Cancer Res Treat.  2020 Jan;52(1):10-23. 10.4143/crt.2019.145.

Cathepsin C Interacts with TNF-α/p38 MAPK Signaling Pathway to Promote Proliferation and Metastasis in Hepatocellular Carcinoma

Affiliations
  • 1Department of Liver Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China

Abstract

Purpose
Although cathepsin C (CTSC) has been reported to maintain malignant biological properties in various cancers, its functions in hepatocellular carcinoma (HCC) remain obscure. We aimed to investigate the potential role of CTSC in HCC.
Materials and Methods
HCC tissue microarrays (n=122) were employed to analyze the correlation between CTSC expression and clinicopathological characteristics through immunohistochemistry staining. Quantitative real-time polymerase chain reaction, western blot assay, Cell Counting Kit-8 assay, colony formation, cell migration, and invasion assays, xenograft mice model were adopted to validate what had been indicated by the bioinformatic web tools.
Results
By bioinformatic tools and tissue microarrays, CTSC was found upregulated in HCC compared with normal liver tissues, and its higher expression was correlated with poor prognosis of HCC patients (hazard ratio, 2.402; 95% confidence interval, 1.493 to 3.865; p < 0.001). By gain/loss-of-function assays, we implicated that CTSC functioned as an oncogene to promote the proliferation and metastasis of HCC cells. Mechanistically, we revealed that CTSC was involved in several cancer-related signaling pathways by Gene Set Enrichment Analysis, among which tumor necrosis factor α (TNF-α)/p38 pathway was verified to be activated by CTSC. Furthermore, we found that TNF-α could activate CTSC expression in a concentration- dependent manner. Ralimetinib, an oral p38 mitogen-activated protein kinase (MAPK) inhibitor could inhibit CTSC expression. These indicated a potential positive feedback loop between CTSC and TNF-α/MAPK (p38) signaling.
Conclusion
Taken together, CTSC plays an important role in the growth and metastasis of HCC and may be a promising therapeutic target upon HCC.

Keyword

Cathepsin C; Hepatocellular carcinoma; Cell proliferation; Metastasis; TNF-α/MAPK (p38) pathway; Ralimetinib

Figure

  • Fig. 1. Cathepsin C (CTSC) is frequently upregulated in hepatocellular carcinoma (HCC) and correlated with poor prognosis of HCC patients. (A) The mRNA level of CTSC in HCC specimens analyzed by GEPIA web tool. (B) Western blot assay detected CTSC expression in both HCC and normal tissues. GAPDH, glyceraldehyde 3-phosphate dehydrogenase. (C) The correlation between CTSC expression and overall survival (OS) analyzed by GEPIA. (D) The differential expression in HCC specimens upon tissue microarrays. T, tumor; N, adjacent normal liver. (E) Kaplan-Meier survival analysis of OS in 122 HCC patients upon tissue microarrays.

  • Fig. 2. The expression of cathepsin C (CTSC) in hepatocellular carcinoma (HCC) cell lines. (A) The mRNA level in HCC cell lines analyzed by Cancer Cell Line Encyclopedia. (B) Western blot assay verified CTSC expression in HCC cell lines. GAPDH, glyceraldehyde 3-phosphate dehydrogenase. (C) The confirmation of CTSC overexpression in SH-HEP-1 and SMMC-7721 cells by quantitative real-time polymerase chain reaction. (D) Knockdown of CTSC in SK-HEP-1 and PLC/PRF/5 cells which was validated by western blot assay. ***p < 0.001.

  • Fig. 3. Cathepsin C (C TSC) promotes proliferation, migration, an d in vasion of hepatocellular carcinoma (HCC) cell lines. (A) The proliferation of SH-HEP-1 and SMMC-7721 cells infected with lentiviral LV-C TSC vector, or SK-HEP-1 and PLC/PRF/5 cells transfected with CTSC siRNA were assessed by Cell Counting Kit-8 assay. (B) The effects of CTSC on viability was assessed by colony formation assays. (C) Effects of overexpressing CTSC on tu morigenesis in vivo. (D) Trans-well chamber assays indicated the migratory ability of HCC cell lines. (E) Boyden chamber assays indicated thein vasiveness of HCC cell lines. *p < 0.05, **p < 0.01, ***p < 0.001.

  • Fig. 4. Tumor necrosis factor α (TNF-α)/mitogen-activated protein kinase (MAPK, p38) pathway is involved in cathepsin C (CTSC) mediated hepatocellular carcinoma progression. (A) The signaling pathway acquired by Gene Set Enrichment Analysis. (B) The activation of TNF-α/MAPK (p38) signaling pathway after overexpressing CTSC in SK-HEP-1 and SMMC-7721 cells, demonstrated by western blot assay. (C) The expression of matrix metalloproteinase (MMP) 3, MMP-9, vimentin after CTSC overexpression in SK-HEP-1 and SMMC-7721 cells detected by western blot assay. (D) The expression of TNF-α after overexpressing CTSC in SK-HEP-1 and SMMC-7721 cells detected by quantitative real-time polymerase chain reaction. (E) The correlation between CTSC and MMP-3, MMP-9, Vimentin, and TNF-α in clinical specimens analyzed by the GEPIA. (F) The western blot assay was used to investigate the effect of TNF-α on CTSC expression. (G, H) The western blot assay was used to investigate the effect of Ralimetinib on CTSC expression. PI3K, phosphoinositide 3-kinase; NF-κB, nuclear factor κB; TNF-α, tumor necrosis factor α; HIF-1, hypoxia-inducible factor 1; VEGF, vascular endothelial growth factor; GAPDH, glyceraldehyde 3-phosphate dehydrogenase. **p < 0.01, ***p < 0.001.


