Abstract

Background: With recorded under-performance of current standard therapeutic strategies as highlighted by high rates of post-treatment (resection or local ablation) recurrence, resistance to chemotherapy, poor overall survival, and an increasing global incidence, hepatocellular carcinoma (HCC) constitutes a medical challenge. Accumulating evidence implicates the presence of HCC stem cells (HCC-SCs) in HCC development, drug-resistance, recurrence, and progression. Therefore, treatment strategies targeting both HCC-SCs and non-CSCs are essential. Methods: Recently, there has been an increasing suggestion of MALAT1 oncogenic activity in HCC; however, its role in HCC stemness remains unexplored. Herein, we investigated the probable role of MALAT1 in the SCs-like phenotype of HCC and explored likely molecular mechanisms by which MALAT1 modulates HCC-SCs-like and metastatic phenotypes. Results: We showed that relative to normal, cirrhotic, or dysplastic liver conditions, MALAT1 was aberrantly expressed in HCC, similar to its overexpression in Huh7, Mahlavu, and SK-Hep1 HCC cells lines, compared to the normal liver cell line THLE-2. We also demonstrated a positive correlation between MALAT1 expression and poor cell differentiation status in HCC using RNAscope. Interestingly, we demonstrated that shRNA-mediated silencing of MALAT1 concomitantly downregulated the expression levels of β-catenin, Stat3, c-Myc, CK19, vimentin, and Twist1 proteins, inhibited HCC oncogenicity, and significantly suppressed the HCC-SCs-related dye-effluxing potential of HCC cells and reduced their ALDH-1 activity, partially due to inhibited MALAT1-β-catenin interaction. Additionally, using TOP/FOP (TCL/LEF-Firefly luciferase) Flash, RT-PCR, and western blot assays, we showed that silencing MALAT1 downregulates β-catenin expression, dysregulates the canonical Wnt signaling pathway, and consequently attenuates HCC tumorsphere formation efficiency, with concurrent reduction in CD133+ and CD90+ HCC cell population, and inhibits tumor growth in SK-Hep1-bearing mice. Conclusions: Taken together, our data indicate that MALAT1/Wnt is a targetable molecular candidate, and the therapeutic targeting of MALAT1/Wnt may constitute a novel promising anticancer strategy for HCC treatment.

Highlights

  • With recorded under-performance of current standard therapeutic strategies as highlighted by high rates of post-treatment recurrence, resistance to chemotherapy, poor overall survival, and an increasing global incidence, hepatocellular carcinoma (HCC) constitutes a medical challenge

  • We observed that the expression of Metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) was highest in HCC, compared to other pathological liver conditions or normal liver; compared to the normal liver samples, MALAT1 expression in the HCC and liver cell dysplasia samples was up-regulated by 3.23-fold (p = 1.38 × 10−6 ; t-test = 6.28), and 3.17-fold (p = 2.59 × 10−6 ; t-test = 6.28), respectively (Figure 1A)

  • Using different human HCC cell lines, we demonstrated that compared to its expression in the epitheloid well-differentiated Huh7 cell (1.5-fold, p < 0.01), the expression of MALAT1 was profoundly enhanced in the fibroblastoid poorly-differentiated Mahlavu (1.7-fold, p < 0.01) and HepG2 human hepatoblastoma cell lines (2.6-fold, p < 0.001), SK-Hep1 (2.8-fold, p < 0.001) HCC cell lines [14], and markedly low expression in the normal liver THLE-2 cell line (0.2-fold, p < 0.001) (Figure 1B)

Read more

Summary

Introduction

With recorded under-performance of current standard therapeutic strategies as highlighted by high rates of post-treatment (resection or local ablation) recurrence, resistance to chemotherapy, poor overall survival, and an increasing global incidence, hepatocellular carcinoma (HCC) constitutes a medical challenge. As genome-wide analyses continue to reveal an increasingly broad landscape of functionally mutated non-coding transcriptomes, there is accruing evidence of the ‘driver’ role of the expression and/or activity of several lncRNAs in malignant transformation and oncogenic phenotypes [4]. This is consistent with common knowledge that lncRNAs, acting as cues for specific cellular states or bioactivities [5], may help identify cellular dysfunctions or pathologies such as malignancies, predict tumor behavior, provide prognostic insight, or inform anticancer therapeutic strategies [4,5]. It was suggested that increased expression of MALAT1 increased the proportion of pancreatic cancer stem cells (CSCs), decreased their sensitivity to anticancer drugs, enhanced their self-renewal capacity, accelerated their tumor angiogenesis, and promoted tumor growth [9], while the downregulation of MALAT1 promoted the proliferation of the glioma stem cell line SHG139S by suppressing the expression of stemness markers, Nestin and Sox2 [10]; the role of MALAT1 in the induction and/or maintenance of the CSCs-like phenotype in HCC remains unexplored, and there is a dearth of data detailing the role of MALAT1 in the maintenance and proliferation of CSCs in HCC.

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.