Skip to main content
Erschienen in: Journal of Experimental & Clinical Cancer Research 1/2020

Open Access 01.12.2020 | Research

MicroRNA-148a-3p inhibits progression of hepatocelluar carcimoma by repressing SMAD2 expression in an Ago2 dependent manner

verfasst von: Zhao Huang, Jingyuan Wen, Jingjing Yu, Jingyu Liao, Sha Liu, Ning Cai, Huifang Liang, Xiaoping Chen, Zeyang Ding, Bixiang Zhang

Erschienen in: Journal of Experimental & Clinical Cancer Research | Ausgabe 1/2020

Abstract

Background

Hepatocellular carcinoma (HCC) is one of the most prevalent common cancer worldwide with high mortality. Transforming growth factor-β (TGF-β) signaling pathway was reported dysregulated during liver cancer formation and progression. As a key component of TGF-β signaling, the role of SMAD2 and its regulatory mechanisms in HCC remain unclear.

Methods

SMAD2 expression in paired HCC specimens were determined by western blot and immunohistochemistry (IHC). quantitative real-time PCR (qRT-PCR) was used to measure mRNA and microRNA (miRNA) expression level. Cell migration, invasion and proliferation ability were evaluated by transwell, CCK8 and EdU assay. In silico websites were used to manifest overall survival rates of HCC patients or to predict miRNAs targeting SMAD2. Dual luciferase reporter assay and anti-Ago2 immunoprecipitation assay were performed to confirm the binding between SMAD2 mRNA and miRNA-148a-3p (miR-148a). Tumorigenesis and lung metastasis mouse model were used to explore the role of miR-148a in vivo. In situ hybridization (ISH) was conducted to determine the expression of miR-148a in liver tissues.

Results

In this study, we found that SMAD2 was highly expressed in HCC and elevated SMAD2 expression predicted shorter overall survival (OS) time for HCC patients. SMAD2 promoted mobility and proliferation of HCC cells in vitro. We further revealed that the expression of miR-148a was negatively correlated with SMAD2 and found that miR-148a repressed SMAD2 expression by downregulating its mRNA through binding with Argonaute 2 (Ago2) in HCC. Transwell, CCK8 and animal experiments exhibited miR-148a inhibited metastasis and proliferation of HCC in vitro and in vivo. Moreover, the phenotype changes caused by miR-148a manipulation were recovered by rescuing SMAD2 expression in HCC cells. ISH assay indicated miR-148a was downregulated in HCC and low expression of miR-148a associated with more aggressive clinic features and poor prognosis.

Conclusion

miR-148a was identified as a repressor of HCC progression by downregulating SMAD2 in an Ago2 dependent manner.
Hinweise
Zhao Huang and Jingyuan Wen contributed equally to this work.

Supplementary information

Supplementary information accompanies this paper at https://​doi.​org/​10.​1186/​s13046-020-01649-0.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
3’UTR
3′-Untranslated region
Ago2
Argonaute 2
ACVR1
Activin A receptor type 1
CCK8
Cell counting kit 8
Co-SMAD
Co-mediator SMAD
DNMT1
DNA methyltransferase 1
HBx
Hepatitis B virus X protein
HBV
Hepatitis B virus
HCC
Hepatocellular carcinoma
HCV
Hepatitis C virus
HPIP
Hematopoietic pre–B cell leukemia transcription factor interacting protein
IHC
Immunohistochemistry
ISH
In situ hybridization
I-SMAD
Inhibitory SMAD
LIHC
Liver hepatocellular carcinoma
MFHs
Mouse fetal hepatoblasts,
miRNA
microRNA
miR-148a
miR-148a-3p
miR-27b
miR-27b-3p
NSCLC
Non-small-cell lung carcinoma
OS
Overall survival
PTPRε
Protein tyrosine phosphatase receptor epsilon
qRT-PCR
Quantitative real-time PCR
RIP
Immunoprecipitation
RISC
RNA-induced silencing complex
R-SMAD
Receptor-regulated SMAD
TGF-β
Transforming growth factor-β

