Machon C, Jandus C, et al. Autocrine adenosine regulates tumor polyfunctional CD73(+)CD4(+) effector T cells devoid of immune checkpoints. Cancer Res. 2018;78:3604–18.CrossRef
12.
go back to reference Li L, Wang L, Li J, Fan Z, Yang L, Zhang Z, et al. Metformin-induced reduction of CD39 and CD73 blocks myeloid-derived suppressor cell activity in patients with ovarian cancer. Cancer Res. 2018;78:1779–91.CrossRef Li L, Wang L, Li J, Fan Z, Yang L, Zhang Z, et al. Metformin-induced reduction of CD39 and CD73 blocks myeloid-derived suppressor cell activity in patients with ovarian cancer. Cancer Res. 2018;78:1779–91.CrossRef
13.
go back to reference Allard B, Pommey S, Smyth MJ, Stagg J. Targeting CD73 enhances the antitumor activity of anti-PD-1 and anti-CTLA-4 mAbs. Clin Cancer Res. 2013;19:5626–35.CrossRef Allard B, Pommey S, Smyth MJ, Stagg J. Targeting CD73 enhances the antitumor activity of anti-PD-1 and anti-CTLA-4 mAbs. Clin Cancer Res. 2013;19:5626–35.CrossRef
14.
go back to reference Young A, Ngiow SF, Barkauskas DS, Sult E, Hay C, Blake SJ, et al. Co-inhibition of CD73 and A2AR adenosine signaling improves anti-tumor immune responses. Cancer Cell. 2016;30:391–403.CrossRef Young A, Ngiow SF, Barkauskas DS, Sult E, Hay C, Blake SJ, et al. Co-inhibition of CD73 and A2AR adenosine signaling improves anti-tumor immune responses. Cancer Cell. 2016;30:391–403.CrossRef
16.
go back to reference Lupia M, Angiolini F, Bertalot G, Freddi S, Sachsenmeier KF, Chisci E, et al. CD73 regulates stemness and epithelial-mesenchymal transition in ovarian cancer-initiating cells. Stem Cell Rep. 2018;10:1412–25.CrossRef Lupia M, Angiolini F, Bertalot G, Freddi S, Sachsenmeier KF, Chisci E, et al. CD73 regulates stemness and epithelial-mesenchymal transition in ovarian cancer-initiating cells. Stem Cell Rep. 2018;10:1412–25.CrossRef
17.
go back to reference Yu J, Wang X, Lu Q, Wang J, Li L, Liao X, et al. Extracellular 5′-nucleotidase (CD73) promotes human breast cancer cells growth through AKT/GSK-3beta/beta-catenin/cyclinD1 signaling pathway. Int J Cancer. 2018;142:959–67.CrossRef Yu J, Wang X, Lu Q, Wang J, Li L, Liao X, et al. Extracellular 5′-nucleotidase (CD73) promotes human breast cancer cells growth through AKT/GSK-3beta/beta-catenin/cyclinD1 signaling pathway. Int J Cancer. 2018;142:959–67.CrossRef
18.
go back to reference Allard B, Turcotte M, Spring K, Pommey S, Royal I, Stagg J. Anti-CD73 therapy impairs tumor angiogenesis. Int J Cancer. 2014;134:1466–73.CrossRef Allard B, Turcotte M, Spring K, Pommey S, Royal I, Stagg J. Anti-CD73 therapy impairs tumor angiogenesis. Int J Cancer. 2014;134:1466–73.CrossRef
19.
go back to reference Beavis PA, Divisekera U, Paget C, Chow MT, John LB, Devaud C, et al. Blockade of A2A receptors potently suppresses the metastasis of CD73+ tumors. Proc Natl Acad Sci U S A. 2013;110:14711–6.CrossRef Beavis PA, Divisekera U, Paget C, Chow MT, John LB, Devaud C, et al. Blockade of A2A receptors potently suppresses the metastasis of CD73+ tumors. Proc Natl Acad Sci U S A. 2013;110:14711–6.CrossRef
20.
go back to reference Buisseret L, Pommey S, Allard B, Garaud S, Bergeron M, Cousineau I, et al. Clinical significance of CD73 in triple-negative breast cancer: multiplex analysis of a phase III clinical trial. Ann Oncol. 2018;29:1056–62.CrossRef Buisseret L, Pommey S, Allard B, Garaud S, Bergeron M, Cousineau I, et al. Clinical significance of CD73 in triple-negative breast cancer: multiplex analysis of a phase III clinical trial. Ann Oncol. 2018;29:1056–62.CrossRef
21.
go back to reference Turcotte M, Spring K, Pommey S, Chouinard G, Cousineau I, George J, et al. CD73 is associated with poor prognosis in high-grade serous ovarian cancer. Cancer Res. 2015;75:4494–503.CrossRef Turcotte M, Spring K, Pommey S, Chouinard G, Cousineau I, George J, et al. CD73 is associated with poor prognosis in high-grade serous ovarian cancer. Cancer Res. 2015;75:4494–503.CrossRef
23.
