Hostname: page-component-848d4c4894-8kt4b Total loading time: 0 Render date: 2024-06-19T06:17:11.979Z Has data issue: false hasContentIssue false

LncRNA MBNL1-AS1 Suppresses Cell Proliferation and Metastasis of Pancreatic Adenocarcinoma through Targeting Carcinogenic miR-301b-3p

Published online by Cambridge University Press:  01 January 2024

Chouman Sulidankazha*
Affiliation:
Department of Pancreatic Surgery, Digestive and Vascular Surgery Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830054, Xinjiang Uygur Autonomous Region, China
Alidake*
Affiliation:
Department of Breast Surgery, Digestive and Vascular Surgery Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830054, Xinjiang Uygur Autonomous Region, China
Hai Lin
Affiliation:
Department of Pancreatic Surgery, Digestive and Vascular Surgery Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830054, Xinjiang Uygur Autonomous Region, China
Tieying He
Affiliation:
Department of Pancreatic Surgery, Digestive and Vascular Surgery Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830054, Xinjiang Uygur Autonomous Region, China
Wei Han
Affiliation:
Department of Pancreatic Surgery, Digestive and Vascular Surgery Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830054, Xinjiang Uygur Autonomous Region, China
Qilong Chen*
Affiliation:
Department of Pancreatic Surgery, Digestive and Vascular Surgery Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830054, Xinjiang Uygur Autonomous Region, China
*
Chouman Sulidankazha and Alidake contributed equally to this work.
Chouman Sulidankazha and Alidake contributed equally to this work.
Correspondence should be addressed to Qilong Chen; chenqilong202204@163.com

Abstract

Pancreatic adenocarcinoma (PAAD) has been a huge challenge to public health due to its increasing incidence, frequent early metastasis, and poor outcome. The molecular basis of tumorigenesis and metastasis in PAAD is largely unclear. Here, we identified a novel tumor-suppressor long noncoding RNA (lncRNA) MBNL1-AS1, in PAAD and revealed its downstream mechanism. Quantitative real-time PCR (qRT-PCR) data showed that MBNL1-AS1 expression was significantly downregulated in PAAD tissues and cells, which was closely associated with metastasis and poor prognosis. Cell counting kit-8 (CCK-8) assay, transwell assay, and western blot verified that overexpression of MBNL1-AS1 suppressed cell proliferation, migration, and epithelial mesenchymal transformation (EMT) behavior in PAAD cells. By using a dual luciferase reporter gene system, we confirmed that miR-301b-3p was a direct target of MBNL1-AS1. Further mechanismic study revealed that upregulation of miR-301b-3p abolished the inhibitory effect of MBNL1-AS1 overexpression on cell proliferation, tumorigenesis, migration and EMT. Our results demonstrate that MBNL1-AS1 plays a tumor-suppressive role in PAAD mainly by downregulating miR-301b-3p, providing a novel therapeutic target for PAAD.

Type
Research Article
Creative Commons
Creative Common License - CCCreative Common License - BY
This is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
Copyright
Copyright © 2023 Chouman Sulidankazha et al.

1. Introduction

Pancreatic adenocarcinoma (PAAD) is a malignant tumor of the digestive tract. Its incidence rate is increasing all over the world, and its prognosis is usually poor [Reference Edderkaoui, Chheda and Soufi1]. Although the treatment for other cancers has made great progress in recent years, the survival rate of PAAD has been stagnant, and the 5-year survival rate remains between 5% and 10% [Reference Tian, Öhlund and Rickelt2]. It is prone to distant metastasis in the early stage, and there are no specific symptoms and signs in the early stage of PAAD, which poses a huge challenge for the improvement of PAAD prognosis. Understanding the molecular mechanism of pancreatic tumorigenesis and metastasis may help to develop new diagnostic and therapeutic methods.

Long noncoding RNAs (lncRNAs) are considered as tumor regulators and have attracted increasing attention in the field of cancer research over the past decades [Reference Gao, Shang and Guo3]. LncRNA is defined as a class of noncoding RNAs (ncRNAs) with a length of more than 200 nucleotides [Reference Wu, Xia and Cao4]. LncRNAs account for a large proportion of the human genome and participate in a set of physiological or pathological process through interacting with DNAs, mRNAs, ncRNAs, and proteins [Reference Li, Han and Hu5]. Numerous studies have showed that lncRNAs are specifically expressed in tumors and regulate multiple tumor biological processes, including sustaining proliferation signaling, evading growth suppressors, enabling replicative immortality, resisting cell death, etc. [Reference Lin, Liao, Liu, Li, Wu and Zeng6Reference Wei, Liang and Li8]. Among the aberrantly expressed lncRNAs, MBNL1-AS1 is downregulated and regarded as a novel tumor-suppressive lncRNA in several common cancers, like colon cancer, breast cancer, and non-small lung cancer [Reference Zhu, Wang, Liu, Li and Yu9Reference Fang, Jiang and Mao11]. MBNL1-AS1 has been proven to repress cell proliferation and migration in these cancers. In addition, MBNL1-AS1 is reported to be upregulated in acute myocardial infarction, and silencing MBNL1-AS1 reduces myocardial injury in animal models [Reference Liu, Lin and Yu12]. From the current research results, we know that MBNL1-AS1 reduces cell survival in both cancer and noncancer cells. However, it is still unclear whether and how MBNL1-AS1 plays tumor-suppressive roles in PAAD.

MicroRNAs (miRNAs) are a class of short ncRNAs consisting of about 22 nucleotides. They usually form incomplete base pairs with the 3′-untranslated region (3′-UTR) of the target gene and play a negative role in regulating gene expression [Reference Fang, Deng, Luo and Zhou13, Reference Sur, Steele, Shi and Ray14]. Specific miRNAs have been reported to regulate of a large amount of cellular biological processes, such as cell proliferation, differentiation, migration, epithelial mesenchymal transformation (EMT), etc. [Reference Wang, Sun and Yu15]. Among them, a lot of functional miRNAs are identified in the cancer progression. For example, miR-106b-5p contributes to the lung metastasis of breast cancer [Reference Wang, Li, Chen, Pan and Shen16]. miR-19a facilitates metastasis and EMT in prostate cancer [Reference Wang, Hu, Qi, Wang and Gao17]. miR-331-3p promotes drug resistance in pancreatic cancer [Reference Zhan, Chen and Tian18]. Recently, miR-301b-3p is reported to function as a tumor promoter in breast cancer, lung cancer, and colorectal cancers [Reference Fan, Li and Zhu19Reference Xiong, Zhang, Tang and Zhu21]. Interestingly, a few evidences have suggested that miR-301b links with gemcitabine resistance and cell invasion in pancreatic cancer, and miR-301b plays its role in pancreatic cancer through downregulating the expression of tumor-suppressor TP6 [Reference Funamizu, Lacy, Kamada, Yanaga and Manome22, Reference Funamizu, Lacy, Parpart, Takai, Hiyoshi and Yanaga23]. Thus, miR-301b might be an effective therapeutic target for PAAD treatment; however, little is known about its detailed regulatory mechanism in PAAD.

Herein, we detected the expressions of MBNL1-AS1 and miR-301b-3p in PAAD and analyzed the correlation between their expressions and metastasis and the overall survival rate of PAAD patients. Moreover, the role of MBNL1-AS1 and miR-301b-3p in PAAD and their interaction mechanisms were revealed in the present work. The study aims to uncover the molecular mechanism of PAAD development and provide potential targets for the diagnosis and treatment of PAAD.

2. Methods and Materials

2.1. Clinical Samples

PAAD tissues and the adjacent normal tissues were collected from 52 cases of PAAD patients in the First Affiliated Hospital of Xinjiang Medical University. All the patients received surgical treatment in our hospital from 2019 to 2021 and were diagnosed with PAAD by postoperative pathology. The patients accepted no treatment before the operation, and their clinicopathological data and follow-up data were complete. The surgical resection tissues were stored at −80°C. All the protocols involved with the human tissue samples were in accordance with the World Medical Association Declaration of Helsinki. The study protocol was approved by the ethics committee affiliated with the First Affiliated Hospital of Xinjiang Medical University (approval number: 2021-032), and the study has been granted an exemption from requiring written informed consent.

2.2. Cell Culture

HPDE6-C7, Capan-2, AsPC-1, SW1990, PANC-1, and SW1990 cells were purchased from the American Type Culture Collection (ATCC, VA., USA) and cultured in Dulbecco’s Modified Eagle Medium (DMEM; Hyclone, UT., USA) supplemented with 10% fetal bovine serum (FBS, Gibco, NY., USA), 100 μg/ml streptomycin, and 100 IU/ml penicillin. The culture medium was kept in an incubator (Liuyi, Beijing, China) with 5% CO2 at 37°C.

2.3. Quantitative Real-Time PCR (qRT-PCR)

qRT-PCR was performed to detect the expression of miR-301b-3p and MBNL1-AS1. Briefly, total RNA was first released from cells by using a TRIzol Kit (Invitrogen, CA., USA) according to the manufacturer’s protocol. Next, total RNA was reverse- transcribed into cDNA using the PrimeScript RT Reagent Kit (TaKaRa, Dalian, China) or One Step PrimeScript® miRNA cDNA Synthesis Kit (TaKaRa). Finally, qPCR was performed using SYBR qPCR reagents (Takara) and PCR operating instrument (BioRad, CA., USA). Sequences of primers were listed as follows: MBNL1-AS1 (forward:5′-TGGATAAGACAGTCCCTACA-3′, reverse: 5′- ATTGGATTGCTTCCCACATA-3′); GAPDH (forward: 5′-TCAAGGCTGAGAACGGGAAG-3′, reverse: 5′-TGGACTCCACGACGTACTCA-3′) as the internal reference of MBNL1-AS1; miR-301b-3p (forward: 5′- CAGTGCTCTGACGAGGTTG-3, reverse: 5′-TGTCCCAGATGCTTTGACA-3′); U6 (forward: 5′-CTCGCTTCGGCAGCACATA-3′, reverse: 5′-AACGATTCACGAATTTGCGT-3′) as the internal reference of miR-301b-3p. The expression was calculated using the 2−ΔΔCt method.

2.4. Western Blot Analysis

Total protein was obtained by RIPA lysis buffer (Solarbio, Beijing, China), and the concentration was measured by a BCA assay kit (Yeasen, Shanghai, China). Then 50 μg protein samples was separated on a sodium dodecyl sulphate-polyacrylamide gel electrophoresis (SDS-PAGE, Beyotime, Shanghai, China) and transferred to polyvinylidene fluoride (PVDF) membranes (Millipore, MD., USA). The membranes were blocked with 5% nonfat milk for 1.5 h and incubated with anti-E-cadherin, N-cadherin, and Vimentin at 4°C overnight. The next day, the membranes were washed and incubated with horseradish peroxidase-labeled IgG antibody at room temperature for 1 h. Images of blots were captured by using the gel imaging system (Bio-Rad, CA., USA), and the original blots are presented in Supplementary file 1.

2.5. Cell Transfection

pcDNA-MBNL1-AS1, miR-301b-3p inhibitor, miR-301b-3p mimic, and its negative control (NC) were purchased from Ribo Bio Co., LTD (Guangzhou, China). PANC-1 and SW1990 cells were seeded on a 96-well plate and cultured for 12 h. Then cells were transfected with pcDNA-MBNL1-AS1, miR-301b-3p inhibitor, miR-301b-3p mimic, or NC by using Lipofectamine 2000 reagent (Invitrogen, CA, USA) according to the manufacturer’s protocol.

2.6. Dual Luciferase Reporter Gene System

According to the binding site between miR-301b-3p and MBNL1-AS1 analyzed by bioinformatic tools, the wild-type sequence of MBNL1-AS1 and the mutant sequence of MBNL1-AS1 were designed, synthesized, and inserted into the vector pRL-TK (Promega, Beijing, China). PANC-1 and SW1990 cells were seeded on a 96-well plate and transfected with the reconstructed vector containing MBNL1-AS1 sequences, miR-301b-3p mimic, or NC mimic, by using Lipofectamine 2000 regents (Invitrogen, CA., USA). 48 h later, the cells were lysed for measurement of luciferase activity.

2.7. Cell Counting Kit (CCK-8) Assay

PANC-1 and SW1990 cells (2 × 103 cells/well) were seeded in 96-well plates, and CCK-8 solution (10 μmol/L) was added into each well at different timepoint (0 h, 24 h, 48 h, and 96 h). After incubating for 2 h at 37°C, the absorbance (OD value) of cells at 450 nm was measured with a microplate reader.

2.8. Apoptosis

Cell apoptosis was detected using the AnnexinV-FITC/PI Apoptosis Detection Kit (BD, NJ, USA). Briefly, cells were digested with 0.25% pancreatin, washed with precooled PBS, and centrifuged at 2000 rpm for 10 min. Cell precipitation was suspended with 300 μL binding buffer, mixed evenly with 5 μL Annexin V-FITC and incubated at room temperature for 15 min in the dark. 5 μL PI was added into cell mixture 5 min before measurement, and 200 μL binding buffer was added into them immediately before measurement. Flow cytometry was utilized for the measurement of the apoptotic rate.

2.9. Transwell Assay

100 μL PANC-1 or SW1990 cells (5 × 104 cells/well) were seeded in the upper chambers of a 24-well Transwell chamber (BD, NJ, USA), while 500 μL DMEM supplemented with 10% FBS was added to the lower chamber. After being cultured for 48 h, migrated cells in the lower chamber were fixed with 4% paraformaldehyde for 15 min and stained with 0.1% crystal violet (Sigma, MO., USA) for 15 min at room temperature. After being washed and dried, cells were photographed under a microscope (Olympus, Tokyo, Japan).

2.10. Xenograft Tumors

PANC-1 cells in the logarithmic growth phase were digested with 0.25% trypsin and resuspended in PBS after washing. 200 μL cell suspension (1 × 106 cells) was subcutaneously injected into the right armpit of nude mice using a 1 mL syringe. Tumor growth was recorded within 28 d after injection. The mice were sacrificed at 28 day and tumors were weighed.

2.11. Statistical Analysis

GraphPad Prism 8.0 (GraphPad Software, CA., USA) was used for data analysis and graphics drawing. Each experiment was independently repeated at least 3 times, and all data were presented as the mean ± standard deviation. Data in two groups was compared by using Student’s t-test, and p < 0.05 was considered statistically significant.

3. Results

3.1. MBNL1-AS1 Was Downregulated in PAAD and Closely Related to PAAD Progression

We first collected 52 cases of surgical resection tissues from PAAD patients and detected the expression of MBNL1-AS1 in PAAD tumor tissues and adjacent normal tissues. The qRT-PCR data in Figure 1(a) indicated that the MBNL1-AS1 expression of tumor tissues was lower than that of normal tissues. Then these tumor tissues were divided into two groups according to the average value of MBNL1-AS1 expression, 28 cases with high MBNL1-AS1 levels and 24 cases with low MBNL1-AS1 levels, and the relationship between MBNL1-AS1 and their clinicopathological features was shown in Table 1. Low expression of MBNL1-AS1 was significantly associated with poor differentiation degree, high TNM stage, and lymph node metastasis, while there was no significant correlation between MBNL1-AS1 and other variables, including age, gender, tumor location, or alcohol history (Table 1). Additionally, Kaplan–Meier analysis data indicated that lower MBNL1-AS1 expression was associated with a poorer prognosis and shorter overall survival of PAAD (Figure 1(b)). Meanwhile, the MBNL1-AS1 expression in PAAD cell lines (Capan-2, AsPC-1, SW1990, and PANC-1) was lower than that in normal pancreatic ductal epithelial cells (Figure 1(c)). Taken together, the data suggested that MBNL1-AS1 was downregulated in PAAD and closely related to PAAD progression and metastasis.

FIGURE 1: MBNL1-AS1 was downregulated in PAAD and closely related to PAAD progression: (a) MBNL1-AS1 expression in PAAD tumor tissues and adjacent normal tissues. (b) The overall survival rate of PAAD patients with high MBNL1-AS1 expression or low MBNL1-AS1 expression. (c) MBNL1-AS1 expression in the normal pancreatic ductal epithelial cells (HPDE6-C7) and PAAD cell lines (capan-2, AsPC-1, SW1990, and PANC-1). *P < 0.05, **P < 0.01, and ***P < 0.001.

TABLE 1: Relationship between MBNL1-AS1 and clinicopathological characteristics of PAAD.

3.2. Overexpression of MBNL1-AS1 Suppressed Cell Proliferation, Migration, and EMT in PAAD

Due to the significance of abnormal expression in SW1990 and PANC-1, the two cell lines were chosen for the following experiments. To investigate the role of MBNL1-AS1 in PAAD, pcDNA-MBNL1-AS1 was transfected into SW1990 and PANC-1 for overexpressing MBNL1-AS1 (Figure 2(a)). Then, CCK-8 assay was performed to evaluate cell proliferation, and the results in Figure 2(b) showed that overexpression of MBNL1-AS1 suppressed the cell viability of the PAAD cells. Next, transwell assay was used to evaluate cell migration, and we found that cell migration ability was also inhibited by overexpression of MBNL1-AS1 (Figure 2(c)). Finally, the expression of EMT markers was measured through western blot (Figure 2(d)). The data indicated that MBNL1-AS1 overexpression elevated the expression of an epithelial marker (E-cadherin) but decreased the expression of mesenchymal marker (N-cadherin and Vimentin), suggesting that MBNL1-AS1 hindered EMT behavior in PAAD cells. Collectively, these results demonstrated that upregulating MBNL1-AS1 suppressed proliferation, migration and EMT in PAAD cells.

FIGURE 2: Overexpression of MBNL1-AS1 suppressed cell proliferation, migration, and EMT in PAAD. (a) Verification of effect of transfection with pcDNA-MBNL1-AS1 in PANC-1 and SW1990. (b) Cell viability of PANC-1 and SW1990 transfected with or without pcDNA-MBNL1-AS1 at different time. (c) Cell migration of PANC-1 and SW1990 transfected with or without pcDNA-MBNL1-AS1. Scale bar = 200 μm. (d) The EMT markers (E-cadherin, N-cadherin, and vimentin) in PANC-1 and SW1990 transfected with or without pcDNA-MBNL1-AS1. OV, means overexpression. Compared to the OV-NC group, *P < 0.05, **P < 0.01, and ***P < 0.001.

3.3. MiR-301b-3p was Targeted by MBNL1-AS1 in PAAD

Through bioinformatics analysis, we found that miR-301b-3p could bind to MBNL1-AS1 theoretically, which was verified by the next dual luciferase reporter gene system (Figure 3(a)). We found that miR-301b-3p mimic reduced the luciferase activity in PAAD cells transfected with the wild-type sequence of MBNL1-AS1, rather than that in cells transfected with the mutant sequence of MBNL1-AS1. And miR-301b-3p expression in PANC-1 and SW1990 could be reduced by overexpressing MBNL1-AS1 (Figure 3(b)).

FIGURE 3: MiR-301b-3p was targeted by MBNL1-AS1 in PAAD. (a) Dual luciferase reporter gene assay was used to verify the targeting relationship between miR-301b-3p and MBNL1-AS1. (b) The effect of pcDNA-MBNL1-AS1 on the miR-301b-3p expression in PANC-1 and SW1990. (c) The miR-301b-3p expression in adjacent normal tissues and PAAD tumor tissues. (d) Linear regression analysis of the relationship between miR-301b-3p and MBNL1-AS1 expression. (e) The overall survival rate of PAAD patients with high miR-301b-3p expression or low miR-301b-3p expression. (f) The miR-301b-3p expression in different cell lines. *P < 0.05, **P < 0.01, and ***P < 0.001.

Subsequently, we detected miR-301b-3p expression in PAAD tissues and normal tissues. As expected, miR-301b-3p was upregulated in PAAD tissues (Figure 3(c)) and was negatively related to MBNL1-AS1 expression (Figure 3(d)). Likewise, we categorized these tumor tissues into a miR-301b-3p high-expression group and a miR-301b-3p low-expression group according to the average level of miR-301b-3p. Contrary to MBNL1-AS1, miR-301b-3p expression was closely related to differentiated degree, TNM stage, and lymph node metastasis (Table 2). Moreover, Kaplan–Meier analysis illustrated that the patients with higher miR-301b-3p expression had a poorer prognosis (Figure 3(e)). In addition, the miR-301b-3p expression in PAAD cell lines was shown to be higher than that in normal pancreatic ductal epithelial cells (Figure 3(f)). Altogether, these data miR-301b-3p was a target of MBNL1-AS1 and involved in the PAAD progression.

TABLE 2: Relationship between miR-301b-3p and clinicopathological characteristics of PAAD.

3.4. MBNL1-AS1 Suppressed Cell Proliferation and Metastasis in PAAD by Targeting miR-301b-3p

To verify whether MBNL1-AS1 suppressed PAAD malignancy by targeting miR-301b-3p, the function recovery experiment was conducted by downregulating miR-301b-3p alone or simultaneously overexpressing miR-301b-3p and MBNL1-AS1 in PANC-1 and SW1990 cells (Figure 4(a)). As shown in Figures 4(b) and 4(c), both MBNL1-AS1 restoration and miR-301b-3p depletion reduced cell viability but promoted cell apoptosis in PANC-1 and SW1990 cells, which were reversed by simultaneously overexpressing miR-301b-3p and MBNL1-AS1. Through the tumorigenesis experiment in vivo, we found that either upregulating MBNL1-AS1 or inhibiting miR-301b-3p restrained tumor growth in vivo, while miR-301b-3p mimic abolished the tumor-suppressive effect of MBNL1-AS1 upregulation (Figures 4(d)4(f)).

FIGURE 4: MBNL1-AS1 suppressed cell proliferation in PAAD by targeting miR-301b-3p. (a) Verification of effect of transfection with miR-301b-3p inhibitor or miR-301b-3p mimic and pcDNA-MBNL1-AS1 in PANC-1 and SW1990 with different intervention. (b) Cell viability in PANC-1 and SW1990 with different intervention at different time. (c) Cell apoptosis in PANC-1 and SW1990 with different intervention. (d–f), The stably transfected PANC-1 cells were subcutaneously into nude mice, tumor growth was recorded within 28 days after transplantation (d). The mice were sacrificed at 28 days and tumors were weighed (e-f). Compared to the NC group, *P < 0.05. Compared to the OV-MBNL1-AS1 group, #P < 0.05.

Additionally, both MBNL1-AS1 upregulation and miR-301b-3p downregulation reduced cell migration ability and EMT behavior in PANC-1 and SW1990 cells (Figures 5(a)5(c)). Nevertheless, overexpressing miR-301b-3p and MBNL1-AS1 simultaneously counteracted the suppressive effect of MBNL1-AS1 restoration on those aggressive processes in PAAD cells.

FIGURE 5: MBNL1-AS1 suppressed cell metastasis in PAAD by targeting miR-301b-3p. (a) Cell migration ability of PANC-1 and SW1990 with different intervention. Scale bar = 200 μm. (b and c), The EMT markers (E-cadherin, N-cadherin and vimentin) in PANC-1 and SW1990 with different intervention. OV means overexpression. Compared to the NC group, *P < 0.05. Compared to the OV-MBNL1-AS1 group, #P < 0.05.

To sum up, it is suggested that MBNL1-AS1 hindered PAAD development partly by targeting the tumor-promoting miR-301b-3p.

4. Discussion

In recent years, PAAD has attracted increasing attention for its growing incidence and high mortality [Reference Tang, Zhang and Liang24, Reference Weng, Zhang and Huang25]. Owing to the rapid growth of PAAD cells, abundant blood vessels and lymphatic vessels around the pancreas, and the incomplete envelope of the pancreas itself, PAAD is prone to metastasis at the early stage [Reference Fang, Wang, Cui and Huang26, Reference Zhang, Chen and Zhou27]. EMT, a transition that epithelial cells lose polarity and connections between cells, acquire the ability of infiltration and migration, and become cells with interstitial morphology and characteristics, is the main driving force of migration [Reference Ji, Ma, Chen and Cao28]. In this study, we explored the driving molecular of cell proliferation, migration, and EMT behavior in PAAD, hoping to improve the understanding of the PAAD development.

Accumulating evidence showed the dysregulation of specific ncRNAs could be observed in almost all cancers, and some of these ncRNAs play critical roles in the cancer development and progression [Reference Mukherjee and Goswami29]. Over the past few decades, a large number of ncRNAs (including lncRNAs and miRNAs) have been recognized as cancer-associated regulators [Reference Luo, Xu, Le, Ge and Wang30Reference Cao, Liu, Huang, Yue and Xi33]. The present work firstly showed that MBNL1-AS1 expression was significantly downregulated in PAAD tissues and cells and demonstrated that overexpression of MBNL1-AS1 suppressed cell proliferation, migration, and EMT behavior in PANC-1 and SW1990 cells. MBNL1-AS1 is a novel lncRNA discovered in recent years. Previous studies have proved that MBNL1-AS1 plays a tumor-suppressive role in several types of cancers, such as breast cancer, lung cancer, and bladder cancer [Reference Cao, Tan and Wei34Reference Wei, Yang and Wang36]. The role of MBNL1-AS1 in PAAD has not been clarified until our study.

According to the competitive endogenous RNA (ceRNA) hypothesis, lncRNAs exert their biology roles usually by sponging miRNAs to regulate genes expression [Reference Wang, Wan, Dong, Wang, Liu and Li37]. To date, some miRNAs have been verified to be the target of MBNL1-AS1, including miR-135a-5p in lung cancer and bladder cancer, miR-412-3p in colon cancer, and miR-338-5p in retinoblastoma [Reference Zhu, Wang, Liu, Li and Yu9, Reference Cao, Tan and Wei34, Reference Wei, Yang and Wang36, Reference Xu, Zhu, Tang and Liu38]. In the present study, miR-301b-3p was demonstrated to be the target molecular of MBNL1-AS1, and its expression was elevated in PAAD tissues and cells. As previously reported, miR-301b-3p is essential for the initiation of diverse common cancers, including PAAD [Reference Funamizu, Lacy, Kamada, Yanaga and Manome22, Reference Chen, Cheng, Zou, Wang, Wang and Chen39Reference Yang, Liu, Cao and Shi41]. MiR-301b-3p promotes cell proliferation and migration by inhibiting a lot of downstream effector genes, which function as tumor suppressors in cells [Reference Xiong, Zhang, Tang and Zhu21, Reference Fan, Jin, Liao, Qiao and Zhao42Reference Zheng and Bai44]. Finally, through a function recovery experiment, we verified that upregulation of miR-301b-3p abolished the inhibitory effect of MBNL1-AS1 overexpression on cell proliferation, apoptosis, tumorgenesis, migration, and EMT. These results preliminarily illustrated the underlying regulatory mechanism of PAAD tumorgenesis and metastasis. However, our present work only illustrated the interaction of MBNL1-AS1 and miR-301b-3p in PAAD because of the limited time and conditions. Actually, many miRNAs could be targeted by MBNL1-AS1. Therefore, further studies are needed to screen other functional miRNAs and improve the molecular mechanisms in the future.

5. Conclusion

In conclusion, we first identified MBNL1-AS1 as a tumor-suppressive lncRNA in PAAD, and MBNL1-AS1 reduces cell proliferation, migration, and EMT behavior of PAAD cells via sponging and downregulating miR-301b-3p. The findings shed light on a significant role of ncRNAs in the PAAD progression and might provide a theoretical basis for the development of PAAD therapies.

Data Availability

The data used in this study are all included in this paper. The original data are available from the corresponding author upon reasonable request.

Conflicts of Interest

The authors declare that there are no conflicts of interest.

Authors’ Contributions

CMS and WH made contributions to the conception and design of the research. CMS and A conducted investigation and data curation. QLC and TYH were responsible for supervision. CMS and A drafted and revised the manuscript. QLC and HL reviewed the manuscript. All authors have read and agreed to the published version of the manuscript. CS and AA contributed equally to this work.

Supplementary Materials

Supplementary file 1: Original western blots: A-B, The original western blots of E-cadherin, N-cadherin, Vimentin, and GAPDH in PANC1 cells (A) and SW1990 cells (B) transfected with or without pcDNA-MBNL1-AS1. C-D, The original western blots of E-cadherin, N-cadherin, Vimentin, and GAPDH in PANC1 cells (C) and SW1990 cells (D) with different intervention. (Supplementary Materials)

References

Edderkaoui, M., Chheda, C., Soufi, B. et al., “An inhibitor of GSK3B and HDACs kills pancreatic cancer cells and slows pancreatic tumor growth and metastasis in mice,Gastroenterology, vol. 155, no. 6, pp. 19851998.e5, 2018.CrossRefGoogle ScholarPubMed
Tian, C., Öhlund, D., Rickelt, S. et al., “Cancer cell-derived matrisome proteins promote metastasis in pancreatic ductal adenocarcinoma,Cancer Research, vol. 80, no. 7, pp. 14611474, 2020.CrossRefGoogle ScholarPubMed
Gao, Y., Shang, S., Guo, S. et al., “Lnc2Cancer 3.0: an updated resource for experimentally supported lncRNA/circRNA cancer associations and web tools based on RNA-seq and scRNA-seq data,Nucleic Acids Research, vol. 49, no. D1, pp. D1251d1258, 2021.CrossRefGoogle ScholarPubMed
Wu, X., Xia, T., Cao, M. et al., “LncRNA BANCR promotes pancreatic cancer tumorigenesis via modulating MiR-195-5p/wnt/β-catenin signaling pathway,Technology in Cancer Research and Treatment, vol. 18, Article ID 153303381988796, 2019.CrossRefGoogle ScholarPubMed
Li, W., Han, S., Hu, P. et al., “LncRNA ZNFTR functions as an inhibitor in pancreatic cancer by modulating ATF3/ZNF24/VEGFA pathway,Cell Death and Disease, vol. 12, no. 9, p. 830, 2021.CrossRefGoogle ScholarPubMed
Lin, J., Liao, S., Liu, Z., Li, E., Wu, X., and Zeng, W., “LncRNA FGD5-AS1 accelerates cell proliferation in pancreatic cancer by regulating miR-520a-3p/KIAA1522 axis,Cancer Biology and Therapy, vol. 22, no. 3, pp. 257266, 2021.CrossRefGoogle ScholarPubMed
Luo, Y., Wang, Q., Teng, L. et al., “LncRNA DANCR promotes proliferation and metastasis in pancreatic cancer by regulating miRNA-33b,FEBS Open Bio, vol. 10, no. 1, pp. 1827, 2020.CrossRefGoogle ScholarPubMed
Wei, C., Liang, Q., Li, X. et al., “Bioinformatics profiling utilized a nine immune-related long noncoding RNA signature as a prognostic target for pancreatic cancer,Journal of Cellular Biochemistry, vol. 120, no. 9, pp. 1491614927, 2019.CrossRefGoogle ScholarPubMed
Zhu, K., Wang, Y., Liu, L., Li, S., and Yu, W., “Long non-coding RNA MBNL1-AS1 regulates proliferation, migration, and invasion of cancer stem cells in colon cancer by interacting with MYL9 via sponging microRNA-412-3p,Clinics and Research in Hepatology and Gastroenterology, vol. 44, no. 1, pp. 101114, 2020.CrossRefGoogle ScholarPubMed
Li, P., Xing, W., Xu, J. et al., “microRNA-301b-3p downregulation underlies a novel inhibitory role of long non-coding RNA MBNL1-AS1 in non-small cell lung cancer,Stem Cell Research and Therapy, vol. 10, no. 1, p. 144, 2019.CrossRefGoogle ScholarPubMed
Fang, J., Jiang, G., Mao, W. et al., “Up-regulation of long noncoding RNA MBNL1-AS1 suppresses breast cancer progression by modulating miR-423-5p/CREBZF axis,Bioengineered, vol. 13, no. 2, pp. 37073723, 2022.CrossRefGoogle ScholarPubMed
Liu, W., Lin, W., and Yu, L., “Long non-coding RNA muscleblind like splicing regulator 1 antisense RNA 1 (LncRNA MBNL1-AS1) promotes the progression of acute myocardial infarction by regulating the microRNA-132-3p/SRY-related high-mobility-group box 4 (SOX4) axis,Bioengineered, vol. 13, no. 1, pp. 14241435, 2022.CrossRefGoogle ScholarPubMed
Fang, Q. Y., Deng, Q. F., Luo, J., and Zhou, C. C., “MiRNA-20a-5p accelerates the proliferation and invasion of non-small cell lung cancer by targeting and downregulating KLF9,European Review for Medical and Pharmacological Sciences, vol. 24, no. 5, pp. 25482556, 2020.Google ScholarPubMed
Sur, S., Steele, R., Shi, X., and Ray, R. B., “miRNA-29b inhibits prostate tumor growth and induces apoptosis by increasing bim expression,Cells, vol. 8, pp. 14551511, 2019.CrossRefGoogle ScholarPubMed
Wang, R., Sun, Y., Yu, W. et al., “Downregulation of miRNA-214 in cancer-associated fibroblasts contributes to migration and invasion of gastric cancer cells through targeting FGF9 and inducing EMT,Journal of Experimental and Clinical Cancer Research, vol. 38, no. 1, p. 20, 2019.CrossRefGoogle ScholarPubMed
Wang, Z., Li, T. E., Chen, M., Pan, J. J., and Shen, K. W., “miR-106b-5p contributes to the lung metastasis of breast cancer via targeting CNN1 and regulating Rho/ROCK1 pathway,Aging (Albany NY), vol. 12, no. 2, pp. 18671887, 2020.CrossRefGoogle Scholar
Wang, Y., Hu, J., Qi, G., Wang, S., and Gao, J., “miR-19a promotes the metastasis and EMT through CUL5 in prostate cancer cell line PC3,J buon, vol. 25, no. 4, pp. 20282035, 2020.Google ScholarPubMed
Zhan, T., Chen, X., Tian, X. et al., “MiR-331-3p links to drug resistance of pancreatic cancer cells by activating WNT/β-Catenin signal via ST7L,Technology in Cancer Research and Treatment, vol. 19, Article ID 153303382094580,2020.CrossRefGoogle ScholarPubMed
Fan, Y., Li, Y., Zhu, Y. et al., “miR-301b-3p regulates breast cancer cell proliferation, migration, and invasion by targeting NR3C2,Journal of Oncology, vol. 2021, Article ID 8810517, 9 pages, 2021.CrossRefGoogle ScholarPubMed
Liu, H., Ma, X., Niu, N. et al., “MIR-301b-3p promotes lung adenocarcinoma cell proliferation, migration and invasion by targeting DLC1,Technology in Cancer Research and Treatment, vol. 20, Article ID 153303382199003, 2021.CrossRefGoogle ScholarPubMed
Xiong, J., Zhang, L., Tang, R., and Zhu, Z., “MicroRNA-301b-3p facilitates cell proliferation and migration in colorectal cancer by targeting HOXB1,Bioengineered, vol. 12, no. 1, pp. 58395849, 2021.CrossRefGoogle ScholarPubMed
Funamizu, N., Lacy, C. R., Kamada, M., Yanaga, K., and Manome, Y., “MicroRNA-200b and -301 are associated with gemcitabine response as biomarkers in pancreatic carcinoma cells,International Journal of Oncology, vol. 54, no. 3, pp. 9911000, 2019.Google ScholarPubMed
Funamizu, N., Lacy, C. R., Parpart, S. T., Takai, A., Hiyoshi, Y., and Yanaga, K., “MicroRNA-301b promotes cell invasiveness through targeting TP63 in pancreatic carcinoma cells,International Journal of Oncology, vol. 44, no. 3, pp. 725734, 2014.CrossRefGoogle ScholarPubMed
Tang, R., Zhang, Y., Liang, C. et al., “The role of m6A-related genes in the prognosis and immune microenvironment of pancreatic adenocarcinoma,PeerJ, vol. 8, p. e9602, 2020.CrossRefGoogle ScholarPubMed
Weng, W., Zhang, Z., Huang, W. et al., “Identification of a competing endogenous RNA network associated with prognosis of pancreatic adenocarcinoma,Cancer Cell International, vol. 20, no. 1, p. 231, 2020.CrossRefGoogle ScholarPubMed
Fang, L., Wang, S. H., Cui, Y. G., and Huang, L., “LINC00941 promotes proliferation and metastasis of pancreatic adenocarcinoma by competitively binding miR-873-3p and thus upregulates ATXN2,European Review for Medical and Pharmacological Sciences, vol. 25, no. 4, pp. 18611868, 2021.Google ScholarPubMed
Zhang, Y., Chen, J., Zhou, N. et al., “FUBP1 mediates the growth and metastasis through TGFβ/Smad signaling in pancreatic adenocarcinoma,International Journal of Molecular Medicine, vol. 47, no. 5, p. 66, 2021.CrossRefGoogle ScholarPubMed
Ji, T., Ma, K., Chen, L., and Cao, T., “PADI1 contributes to EMT in PAAD by activating the ERK1/2-p38 signaling pathway,Journal of Gastrointestinal Oncology, vol. 12, no. 3, pp. 11801190, 2021.CrossRefGoogle ScholarPubMed
Mukherjee, M. and Goswami, S., “Identification of key deregulated RNA-binding proteins in pancreatic cancer by meta-analysis and prediction of their role as modulators of oncogenesis,Frontiers in Cell and Developmental Biology, vol. 9, Article ID 713852, 2021.CrossRefGoogle ScholarPubMed
Luo, H., Xu, C., Le, W., Ge, B., and Wang, T., “lncRNA CASC11 promotes cancer cell proliferation in bladder cancer through miRNA-150,Journal of Cellular Biochemistry, vol. 120, no. 8, pp. 1348713493, 2019.CrossRefGoogle ScholarPubMed
Luan, X. and Wang, Y., “LncRNA XLOC_006390 facilitates cervical cancer tumorigenesis and metastasis as a ceRNA against miR-331-3p and miR-338-3p,J Gynecol Oncol, vol. 29, no. 6, p. e95, 2018.CrossRefGoogle Scholar
Kong, X., Duan, Y., Sang, Y. et al., “LncRNA-CDC6 promotes breast cancer progression and function as ceRNA to target CDC6 by sponging microRNA-215,Journal of Cellular Physiology, vol. 234, no. 6, pp. 91059117, 2019.CrossRefGoogle ScholarPubMed
Cao, H. L., Liu, Z. J., Huang, P. L., Yue, Y. L., and Xi, J. N., “lncRNA-RMRP promotes proliferation, migration and invasion of bladder cancer via miR-206,European Review for Medical and Pharmacological Sciences, vol. 23, no. 3, pp. 10121021, 2019.Google ScholarPubMed
Cao, G., Tan, B., Wei, S. et al., “Down-regulation of MBNL1-AS1 contributes to tumorigenesis of NSCLC via sponging miR-135a-5p,Biomedicine and Pharmacotherapy, vol. 125, Article ID 109856, 2020.CrossRefGoogle ScholarPubMed
Jin, Y., Xu, L., Zhao, B. et al., “Tumour-suppressing functions of the lncRNA MBNL1-AS1/miR-889-3p/KLF9 axis in human breast cancer cells,Cell Cycle, vol. 21, pp. 113, 2022.CrossRefGoogle ScholarPubMed
Wei, X., Yang, X., Wang, B. et al., “LncRNA MBNL1-AS1 represses cell proliferation and enhances cell apoptosis via targeting miR-135a-5p/PHLPP2/FOXO1 axis in bladder cancer,Cancer Medicine, vol. 9, no. 2, pp. 724736, 2020.CrossRefGoogle ScholarPubMed
Wang, L. X., Wan, C., Dong, Z. B., Wang, B. H., Liu, H. Y., and Li, Y., “Integrative analysis of long noncoding RNA (lncRNA), microRNA (miRNA) and mRNA expression and construction of a competing endogenous RNA (ceRNA) network in metastatic melanoma,Medical Science Monitor, vol. 25, pp. 28962907, 2019.CrossRefGoogle ScholarPubMed
Xu, L., Zhu, S., Tang, A., and Liu, W., “LncRNA MBLN1-AS1 inhibits the progression of retinoblastoma through targeting miR-338-5p-Wnt/β-catenin signaling pathway,Inflammation Research, vol. 70, no. 2, pp. 217227, 2021.CrossRefGoogle ScholarPubMed
Chen, J., Cheng, L., Zou, W., Wang, R., Wang, X., and Chen, Z., “ADAMTS9-AS1 constrains breast cancer cell invasion and proliferation via sequestering miR-301b-3p,Frontiers in Cell and Developmental Biology, vol. 9, Article ID 719993, 2021.CrossRefGoogle ScholarPubMed
Wang, L., Lin, C., Sun, N., Wang, Q., Ding, X., and Sun, Y., “Long non-coding RNA CASC19 facilitates non-small cell lung cancer cell proliferation and metastasis by targeting the miR-301b-3p/LDLR axis,The Journal of Gene Medicine, vol. 22, no. 12, p. e3254, 2020.CrossRefGoogle ScholarPubMed
Yang, R., Liu, Z., Cao, H., and Shi, Y., “LINC01089, suppressed by YY1, inhibits lung cancer progression by targeting miR-301b-3p/HPDG axis,Cell Biology and Toxicology, vol. 38, no. 6, pp. 10631077, 2022.CrossRefGoogle ScholarPubMed
Fan, H., Jin, X., Liao, C., Qiao, L., and Zhao, W., “MicroRNA-301b-3p accelerates the growth of gastric cancer cells by targeting zinc finger and BTB domain containing 4,Pathology, Research and Practice, vol. 215, no. 11, Article ID 152667, 2019.CrossRefGoogle ScholarPubMed
Guo, Y., Yao, B., Zhu, Q. et al., “MicroRNA-301b-3p contributes to tumour growth of human hepatocellular carcinoma by repressing vestigial like family member 4,Journal of Cellular and Molecular Medicine, vol. 23, no. 8, pp. 50375047, 2019.CrossRefGoogle ScholarPubMed
Zheng, H. and Bai, L., “Hypoxia induced microRNA-301b-3p overexpression promotes proliferation, migration and invasion of prostate cancer cells by targeting LRP1B,Experimental and Molecular Pathology, vol. 111, Article ID 104301, 2019.CrossRefGoogle ScholarPubMed
Figure 0

FIGURE 1: MBNL1-AS1 was downregulated in PAAD and closely related to PAAD progression: (a) MBNL1-AS1 expression in PAAD tumor tissues and adjacent normal tissues. (b) The overall survival rate of PAAD patients with high MBNL1-AS1 expression or low MBNL1-AS1 expression. (c) MBNL1-AS1 expression in the normal pancreatic ductal epithelial cells (HPDE6-C7) and PAAD cell lines (capan-2, AsPC-1, SW1990, and PANC-1). *P < 0.05, **P < 0.01, and ***P < 0.001.

Figure 1

TABLE 1: Relationship between MBNL1-AS1 and clinicopathological characteristics of PAAD.

Figure 2

FIGURE 2: Overexpression of MBNL1-AS1 suppressed cell proliferation, migration, and EMT in PAAD. (a) Verification of effect of transfection with pcDNA-MBNL1-AS1 in PANC-1 and SW1990. (b) Cell viability of PANC-1 and SW1990 transfected with or without pcDNA-MBNL1-AS1 at different time. (c) Cell migration of PANC-1 and SW1990 transfected with or without pcDNA-MBNL1-AS1. Scale bar = 200 μm. (d) The EMT markers (E-cadherin, N-cadherin, and vimentin) in PANC-1 and SW1990 transfected with or without pcDNA-MBNL1-AS1. OV, means overexpression. Compared to the OV-NC group, *P < 0.05, **P < 0.01, and ***P < 0.001.

Figure 3

FIGURE 3: MiR-301b-3p was targeted by MBNL1-AS1 in PAAD. (a) Dual luciferase reporter gene assay was used to verify the targeting relationship between miR-301b-3p and MBNL1-AS1. (b) The effect of pcDNA-MBNL1-AS1 on the miR-301b-3p expression in PANC-1 and SW1990. (c) The miR-301b-3p expression in adjacent normal tissues and PAAD tumor tissues. (d) Linear regression analysis of the relationship between miR-301b-3p and MBNL1-AS1 expression. (e) The overall survival rate of PAAD patients with high miR-301b-3p expression or low miR-301b-3p expression. (f) The miR-301b-3p expression in different cell lines. *P < 0.05, **P < 0.01, and ***P < 0.001.

Figure 4

TABLE 2: Relationship between miR-301b-3p and clinicopathological characteristics of PAAD.

Figure 5

FIGURE 4: MBNL1-AS1 suppressed cell proliferation in PAAD by targeting miR-301b-3p. (a) Verification of effect of transfection with miR-301b-3p inhibitor or miR-301b-3p mimic and pcDNA-MBNL1-AS1 in PANC-1 and SW1990 with different intervention. (b) Cell viability in PANC-1 and SW1990 with different intervention at different time. (c) Cell apoptosis in PANC-1 and SW1990 with different intervention. (d–f), The stably transfected PANC-1 cells were subcutaneously into nude mice, tumor growth was recorded within 28 days after transplantation (d). The mice were sacrificed at 28 days and tumors were weighed (e-f). Compared to the NC group, *P < 0.05. Compared to the OV-MBNL1-AS1 group, #P < 0.05.

Figure 6

FIGURE 5: MBNL1-AS1 suppressed cell metastasis in PAAD by targeting miR-301b-3p. (a) Cell migration ability of PANC-1 and SW1990 with different intervention. Scale bar = 200 μm. (b and c), The EMT markers (E-cadherin, N-cadherin and vimentin) in PANC-1 and SW1990 with different intervention. OV means overexpression. Compared to the NC group, *P < 0.05. Compared to the OV-MBNL1-AS1 group, #P < 0.05.

Supplementary material: File

Sulidankazha et al. supplementary material

Sulidankazha et al. supplementary material
Download Sulidankazha et al. supplementary material(File)
File 119.6 KB