Background

Liver cancer is the sixth most common cancer in incidence and the fourth leading cause of cancer-related death worldwide [1, 2]. Hepatocellular carcinoma (HCC) is the major type of liver cancer, accounting for 75–85% of primary liver cancers [3]. The main risks for HCC are hepatitis B virus (HBV), hepatitis C virus (HCV), aflatoxin contamination in food, alcohol abuse, obesity and so on [3]. Treatments for liver cancer patients contain surgical therapies, tumor ablation, transarterial therapies and systemic therapies [2]. However, the prognosis remains miserable, 5-years survival rate for liver cancer patients is 18%, only inferior to pancreatic cancer [1].
TGF-β signaling pathway has been reported playing a multifaceted role in tumor development and progression. It promotes homeostasis and suppresses tumor progression by inducing cytostasis, differentiation, apoptosis, inflammation depression and stroma-derived mitogens in normal and premalignant cells. Meanwhile, in advanced cancer cells, TGF-β initiates immune evasion, growth factor production, differentiation to invasive phenotype, metastatic dissemination and metastatic colonies establishment [4]. Eight SMAD proteins were identified participating in TGF-β signaling: the receptor-regulated SMAD (R-SMAD) (SMAD1, 2, 3, 5 and 8), the co-mediator SMAD (Co-SMAD) (SMAD4), and the inhibitory SMAD (I-SMAD) (SMAD6 and 7) [5]. As R-SMAD, SMAD2 and SMAD3 are phosphorylated by receptors of TGF-β branch, whereas SMAD1, 5 and 8 are phosphorylated by other branches of receptors such as BMP receptors [6]. Upon TGF-β stimulation, SMAD2 and SMAD3 are phosphorylated and activated by type I receptor kinase, and bind with SMAD4 to form activated SMAD complexes [5]. Regardless of the similar working mechanisms and high identity of protein structures [7], the role of SMAD2 and SMAD3 are often different, even opposite. Distinct effects of SMAD2 and SMAD3 were reported in breast cancer bone metastasis [8], pancreatic ductal adenocarcinoma cell proliferation and migration [9], HaCaT keratinocyte cell growth [10], TGF-β autoinduction in clostridium butyricum-activated dendritic cells [11] and TGF-β induced transcription [12]. Our lab has previous reported SMAD3 promoted HCC metastasis by upregulating protein tyrosine phosphatase receptor epsilon (PTPRε) expression and high expression of SMAD3 was associated with poor prognosis in HCC [13], but the impacts of SMAD2 on HCC remain largely unclear.
MicroRNAs (miRNAs) are short endogenous RNAs 19–25 nucleotides in length that post-transcriptionally regulate mRNA expression [14]. Instead of directly silencing targeted mRNA, miRNAs in human and other bilaterian animals promote mRNA decay or repress mRNA translation mainly by recruiting RNA-induced silencing complex (RISC). As the heart of RISC, Argonaute2 (Ago2) recruits TNRC6, PABPC, and deadenylase complexes, i.e., the PAN2-PAN3 complex or CCR4-NOT complex to destabilize or repress mRNA [1517]. Numerous miRNAs have found dysregulated in HCC and involved in HCC diagnosis, prognosis and therapeutics, such as, miR-16, miR-92a and miR-500 [1820]. Lower miR-148a expression in HCC tissues was observed than that in adjacent noncancerous hepatic tissues and miR-148a-3p expression was correlated to clinical TNM stage, metastasis, status of capsular infiltration and numbers of tumor nodes in HCC [21]. Xu et al. reported that hepatitis B virus X protein (HBx) repressed miR-148a expression and further upregulated the expression of hematopoietic pre–B cell leukemia transcription factor interacting protein (HPIP) to enhance tumorigenesis [22]. Gailhouste et al. revealed that miR-148a promoted hepatospecific phenotype of mouse fetal hepatoblasts (MFHs) and suppressed the invasiveness of transformed cells [23].
Our study revealed that SMAD2 exhibited oncogenic role in HCC and found that miR-148a was an upstream regulator of SMAD2 by decaying SMAD2 mRNA in an Ago2 dependent manner. miR-148a inhibited the mobility and proliferation of HCC cells and low expression of miR-148a in HCC was associated with shorter overall survival time. These results indicated miR-148a could be a promising diagnostic marker and therapeutic target for HCC patients.

Methods

Patients and tissue specimens

Human tumor and adjacent non-tumor tissues were collected from HCC patients underwent hepatectomy at the Hepatic Surgery Center, Tongji Hospital of Huazhong University of Science and Technology (HUST) (Wuhan, China). All procedures were approved by the Ethics Committee of Tongji Hospital, HUST and conducted according to the Declaration of Helsinki Principles. Prior written and informed consent was obtained from each patient.

Cell lines and culture

HCC cell lines MHCC-97H, HCC-LM3 were obtained from Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China. Hep3B, Huh7, PLC/PRF-5 (ALEX), HLF, Bel7402 and normal liver cell line HL7702 were purchased from China Center for Type Culture Collection (CCTCC, Wuhan, China). ALEX was purchased from cell bank of Chinese Academy of Sciences (Shanghai, China) [24]. All cell lines were maintained in Dulbecco Modified Eagle Medium (DMEM) (Hyclone, UT, USA) supplemented with 10% fetal bovine serum (FBS) (Gibico) at 37 °C in a 5% CO2 cell incubator.

Cell counting kit 8 assay and EdU incorporation assay

Cell Counting Kit 8 assay (Dojindo, Kumamoto, Japan) was performed according to manufactures’ protocol. Briefly, 1500 indicated cells were seeded into 96-well plates. Culture medium was changed to 100 μl 10% CCK8 solution at the indicated time and incubated in cell incubator for 2 h. Optical density (O.D.) was measure by Universal Microplate Reader ELx 800 (BIO-TEK, USA) at 450 nm wave length. For each group, the absorbance values were measured by five replicates. After extracting blank value, an average of gross O.D. values was used for data analysis.
HCC cells (4000 cells/ well) were seeded into 96-well plates and cultured overnight for EdU incorporation assay by using Cell-Light™ EdU Apollo567 In Vitro Imaging Kit (Ribobio, Guangzhou, China) according to the manufacturer’s instructions. Briefly, 100 μl 50 μM EdU solutions were added into cells and cultured for 2 h. Cells were rinsed with PBS, fixed with 4% paraformaldehyde and incubated with 0.5% TritonX-100. Then, cells were stained with 100 μl 1X Apollo solution for 30 min, nucleus were stained with 1X Hoechst33342 solution. Representative images were captured with EVOS FL auto imaging system (life technologies, USA) and positive cells were counted by Image Pro. Plus version 6.0.

Dual luciferase assay

The sequence of 8284–8390, containing predicted binding site with miR-148a, in the 3′-untranslated region (3’UTR) of SMAD2 or 3′UTR-SMAD2-mutant was cloned into the psiCHECK™-2-vector (Promega, Madison, WI, United States). About 1 × 105 cells/well were seeded into 24-well plates. After 24 h, the recombinant plasmid pSicheck-2-3′UTR-SMAD2 or pSicheck-2-3′UTR-SMAD2-mutant were co-transfected into cells with miR-148a mimic, miR-148a inhibitor or their respective negative control (nc) using Lipofectamine 3000 (Invitrogen). Forty-eight h later, total protein from cells were extracted by Passive Lysis Buffer (Promega) and the luciferase activity was determined using Dual-Luciferase® Reporter 1000 Assay System (Promega) by GloMax 20/20. Firefly luciferase values were normalized against Renilla luciferase activity, and the ratio of firefly/Renilla luciferase activity was presented.

Anti-Ago2 RNA binding protein immuno-precipitation (RIP) assay

Rabbit anti-Ago2 IgG was purchased from Abcam (ab32381). Magna RIP™ RNA-Binding Protein Immunoprecipitation Kit (Millipore, Darmstadt, Germany) was used to enrich Ago2 binding RNA. The enriched RNA was subjected to qRT-PCR. 2-ΔCT was calculated and normalized to the 2-ΔCT of 10% input.

Animal experiments

4–5 weeks old male BALB/cA-nude mice were purchased from Beijing HFK Bioscience Co. Ltd. and maintained at SPF conditions. All animal experiments were approved by the Ethics Committee of Tongji Hospital, HUST. The whole procedure was in accordance with the “Guide for the Care and Use of Laboratory Animals” (NIH publication 86–23 revised 1985). For xenograft tumor model, 1× 106 indicated tumor cells were suspended in 100 μl serum free DMEM and inoculated subcutaneously into the flanks of nude mice. Thirty days later, all mice were sacrificed to compare the volume and weight of tumors. For lung metastasis model, 1 × 106 cells were suspended in 100 μl serum free DMEM. Then cells were injected via the tail vein of nude mice. After 2 months of injection, mice were sacrificed and lungs were resected to calculate metastatic nodules.

In situ hybridization (ISH) and immunohistochemistry analysis (IHC)

ISH was performed using the ISH Kit (Boster, Bio-Engineering Company, Wuhan, China). All procedures followed the manufacturer’s instructions. Samples were stained with hematoxylin, dehydrated with alcohol, washed with xylene, sealed with flavor sealing tablets. Oligo (5’Digoxin-ACAAAGTTCTGTAGTGCACTGA) was used as ISH probe for miR-148a. IHC staining was performed as described previously [24]. Primary antibody for SMAD2 (1:100, 12,570–1-AP) and Ki67 (1:100, 27,309–1-AP) were purchased from Proteintech. The representative images of ISH and IHC were captured and processed using DM2300 microscope and ScopeImage 9.0 software (Nanjing Jiangnan Novel Optics Co., Ltd., China). ISH and IHC staining scores were independently determined by 3 pathologists without prior knowledge of patient information. The overall score defined by multiplying the percentage of positive cells by the staining intensity score as described previously [24].

In silico analysis and prediction websites

Overall survival rates of HCC patients with different SMAD2 or SMAD3 level were analyzed by GEPIA [25] and Kaplan Meier-plotter [26] websites. TargetScan [27], miRTarBase [28] and miRcode [29] websites were used to predicted miRNAs targeting SMAD2 mRNA. StarBase [30] website was used to analyzed correlations between expression of miRNAs and SMAD2 mRNA, or overall survival rate of liver hepatocellular carcinoma (LIHC) patients with different miRNAs expression.

Statistical analyses

We used Prism 7.0 (GraphPad Software, La Jolla, CA, USA) or SPSS 13.0 (SPSS, Chicago, IL, USA) to analyze the data. Quantitative data were analyzed by Student’s t test (unpaired two-tailed comparison) or Pearson’s correlation test. Kaplan-Meier and log-rank analysis were applied to evaluate survival between two groups. Categorical data were analyzed by chi-square test. All values were presented as mean ± SEM. P values less than 0.05 were considered statistically significant.
Supplementary materials and methods were provided in additional information.

Result

SMAD2 and SMAD3 are upregulated in HCC specimens and their high expression predict poor prognosis

We firstly examined the expression of SMAD2 and SMAD3 in 75 pairs of HCC and adjacent non-cancerous tissues by western blot (Fig. 1a and Supplementary Figure 1). The average band intensities of SMAD2 and SMAD3 were significantly higher in HCC tissues than adjacent non-cancerous tissues (Fig. 1b and Supplementary Figure 2A) and their expression levels were correlated positively in HCC samples (Supplementary Figure 2B). We then explored the relevance between SMAD2 or SMAD3 expression and patient prognosis using GEPIA and Kaplan Meier-plotter websites, higher expression of SMAD2 predicted shorter overall survival (OS) in both patient cohort (Fig. 1c and d). While elevated expression of SMAD3 was associated with poorer prognosis only in GEPIA patient cohort (Supplementary Figure 2C and 2D). Given the role of SMAD3 in HCC had been investigated in detail [13], we focused on exploring the impacts of SMAD2 in HCC.

SMAD2 promotes migration, invasion and proliferation of HCC cells

The endogenous SMAD2 expression was found relatively low in Hep3B, ALEX, HLF and Bel7402 cells, meanwhile high in Huh7, MHCC-97H and HCC-LM3 cells (Fig. 2a). We successfully knocked down SMAD2 in MHCC-97H and Huh7 cells by two independent shRNA (MHCC-97H/sh SMAD2, Huh/sh SMAD2), and overexpressed SMAD2 in HLF and Hep3B cells (HLF/SMAD2, Hep3B/SMAD2) (Fig. 2b and Supplementary Figure 3A). Transwell assay indicated that overexpression of SMAD2 promoted and knockdown of SMAD2 inhibited migration and invasion in HCC cells (Fig. 2c and Supplementary Figure 3B). CCK8 and EdU assays showed that HCC cells with higher SMAD2 expression exhibited stronger proliferation ability compared with counterpart lower SMAD2 expression cells (Fig. 2d, e, Supplementary Figure 3C and 3D).

The mRNA of SMAD2 is targeted by miR-148a

To identify the candidate upstream miRNAs of SMAD2, we used in silico websites to predict miRNAs which potentially bind with SMAD2 mRNA. Eleven miRNAs were co-predicted in TargetScan, miRTarBase and miRcode websites (Fig. 3a and Supplementary Table 1). Then, we overexpressed these 11 miRNAs respectively in MHCC-97H and Huh7 cells by miRNA mimics, qRT-PCR revealed that SMAD2 mRNA level was downregulated after transfected with these miRNA mimics (Fig. 3b). Given miRNA mainly exerts its role by binding and decaying targeted mRNA [14, 17], we analyzed the mRNA expression relevance between SMAD2 and these miRNA in HCC samples by StarBase website. The mRNA level of miR-27b-3p (miR-27b) and miR-148a were found in negative correlation with SMAD2 mRNA (Fig. 3c, Supplementary Figure 4). We further analyzed the relevance between these 2 miRNAs and HCC patients’ OS. Patients with higher miR-148a expression exhibited longer OS, while miR-27b showed no significant difference (Fig. 3d). Therefore, we speculated miR-148a was a potential regulator of SMAD2 in HCC.

MiR-148a inhibits the expression of SMAD2 by downregulating its mRNA

We stably overexpressed miR-148a in MHCC-97H and Huh7 cells (MHCC-97H/miR-148a, Huh7/miR-148a) with high endogenous SMAD2 level and knocked down miR-148a in HLF and Hep3B cells (HLF/KD miR-148a, Hep3B/KD miR-148a-) with low endogenous SMAD2 level by lentivirus (Fig. 4a). qRT-PCR and western blot demonstrated that overexpressing miR-148a downregulated and knocking down miR-148a upregulated the SMAD2 mRNA and protein level (Fig. 4b and c). TargetScan predicted that the position 8334–8340 of SMAD2 3′ UTR was potential binding site of miR-148a. We constructed dual luciferase reporter plasmids pSicheck2-SMAD2–3’UTR and pSicheck2-SMAD2–3’UTR-mutant (Fig. 4d). Dual luciferase reporter assay showed that miR-148a mimic inhibited and miR-148a inhibitor enhanced reporter luciferase activity and the changes were abolished after mutating the binding site (Fig. 4e). Anti-Ago2 RNA immunoprecipitation (RIP) assays indicated that Ago2 enriched miR-148a and 3’UTR of SMAD2. The enrichments increased in miR-148a overexpressing MHCC-97H cells and decreased in miR-148a knocking down HLF cells (Fig. 4f).

MiR-148a inhibits migration, invasion and proliferation ability of HCC cells in vitro

To investigated the role of miR-148a in HCC cells, we performed transwell and CCK8 assays after manipulating miR-148a expression. Overexpressing miR-148a inhibited migration and invasion in MHCC-97H and Huh7 cells, knocking down miR-148a promoted migration and invasion in HLF and Hep3B cells (Fig. 5a and Supplementary Figure 5A). CCK8 assay showed that overexpression of miR-148a inhibited and knockdown of miR-148a promoted proliferation in HCC cells (Fig. 5b and Supplementary Figure 5B).

MiR-148a inhibits tumorigenesis and lung metastasis of HCC cells in vivo

HCC cells were injected to nude mice subcutaneously to investigated the role of miR-148a on tumorigenesis. The volume and weight of subcutaneous tumors significantly decreased after overexpressing miR-148a in MHCC-97H cells and increased after knocking down miR-148a in HLF cells (Fig. 5c and Supplementary Figure 5C). IHC staining of SMAD2 and Ki67 indicated that HCC cells with higher miR-148a expression exhibited lower SMAD2 expression and weaker proliferation abilities (Fig. 5d). We then established HCC lung metastasis animal model by injecting cancer cells via tail veins. Less metastatic nodules were observed in post-mertem lungs of mice injected with miR-148a overexpressing MHCC-97H cells compared with control cells, and H&E staining was further performed to verify the metastatic lesion formation. Similar results were obtained in mice injected with miR-148a knocking down HLF groups (Fig. 5e and Supplementary 5D). 

The role of miR-148a in HCC cells was mediated by SMAD2 and low expression of miR-148a associated with more aggressive clinic features

We further overexpressed SMAD2 in MHCC-97H/miR-148a, Huh7/miR-148a cells using pcDNA3.1-SMAD2 and stably knocked down SMAD2 in HLF/KD miR-148a, Hep3B/KD miR-148a cells (Fig. 6a and b). Transwell and CCK8 assay showed that the impaired migration, invasion and proliferation abilities in MHCC-97H/miR-148a, Huh7/miR-148a cells were recovered by overexpressing SMAD2. In contrast, the enhanced migration, invasion and proliferation abilities in HLF/KD miR-148a, Hep3B/KD miR-148a cells were impeded by knockdown of SMAD2 (Fig. 6c-f).
ISH were performed in 77 pairs of HCC samples (Supplementary Table 2) to investigated the role of miR-148a in clinic. The average miR-148a staining score was significantly lower in HCC samples compared with adjacent non-cancerous tissues (Fig. 6g). Chi-squared analysis indicated that lower expression of miR-148a was significantly associated with higher AFP level, HBV infection and higher Child-Pugh score (Supplementary Table 3).