Reference

References

1. Bertuccio P, Turati F, Carioli G, Rodriguez T, La Vecchia C, Malvezzi M, et al. Global trends and predictions in hepatocellular carcinoma mortality. J Hepatol. 2017; 67:302–9.
Article
2. McGlynn KA, Petrick JL, London WT. Global epidemiology of hepatocellular carcinoma: an emphasis on demographic and regional variability. Clin Liver Dis. 2015; 19:223–38.
3. Ozakyol A. Global epidemiology of hepatocellular carcinoma (HCC epidemiology). J Gastrointest Cancer. 2017; 48:238–40.
Article
4. Wallace MC, Preen D, Jeffrey GP, Adams LA. The evolving epidemiology of hepatocellular carcinoma: a global perspective. Expert Rev Gastroenterol Hepatol. 2015; 9:765–79.
Article
5. Yagi R, Midorikawa Y, Moriguchi M, Nakayama H, Aramaki O, Yamazaki S, et al. Liver resection for recurrent hepatocellular carcinoma to improve survivability: a proposal of indication criteria. Surgery. 2018; 163:1250–6.
Article
6. Zhu J, Yin T, Xu Y, Lu XJ. Therapeutics for advanced hepatocellular carcinoma: Recent advances, current dilemma, and future directions. J Cell Physiol. 2019; 234:12122–32.
Article
7. Olson OC, Joyce JA. Cysteine cathepsin proteases: regulators of cancer progression and therapeutic response. Nat Rev Cancer. 2015; 15:712–29.
Article
8. Ishidoh K, Kominami E. Processing and activation of lysosomal proteinases. Biol Chem. 2002; 383:1827–31.
Article
9. Ruan J, Zheng H, Rong X, Rong X, Zhang J, Fang W, et al. Over-expression of cathepsin B in hepatocellular carcinomas predicts poor prognosis of HCC patients. Mol Cancer. 2016; 15:17.
Article
10. Lee TK, Cheung VC, Lu P, Lau EY, Ma S, Tang KH, et al. Blockade of CD47-mediated cathepsin S/protease-activated receptor 2 signaling provides a therapeutic target for hepatocellular carcinoma. Hepatology. 2014; 60:179–91.
Article
11. Ruffell B, Affara NI, Cottone L, Junankar S, Johansson M, DeNardo DG, et al. Cathepsin C is a tissue-specific regulator of squamous carcinogenesis. Genes Dev. 2013; 27:2086–98.
Article
12. Joyce JA, Hanahan D. Multiple roles for cysteine cathepsins in cancer. Cell Cycle. 2004; 3:1516–619.
Article
13. Gocheva V, Zeng W, Ke D, Klimstra D, Reinheckel T, Peters C, et al. Distinct roles for cysteine cathepsin genes in multistage tumorigenesis. Genes Dev. 2006; 20:543–56.
Article
14. Khaket TP, Singh MP, Khan I, Bhardwaj M, Kang SC. Targeting of cathepsin C induces autophagic dysregulation that directs ER stress mediated cellular cytotoxicity in colorectal cancer cells. Cell Signal. 2018; 46:92–102.
Article
15. Mikhaylov G, Mikac U, Magaeva AA, Itin VI, Naiden EP, Psakhye I, et al. Ferri-liposomes as an MRI-visible drug-delivery system for targeting tumours and their microenvironment. Nat Nanotechnol. 2011; 6:594–602.
Article
16. Xia P, Zhang R, Ge G. C/EBPbeta mediates TNF-alphainduced cancer cell migration by inducing MMP expression dependent on p38 MAPK. J Cell Biochem. 2015; 116:2766–77.
17. Park EJ, Lee JH, Yu GY, He G, Ali SR, Holzer RG, et al. Dietary and genetic obesity promote liver inflammation and tumorigenesis by enhancing IL-6 and TNF expression. Cell. 2010; 140:197–208.
Article
18. Jing Y, Sun K, Liu W, Sheng D, Zhao S, Gao L, et al. Tumor necrosis factor-alpha promotes hepatocellular carcinogenesis through the activation of hepatic progenitor cells. Cancer Lett. 2018; 434:22–32.
19. Xu ZW, Yan SX, Wu HX, Chen JY, Zhang Y, Li Y, et al. The influence of TNF-alpha and Ang II on the proliferation, migration and invasion of HepG2 cells by regulating the expression of GRK2. Cancer Chemother Pharmacol. 2017; 79:747–58.
20. Vasiljeva O, Papazoglou A, Kruger A, Brodoefel H, Korovin M, Deussing J, et al. Tumor cell-derived and macrophagederived cathepsin B promotes progression and lung metastasis of mammary cancer. Cancer Res. 2006; 66:5242–50.
Article
21. Alapati K, Gopinath S, Malla RR, Dasari VR, Rao JS. uPAR and cathepsin B knockdown inhibits radiation-induced PKC integrated integrin signaling to the cytoskeleton of glioma-initiating cells. Int J Oncol. 2012; 41:599–610.
Article
22. Li A, Shi D, Xu B, Wang J, Tang YL, Xiao W, et al. S100A6 promotes cell proliferation in human nasopharyngeal carcinoma via the p38/MAPK signaling pathway. Mol Carcinog. 2017; 56:972–84.
Article
23. Urosevic J, Garcia-Albeniz X, Planet E, Real S, Cespedes MV, Guiu M, et al. Colon cancer cells colonize the lung from established liver metastases through p38 MAPK signalling and PTHLH. Nat Cell Biol. 2014; 16:685–94.
24. Tang YM, Cao QY, Guo XY, Dong SH, Duan JA, Wu QN, et al. Inhibition of p38 and ERK1/2 pathways by Sparstolonin B suppresses inflammation-induced melanoma metastasis. Biomed Pharmacother. 2018; 98:382–9.
Article
25. Bibikova E, Youn MY, Danilova N, Ono-Uruga Y, KontoGhiorghi Y, Ochoa R, et al. TNF-mediated inflammation represses GATA1 and activates p38 MAP kinase in RPS19-deficient hematopoietic progenitors. Blood. 2014; 124:3791–8.
Article
26. Liu Q, Zhang Y, Liu S, Liu Y, Yang X, Liu G, et al. Cathepsin C promotes microglia M1 polarization and aggravates neuroinflammation via activation of Ca(2+)-dependent PKC/p38MAPK/NF-kappaB pathway. J Neuroinflammation. 2019; 16:10.
Article
27. Jin X, Mo Q, Zhang Y, Gao Y, Wu Y, Li J, et al. The p38 MAPK inhibitor BIRB796 enhances the antitumor effects of VX680 in cervical cancer. Cancer Biol Ther. 2016; 17:566–76.
Article
28. Guo X, Ma N, Wang J, Song J, Bu X, Cheng Y, et al. Increased p38-MAPK is responsible for chemotherapy resistance in human gastric cancer cells. BMC Cancer. 2008; 8:375.
Article
29. Lim SJ, Lee YJ, Lee E. p38MAPK inhibitor SB203580 sensitizes human SNU-C4 colon cancer cells to exisulind-induced apoptosis. Oncol Rep. 2006; 16:1131–5.
Article
30. Patnaik A, Haluska P, Tolcher AW, Erlichman C, Papadopoulos KP, Lensing JL, et al. A first-in-human phase I study of the oral p38 MAPK inhibitor, ralimetinib (LY2228820 Dimesylate), in patients with advanced cancer. Clin Cancer Res. 2016; 22:1095–102.
Article
Full Text Links
  • CRT
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr