Skip to main content
Top
Published in: Journal of Experimental & Clinical Cancer Research 1/2024

Open Access 01-12-2024 | Hepatocellular Carcinoma | Research

DCAF1 interacts with PARD3 to promote hepatocellular carcinoma progression and metastasis by activating the Akt signaling pathway

Authors: Jinyao Zhang, Yuze Shi, Ke Ding, Weiwei Yu, Jianbo He, Beicheng Sun

Published in: Journal of Experimental & Clinical Cancer Research | Issue 1/2024

Login to get access

Abstract

Background

Hepatocellular carcinoma (HCC) is a fatal malignancy with poor prognosis due to lack of effective clinical interference. DCAF1 plays a vital role in regulating cell growth and proliferation, and is involved in the progression of various malignancies. However, the function of DCAF1 in HCC development and the underlying mechanism are still unknown. This study aimed to explore the effect of DCAF1 in HCC and the corresponding molecular mechanism.

Methods

Quantitative real-time PCR, Western blot and immunostaining were used to determine DCAF1 expression in tumor tissues and cell lines. Subsequently, in vitro and in vivo experiments were conducted to explore the function of DCAF1 in tumor growth and metastasis in HCC. Coimmunoprecipitation, mass spectrometry and RNA sequencing were performed to identify the underlying molecular mechanisms.

Results

In this study, we found that DCAF1 was observably upregulated and associated with poor prognosis in HCC. Knockdown of DCAF1 inhibited tumor proliferation and metastasis and promoted tumor apoptosis, whereas overexpressing DCAF1 yielded opposite effects. Mechanistically, DCAF1 could activate the Akt signaling pathway by binding to PARD3 and enhancing its expression. We also found that the combined application of DCAF1 knockdown and Akt inhibitor could significantly suppress subcutaneous xenograft tumor growth.

Conclusions

Our study illustrates that DCAF1 plays a crucial role in HCC development and the DCAF1/PARD3/Akt axis presents a potentially effective therapeutic strategy for HCC.

Graphical Abstract

Appendix
Available only for authorised users
Literature
1.
go back to reference Siegel RL, Miller KD, Wagle NS, Jemal A. Cancer statistics, 2023. CA Cancer J Clin. 2023;73(1):17–48.CrossRefPubMed Siegel RL, Miller KD, Wagle NS, Jemal A. Cancer statistics, 2023. CA Cancer J Clin. 2023;73(1):17–48.CrossRefPubMed
2.
go back to reference Llovet JM, Kelley RK, Villanueva A, Singal AG, Pikarsky E, Roayaie S, et al. Hepatocellular carcinoma. Nat Rev Dis Primers. 2021;7(1):6.CrossRefPubMed Llovet JM, Kelley RK, Villanueva A, Singal AG, Pikarsky E, Roayaie S, et al. Hepatocellular carcinoma. Nat Rev Dis Primers. 2021;7(1):6.CrossRefPubMed
3.
go back to reference Le Rouzic E, Belaïdouni N, Estrabaud E, Morel M, Rain JC, Transy C, et al. HIV1 Vpr arrests the cell cycle by recruiting DCAF1/VprBP, a receptor of the Cul4-DDB1 ubiquitin ligase. Cell Cycle. 2007;6(2):182–8.CrossRefPubMed Le Rouzic E, Belaïdouni N, Estrabaud E, Morel M, Rain JC, Transy C, et al. HIV1 Vpr arrests the cell cycle by recruiting DCAF1/VprBP, a receptor of the Cul4-DDB1 ubiquitin ligase. Cell Cycle. 2007;6(2):182–8.CrossRefPubMed
4.
go back to reference Zhou Z, Song X, Wavelet CM, Wan Y. Cullin 4-DCAF Proteins in Tumorigenesis. Adv Exp Med Biol. 2020;1217:241–59.CrossRefPubMed Zhou Z, Song X, Wavelet CM, Wan Y. Cullin 4-DCAF Proteins in Tumorigenesis. Adv Exp Med Biol. 2020;1217:241–59.CrossRefPubMed
5.
go back to reference Han XR, Sasaki N, Jackson SC, Wang P, Li Z, Smith MD, et al. CRL4(DCAF1/VprBP) E3 ubiquitin ligase controls ribosome biogenesis, cell proliferation, and development. Sci Adv. 2020;6(51):eabd6078. Han XR, Sasaki N, Jackson SC, Wang P, Li Z, Smith MD, et al. CRL4(DCAF1/VprBP) E3 ubiquitin ligase controls ribosome biogenesis, cell proliferation, and development. Sci Adv. 2020;6(51):eabd6078.
6.
go back to reference Hrecka K, Hao C, Shun MC, Kaur S, Swanson SK, Florens L, et al. HIV-1 and HIV-2 exhibit divergent interactions with HLTF and UNG2 DNA repair proteins. Proc Natl Acad Sci U S A. 2016;113(27):E3921–30.CrossRefPubMedPubMedCentral Hrecka K, Hao C, Shun MC, Kaur S, Swanson SK, Florens L, et al. HIV-1 and HIV-2 exhibit divergent interactions with HLTF and UNG2 DNA repair proteins. Proc Natl Acad Sci U S A. 2016;113(27):E3921–30.CrossRefPubMedPubMedCentral
7.
go back to reference Yurkovetskiy L, Guney MH, Kim K, Goh SL, McCauley S, Dauphin A, et al. Primate immunodeficiency virus proteins Vpx and Vpr counteract transcriptional repression of proviruses by the HUSH complex. Nat Microbiol. 2018;3(12):1354–61.CrossRefPubMedPubMedCentral Yurkovetskiy L, Guney MH, Kim K, Goh SL, McCauley S, Dauphin A, et al. Primate immunodeficiency virus proteins Vpx and Vpr counteract transcriptional repression of proviruses by the HUSH complex. Nat Microbiol. 2018;3(12):1354–61.CrossRefPubMedPubMedCentral
8.
go back to reference Liu J, Yuan B, Cao J, Luo H, Gu S, Zhang M, et al. AMBRA1 Promotes TGFβ Signaling via Nonproteolytic Polyubiquitylation of Smad4. Cancer Res. 2021;81(19):5007–20.CrossRefPubMed Liu J, Yuan B, Cao J, Luo H, Gu S, Zhang M, et al. AMBRA1 Promotes TGFβ Signaling via Nonproteolytic Polyubiquitylation of Smad4. Cancer Res. 2021;81(19):5007–20.CrossRefPubMed
9.
go back to reference Tang J, Wu Z, Wang X, Hou Y, Bai Y, Tian Y. Hypoxia-Regulated lncRNA USP2-AS1 Drives Head and Neck Squamous Cell Carcinoma Progression. Cells. 2022;11(21):3407. Tang J, Wu Z, Wang X, Hou Y, Bai Y, Tian Y. Hypoxia-Regulated lncRNA USP2-AS1 Drives Head and Neck Squamous Cell Carcinoma Progression. Cells. 2022;11(21):3407.
10.
go back to reference Wang X, Arceci A, Bird K, Mills CA, Choudhury R, Kernan JL, et al. VprBP/DCAF1 Regulates the Degradation and Nonproteolytic Activation of the Cell Cycle Transcription Factor FoxM1. Mol Cell Biol. 2017;37(13):e00609-16. Wang X, Arceci A, Bird K, Mills CA, Choudhury R, Kernan JL, et al. VprBP/DCAF1 Regulates the Degradation and Nonproteolytic Activation of the Cell Cycle Transcription Factor FoxM1. Mol Cell Biol. 2017;37(13):e00609-16.
11.
go back to reference Ren W, Shen S, Sun Z, Shu P, Shen X, Bu C, et al. Jak-STAT3 pathway triggers DICER1 for proteasomal degradation by ubiquitin ligase complex of CUL4A(DCAF1) to promote colon cancer development. Cancer Lett. 2016;375(2):209–20.CrossRefPubMed Ren W, Shen S, Sun Z, Shu P, Shen X, Bu C, et al. Jak-STAT3 pathway triggers DICER1 for proteasomal degradation by ubiquitin ligase complex of CUL4A(DCAF1) to promote colon cancer development. Cancer Lett. 2016;375(2):209–20.CrossRefPubMed
12.
go back to reference Poulose N, Forsythe N, Polonski A, Gregg G, Maguire S, Fuchs M, et al. VPRBP functions downstream of the androgen receptor and OGT to Restrict p53 activation in prostate cancer. Mol Cancer Res. 2022;20(7):1047–60.CrossRefPubMedPubMedCentral Poulose N, Forsythe N, Polonski A, Gregg G, Maguire S, Fuchs M, et al. VPRBP functions downstream of the androgen receptor and OGT to Restrict p53 activation in prostate cancer. Mol Cancer Res. 2022;20(7):1047–60.CrossRefPubMedPubMedCentral
13.
14.
15.
go back to reference Wu J, Tan HY, Chan YT, Lu Y, Feng Z, Yuan H, et al. PARD3 drives tumorigenesis through activating Sonic Hedgehog signalling in tumour-initiating cells in liver cancer. J Exp Clin Cancer Res. 2024;43(1):42.CrossRefPubMedPubMedCentral Wu J, Tan HY, Chan YT, Lu Y, Feng Z, Yuan H, et al. PARD3 drives tumorigenesis through activating Sonic Hedgehog signalling in tumour-initiating cells in liver cancer. J Exp Clin Cancer Res. 2024;43(1):42.CrossRefPubMedPubMedCentral
16.
go back to reference Gou H, Wong CC, Chen H, Shang H, Su H, Zhai J, et al. TRIP6 disrupts tight junctions to promote metastasis and drug resistance and is a therapeutic target in colorectal cancer. Cancer Lett. 2023;578: 216438.CrossRefPubMed Gou H, Wong CC, Chen H, Shang H, Su H, Zhai J, et al. TRIP6 disrupts tight junctions to promote metastasis and drug resistance and is a therapeutic target in colorectal cancer. Cancer Lett. 2023;578: 216438.CrossRefPubMed
17.
go back to reference Sun X, Jin Z, Song X, Wang J, Li Y, Qian X, et al. Evaluation of KIF23 variant 1 expression and relevance as a novel prognostic factor in patients with hepatocellular carcinoma. BMC Cancer. 2015;15:961.CrossRefPubMedPubMedCentral Sun X, Jin Z, Song X, Wang J, Li Y, Qian X, et al. Evaluation of KIF23 variant 1 expression and relevance as a novel prognostic factor in patients with hepatocellular carcinoma. BMC Cancer. 2015;15:961.CrossRefPubMedPubMedCentral
18.
go back to reference Mittal V. Epithelial mesenchymal transition in tumor metastasis. Annu Rev Pathol. 2018;13:395–412.CrossRefPubMed Mittal V. Epithelial mesenchymal transition in tumor metastasis. Annu Rev Pathol. 2018;13:395–412.CrossRefPubMed
19.
go back to reference Yang Y, Ren P, Liu X, Sun X, Zhang C, Du X, et al. PPP1R26 drives hepatocellular carcinoma progression by controlling glycolysis and epithelial-mesenchymal transition. J Exp Clin Cancer Res. 2022;41(1):101.CrossRefPubMedPubMedCentral Yang Y, Ren P, Liu X, Sun X, Zhang C, Du X, et al. PPP1R26 drives hepatocellular carcinoma progression by controlling glycolysis and epithelial-mesenchymal transition. J Exp Clin Cancer Res. 2022;41(1):101.CrossRefPubMedPubMedCentral
20.
go back to reference Yang J, Huang Y, Song M, Pan Q, Zhao J, He J, et al. SPC25 promotes proliferation and stemness of hepatocellular carcinoma cells via the DNA-PK/AKT/Notch1 signaling pathway. Int J Biol Sci. 2022;18(14):5241–59.CrossRefPubMedPubMedCentral Yang J, Huang Y, Song M, Pan Q, Zhao J, He J, et al. SPC25 promotes proliferation and stemness of hepatocellular carcinoma cells via the DNA-PK/AKT/Notch1 signaling pathway. Int J Biol Sci. 2022;18(14):5241–59.CrossRefPubMedPubMedCentral
21.
go back to reference Sun F, Wang J, Sun Q, Li F, Gao H, Xu L, et al. Interleukin-8 promotes integrin β3 upregulation and cell invasion through PI3K/Akt pathway in hepatocellular carcinoma. J Exp Clin Cancer Res. 2019;38(1):449.CrossRefPubMedPubMedCentral Sun F, Wang J, Sun Q, Li F, Gao H, Xu L, et al. Interleukin-8 promotes integrin β3 upregulation and cell invasion through PI3K/Akt pathway in hepatocellular carcinoma. J Exp Clin Cancer Res. 2019;38(1):449.CrossRefPubMedPubMedCentral
23.
go back to reference Gao W, Guo H, Niu M, Zheng X, Zhang Y, Xue X, et al. circPARD3 drives malignant progression and chemoresistance of laryngeal squamous cell carcinoma by inhibiting autophagy through the PRKCI-Akt-mTOR pathway. Mol Cancer. 2020;19(1):166.CrossRefPubMedPubMedCentral Gao W, Guo H, Niu M, Zheng X, Zhang Y, Xue X, et al. circPARD3 drives malignant progression and chemoresistance of laryngeal squamous cell carcinoma by inhibiting autophagy through the PRKCI-Akt-mTOR pathway. Mol Cancer. 2020;19(1):166.CrossRefPubMedPubMedCentral
24.
go back to reference Bamodu OA, Chang HL, Ong JR, Lee WH, Yeh CT, Tsai JT. Elevated PDK1 Expression Drives PI3K/AKT/MTOR Signaling Promotes Radiation-Resistant and Dedifferentiated Phenotype of Hepatocellular Carcinoma. Cells. 2020;9(3):746. Bamodu OA, Chang HL, Ong JR, Lee WH, Yeh CT, Tsai JT. Elevated PDK1 Expression Drives PI3K/AKT/MTOR Signaling Promotes Radiation-Resistant and Dedifferentiated Phenotype of Hepatocellular Carcinoma. Cells. 2020;9(3):746.
25.
go back to reference Zhang F, Li K, Pan M, Li W, Wu J, Li M, et al. miR-589 promotes gastric cancer aggressiveness by a LIFR-PI3K/AKT-c-Jun regulatory feedback loop. J Exp Clin Cancer Res. 2018;37(1):152.CrossRefPubMedPubMedCentral Zhang F, Li K, Pan M, Li W, Wu J, Li M, et al. miR-589 promotes gastric cancer aggressiveness by a LIFR-PI3K/AKT-c-Jun regulatory feedback loop. J Exp Clin Cancer Res. 2018;37(1):152.CrossRefPubMedPubMedCentral
26.
go back to reference Nakanishi K, Sakamoto M, Yamasaki S, Todo S, Hirohashi S. Akt phosphorylation is a risk factor for early disease recurrence and poor prognosis in hepatocellular carcinoma. Cancer. 2005;103(2):307–12.CrossRefPubMed Nakanishi K, Sakamoto M, Yamasaki S, Todo S, Hirohashi S. Akt phosphorylation is a risk factor for early disease recurrence and poor prognosis in hepatocellular carcinoma. Cancer. 2005;103(2):307–12.CrossRefPubMed
27.
go back to reference Bang J, Jun M, Lee S, Moon H, Ro SW. Targeting EGFR/PI3K/AKT/mTOR Signaling in Hepatocellular Carcinoma. Pharmaceutics. 2023;15(8):2130. Bang J, Jun M, Lee S, Moon H, Ro SW. Targeting EGFR/PI3K/AKT/mTOR Signaling in Hepatocellular Carcinoma. Pharmaceutics. 2023;15(8):2130.
28.
go back to reference Formisano L, Napolitano F, Rosa R, D’Amato V, Servetto A, Marciano R, et al. Mechanisms of resistance to mTOR inhibitors. Crit Rev Oncol Hematol. 2020;147: 102886.CrossRefPubMed Formisano L, Napolitano F, Rosa R, D’Amato V, Servetto A, Marciano R, et al. Mechanisms of resistance to mTOR inhibitors. Crit Rev Oncol Hematol. 2020;147: 102886.CrossRefPubMed
29.
go back to reference Jin H, Wang L, Bernards R. Rational combinations of targeted cancer therapies: background, advances and challenges. Nat Rev Drug Discov. 2023;22(3):213–34.CrossRefPubMed Jin H, Wang L, Bernards R. Rational combinations of targeted cancer therapies: background, advances and challenges. Nat Rev Drug Discov. 2023;22(3):213–34.CrossRefPubMed
30.
go back to reference Wang Y, Minden A. Current Molecular Combination Therapies Used for the Treatment of Breast Cancer. Int J Mol Sci. 2022;23(19):11046. Wang Y, Minden A. Current Molecular Combination Therapies Used for the Treatment of Breast Cancer. Int J Mol Sci. 2022;23(19):11046.
Metadata
Title
DCAF1 interacts with PARD3 to promote hepatocellular carcinoma progression and metastasis by activating the Akt signaling pathway
Authors
Jinyao Zhang
Yuze Shi
Ke Ding
Weiwei Yu
Jianbo He
Beicheng Sun
Publication date
01-12-2024
Publisher
BioMed Central
Published in
Journal of Experimental & Clinical Cancer Research / Issue 1/2024
Electronic ISSN: 1756-9966
DOI
https://doi.org/10.1186/s13046-024-03055-2

Other articles of this Issue 1/2024

Journal of Experimental & Clinical Cancer Research 1/2024 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