Discussion

As R-SMAD proteins, the impacts of SMAD2 and SMAD3 on cancer initiation and progression were widely investigated. Interestingly, SMAD2 and SMAD3 were found play distinct, even opposite roles under certain context [1012]. In non-small-cell lung carcinoma (NSCLC), cancer metastasis was associated with inactivation of SMAD2-mediated and activation of SMAD3-mediated transcriptional programs [31]. During breast cancer bone metastasis progression, the TGF-β induced bone metastatic genes expression were found depend on SMAD3 but not SMAD2, and knockdown of SMAD3 in MDA-MB-231 cells inhibited bone metastasis, while SMAD3 knockdown led to a more aggressive phenotype [8]. In HCC, SMAD3 was reported suppressing carcinogenesis in chemically inducing animal models [32] and sustained SMAD3 activation promoted cancer metastasis [13]. However, the role of SMAD2 in HCC was poorly focused and remained obscure. Our results showed that the expression of SMAD2 was elevated in HCC specimens and high expression of SMAD2 in HCC associated with poor prognosis. SMAD2 promoted proliferation, migration and invasion of HCC cells.
miRNAs can affect TGF-β signaling process by directly targeting canonical members in the signaling pathway or targeting its effector genes [33]. In human, instead of directly slicing targeting mRNA by associated with an Ago protein that retained the catalytic ability of endonucleolytic cleavage, miRNAs mainly partially paired with targeted mRNA, and recruited RISC complexes to enhance mRNA decay or translational repression [17]. In our study, we predicted the mRNA of SMAD2 can be targeted by serials of miRNAs. Among these miRNAs, we identified miR-148a as an upstream regulator of SMAD2 by analyzing the expression relevance between miRNAs and SMAD2, and the association between miRNAs expression and OS in HCC. miR-148a was found downregulated in HCC tissue and low expression of miR-148a was associated with shorter OS time, which is consistent with previous study [21]. We further found that miR-148a downregulating SMAD2 expression through binding with Ago2, the core protein of RISC complexes.
Endeavors have been paid on exploring the effects of miR-148a on the HCC formation and progression. Researchers revealed that miR-148a promoted mouse fetal hepatoblasts (MFHs) differentiating to mature hepatocytes by directly targeting DNA methyltransferase 1 (DNMT1) [23] and induced hepatocytic differentiation to HCC by inhibiting IKKα/NUMB/NOTCH signaling [34]. While, Xiaojie Xu et al. found that Hepatitis B virus X protein (HBx) repressed miRNA-148a to enhance tumorigenesis in mouse animal model of HCC [22]. The role of miRNA-148a as an HCC metastasis repressor seems consistent across different studies, miR-148a-3p suppressed the invasiveness of HCC cell by regulating c-Met [23], Wnt1 [35] or activin A receptor type 1 (ACVR1) [36]. Our results showed that miR-148a inhibited proliferation and migration of HCC cells in vivo and in vitro. And the impacts of miR-148a in HCC cells were mediated by SMAD2.

Conclusions

This study demonstrated that SMAD2 was highly expressed in HCC specimens, elevated SMAD2 expression was associated with shorter overall survival time for HCC patients. SMAD2 played its tumor promoter role by enhancing migration, invasion and proliferation abilities of HCC cells. Besides, the expression of miR-148a was found negatively related with SMAD2 in HCC. Low expression of miR-148a was associated with more aggressive clinical features and predicted poorer prognosis. miR-148a inhibited HCC cells migration, invasion and proliferation in vitro, suppress tumorigenesis and metastasis in vivo. In mechanism, miR-148a recruited Ago2, bound with and decayed SMAD2 mRNA to inhibit SMAD2 expression. In conclusion, miR-148a was identified as a regulator of oncogenic SMAD2 and may serve as a promising prognostic marker or therapeutic target for HCC patients.

Supplementary information

Supplementary information accompanies this paper at https://​doi.​org/​10.​1186/​s13046-020-01649-0.

Acknowledgements

As the first author of this study, I (Zhao Huang) want to thank my wife and her mother for taking care of our baby when I was engaged in this manuscript and thank my friends for supporting me in my daily expense.
All experiments containing patient specimens or data were approved by the Ethics Committee of Tongji Hospital, HUST and were performed in accordance with the Declaration of Helsinki Principles. Prior written and informed consent was obtained from each patient.
All materials and images are original. No consent needs to declare.

Competing interests

The authors declare no competing interest.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Villanueva A. Hepatocellular carcinoma. N Engl J Med. 2019;380:1450–62.CrossRef Villanueva A. Hepatocellular carcinoma. N Engl J Med. 2019;380:1450–62.CrossRef
2.
Zurück zum Zitat Yang JD, Hainaut P, Gores GJ, Amadou A, Plymoth A, Roberts LR. A global view of hepatocellular carcinoma: trends, risk, prevention and management. Nat Rev Gastroenterol Hepatol. 2019;16:589–604.CrossRef Yang JD, Hainaut P, Gores GJ, Amadou A, Plymoth A, Roberts LR. A global view of hepatocellular carcinoma: trends, risk, prevention and management. Nat Rev Gastroenterol Hepatol. 2019;16:589–604.CrossRef
3.
Zurück zum Zitat Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018;68:394–424.CrossRef Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018;68:394–424.CrossRef
5.
Zurück zum Zitat Shi Y, Massague J. Mechanisms of TGF-beta signaling from cell membrane to the nucleus. Cell. 2003;113:685–700.CrossRef Shi Y, Massague J. Mechanisms of TGF-beta signaling from cell membrane to the nucleus. Cell. 2003;113:685–700.CrossRef
6.
Zurück zum Zitat Massague J. A very private TGF-beta receptor embrace. Mol Cell. 2008;29:149–50.CrossRef Massague J. A very private TGF-beta receptor embrace. Mol Cell. 2008;29:149–50.CrossRef
7.
Zurück zum Zitat Macias MJ, Martin-Malpartida P, Massague J. Structural determinants of Smad function in TGF-beta signaling. Trends Biochem Sci. 2015;40:296–308.CrossRef Macias MJ, Martin-Malpartida P, Massague J. Structural determinants of Smad function in TGF-beta signaling. Trends Biochem Sci. 2015;40:296–308.CrossRef
8.
Zurück zum Zitat Petersen M, Pardali E, van der Horst G, Cheung H, van den Hoogen C, van der Pluijm G, Ten Dijke P. Smad2 and Smad3 have opposing roles in breast cancer bone metastasis by differentially affecting tumor angiogenesis. Oncogene. 2010;29:1351–61.CrossRef Petersen M, Pardali E, van der Horst G, Cheung H, van den Hoogen C, van der Pluijm G, Ten Dijke P. Smad2 and Smad3 have opposing roles in breast cancer bone metastasis by differentially affecting tumor angiogenesis. Oncogene. 2010;29:1351–61.CrossRef
9.
Zurück zum Zitat Ungefroren H, Groth S, Sebens S, Lehnert H, Gieseler F, Fandrich F. Differential roles of Smad2 and Smad3 in the regulation of TGF-beta1-mediated growth inhibition and cell migration in pancreatic ductal adenocarcinoma cells: control by Rac1. Mol Cancer. 2011;10:67.CrossRef Ungefroren H, Groth S, Sebens S, Lehnert H, Gieseler F, Fandrich F. Differential roles of Smad2 and Smad3 in the regulation of TGF-beta1-mediated growth inhibition and cell migration in pancreatic ductal adenocarcinoma cells: control by Rac1. Mol Cancer. 2011;10:67.CrossRef
10.
Zurück zum Zitat Brown KA, Pietenpol JA, Moses HL. A tale of two proteins: differential roles and regulation of Smad2 and Smad3 in TGF-beta signaling. J Cell Biochem. 2007;101:9–33.CrossRef Brown KA, Pietenpol JA, Moses HL. A tale of two proteins: differential roles and regulation of Smad2 and Smad3 in TGF-beta signaling. J Cell Biochem. 2007;101:9–33.CrossRef
11.
Zurück zum Zitat Kashiwagi I, Morita R, Schichita T, Komai K, Saeki K, Matsumoto M, Takeda K, et al. Smad2 and Smad3 inversely regulate TGF-beta autoinduction in Clostridium butyricum-activated dendritic cells. Immunity. 2015;43:65–79.CrossRef Kashiwagi I, Morita R, Schichita T, Komai K, Saeki K, Matsumoto M, Takeda K, et al. Smad2 and Smad3 inversely regulate TGF-beta autoinduction in Clostridium butyricum-activated dendritic cells. Immunity. 2015;43:65–79.CrossRef
12.
Zurück zum Zitat Labbe E, Silvestri C, Hoodless PA, Wrana JL, Attisano L. Smad2 and Smad3 positively and negatively regulate TGF beta-dependent transcription through the forkhead DNA-binding protein FAST2. Mol Cell. 1998;2:109–20.CrossRef Labbe E, Silvestri C, Hoodless PA, Wrana JL, Attisano L. Smad2 and Smad3 positively and negatively regulate TGF beta-dependent transcription through the forkhead DNA-binding protein FAST2. Mol Cell. 1998;2:109–20.CrossRef
13.
Zurück zum Zitat Liao Z, Chen L, Zhang X, Zhang H, Tan X, Dong K, Lu X, et al. PTPRepsilon acts as a metastatic promoter in hepatocellular carcinoma by facilitating recruitment of SMAD3 to TGF-beta receptor 1. Hepatology. 2020. https://doi.org/10.1002/hep.31104. Liao Z, Chen L, Zhang X, Zhang H, Tan X, Dong K, Lu X, et al. PTPRepsilon acts as a metastatic promoter in hepatocellular carcinoma by facilitating recruitment of SMAD3 to TGF-beta receptor 1. Hepatology. 2020. https://​doi.​org/​10.​1002/​hep.​31104.
14.
Zurück zum Zitat Lu TX, Rothenberg ME. MicroRNA. J Allergy Clin Immunol. 2018;141:1202–7.CrossRef Lu TX, Rothenberg ME. MicroRNA. J Allergy Clin Immunol. 2018;141:1202–7.CrossRef
15.
Zurück zum Zitat Chipman LB, Pasquinelli AE. miRNA targeting: growing beyond the seed. Trends Genet. 2019;35:215–22.CrossRef Chipman LB, Pasquinelli AE. miRNA targeting: growing beyond the seed. Trends Genet. 2019;35:215–22.CrossRef
16.
Zurück zum Zitat Elkayam E, Kuhn CD, Tocilj A, Haase AD, Greene EM, Hannon GJ, Joshua-Tor L. The structure of human argonaute-2 in complex with miR-20a. Cell. 2012;150:100–10.CrossRef Elkayam E, Kuhn CD, Tocilj A, Haase AD, Greene EM, Hannon GJ, Joshua-Tor L. The structure of human argonaute-2 in complex with miR-20a. Cell. 2012;150:100–10.CrossRef
18.
Zurück zum Zitat Yang N, Ekanem NR, Sakyi CA, Ray SD. Hepatocellular carcinoma and microRNA: new perspectives on therapeutics and diagnostics. Adv Drug Deliv Rev. 2015;81:62–74.CrossRef Yang N, Ekanem NR, Sakyi CA, Ray SD. Hepatocellular carcinoma and microRNA: new perspectives on therapeutics and diagnostics. Adv Drug Deliv Rev. 2015;81:62–74.CrossRef
19.
Zurück zum Zitat Mott JL. MicroRNAs involved in tumor suppressor and oncogene pathways: implications for hepatobiliary neoplasia. Hepatology. 2009;50:630–7.CrossRef Mott JL. MicroRNAs involved in tumor suppressor and oncogene pathways: implications for hepatobiliary neoplasia. Hepatology. 2009;50:630–7.CrossRef
20.
Zurück zum Zitat Giordano S, Columbano A. MicroRNAs: new tools for diagnosis, prognosis, and therapy in hepatocellular carcinoma? Hepatology. 2013;57:840–7.CrossRef Giordano S, Columbano A. MicroRNAs: new tools for diagnosis, prognosis, and therapy in hepatocellular carcinoma? Hepatology. 2013;57:840–7.CrossRef
21.
Zurück zum Zitat Pan L, Huang S, He R, Rong M, Dang Y, Chen G. Decreased expression and clinical significance of miR-148a in hepatocellular carcinoma tissues. Eur J Med Res. 2014;19:68.CrossRef Pan L, Huang S, He R, Rong M, Dang Y, Chen G. Decreased expression and clinical significance of miR-148a in hepatocellular carcinoma tissues. Eur J Med Res. 2014;19:68.CrossRef
22.
Zurück zum Zitat Xu X, Fan Z, Kang L, Han J, Jiang C, Zheng X, Zhu Z, et al. Hepatitis B virus X protein represses miRNA-148a to enhance tumorigenesis. J Clin Invest. 2013;123:630–45.PubMedPubMedCentral Xu X, Fan Z, Kang L, Han J, Jiang C, Zheng X, Zhu Z, et al. Hepatitis B virus X protein represses miRNA-148a to enhance tumorigenesis. J Clin Invest. 2013;123:630–45.PubMedPubMedCentral
23.
Zurück zum Zitat Gailhouste L, Gomez-Santos L, Hagiwara K, Hatada I, Kitagawa N, Kawaharada K, Thirion M, et al. miR-148a plays a pivotal role in the liver by promoting the hepatospecific phenotype and suppressing the invasiveness of transformed cells. Hepatology. 2013;58:1153–65.CrossRef Gailhouste L, Gomez-Santos L, Hagiwara K, Hatada I, Kitagawa N, Kawaharada K, Thirion M, et al. miR-148a plays a pivotal role in the liver by promoting the hepatospecific phenotype and suppressing the invasiveness of transformed cells. Hepatology. 2013;58:1153–65.CrossRef
24.
Zurück zum Zitat Ding ZY, Jin GN, Wang W, Chen WX, Wu YH, Ai X, Chen L, et al. Reduced expression of transcriptional intermediary factor 1 gamma promotes metastasis and indicates poor prognosis of hepatocellular carcinoma. Hepatology. 2014;60:1620–36.CrossRef Ding ZY, Jin GN, Wang W, Chen WX, Wu YH, Ai X, Chen L, et al. Reduced expression of transcriptional intermediary factor 1 gamma promotes metastasis and indicates poor prognosis of hepatocellular carcinoma. Hepatology. 2014;60:1620–36.CrossRef
25.
Zurück zum Zitat Tang Z, Li C, Kang B, Gao G, Li C, Zhang Z. GEPIA: a web server for cancer and normal gene expression profiling and interactive analyses. Nucleic Acids Res. 2017;45:W98–W102.CrossRef Tang Z, Li C, Kang B, Gao G, Li C, Zhang Z. GEPIA: a web server for cancer and normal gene expression profiling and interactive analyses. Nucleic Acids Res. 2017;45:W98–W102.CrossRef
26.
Zurück zum Zitat Menyhart O, Nagy A, Gyorffy B. Determining consistent prognostic biomarkers of overall survival and vascular invasion in hepatocellular carcinoma. R Soc Open Sci. 2018;5:181006.CrossRef Menyhart O, Nagy A, Gyorffy B. Determining consistent prognostic biomarkers of overall survival and vascular invasion in hepatocellular carcinoma. R Soc Open Sci. 2018;5:181006.CrossRef
27.
Zurück zum Zitat Lewis BP, Burge CB, Bartel DP. Conserved seed pairing, often flanked by adenosines, indicates that thousands of human genes are microRNA targets. Cell. 2005;120:15–20.CrossRef Lewis BP, Burge CB, Bartel DP. Conserved seed pairing, often flanked by adenosines, indicates that thousands of human genes are microRNA targets. Cell. 2005;120:15–20.CrossRef
28.
Zurück zum Zitat Chou CH, Shrestha S, Yang CD, Chang NW, Lin YL, Liao KW, Huang WC, et al. miRTarBase update 2018: a resource for experimentally validated microRNA-target interactions. Nucleic Acids Res. 2018;46:D296–302.CrossRef Chou CH, Shrestha S, Yang CD, Chang NW, Lin YL, Liao KW, Huang WC, et al. miRTarBase update 2018: a resource for experimentally validated microRNA-target interactions. Nucleic Acids Res. 2018;46:D296–302.CrossRef
29.
Zurück zum Zitat Jeggari A, Marks DS, Larsson E. miRcode: a map of putative microRNA target sites in the long non-coding transcriptome. Bioinformatics. 2012;28:2062–3.CrossRef Jeggari A, Marks DS, Larsson E. miRcode: a map of putative microRNA target sites in the long non-coding transcriptome. Bioinformatics. 2012;28:2062–3.CrossRef
30.
Zurück zum Zitat 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
31.
Zurück zum Zitat Ying Z, Tian H, Li Y, Lian R, Li W, Wu S, Zhang HZ, et al. CCT6A suppresses SMAD2 and promotes prometastatic TGF-beta signaling. J Clin Invest. 2017;127:1725–40.CrossRef Ying Z, Tian H, Li Y, Lian R, Li W, Wu S, Zhang HZ, et al. CCT6A suppresses SMAD2 and promotes prometastatic TGF-beta signaling. J Clin Invest. 2017;127:1725–40.CrossRef
32.
Zurück zum Zitat Millet C, Zhang YE. Roles of Smad3 in TGF-beta signaling during carcinogenesis. Crit Rev Eukaryot Gene Expr. 2007;17:281–93.CrossRef Millet C, Zhang YE. Roles of Smad3 in TGF-beta signaling during carcinogenesis. Crit Rev Eukaryot Gene Expr. 2007;17:281–93.CrossRef
33.
Zurück zum Zitat Butz H, Racz K, Hunyady L, Patocs A. Crosstalk between TGF-beta signaling and the microRNA machinery. Trends Pharmacol Sci. 2012;33:382–93.CrossRef Butz H, Racz K, Hunyady L, Patocs A. Crosstalk between TGF-beta signaling and the microRNA machinery. Trends Pharmacol Sci. 2012;33:382–93.CrossRef
34.
Zurück zum Zitat Jung KH, Zhang J, Zhou C, Shen H, Gagea M, Rodriguez-Aguayo C, Lopez-Berestein G, et al. Differentiation therapy for hepatocellular carcinoma: multifaceted effects of miR-148a on tumor growth and phenotype and liver fibrosis. Hepatology. 2016;63:864–79.CrossRef Jung KH, Zhang J, Zhou C, Shen H, Gagea M, Rodriguez-Aguayo C, Lopez-Berestein G, et al. Differentiation therapy for hepatocellular carcinoma: multifaceted effects of miR-148a on tumor growth and phenotype and liver fibrosis. Hepatology. 2016;63:864–79.CrossRef
35.
Zurück zum Zitat Yan H, Dong X, Zhong X, Ye J, Zhou Y, Yang X, Shen J, et al. Inhibitions of epithelial to mesenchymal transition and cancer stem cells-like properties are involved in miR-148a-mediated anti-metastasis of hepatocellular carcinoma. Mol Carcinog. 2014;53:960–9.CrossRef Yan H, Dong X, Zhong X, Ye J, Zhou Y, Yang X, Shen J, et al. Inhibitions of epithelial to mesenchymal transition and cancer stem cells-like properties are involved in miR-148a-mediated anti-metastasis of hepatocellular carcinoma. Mol Carcinog. 2014;53:960–9.CrossRef
36.
Zurück zum Zitat Li L, Liu Y, Guo Y, Liu B, Zhao Y, Li P, Song F, et al. Regulatory MiR-148a-ACVR1/BMP circuit defines a cancer stem cell-like aggressive subtype of hepatocellular carcinoma. Hepatology. 2015;61:574–84.CrossRef Li L, Liu Y, Guo Y, Liu B, Zhao Y, Li P, Song F, et al. Regulatory MiR-148a-ACVR1/BMP circuit defines a cancer stem cell-like aggressive subtype of hepatocellular carcinoma. Hepatology. 2015;61:574–84.CrossRef
Metadaten
Titel
MicroRNA-148a-3p inhibits progression of hepatocelluar carcimoma by repressing SMAD2 expression in an Ago2 dependent manner
verfasst von
Zhao Huang
Jingyuan Wen
Jingjing Yu
Jingyu Liao
Sha Liu
Ning Cai
Huifang Liang
Xiaoping Chen
Zeyang Ding
Bixiang Zhang
Publikationsdatum
01.12.2020
Verlag
BioMed Central
Erschienen in
Journal of Experimental & Clinical Cancer Research / Ausgabe 1/2020
Elektronische ISSN: 1756-9966
DOI
https://doi.org/10.1186/s13046-020-01649-0

Weitere Artikel der Ausgabe 1/2020

Journal of Experimental & Clinical Cancer Research 1/2020 Zur Ausgabe

Umsetzung der POMGAT-Leitlinie läuft

03.05.2024 DCK 2024 Kongressbericht

Seit November 2023 gibt es evidenzbasierte Empfehlungen zum perioperativen Management bei gastrointestinalen Tumoren (POMGAT) auf S3-Niveau. Vieles wird schon entsprechend der Empfehlungen durchgeführt. Wo es im Alltag noch hapert, zeigt eine Umfrage in einem Klinikverbund.

CUP-Syndrom: Künstliche Intelligenz kann Primärtumor finden

30.04.2024 Künstliche Intelligenz Nachrichten

Krebserkrankungen unbekannten Ursprungs (CUP) sind eine diagnostische Herausforderung. KI-Systeme können Pathologen dabei unterstützen, zytologische Bilder zu interpretieren, um den Primärtumor zu lokalisieren.

Sind Frauen die fähigeren Ärzte?

30.04.2024 Gendermedizin Nachrichten

Patienten, die von Ärztinnen behandelt werden, dürfen offenbar auf bessere Therapieergebnisse hoffen als Patienten von Ärzten. Besonders gilt das offenbar für weibliche Kranke, wie eine Studie zeigt.

Adjuvante Immuntherapie verlängert Leben bei RCC

25.04.2024 Nierenkarzinom Nachrichten

Nun gibt es auch Resultate zum Gesamtüberleben: Eine adjuvante Pembrolizumab-Therapie konnte in einer Phase-3-Studie das Leben von Menschen mit Nierenzellkarzinom deutlich verlängern. Die Sterberate war im Vergleich zu Placebo um 38% geringer.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.