go back to reference Forner A, Reig ME, de Lope CR, Bruix J. Current strategy for staging and treatment: the BCLC update and future prospects. Semin Liver Dis. 2010;30:61–74.CrossRef Forner A, Reig ME, de Lope CR, Bruix J. Current strategy for staging and treatment: the BCLC update and future prospects. Semin Liver Dis. 2010;30:61–74.CrossRef
24.
go back to reference Guo W, Sun YF, Shen MN, Ma XL, Wu J, Zhang CY, et al. Circulating tumor cells with stem-like phenotypes for diagnosis, prognosis, and therapeutic response evaluation in hepatocellular carcinoma. Clin Cancer Res. 2018;24:2203–13.CrossRef Guo W, Sun YF, Shen MN, Ma XL, Wu J, Zhang CY, et al. Circulating tumor cells with stem-like phenotypes for diagnosis, prognosis, and therapeutic response evaluation in hepatocellular carcinoma. Clin Cancer Res. 2018;24:2203–13.CrossRef
25.
go back to reference Cha I, Lee SH, Jeon TJ. Chemoattractant-mediated Rap1 activation requires GPCR/G proteins. Mol Cells. 2010;30:563–7.CrossRef Cha I, Lee SH, Jeon TJ. Chemoattractant-mediated Rap1 activation requires GPCR/G proteins. Mol Cells. 2010;30:563–7.CrossRef
26.
go back to reference Kortholt A, Bolourani P, Rehmann H, Keizer-Gunnink I, Weeks G, Wittinghofer A, et al. A Rap/phosphatidylinositol 3-kinase pathway controls pseudopod formation [corrected]. Mol Biol Cell. 2010;21:936–45.CrossRef Kortholt A, Bolourani P, Rehmann H, Keizer-Gunnink I, Weeks G, Wittinghofer A, et al. A Rap/phosphatidylinositol 3-kinase pathway controls pseudopod formation [corrected]. Mol Biol Cell. 2010;21:936–45.CrossRef
28.
go back to reference Xiong Y, Ye C, Yang N, Li M, Liu H. Ubc9 binds to ADAP and is required for Rap1 membrane recruitment, Rac1 activation, and integrin-mediated T cell adhesion. J Immunol. 2017;199:4142–54.CrossRef Xiong Y, Ye C, Yang N, Li M, Liu H. Ubc9 binds to ADAP and is required for Rap1 membrane recruitment, Rac1 activation, and integrin-mediated T cell adhesion. J Immunol. 2017;199:4142–54.CrossRef
29.
go back to reference Thorpe LM, Yuzugullu H, Zhao JJ. PI3K in cancer: divergent roles of isoforms, modes of activation and therapeutic targeting. Nat Rev Cancer. 2015;15:7–24.CrossRef Thorpe LM, Yuzugullu H, Zhao JJ. PI3K in cancer: divergent roles of isoforms, modes of activation and therapeutic targeting. Nat Rev Cancer. 2015;15:7–24.CrossRef
30.
go back to reference Zhu J, Zeng Y, Li W, Qin H, Lei Z, Shen D, et al. CD73/NT5E is a target of miR-30a-5p and plays an important role in the pathogenesis of non-small cell lung cancer. Mol Cancer. 2017;16:34.CrossRef Zhu J, Zeng Y, Li W, Qin H, Lei Z, Shen D, et al. CD73/NT5E is a target of miR-30a-5p and plays an important role in the pathogenesis of non-small cell lung cancer. Mol Cancer. 2017;16:34.CrossRef
31.
go back to reference Xie M, Qin H, Luo Q, Huang Q, He X, Yang Z, et al. MicroRNA-30a regulates cell proliferation and tumor growth of colorectal cancer by targeting CD73. BMC Cancer. 2017;17:305.CrossRef Xie M, Qin H, Luo Q, Huang Q, He X, Yang Z, et al. MicroRNA-30a regulates cell proliferation and tumor growth of colorectal cancer by targeting CD73. BMC Cancer. 2017;17:305.CrossRef
32.
go back to reference Li JH, Liu S, Zhou H, Qu LH, Yang JH. starBase v2.0: decoding miRNA-ceRNA, miRNA-ncRNA and protein-RNA interaction networks from large-scale CLIP-Seq data. Nucleic Acids Res. 2014;42:D92–7.CrossRef Li JH, Liu S, Zhou H, Qu LH, Yang JH. starBase v2.0: decoding miRNA-ceRNA, miRNA-ncRNA and protein-RNA interaction networks from large-scale CLIP-Seq data. Nucleic Acids Res. 2014;42:D92–7.CrossRef
33.
go back to reference Li S, Yao J, Xie M, LiuY ZM. Exosomal miRNAs in hepatocellular carcinoma development and clinical responses. J Hematol Oncol. 2018;11:54.CrossRef Li S, Yao J, Xie M, LiuY ZM. Exosomal miRNAs in hepatocellular carcinoma development and clinical responses. J Hematol Oncol. 2018;11:54.CrossRef
34.
go back to reference Wei R, Huang GL, Zhang MY, Li BK, Zhang HZ, Shi M, et al. Clinical significance and prognostic value of microRNA expression signature in hepatocellular carcinoma. Clin Cancer Res. 2013;19:4780–91.CrossRef Wei R, Huang GL, Zhang MY, Li BK, Zhang HZ, Shi M, et al. Clinical significance and prognostic value of microRNA expression signature in hepatocellular carcinoma. Clin Cancer Res. 2013;19:4780–91.CrossRef
35.
go back to reference Kordass T, Osen W, Eichmuller SB. Controlling the immune suppressor: transcription factors and microRNAs regulating CD73/NT5E. Front Immunol. 2018;9:813.CrossRef Kordass T, Osen W, Eichmuller SB. Controlling the immune suppressor: transcription factors and microRNAs regulating CD73/NT5E. Front Immunol. 2018;9:813.CrossRef
36.
go back to reference Jayachandran A, Dhungel B, Steel JC. Epithelial-to-mesenchymal plasticity of cancer stem cells: therapeutic targets in hepatocellular carcinoma. J Hematol Oncol. 2016;9:74.CrossRef Jayachandran A, Dhungel B, Steel JC. Epithelial-to-mesenchymal plasticity of cancer stem cells: therapeutic targets in hepatocellular carcinoma. J Hematol Oncol. 2016;9:74.CrossRef
37.
go back to reference Breitbach M, Kimura K, Luis TC, Fuegemann CJ, Woll PS, Hesse M, et al. In vivo labeling by CD73 marks multipotent stromal cells and highlights endothelial heterogeneity in the bone marrow niche. Cell Stem Cell. 2018;22:262–76.CrossRef Breitbach M, Kimura K, Luis TC, Fuegemann CJ, Woll PS, Hesse M, et al. In vivo labeling by CD73 marks multipotent stromal cells and highlights endothelial heterogeneity in the bone marrow niche. Cell Stem Cell. 2018;22:262–76.CrossRef
38.
go back to reference Suto EG, Mabuchi Y, Suzuki N, Suzuki K, Ogata Y, Taguchi M, et al. Prospectively isolated mesenchymal stem/stromal cells are enriched in the CD73(+) population and exhibit efficacy after transplantation. Sci Rep. 2017;7:4838.CrossRef Suto EG, Mabuchi Y, Suzuki N, Suzuki K, Ogata Y, Taguchi M, et al. Prospectively isolated mesenchymal stem/stromal cells are enriched in the CD73(+) population and exhibit efficacy after transplantation. Sci Rep. 2017;7:4838.CrossRef
39.
go back to reference Zhou Q, Lui VW, Yeo W. Targeting the PI3K/Akt/mTOR pathway in hepatocellular carcinoma. Future Oncol. 2011;7:1149–67.CrossRef Zhou Q, Lui VW, Yeo W. Targeting the PI3K/Akt/mTOR pathway in hepatocellular carcinoma. Future Oncol. 2011;7:1149–67.CrossRef
40.
go back to reference Cao MQ, You AB, Zhu XD, Zhang W, Zhang YY, Zhang SZ, et al. miR-182-5p promotes hepatocellular carcinoma progression by repressing FOXO3a. J Hematol Oncol. 2018;11:12.CrossRef Cao MQ, You AB, Zhu XD, Zhang W, Zhang YY, Zhang SZ, et al. miR-182-5p promotes hepatocellular carcinoma progression by repressing FOXO3a. J Hematol Oncol. 2018;11:12.CrossRef
41.
go back to reference Frische EW, Zwartkruis FJ. Rap1, a mercenary among the Ras-like GTPases. Dev Biol. 2010;340:1–9.CrossRef Frische EW, Zwartkruis FJ. Rap1, a mercenary among the Ras-like GTPases. Dev Biol. 2010;340:1–9.CrossRef
42.
go back to reference Onodera Y, Nam JM, Bissell MJ. Increased sugar uptake promotes oncogenesis via EPAC/RAP1 and O-GlcNAc pathway. J Clin Invest. 2014;124:367–84.CrossRef Onodera Y, Nam JM, Bissell MJ. Increased sugar uptake promotes oncogenesis via EPAC/RAP1 and O-GlcNAc pathway. J Clin Invest. 2014;124:367–84.CrossRef
Metadata
Title
CD73 promotes hepatocellular carcinoma progression and metastasis via activating PI3K/AKT signaling by inducing Rap1-mediated membrane localization of P110β and predicts poor prognosis
Authors
Xiao-Lu Ma
Min-Na Shen
Bo Hu
Bei-Li Wang
Wen-Jing Yang
Li-Hua Lv
Hao Wang
Yan Zhou
An-Li Jin
Yun-Fan Sun
Chuan-Yan Zhang
Shuang-Jian Qiu
Bai-Shen Pan
Jian Zhou
Jia Fan
Xin-Rong Yang
Wei Guo
Publication date
01-12-2019
Publisher
BioMed Central
Published in
Journal of Hematology & Oncology / Issue 1/2019
Electronic ISSN: 1756-8722
DOI
https://doi.org/10.1186/s13045-019-0724-7

Other articles of this Issue 1/2019

Journal of Hematology & Oncology 1/2019 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine