Skip to main content
Erschienen in: BMC Oral Health 1/2022

Open Access 01.12.2022 | Research

MicroRNA-18a regulates the metastatic properties of oral squamous cell carcinoma cells via HIF-1α expression

verfasst von: Shihyun Kim, Suyeon Park, Ji-Hyeon Oh, Sang Shin Lee, Yoon Lee, Jongho Choi

Erschienen in: BMC Oral Health | Ausgabe 1/2022

Abstract

Background

Rapid metastasis of oral squamous cell carcinoma (OSCC) is associated with a poor prognosis and a high mortality rate. However, the molecular mechanisms underlying OSCC metastasis have not been fully elucidated. Although deregulated expression of microRNA (miRNA) has a crucial role in malignant cancer progression, the biological function of miRNA in OSCC progression remains unclear. This study aimed to investigate the function of miRNA-18a in OSCC metastatic regulation via hypoxia-inducible factor 1α (HIF-1α).

Methods

miRNA-18a-5p (miRNA-18a) expressions in patients with OSCC (n = 39) and in OSCC cell lines (e.g., YD-10B and HSC-2 cells) were analyzed using quantitative real-time polymerase chain reaction. HIF-1α protein expressions in OSCC cells treated with miRNA-18a mimics or combined with cobalt chloride were analyzed using western blotting. The miRNA-18a expression-dependent proliferation and invasion abilities of OSCC cells were analyzed using MTT assay, EdU assay, and a Transwell® insert system.

Results

miRNA-18a expression was significantly lower in OSCC tissue than in the adjacent normal tissue. In OSCC cell lines, HIF-1α expression was significantly decreased by miRNA-18a mimic treatment. Furthermore, the migration and invasion abilities of OSCC cells were significantly decreased by miRNA-18a mimics and significantly increased by the overexpression of HIF-1α under hypoxic conditions relative to those abilities in cells treated only with miRNA-18a mimics.

Conclusions

miRNA-18a negatively affects HIF-1α expression and inhibits the metastasis of OSCC, thereby suggesting its potential as a therapeutic target for antimetastatic strategies in OSCC.
Begleitmaterial
Additional file 2. Figure S1. The original picture of western blots of Figs. 2A were shown in Figs. S1 (the red box indicated the representative picture in duplicate). Protein expression of HIF-1α in YD-10B and HSC-2 cells under hypoxic conditions by western blot analysis. All membranes were cut into small pieces and incubated with each antibodies. GAPDH was used as the control gene. Merge images indicate combination with bright field and chemiluminescence. The chemiluminescence images were time exposures of minimum (Low exposure) and maximum (High exposure) duration by the Fusion Solo Vilber Lourmat system. All images were unprocessed files.Figure S2. The original picture of western blots of Figs. 3C were shown in Figs. S2 (the red box indicated the representative picture in duplicate). Protein expression of HIF-1α and PCNA induced by miRNA-18a mimics in YD-10B cells by western blot analysis. All membranes were cut into small pieces and incubated with each antibodies. GAPDH was used as the control gene. Merge images indicate combination with bright field and chemiluminescence. The chemiluminescence images were time exposures of minimum (Low exposure) and maximum (High exposure) duration by the Fusion Solo Vilber Lourmat system. All images were unprocessed files. Figure S3. The original picture of western blots of Figs. 3C were shown in Figs. S3 (the red box indicated the representative picture in duplicate). Protein expression of HIF-1α and PCNA induced by miRNA-18a mimics in HSC-2 cells by western blot analysis. All membranes were cut into small pieces and incubated with each antibodies. GAPDH was used as the control gene. Merge images indicate combination with bright field and chemiluminescence. The chemiluminescence images were time exposures of minimum (Low exposure) and maximum (High exposure) duration by the Fusion Solo Vilber Lourmat system. All images were unprocessed files. Figure S4. The original picture of western blots of Figs. 3D were shown in Figs. S4 (the red box indicated the representative picture in duplicate). Protein expression of HIF-1α and PCNA induced by miRNA-18a mimics or miRNA-18a and CoCl2 in YD-10B cells by western blot analysis. All membranes were cut into small pieces and incubated with each antibodies. GAPDH was used as the control gene. Merge images indicate combination with bright field and chemiluminescence. The chemiluminescence images were time exposures of minimum (Low exposure) and maximum (High exposure) duration by the Fusion Solo Vilber Lourmat system. All images were unprocessed files. Figure S5. The original picture of western blots of Figs. 3D were shown in Figs. S5 (the red box indicated the representative picture in duplicate). Protein expression of HIF-1α and PCNA induced by miRNA-18a mimics or miRNA-18a and CoCl2 in HSC-2 cells by western blot analysis. All membranes were cut into small pieces and incubated with each antibodies. GAPDH was used as the control gene. Merge images indicate combination with bright field and chemiluminescence. The chemiluminescence images were time exposures of minimum (Low exposure) and maximum (High exposure) duration by the Fusion Solo Vilber Lourmat system. All images were unprocessed files.
Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1186/​s12903-022-02425-6.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
OSCC
Oral squamous cell carcinoma
miRNA
MicroRNA
HIF-1α
Hypoxia-inducible factor 1α
CoCl2
Cobalt chloride
PCNA
Proliferating cell nuclear antigen

Background

Oral squamous cell carcinoma (OSCC), the most common type of head and neck cancer worldwide, affects multiple sites, including the oral cavity, larynx, pharynx, and neck [1]. Approximately, 65,000 new OSCC cases are estimated to occur annually in the United States. [2]. In South Korea, 3310 patients were newly diagnosed with OSCC and died from the disease in 2015 [3]. Late diagnosis strongly influences the high mortality rate in patients with OSCC [4]. The presence of lymph node metastasis is one of the critical factors that determine the prognosis in such patients [5, 6]. Metastasis is associated with the migration and invasion of cancer cells from the primary tumor and is regulated in multiple steps and by various underlying molecular mechanisms. In OSCC, metastasis occurs predominantly in the cervical lymph nodes, which is an important determinant in OSCC diagnosis and patient survival [7, 8]. Despite the advancements in the detection and elimination of lymph node metastasis, this strategy has its own limitations, and the molecular mechanism underlying OSCC metastasis remains unclear.
MicroRNAs (miRNAs) are single-stranded noncoding RNAs composed of 21–25 nucleotides that negatively regulate gene expression in post-transcriptional processes by regulating messenger RNA (mRNA) degradation or translational suppression [911]. In OSCC, miRNAs have been reported to play a crucial role in the progression, metastasis, and the low long-term survival rates of the patients [9, 12]. Particularly, the miRNA-17-92 cluster, including miRNA-17, -18a, -19a/b, -20a, and -92a, has been reported to function in tumorigenesis and metastasis in several cancers, including thyroid, colon, lung, and renal cell carcinomas [1316]. In this cluster, miRNA-17 is highly expressed and is related to recurrence in patients with OSCC according to its participation in cell cycle distribution [17]. Chang et al. detected the regulation of miRNA-17 and -20a in OSCC cells, which promoted the migration ability of OSCC cells. Clinically, the expression of these miRNAs is negatively associated with tumor node metastasis stage and lymphatic metastasis via direct binding to integrin β8 [18]. However, the expression level of this cluster in OSCC has not been verified, and it is unclear which miRNA in this cluster is involved in OSCC metastasis.
Hypoxia, an insufficient oxygen level in whole organisms or tissues, is a well-known critical factor in tumor microenvironments and pathophysiology. Hypoxia in tissues triggers various cellular molecular responses, including rapid cell division, metabolism, and abnormal blood vessel formation [19, 20]. Hypoxia also induces tumor progression and is one of the causes for chemotherapy and radiotherapy resistance in tumors [21]. Under hypoxic conditions, a family of transcription factors known as hypoxia-inducible factors (HIFs) is activated and plays a critical role in orchestrating multicellular signaling pathways by changing the expressions of hundreds of genes [22]. HIFs are heterodimeric transcription factors composed of HIF-1α and constitutively expressed HIF-1β. In particular, HIF-1α is a key molecule induced under hypoxic conditions during excessive tumor growth and can control the expression of target genes involved in tumor angiogenesis, proliferation, invasion, and metastasis in various cancers [2326]. HIF-1α expression can regulate a panel of miRNAs, and certain miRNAs are able to target the protein expression of HIF-1α [27]. Wu et al. showed that hypoxia affected the induction level of type 1 collagen prolyl-4-hydroxylase, which led to the accumulation of Ago2 via an HIF-1α-dependent pathway [28]. In addition, Kelly et al. found that hypoxia-induced miRNA-210 expression suppressed the activity of glycerol-3-phosphate dehydrogenase 1-like, a regulator of HIF-1α, which resulted in an increase in HIF-1α stability and the expression of HIF-1α target genes [29]. However, other than the expression of HIF-1α, the precise molecular mechanisms underlying the clinical progression of OSCC are not well understood.
This study aimed to analyze the expression of HIF-1α and validate HIF-1α target miRNA candidates in OSCC. Furthermore, this study intended to determine if the interaction between HIF-1α and miRNAs regulates OSCC progression and metastasis.

Methods

Human tissues

In total, 39 paired normal and tumor tissue specimens were obtained from patients with OSCC. The biospecimens and data used in this study were obtained from the Biobank of Inje Paik University, Pusan National University and Keimyung University Dongsan Hospital (a member of the Korea Biobank Network), or Gangneung-Wonju Dental Hospital National University, Gangneung, South Korea according to the approved protocols. All studies received ethical approval from the Committees for Ethical Review of Research at Gangneung-Wonju National University (Institutional Review Board [IRB] No.: GWNUIRB-2020-26-1).

Cell culture and transfection

The human OSCC cell lines YD-10B and HSC-2 were purchased from the Korean Cell Line Bank (Seoul, South Korea) and the Japanese Collection of Research Bioresources Cell Bank (Ibaraki, Osaka, Japan), respectively. These cell lines were cultured in Dulbecco’s modified Eagle’s medium with high glucose (Invitrogen, Carlsbad, CA, USA) supplemented with 1% penicillin/streptomycin and 10% fetal bovine serum (FBS; Gibco, Waltham, MA, USA). The cells were cultured in a humidified atmosphere at 37 °C with 5% CO2. To expose YD-10B and HSC-2 cells to hypoxic conditions, they were cultured in an HERA cell 150i incubator with 94% N2, 1% O2, and 5% CO2 at 37 °C. For the transfection of miRNA-18a, the cells were first cultured with reduced serum Opti-MEM (Gibco). After the cells reached 60% confluence, hsa-miRNA-negative control (25 nM; Bioneer, Daejeon, Korea) or hsa-miRNA-18a-5p (25 nM; Bioneer) was transfected into the cells using Lipofectamine RNAiMAX Transfection Reagent (Invitrogen). The cells were further cultured with 300 mM cobalt chloride (CoCl2) for 24 h after transfection. The mature sequence of hsa-miRNA-18a-5p used was as follows: 5′-UAA GGU GCA UCU AGU GCA GAU AG -3′.

RNA extraction and quantitative real-time polymerase chain reaction (qRT-PCR) analysis

Total RNA was extracted using the TRIzol reagent (Invitrogen). For HIF-1α or miRNA-18a quantification, cDNA was synthesized using Superscript III Reverse Transcriptase (Invitrogen) or miRNA First-Strand Synthesis Kit (Takara Bio Inc., Shiga, Japan) according to the respective manufacturer’s instructions. The expression of HIF-1α or mature miRNAs was detected using a CFX96 Touch Real-Time PCR system (Bio-Rad Laboratories, Hercules, CA, USA) and SYBR Premix ExTaq (Takara Bio Inc., Shiga, Japan). The amplification conditions were as follows: initial denaturation at 95 °C for 10 s, followed by 40 cycles of denaturation at 95 °C for 5 s, and annealing and extension at 60 °C for 20 s. The primers used in the present study were as follows: forward-HIF-1α: 5′-CCA GTT ACG TTC CTT CGA TCA G-3′; reverse-HIF-1α: 5′-GTA GTG GTG GCA TTA GCA GTA G-3′; forward-hsa-miRNA-17: 5′-TGC TTA CAG TGC AGG TAG-3′; forward-hsa-miRNA-18a: 5′-AGG TGC ATC TAG TGC AG-3′; forward-hsa-miRNA-19a: 5′-GTT TTG CAT AGT TGC ACT A-3′; and forward-has-miRNA-20a: 5′-GTG CTT ATA GTG CAG GTA-3′. In addition, normalization was performed using the following primers: forward-GAPDH: 5′-CAA AGT TGT CAT GGA TGA CC-3′; reverse-GAPDH: 5′-CCA TGG AGA AGG CTG GGG-3′; and reverse-U6: 5′-AAA ATA TGG AAC GCT TCA CGA-3′. Relative expression levels were calculated using the 2−ΔΔCt method. All experiments were performed in triplicate.

Western blots

The cells were lysed with lysis buffer (RIPA buffer; Sigma), including Complete Protease Inhibitor Cocktail (Roche Diagnostics, Basel, Switzerland). The protein concentration of the lysates was calculated using the BCA Protein Assay Kit (Pierce, Rockford, IL, USA). Subsequently, 50 µg of the total lysate was mixed with 5 × loading dye (Bio-Rad) and separated using 8–12% sodium dodecyl sulfate polyacrylamide gel electrophoresis. After being electrophoresed, the proteins were transferred to a nitrocellulose membrane using a Wet/Tank Blotting System (Bio-Rad). The membranes were incubated with 5% bovine serum albumin (Sigma) in phosphate-buffered saline with 0.01% Tween-20 for 30 min at room temperature. Subsequently, they were incubated with rabbit monoclonal antibodies specific to anti-HIF-1α (1:1000 dilution; GeneTex, Inc., Irvine, CA, USA), GAPDH (1:3000 dilution; Ab Frontier, Seoul, Republic of Korea), and anti-mouse-Proliferating Cell Nuclear Antigen (PCNA) (1:1000 dilution; Cell Signaling Technology, Danvers, MA, USA) for 3 h at room temperature. Secondary antibodies were blocked using horseradish peroxidase (HRP)-conjugated goat anti-rabbit immunoglobulin G (IgG) (1:5000 dilution; Cell Signaling Technology) and HRP-conjugated goat anti-mouse IgG (1:10,000 dilution; Cell Signaling Technology). Specific bands were detected using an imaging system (VILBER Smart Imaging, Eberhardzell, Germany) and chemiluminescence reagent (Millipore Corporation, St. Louis, MO, USA). ImageJ image processing software (https://​imagej.​nih.​gov/​ij/​) was used to analyze the intensity of each detected band, which was quantified as the fold change. All experiments were performed at least in duplicate.

Cell proliferation assay

To analyze the proliferation ability of the OSCC cell lines, a 3-(4,5-dimethylyhiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) assay was performed to assess cell viability in proliferating cells and a Click-iT 5-ethynyl-2′-deoxyuurindine (EdU) assay was conducted to assess DNA synthesis in proliferating cells (each according to the manufacturer’s instructions). In the MTT assay, 3 × 103 cells were seeded in a 96-well cell culture plate (Corning Inc., Corning, NY, USA) and cultivated with miRNA-18a or CoCl2 at 37 °C in an incubator with a humidified atmosphere containing 5% CO2. After 24 h, 200 µl of dimethyl sulfoxide (DMSO; Sigma) was added for 10 min, and the resulting formazan was dissolved in DMSO. The absorbance was then detected at 570 nm using a SpectraMax iD3 Multimode Microplate Reader (Molecular Devices, San Jose, CA, USA). In the EdU assay, 3 × 105 cells were seeded on coverslips in cell culture dishes and cultured with miRNA-18a or CoCl2 at 37 °C in an incubator with a humidified atmosphere containing 5% CO2. The cells were maintained in EdU solution for 2 h, after which they were fixed using 4% paraformaldehyde for 20 min and permeabilized in 0.5% Triton X-100 for 15 min. Subsequently, the coverslips were stained using an EdU Cell Proliferation Kit, Alexa Fluor 488 (Abcam, Cambridge, UK) according to the manufacturer’s protocol. Each process was performed in a dark environment to protect the fluorescence from the light. The cells were imaged using fluorescence microscopy (Olympus, Tokyo, Japan), and the images were chosen randomly from seven parts of the coverslip. All experiments were performed at least in triplicate.

Transwell migration and invasion assays

To analyze the migration and invasion abilities of the OSCC cells, a 24-well plate cell culture insert system (8.0-µm pore size; Corning, Inc.) was used. To determine the invasion abilities of the OSCC cells, the inserts were precoated with 5% collagen mixed with 40 µl of reduced serum Opti-MEM (Gibco). After transfection with the negative control or miRNA-18a, the reduced serum Opti-MEM medium was added to the upper insert and culture medium containing 10% FBS was added to the lower plate for 24 h. The inserts were then fixed with methanol and stained using Mayer’s Hematoxylin Histological Staining Reagent (Dako, Santa Clara, CA, USA) for 20 min at room temperature. The noninvaded cells in the upper chamber were scraped out using a cotton swap, and the number of invaded cells was counted using an inverted light microscope (Olympus, Tokyo, Japan) to make observations. The images of the migrated and invaded cells were randomly selected from seven parts of each insert, and the number of cells was calculated using ImageJ. All experiments were performed at least in triplicate.

Statistical analysis

All results were presented as mean ± standard error of the mean. To confirm the normal distribution of the analysis data, Levene’s test was performed. If the data were normally distributed, student’s t-test was performed, and if they were not normally distributed, the Mann–Whitney test was performed. In this study, the differences were considered to be statistically significant for p values < 0.05.

Results

miRNA-18a and -20a target HIF-1α directly in patients with OSCC

To determine miRNA candidates that might target HIF-1α in OSCC, we used three public algorithm program sets, namely, Targetscan, MiRDB, and PICTAR, to predict targets of miRNA. We found that miRNA-4495, -17, -18a, -1276, -19a, -20a, and -5692a potentially targeted HIF-1α (Fig. 1A). To verify these targets, we matched the miRNA sequences with the 3′-UTR region of HIF-1α mRNA. Sequence alignment showed that parts of miRNA-17, -18a, -19a, and -20a possessed complementary binding sites for the 3′-UTR of HIF-1α, whereas miRNA-4495, -1276, and -5692 did not (Fig. 1B). In clinical OSCC samples, the expressions of miRNA-18a and -20a were significantly decreased and increased, respectively, in the OSCC tissue relative to their expressions in the normal tissue (p < 0.05; Fig. 1C). Additionally, the expressions of HIF-1α and p53 (a tumor suppressor gene) were significantly increased and decreased, respectively, in the OSCC tissue relative to their expressions in the normal tissue (p < 0.05; Fig. 1D). Moreover, the expression of HIF-1α was significantly correlated with OSCC TNM staging including tumor size and lymph node metastasis  (p < 0.05; Additional file 1: Table S1). Collectively, these results suggest that miRNA-18a and -20a are direct targets of HIF-1α in OSCC although their expression patterns differed.

Hypoxia suppresses the expression of miR-18a in OSCC cell lines

Some miRNAs are differentially expressed under hypoxia and are known as hypoxia-regulated miRNAs, which have been identified in several cancer types [30]. To determine if miRNA-18a and -20a are hypoxia-regulated miRNAs, we confirmed the expressions of HIF-1α and two miRNAs in YD-10B and HSC-2 cells cultured under normoxic or hypoxic conditions. The protein expression of HIF-1α was significantly increased in YD-10B and HSC-2 cells cultured under hypoxic conditions relative to its expression under normoxic conditions (p < 0.05; Fig. 2A, B). In contrast, the mRNA expression of miRNA-18a was significantly decreased in YD-10B and HSC-2 cells cultured under hypoxic conditions relative to its expression under normoxic conditions (p < 0.05; Fig. 2C). However, the mRNA expression of miRNA-20a was not altered in the two cell lines cultured under hypoxic conditions (Fig. 2D).

miRNA-18a regulates the expression of HIF-1α in OSCC cell lines

To determine if miRNA-18a directly regulates the expression of HIF-1α in OSCC cell lines, we confirmed HIF-1α expression in YD-10B and HSC-2 cells treated with miRNA-18a mimics. miRNA scrambled control (control) or miRNA mimics were transfected into each OSCC cell line. The qPCR analysis showed an approximately five-fold increase in miRNA-18a expression in YD-10B and HSC-2 cells treated with miRNA-18a mimics compared with its expression in control cells (p < 0.05; Fig. 3A), which resulted in significantly decreased mRNA expression of HIF-1α relative to that in control cells (p < 0.05; Fig. 3B). Indeed, miRNA-mediated translation expression is usually coupled with the target degradation of mRNA [31]. Furthermore, we observed that the protein expression of HIF-1α was significantly decreased in YD-10B and HSC-2 cells treated with miRNA-18a mimics relative to its expression in control cells (p < 0.05; Fig. 3C). To test the re-expression of HIF-1α, we used a combination treatment of YD-10B and HSC-2 cells, i.e., treatment with CoCl2 after miRNA-18a mimic treatment. Accordingly, HIF-1α protein expression was significantly increased in YD-10B and HSC-2 cells treated with miRNA-18a mimics and CoCl2 relative to its expression in cells treated only with miRNA-18a mimics (p < 0.05; Fig. 3D). However, the miRNA-18a mimics and CoCl2 had no effect on the protein expression of PCNA (Fig. 3C, D). These results suggest that the expression of HIF-1α was regulated by miRNA-18a in YD-10B and HSC-2 cells.

miRNA-18a inhibits the metastatic, but not proliferative, properties of OSCC cells

Overexpression of miRNA-18a has previously been reported to promote the proliferation ability of OSCC cells in vitro [32]. To further examine the effect of miRNA-18a expression on the proliferation ability of OSCC cells, we performed EdU and MTT assays to analyze the proliferative abilities of YD-10B and HSC-2 cells treated with miRNA-18a mimics. Compared with the control cells, EdU incorporation into cells treated with miRNA-18a mimics had no significant effect (Fig. 4A, B). Consistently, there was no significant difference in the MTT absorbance of YD-10B and HSC-2 cells treated with miRNA-18a mimics relative to that of the control cells (Fig. 4C). Additionally, a Transwell migration assay performed to determine if miRNA-18a had an effect on the metastatic properties of OSCC cells showed that the number of migrated cells decreased significantly in miRNA-18a mimic-treated YD-10B and HSC-2 cells relative to the number in the control cells (p < 0.05; Fig. 4D) (Additional files 1 and 2). Moreover, the number of invaded cells decreased significantly in YD-10B and HSC-2 cells treated with miRNA-18a mimics (p < 0.05; Fig. 4E). Collectively, these findings indicate that miRNA-18a plays a role in the metastatic properties of OSCC.

miRNA-18a regulates the metastatic properties of OSCC primarily via HIF-1α

To determine if the proliferative properties induced by miRNA-18a were mediated by HIF-1α expression, we investigated the effects of miRNA-18a mimic treatment alone or in combination with CoCl2. The proliferation assay showed that EdU incorporation into YD-10B and HSC-2 cells treated with miRNA-18a mimics and CoCl2 was not significantly different from that in cells treated with miRNA-18a mimics alone (Fig. 5A, B). However, there was no significant difference in the MTT absorbances of YD-10B and HSC-2 cells treated with both miRNA-18a mimics and CoCl2 relative to those of cells treated with miRNA-18a alone (Fig. 5C). To determine if the metastatic properties induced by miRNA-18a were mediated by HIF-1α expression, we confirmed the migration and invasion ability of YD-10B and HSC-2 cells treated with miRNA-18a mimics alone or in combination with CoCl2. The number of migrated cells increased significantly in YD-10B and HSC-2 cells treated with both miRNA-18a mimics and CoCl2 relative to that in cells treated with miRNA-18a mimics alone (p < 0.05; Fig. 5D). Furthermore, the number of invaded cells increased significantly in YD-10B and HSC-2 cells treated with miRNA-18a mimics and CoCl2 in combination relative to that in cells treated only with miRNA-18a mimics (p < 0.05; Fig. 5E). These results allude that the miRNA-18a-induced metastatic properties of OSCC were mediated by HIF-1α expression although the results were inconsistent with the proliferative properties of OSCC cells.

Discussion

Despite the advancements in diagnosis and treatment, the 5-year survival rate of patients with OSCC remains low; thus, the disease is an unresolved public health problem [33]. Several studies have highlighted the fundamental role of miRNAs in tumor progression [34, 35]. The miRNA-17-92 cluster is a well-known collection of miRNAs, the deregulation of which has been investigated in various tumor types [36, 37]. Many studies have highlighted the conflicting pro-oncogenic or antioncogenic roles of these miRNAs in different cancer types, and this disagreement probably reflects the polycistronic structure of the clusters [38, 39]. In this study, the expression of the miRNA-17-92 cluster, including miRNA-17, -18a, -19a, and -20a, was inconsistent in patients with OSCC. For example, miRNA-18a was rarely expressed in patients with OSCC, whereas miRNA-20a was highly expressed. Thus, we hypothesized that these two miRNAs have different functions: miRNA-18a acts as an antioncogene, whereas miRNA-20a acts as a pro-oncogene and participates in the progression and tumorigenesis of OSCC.
As one of the most multifunctional and conserved miRNAs, the altered expression of miRNA-18a has been detected in various pathological and physiological processes, including cell proliferation, apoptosis, cancer invasion, and metastasis [40]. Relative to the other members of this cluster, miRNA-18a is less well-studied, and our limited understanding of it comes primarily from expression studies in human cancer cells. For example, Liu et al. showed that miRNA-18a is upregulated in hepatocellular carcinoma cells and that its expression promotes the proliferation of these cells by blocking the translation of estrogen receptor alpha [41]. In addition, the inhibition of miRNA-18a is known to induce the proliferation of thyroid cancer cells, such as ARO and FRO cells, because of caspase cascade activation, which results in apoptosis [15]. Thus, miRNA-18a may be a potential therapeutic target for cancer therapy. However, the functional role of miRNA-18a in OSCC progression is yet to be confirmed [42]. Studies have shown that the expression is upregulated in hypopharyngeal and laryngeal squamous cell carcinomas (SCCs) but downregulated in oropharyngeal SCC and that this differential expression markedly affects human papillomavirus (HPV) status [4345]. In the present study, to exclude the expression of HPV-status-dependent miRNAs, two types of OSCC cell lines, YD-10B and HSC-2 cells (i.e., HPV-negative cell lines [4648]), were used to determine the effects of miRNA-18a in OSCC progression. Unexpectedly, we found that miRNA-18a expression was not associated with the viability of either OSCC cell line. Similarly, Liu et al. demonstrated that miRNA-18a had no effect on cell viability in cervical cancer or on apoptosis after transfection but did affect cell viability in cervical cancer after radiotherapy [49]. Thus, miRNA-18a apparently participates in cellular senescence by regulating DNA double-strand breaks rather than proliferation and apoptosis [50]. However, further investigation into the cellular responses to DNA double-strand breaks and cellular senescence in relation to miRNA-18a expression during OSCC progression is required.
Solid tumor cells commonly encounter low nutrient and oxygen conditions in their region of origin during tumor development and progression, and hypoxic conditions in solid tumors are known to promote the activation of HIF-1α [51]. Hypoxic conditions mediate tumor growth, metabolism, and metastasis via several mechanisms, including inactivation of tumor suppressor genes and activation of oncogenes via miRNAs [27] known as hypoxamiRNAs [52]. Using bioinformatics analysis, we found evidence showing that four candidate miRNAs among the miRNA-17-92 cluster target HIF-1α, and miRNA-18a was verified as a target of HIF-1α expression. Indeed, the expression of miRNA-18a was markedly altered in OSCC cell lines under hypoxic conditions; thus, we hypothesized that it has a role in the regulation of OSCC cell behavior. In addition, overexpression of miRNA-18a inhibited the migration and invasion ability of OSCC cells under hypoxic conditions. Based on the effects of miRNA-18a under hypoxic conditions and the association between miRNA-18a and HIF-1α, it is possible that miRNA-18a affects the migration and invasion abilities of OSCC cells via HIF-1α.
One limitation of this study is that the mechanism underlying the migration and invasion ability of OSCC cells that depended on the altered expression of miRNA-18 and/or HIF-1α under hypoxic conditions was not investigated in vivo. Therefore, further studies should be conducted to address this limitation.

Conclusion

This study showed that miRNA-18a is downregulated in patients with OSCC and that it affects the migration and invasion of OSCC cells (YD-10B and HSC-2 cell lines) under hypoxic conditions. Furthermore, miRNA-18a was identified to be a target miRNA of HIF-1α. Collectively, these results indicate that miRNA-18a is a potential target for antimetastatic strategies in OSCC. However, the interaction between miRNA-18a and HIF-α and how it regulates the molecular mechanisms underlying metastasis should be further investigated to clarify the effect of miRNA-18a on OSCC metastasis.

Acknowledgements

This research was supported by a National Research Foundation of Korea (NRF) grant funded by the Korean government (MSIT) (2020R1C1C1004007) and the Co-operative Research Program (CR1902) of Gangneung-Wonju National University Dental Hospital.

Declarations

All experiments were approved by the Committees for Ethical Review of Research at Gangneung-Wonju National University (IRB No.: GWNUIRB-2020-26-1). All participants provided informed consent before participating in the study and were assured that their information would be kept confidential. All methods were performed in accordance with the Declaration of Helsinki.
Not applicable.

Competing interests

The authors declare that they have no competing interests.
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.
Anhänge

Supplementary Information

Additional file 2. Figure S1. The original picture of western blots of Figs. 2A were shown in Figs. S1 (the red box indicated the representative picture in duplicate). Protein expression of HIF-1α in YD-10B and HSC-2 cells under hypoxic conditions by western blot analysis. All membranes were cut into small pieces and incubated with each antibodies. GAPDH was used as the control gene. Merge images indicate combination with bright field and chemiluminescence. The chemiluminescence images were time exposures of minimum (Low exposure) and maximum (High exposure) duration by the Fusion Solo Vilber Lourmat system. All images were unprocessed files.Figure S2. The original picture of western blots of Figs. 3C were shown in Figs. S2 (the red box indicated the representative picture in duplicate). Protein expression of HIF-1α and PCNA induced by miRNA-18a mimics in YD-10B cells by western blot analysis. All membranes were cut into small pieces and incubated with each antibodies. GAPDH was used as the control gene. Merge images indicate combination with bright field and chemiluminescence. The chemiluminescence images were time exposures of minimum (Low exposure) and maximum (High exposure) duration by the Fusion Solo Vilber Lourmat system. All images were unprocessed files. Figure S3. The original picture of western blots of Figs. 3C were shown in Figs. S3 (the red box indicated the representative picture in duplicate). Protein expression of HIF-1α and PCNA induced by miRNA-18a mimics in HSC-2 cells by western blot analysis. All membranes were cut into small pieces and incubated with each antibodies. GAPDH was used as the control gene. Merge images indicate combination with bright field and chemiluminescence. The chemiluminescence images were time exposures of minimum (Low exposure) and maximum (High exposure) duration by the Fusion Solo Vilber Lourmat system. All images were unprocessed files. Figure S4. The original picture of western blots of Figs. 3D were shown in Figs. S4 (the red box indicated the representative picture in duplicate). Protein expression of HIF-1α and PCNA induced by miRNA-18a mimics or miRNA-18a and CoCl2 in YD-10B cells by western blot analysis. All membranes were cut into small pieces and incubated with each antibodies. GAPDH was used as the control gene. Merge images indicate combination with bright field and chemiluminescence. The chemiluminescence images were time exposures of minimum (Low exposure) and maximum (High exposure) duration by the Fusion Solo Vilber Lourmat system. All images were unprocessed files. Figure S5. The original picture of western blots of Figs. 3D were shown in Figs. S5 (the red box indicated the representative picture in duplicate). Protein expression of HIF-1α and PCNA induced by miRNA-18a mimics or miRNA-18a and CoCl2 in HSC-2 cells by western blot analysis. All membranes were cut into small pieces and incubated with each antibodies. GAPDH was used as the control gene. Merge images indicate combination with bright field and chemiluminescence. The chemiluminescence images were time exposures of minimum (Low exposure) and maximum (High exposure) duration by the Fusion Solo Vilber Lourmat system. All images were unprocessed files.
Literatur
2.
3.
Zurück zum Zitat Jung KW, Won YJ, Kong HJ, Lee ES. Cancer statistics in Korea: incidence, mortality, survival, and prevalence in 2015. Cancer Res Treat. 2018;50(2):303–16.PubMedPubMedCentralCrossRef Jung KW, Won YJ, Kong HJ, Lee ES. Cancer statistics in Korea: incidence, mortality, survival, and prevalence in 2015. Cancer Res Treat. 2018;50(2):303–16.PubMedPubMedCentralCrossRef
4.
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(6):394–424.PubMedCrossRef 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(6):394–424.PubMedCrossRef
5.
Zurück zum Zitat Kuroshima T, Onozato Y, Oikawa Y, Ohsako T, Kugimoto T, Hirai H, Tomioka H, Michi Y, Miura M, Yoshimura R, et al. Prognostic impact of lingual lymph node metastasis in patients with squamous cell carcinoma of the tongue: a retrospective study. Sci Rep. 2021;11(1):20535.PubMedPubMedCentralCrossRef Kuroshima T, Onozato Y, Oikawa Y, Ohsako T, Kugimoto T, Hirai H, Tomioka H, Michi Y, Miura M, Yoshimura R, et al. Prognostic impact of lingual lymph node metastasis in patients with squamous cell carcinoma of the tongue: a retrospective study. Sci Rep. 2021;11(1):20535.PubMedPubMedCentralCrossRef
6.
Zurück zum Zitat Brands MT, Brennan PA, Verbeek ALM, Merkx MAW, Geurts SME. Follow-up after curative treatment for oral squamous cell carcinoma. A critical appraisal of the guidelines and a review of the literature. Eur J Surg Oncol. 2018;44(5):559–65.PubMedCrossRef Brands MT, Brennan PA, Verbeek ALM, Merkx MAW, Geurts SME. Follow-up after curative treatment for oral squamous cell carcinoma. A critical appraisal of the guidelines and a review of the literature. Eur J Surg Oncol. 2018;44(5):559–65.PubMedCrossRef
7.
Zurück zum Zitat Layland MK, Sessions DG, Lenox J. The influence of lymph node metastasis in the treatment of squamous cell carcinoma of the oral cavity, oropharynx, larynx, and hypopharynx: N0 versus N+. Laryngoscope. 2005;115(4):629–39.PubMedCrossRef Layland MK, Sessions DG, Lenox J. The influence of lymph node metastasis in the treatment of squamous cell carcinoma of the oral cavity, oropharynx, larynx, and hypopharynx: N0 versus N+. Laryngoscope. 2005;115(4):629–39.PubMedCrossRef
8.
Zurück zum Zitat Woolgar JA, Scott J, Vaughan ED, Brown JS, West CR, Rogers S. Survival, metastasis and recurrence of oral cancer in relation to pathological features. Ann R Coll Surg Engl. 1995;77(5):325–31.PubMedPubMedCentral Woolgar JA, Scott J, Vaughan ED, Brown JS, West CR, Rogers S. Survival, metastasis and recurrence of oral cancer in relation to pathological features. Ann R Coll Surg Engl. 1995;77(5):325–31.PubMedPubMedCentral
9.
Zurück zum Zitat Lu J, Getz G, Miska EA, Alvarez-Saavedra E, Lamb J, Peck D, Sweet-Cordero A, Ebert BL, Mak RH, Ferrando AA, et al. MicroRNA expression profiles classify human cancers. Nature. 2005;435(7043):834–8.PubMedCrossRef Lu J, Getz G, Miska EA, Alvarez-Saavedra E, Lamb J, Peck D, Sweet-Cordero A, Ebert BL, Mak RH, Ferrando AA, et al. MicroRNA expression profiles classify human cancers. Nature. 2005;435(7043):834–8.PubMedCrossRef
10.
Zurück zum Zitat He L, Thomson JM, Hemann MT, Hernando-Monge E, Mu D, Goodson S, Powers S, Cordon-Cardo C, Lowe SW, Hannon GJ, et al. A microRNA polycistron as a potential human oncogene. Nature. 2005;435(7043):828–33.PubMedPubMedCentralCrossRef He L, Thomson JM, Hemann MT, Hernando-Monge E, Mu D, Goodson S, Powers S, Cordon-Cardo C, Lowe SW, Hannon GJ, et al. A microRNA polycistron as a potential human oncogene. Nature. 2005;435(7043):828–33.PubMedPubMedCentralCrossRef
11.
Zurück zum Zitat Lim LP, Glasner ME, Yekta S, Burge CB, Bartel DP. Vertebrate microRNA genes. Science. 2003;299(5612):1540.PubMedCrossRef Lim LP, Glasner ME, Yekta S, Burge CB, Bartel DP. Vertebrate microRNA genes. Science. 2003;299(5612):1540.PubMedCrossRef
12.
Zurück zum Zitat Carron J, Torricelli C, Silva JK, Queiroz GSR, Ortega MM, Lima CSP, Lourenço GJ. microRNAs deregulation in head and neck squamous cell carcinoma. Head Neck. 2021;43(2):645–67.PubMedCrossRef Carron J, Torricelli C, Silva JK, Queiroz GSR, Ortega MM, Lima CSP, Lourenço GJ. microRNAs deregulation in head and neck squamous cell carcinoma. Head Neck. 2021;43(2):645–67.PubMedCrossRef
13.
Zurück zum Zitat Tsuchida A, Ohno S, Wu W, Borjigin N, Fujita K, Aoki T, Ueda S, Takanashi M, Kuroda M. miR-92 is a key oncogenic component of the miR-17-92 cluster in colon cancer. Cancer Sci. 2011;102(12):2264–71.PubMedCrossRef Tsuchida A, Ohno S, Wu W, Borjigin N, Fujita K, Aoki T, Ueda S, Takanashi M, Kuroda M. miR-92 is a key oncogenic component of the miR-17-92 cluster in colon cancer. Cancer Sci. 2011;102(12):2264–71.PubMedCrossRef
14.
Zurück zum Zitat Chow TF, Mankaruos M, Scorilas A, Youssef Y, Girgis A, Mossad S, Metias S, Rofael Y, Honey RJ, Stewart R, et al. The miR-17-92 cluster is over expressed in and has an oncogenic effect on renal cell carcinoma. J Urol. 2010;183(2):743–51.PubMedCrossRef Chow TF, Mankaruos M, Scorilas A, Youssef Y, Girgis A, Mossad S, Metias S, Rofael Y, Honey RJ, Stewart R, et al. The miR-17-92 cluster is over expressed in and has an oncogenic effect on renal cell carcinoma. J Urol. 2010;183(2):743–51.PubMedCrossRef
15.
Zurück zum Zitat Takakura S, Mitsutake N, Nakashima M, Namba H, Saenko VA, Rogounovitch TI, Nakazawa Y, Hayashi T, Ohtsuru A, Yamashita S. Oncogenic role of miR-17-92 cluster in anaplastic thyroid cancer cells. Cancer Sci. 2008;99(6):1147–54.PubMedCrossRef Takakura S, Mitsutake N, Nakashima M, Namba H, Saenko VA, Rogounovitch TI, Nakazawa Y, Hayashi T, Ohtsuru A, Yamashita S. Oncogenic role of miR-17-92 cluster in anaplastic thyroid cancer cells. Cancer Sci. 2008;99(6):1147–54.PubMedCrossRef
16.
Zurück zum Zitat Hayashita Y, Osada H, Tatematsu Y, Yamada H, Yanagisawa K, Tomida S, Yatabe Y, Kawahara K, Sekido Y, Takahashi T. A polycistronic microRNA cluster, miR-17-92, is overexpressed in human lung cancers and enhances cell proliferation. Cancer Res. 2005;65(21):9628–32.PubMedCrossRef Hayashita Y, Osada H, Tatematsu Y, Yamada H, Yanagisawa K, Tomida S, Yatabe Y, Kawahara K, Sekido Y, Takahashi T. A polycistronic microRNA cluster, miR-17-92, is overexpressed in human lung cancers and enhances cell proliferation. Cancer Res. 2005;65(21):9628–32.PubMedCrossRef
17.
Zurück zum Zitat Huang Q, Shen YJ, Hsueh CY, Guo Y, Zhang YF, Li JY, Zhou L. miR-17-5p drives G2/M-phase accumulation by directly targeting CCNG2 and is related to recurrence of head and neck squamous cell carcinoma. BMC Cancer. 2021;21(1):1074.PubMedPubMedCentralCrossRef Huang Q, Shen YJ, Hsueh CY, Guo Y, Zhang YF, Li JY, Zhou L. miR-17-5p drives G2/M-phase accumulation by directly targeting CCNG2 and is related to recurrence of head and neck squamous cell carcinoma. BMC Cancer. 2021;21(1):1074.PubMedPubMedCentralCrossRef
18.
Zurück zum Zitat Chang CC, Yang YJ, Li YJ, Chen ST, Lin BR, Wu TS, Lin SK, Kuo MY, Tan CT. MicroRNA-17/20a functions to inhibit cell migration and can be used a prognostic marker in oral squamous cell carcinoma. Oral Oncol. 2013;49(9):923–31.PubMedCrossRef Chang CC, Yang YJ, Li YJ, Chen ST, Lin BR, Wu TS, Lin SK, Kuo MY, Tan CT. MicroRNA-17/20a functions to inhibit cell migration and can be used a prognostic marker in oral squamous cell carcinoma. Oral Oncol. 2013;49(9):923–31.PubMedCrossRef
19.
Zurück zum Zitat Chipurupalli S, Kannan E, Tergaonkar V, D’Andrea R, Robinson N. Hypoxia induced ER stress response as an adaptive mechanism in cancer. Int J Mol Sci. 2019;20(3):749.PubMedCentralCrossRef Chipurupalli S, Kannan E, Tergaonkar V, D’Andrea R, Robinson N. Hypoxia induced ER stress response as an adaptive mechanism in cancer. Int J Mol Sci. 2019;20(3):749.PubMedCentralCrossRef
21.
Zurück zum Zitat Joseph JP, Harishankar MK, Pillai AA, Devi A. Hypoxia induced EMT: a review on the mechanism of tumor progression and metastasis in OSCC. Oral Oncol. 2018;80:23–32.PubMedCrossRef Joseph JP, Harishankar MK, Pillai AA, Devi A. Hypoxia induced EMT: a review on the mechanism of tumor progression and metastasis in OSCC. Oral Oncol. 2018;80:23–32.PubMedCrossRef
22.
Zurück zum Zitat Wang J, Ni Z, Duan Z, Wang G, Li F. Altered expression of hypoxia-inducible factor-1α (HIF-1α) and its regulatory genes in gastric cancer tissues. PLoS ONE. 2014;9(6):e99835.PubMedPubMedCentralCrossRef Wang J, Ni Z, Duan Z, Wang G, Li F. Altered expression of hypoxia-inducible factor-1α (HIF-1α) and its regulatory genes in gastric cancer tissues. PLoS ONE. 2014;9(6):e99835.PubMedPubMedCentralCrossRef
23.
Zurück zum Zitat Yang H, Geng YH, Wang P, Zhou YT, Yang H, Huo YF, Zhang HQ, Li Y, He HY, Tian XX, et al. Extracellular ATP promotes breast cancer invasion and epithelial-mesenchymal transition via hypoxia-inducible factor 2α signaling. Cancer Sci. 2019;110(8):2456–70.PubMedPubMedCentralCrossRef Yang H, Geng YH, Wang P, Zhou YT, Yang H, Huo YF, Zhang HQ, Li Y, He HY, Tian XX, et al. Extracellular ATP promotes breast cancer invasion and epithelial-mesenchymal transition via hypoxia-inducible factor 2α signaling. Cancer Sci. 2019;110(8):2456–70.PubMedPubMedCentralCrossRef
24.
Zurück zum Zitat Gilkes DM, Semenza GL. Role of hypoxia-inducible factors in breast cancer metastasis. Future Oncol. 2013;9(11):1623–36.PubMedCrossRef Gilkes DM, Semenza GL. Role of hypoxia-inducible factors in breast cancer metastasis. Future Oncol. 2013;9(11):1623–36.PubMedCrossRef
25.
Zurück zum Zitat Semenza GL. Hydroxylation of HIF-1. Oxygen sensing at the molecular level. Physiology (Bethesda). 2004;19:176–82.PubMed Semenza GL. Hydroxylation of HIF-1. Oxygen sensing at the molecular level. Physiology (Bethesda). 2004;19:176–82.PubMed
28.
Zurück zum Zitat Wu C, So J, Davis-Dusenbery BN, Qi HH, Bloch DB, Shi Y, Lagna G, Hata A. Hypoxia potentiates microRNA-mediated gene silencing through posttranslational modification of Argonaute2. Mol Cell Biol. 2011;31(23):4760–74.PubMedPubMedCentralCrossRef Wu C, So J, Davis-Dusenbery BN, Qi HH, Bloch DB, Shi Y, Lagna G, Hata A. Hypoxia potentiates microRNA-mediated gene silencing through posttranslational modification of Argonaute2. Mol Cell Biol. 2011;31(23):4760–74.PubMedPubMedCentralCrossRef
29.
Zurück zum Zitat Kelly TJ, Souza AL, Clish CB, Puigserver P. A hypoxia-induced positive feedback loop promotes hypoxia-inducible factor 1alpha stability through miR-210 suppression of glycerol-3-phosphate dehydrogenase 1-like. Mol Cell Biol. 2011;31(13):2696–706.PubMedPubMedCentralCrossRef Kelly TJ, Souza AL, Clish CB, Puigserver P. A hypoxia-induced positive feedback loop promotes hypoxia-inducible factor 1alpha stability through miR-210 suppression of glycerol-3-phosphate dehydrogenase 1-like. Mol Cell Biol. 2011;31(13):2696–706.PubMedPubMedCentralCrossRef
30.
Zurück zum Zitat Blick C, Ramachandran A, McCormick R, Wigfield S, Cranston D, Catto J, Harris AL. Identification of a hypoxia-regulated miRNA signature in bladder cancer and a role for miR-145 in hypoxia-dependent apoptosis. Br J Cancer. 2015;113(4):634–44.PubMedPubMedCentralCrossRef Blick C, Ramachandran A, McCormick R, Wigfield S, Cranston D, Catto J, Harris AL. Identification of a hypoxia-regulated miRNA signature in bladder cancer and a role for miR-145 in hypoxia-dependent apoptosis. Br J Cancer. 2015;113(4):634–44.PubMedPubMedCentralCrossRef
31.
Zurück zum Zitat Omer AD, Janas MM, Novina CD. The chicken or the egg: microRNA-mediated regulation of mRNA translation or mRNA stability. Mol Cell. 2009;35(6):739–40.PubMedCrossRef Omer AD, Janas MM, Novina CD. The chicken or the egg: microRNA-mediated regulation of mRNA translation or mRNA stability. Mol Cell. 2009;35(6):739–40.PubMedCrossRef
32.
Zurück zum Zitat Huang C, Song H, Lai L. The role and mechanism of microRNA-18a-5p in oral squamous cell carcinoma. Mol Med Rep. 2019;20(2):1637–44.PubMedPubMedCentral Huang C, Song H, Lai L. The role and mechanism of microRNA-18a-5p in oral squamous cell carcinoma. Mol Med Rep. 2019;20(2):1637–44.PubMedPubMedCentral
33.
Zurück zum Zitat Ndaw S, Denis F, Marsan P, d’Almeida A, Robert A. Biological monitoring of occupational exposure to 5-fluorouracil: urinary α-fluoro-β-alanine assay by high performance liquid chromatography tandem mass spectrometry in health care personnel. J Chromatogr B Anal Technol Biomed Life Sci. 2010;878(27):2630–4.CrossRef Ndaw S, Denis F, Marsan P, d’Almeida A, Robert A. Biological monitoring of occupational exposure to 5-fluorouracil: urinary α-fluoro-β-alanine assay by high performance liquid chromatography tandem mass spectrometry in health care personnel. J Chromatogr B Anal Technol Biomed Life Sci. 2010;878(27):2630–4.CrossRef
34.
Zurück zum Zitat Wang Y, Wang L, Chen C, Chu X. New insights into the regulatory role of microRNA in tumor angiogenesis and clinical implications. Mol Cancer. 2018;17(1):22.PubMedPubMedCentralCrossRef Wang Y, Wang L, Chen C, Chu X. New insights into the regulatory role of microRNA in tumor angiogenesis and clinical implications. Mol Cancer. 2018;17(1):22.PubMedPubMedCentralCrossRef
35.
Zurück zum Zitat Rupaimoole R, Calin GA, Lopez-Berestein G, Sood AK. miRNA Deregulation in cancer cells and the tumor microenvironment. Cancer Discov. 2016;6(3):235–46.PubMedPubMedCentralCrossRef Rupaimoole R, Calin GA, Lopez-Berestein G, Sood AK. miRNA Deregulation in cancer cells and the tumor microenvironment. Cancer Discov. 2016;6(3):235–46.PubMedPubMedCentralCrossRef
36.
Zurück zum Zitat Moi L, Braaten T, Al-Shibli K, Lund E, Busund LR. Differential expression of the miR-17-92 cluster and miR-17 family in breast cancer according to tumor type; results from the Norwegian Women and Cancer (NOWAC) study. J Transl Med. 2019;17(1):334.PubMedPubMedCentralCrossRef Moi L, Braaten T, Al-Shibli K, Lund E, Busund LR. Differential expression of the miR-17-92 cluster and miR-17 family in breast cancer according to tumor type; results from the Norwegian Women and Cancer (NOWAC) study. J Transl Med. 2019;17(1):334.PubMedPubMedCentralCrossRef
37.
Zurück zum Zitat Fang LL, Wang XH, Sun BF, Zhang XD, Zhu XH, Yu ZJ, Luo H. Expression, regulation and mechanism of action of the miR-17-92 cluster in tumor cells (Review). Int J Mol Med. 2017;40(6):1624–30.PubMedPubMedCentral Fang LL, Wang XH, Sun BF, Zhang XD, Zhu XH, Yu ZJ, Luo H. Expression, regulation and mechanism of action of the miR-17-92 cluster in tumor cells (Review). Int J Mol Med. 2017;40(6):1624–30.PubMedPubMedCentral
38.
Zurück zum Zitat Donayo AO, Johnson RM, Tseng HW, Izreig S, Gariepy A, Mayya VK, Wu E, Alam R, Lussier C, Jones RG, et al. Oncogenic biogenesis of pri-miR-17∼92 reveals hierarchy and competition among polycistronic microRNAs. Mol Cell. 2019;75(2):340-56.e310.PubMedCrossRef Donayo AO, Johnson RM, Tseng HW, Izreig S, Gariepy A, Mayya VK, Wu E, Alam R, Lussier C, Jones RG, et al. Oncogenic biogenesis of pri-miR-17∼92 reveals hierarchy and competition among polycistronic microRNAs. Mol Cell. 2019;75(2):340-56.e310.PubMedCrossRef
40.
41.
Zurück zum Zitat Liu WH, Yeh SH, Lu CC, Yu SL, Chen HY, Lin CY, Chen DS, Chen PJ. MicroRNA-18a prevents estrogen receptor-alpha expression, promoting proliferation of hepatocellular carcinoma cells. Gastroenterology. 2009;136(2):683–93.PubMedCrossRef Liu WH, Yeh SH, Lu CC, Yu SL, Chen HY, Lin CY, Chen DS, Chen PJ. MicroRNA-18a prevents estrogen receptor-alpha expression, promoting proliferation of hepatocellular carcinoma cells. Gastroenterology. 2009;136(2):683–93.PubMedCrossRef
42.
Zurück zum Zitat Lamperska KM, Kozlowski P, Kolenda T, Teresiak A, Blizniak R, Przybyla W, Masternak MM, Golusinski P, Golusinski W. Unpredictable changes of selected miRNA in expression profile of HNSCC. Cancer Biomark. 2016;16(1):55–64.PubMedCrossRef Lamperska KM, Kozlowski P, Kolenda T, Teresiak A, Blizniak R, Przybyla W, Masternak MM, Golusinski P, Golusinski W. Unpredictable changes of selected miRNA in expression profile of HNSCC. Cancer Biomark. 2016;16(1):55–64.PubMedCrossRef
43.
Zurück zum Zitat Kikkawa N, Hanazawa T, Fujimura L, Nohata N, Suzuki H, Chazono H, Sakurai D, Horiguchi S, Okamoto Y, Seki N. miR-489 is a tumour-suppressive miRNA target PTPN11 in hypopharyngeal squamous cell carcinoma (HSCC). Br J Cancer. 2010;103(6):877–84.PubMedPubMedCentralCrossRef Kikkawa N, Hanazawa T, Fujimura L, Nohata N, Suzuki H, Chazono H, Sakurai D, Horiguchi S, Okamoto Y, Seki N. miR-489 is a tumour-suppressive miRNA target PTPN11 in hypopharyngeal squamous cell carcinoma (HSCC). Br J Cancer. 2010;103(6):877–84.PubMedPubMedCentralCrossRef
44.
Zurück zum Zitat Avissar M, McClean MD, Kelsey KT, Marsit CJ. MicroRNA expression in head and neck cancer associates with alcohol consumption and survival. Carcinogenesis. 2009;30(12):2059–63.PubMedPubMedCentralCrossRef Avissar M, McClean MD, Kelsey KT, Marsit CJ. MicroRNA expression in head and neck cancer associates with alcohol consumption and survival. Carcinogenesis. 2009;30(12):2059–63.PubMedPubMedCentralCrossRef
45.
Zurück zum Zitat Gao G, Gay HA, Chernock RD, Zhang TR, Luo J, Thorstad WL, Lewis JS Jr, Wang X. A microRNA expression signature for the prognosis of oropharyngeal squamous cell carcinoma. Cancer. 2013;119(1):72–80.PubMedCrossRef Gao G, Gay HA, Chernock RD, Zhang TR, Luo J, Thorstad WL, Lewis JS Jr, Wang X. A microRNA expression signature for the prognosis of oropharyngeal squamous cell carcinoma. Cancer. 2013;119(1):72–80.PubMedCrossRef
46.
Zurück zum Zitat Aggarwal N, Yadav J, Thakur K, Bibban R, Chhokar A, Tripathi T, Bhat A, Singh T, Jadli M, Singh U, et al. Human papillomavirus infection in head and neck squamous cell carcinomas: transcriptional triggers and changed disease patterns. Front Cell Infect Microbiol. 2020;10:537650.PubMedPubMedCentralCrossRef Aggarwal N, Yadav J, Thakur K, Bibban R, Chhokar A, Tripathi T, Bhat A, Singh T, Jadli M, Singh U, et al. Human papillomavirus infection in head and neck squamous cell carcinomas: transcriptional triggers and changed disease patterns. Front Cell Infect Microbiol. 2020;10:537650.PubMedPubMedCentralCrossRef
47.
Zurück zum Zitat Kim YR, Lee B, Byun MR, Lee JK, Choi JW. Evaluation of pemetrexed and etoposide as therapeutic regimens for human papillomavirus-positive oral and oropharyngeal cancer. PLoS ONE. 2018;13(7):e0200509.PubMedPubMedCentralCrossRef Kim YR, Lee B, Byun MR, Lee JK, Choi JW. Evaluation of pemetrexed and etoposide as therapeutic regimens for human papillomavirus-positive oral and oropharyngeal cancer. PLoS ONE. 2018;13(7):e0200509.PubMedPubMedCentralCrossRef
48.
Zurück zum Zitat Gaykalova DA, Manola JB, Ozawa H, Zizkova V, Morton K, Bishop JA, Sharma R, Zhang C, Michailidi C, Considine M, et al. NF-κB and stat3 transcription factor signatures differentiate HPV-positive and HPV-negative head and neck squamous cell carcinoma. Int J Cancer. 2015;137(8):1879–89.PubMedPubMedCentralCrossRef Gaykalova DA, Manola JB, Ozawa H, Zizkova V, Morton K, Bishop JA, Sharma R, Zhang C, Michailidi C, Considine M, et al. NF-κB and stat3 transcription factor signatures differentiate HPV-positive and HPV-negative head and neck squamous cell carcinoma. Int J Cancer. 2015;137(8):1879–89.PubMedPubMedCentralCrossRef
49.
Zurück zum Zitat Liu S, Pan X, Yang Q, Wen L, Jiang Y, Zhao Y, Li G. MicroRNA-18a enhances the radiosensitivity of cervical cancer cells by promoting radiation-induced apoptosis. Oncol Rep. 2015;33(6):2853–62.PubMedCrossRef Liu S, Pan X, Yang Q, Wen L, Jiang Y, Zhao Y, Li G. MicroRNA-18a enhances the radiosensitivity of cervical cancer cells by promoting radiation-induced apoptosis. Oncol Rep. 2015;33(6):2853–62.PubMedCrossRef
50.
Zurück zum Zitat Wu CW, Dong YJ, Liang QY, He XQ, Ng SS, Chan FK, Sung JJ, Yu J. MicroRNA-18a attenuates DNA damage repair through suppressing the expression of ataxia telangiectasia mutated in colorectal cancer. PLoS ONE. 2013;8(2):e57036.PubMedPubMedCentralCrossRef Wu CW, Dong YJ, Liang QY, He XQ, Ng SS, Chan FK, Sung JJ, Yu J. MicroRNA-18a attenuates DNA damage repair through suppressing the expression of ataxia telangiectasia mutated in colorectal cancer. PLoS ONE. 2013;8(2):e57036.PubMedPubMedCentralCrossRef
51.
Zurück zum Zitat Yasuda H. Solid tumor physiology and hypoxia-induced chemo/radio-resistance: novel strategy for cancer therapy: nitric oxide donor as a therapeutic enhancer. Nitric Oxide. 2008;19(2):205–16.PubMedCrossRef Yasuda H. Solid tumor physiology and hypoxia-induced chemo/radio-resistance: novel strategy for cancer therapy: nitric oxide donor as a therapeutic enhancer. Nitric Oxide. 2008;19(2):205–16.PubMedCrossRef
52.
Zurück zum Zitat Madanecki P, Kapoor N, Bebok Z, Ochocka R, Collawn JF, Bartoszewski R. Regulation of angiogenesis by hypoxia: the role of microRNA. Cell Mol Biol Lett. 2013;18(1):47–57.PubMedCrossRef Madanecki P, Kapoor N, Bebok Z, Ochocka R, Collawn JF, Bartoszewski R. Regulation of angiogenesis by hypoxia: the role of microRNA. Cell Mol Biol Lett. 2013;18(1):47–57.PubMedCrossRef
Metadaten
Titel
MicroRNA-18a regulates the metastatic properties of oral squamous cell carcinoma cells via HIF-1α expression
verfasst von
Shihyun Kim
Suyeon Park
Ji-Hyeon Oh
Sang Shin Lee
Yoon Lee
Jongho Choi
Publikationsdatum
01.12.2022
Verlag
BioMed Central
Erschienen in
BMC Oral Health / Ausgabe 1/2022
Elektronische ISSN: 1472-6831
DOI
https://doi.org/10.1186/s12903-022-02425-6

Weitere Artikel der Ausgabe 1/2022

BMC Oral Health 1/2022 Zur Ausgabe

„Übersichtlicher Wegweiser“: Lauterbachs umstrittener Klinik-Atlas ist online

17.05.2024 Klinik aktuell Nachrichten

Sie sei „ethisch geboten“, meint Gesundheitsminister Karl Lauterbach: mehr Transparenz über die Qualität von Klinikbehandlungen. Um sie abzubilden, lässt er gegen den Widerstand vieler Länder einen virtuellen Klinik-Atlas freischalten.

Klinikreform soll zehntausende Menschenleben retten

15.05.2024 Klinik aktuell Nachrichten

Gesundheitsminister Lauterbach hat die vom Bundeskabinett beschlossene Klinikreform verteidigt. Kritik an den Plänen kommt vom Marburger Bund. Und in den Ländern wird über den Gang zum Vermittlungsausschuss spekuliert.

Darf man die Behandlung eines Neonazis ablehnen?

08.05.2024 Gesellschaft Nachrichten

In einer Leseranfrage in der Zeitschrift Journal of the American Academy of Dermatology möchte ein anonymer Dermatologe bzw. eine anonyme Dermatologin wissen, ob er oder sie einen Patienten behandeln muss, der eine rassistische Tätowierung trägt.

Ein Drittel der jungen Ärztinnen und Ärzte erwägt abzuwandern

07.05.2024 Klinik aktuell Nachrichten

Extreme Arbeitsverdichtung und kaum Supervision: Dr. Andrea Martini, Sprecherin des Bündnisses Junge Ärztinnen und Ärzte (BJÄ) über den Frust des ärztlichen Nachwuchses und die Vorteile des Rucksack-Modells.

Update Zahnmedizin

Bestellen Sie unseren kostenlosen Newsletter und bleiben Sie gut informiert – ganz bequem per eMail.