Regulation of radiosensitivity by 4‑methylumbelliferone via the suppression of interleukin‑1 in fibrosarcoma cells

  • Authors:
    • Ryo Saga
    • Kazuki Hasegawa
    • Kosho Murata
    • Mitsuru Chiba
    • Toshiya Nakamura
    • Kazuhiko Okumura
    • Eichi Tsuruga
    • Yoichiro Hosokawa
  • View Affiliations

  • Published online on: January 29, 2019     https://doi.org/10.3892/ol.2019.9990
  • Pages: 3555-3561
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Tumor recurrence and distant metastasis following radiotherapy, which can lead to poor prognosis, are caused by residual cancer cells that acquire radioresistance. Chemotherapy or a combination of targeted inhibitors can potentially enhance radiation sensitivity and prevent metastasis. It was previously reported that co‑administration of the hyaluronan synthesis inhibitor 4‑methylumbelliferone (4‑MU) enhanced the lethality of X‑ray irradiation in HT1080 human fibrosarcoma cells and decreased their invasiveness to a greater extent than either treatment alone. To clarify the molecular basis of these effects, the present study conducted mRNA expression profiling by cDNA microarray to identify the signaling pathways that are altered under this combination treatment. The activation state of the signaling pathways was classified by z‑scores in the Ingenuity Pathway Analysis. The results revealed that the pro‑inflammatory cytokines interleukin (IL)‑6 and IL‑8 were activated by 2 Gy X‑ray irradiation, an effect that was abolished by co‑administration of 4‑MU. Similar trends were observed for the upstream signaling component IL‑1. These results indicate that the radiosensitivity of fibrosarcoma cells is improved by suppressing inflammation through the administration of 4‑MU.

Introduction

The advancement of technology has resulted in highly accurate radiation therapy for primary solid tumors (1); however, recurrence or distant metastasis due to residual cancer cells resistant to irradiation remains a major problem that leads to poor outcome (2) Radioresistance in SAS oral squamous cell carcinoma cells, HEp-2 laryngeal cancer cells, and lung cancer cells can be induced by fractionated radiotherapy (36). Combining chemotherapy (e.g. cisplatin, 5-fluorouracil, and paclitaxel) with targeted inhibitors can potentially overcome radioresistance and extend progression-free and overall survival (712). However, few studies have examined the efficacy of combined treatments in preventing distant metastasis (10).

4-Methylumbelliferone (4-MU) is a hyaluronan synthesis inhibitor (13,14) that has demonstrated anti-tumor and -invasion/metastasis effects in various cancer cell types and mouse models of prostate and liver cancer that are exerted via suppression of hyaluronan synthase (HAS) expression (15,16). 4-MU was also shown to suppress inflammatory cytokines and chemokines such as interleukin (IL)-6 and −8 (17). Elevated levels of nuclear factor (NF)-κB, a cytokine and transcription factor that regulates proinflammatory molecules such as IL-1β and IL-6 and tumor necrosis factor (TNF)α induce resistance to apoptosis (18) and radiotherapy (19) in cancer cells. IL-6 is a potential therapeutic target owing to its close association with cancer stem cells (20,21).

We speculated that combined administration of 4-MU which has anti-inflammatory effects and radiotherapy can not only prevent distant metastasis but also sensitize radioresistant cells to the effects of X-ray radiation. In our previous study, HT1080 human fibrosarcoma cells were exposed to 2 Gy X-ray radiation in the presence of 100 µM 4-MU (22); this inhibited colony-forming ability and metastatic potential, which was accompanied by downregulation of matrix metalloproteinases-2 and −9. However, the mechanistic basis of these effects is unclear. We addressed this in the present study by performing mRNA profiling to identify factors related to the anti-tumor and -invasion effects of 4-MU in HT1080 cells.

Materials and methods

Reagents

4-MU was purchased from Nacalai Tesque (Kyoto, Japan) and diluted in dimethylsulfoxide (Wako Pure Chemical Industries, Ltd., Osaka, Japan); the working concentration was 500 µM. The reason for using 500 µM 4-MU was that clear effects of 4-MU could be observed and the cytotoxic effect was low for normal fibroblast cells at a 500 µM concentration (22). Monoclonal phycoerythin (PE)-conjugated anti-human cluster of differentiation (CD)126 antibody (cat. no. 352804); mouse monoclonal PE-IgG1, κ isotype control (cat. no. 400114); monoclonal allophycocyanin (APC)-conjugated anti-human CD130 (gp130) antibody (cat. no. 362005); mouse monoclonal APC-IgG2a, κ isotype control (cat. no. 400221); fluorescein isothiocyanate (FITC)-annexin V (cat. no. 640905); and propidium iodide (PI) (cat. no. 421301) were from BioLegend (San Diego, CA, USA). PE-conjugated polyclonal anti-human type 1 IL-1 receptor (IL-1R) antibody (cat. no. FAB269P) and PE-conjugated goat IgG (cat. no. IC108P) were R&D Systems, Inc., (Minneapolis, MN, USA).

Cell culture

HT1080 human fibrosarcoma cells from American Type Culture Collection (Manassas, VA, USA) were cultured in Roswell Park Memorial Institute 1640 medium (Thermo Fisher Scientific, Inc., Waltham, MA, USA) supplemented with 10% heat-inactivated fetal bovine serum (FBS; Japan Bio Serum, Fukuyama, Japan) and 1% penicillin/streptomycin (Life Technologies) at 37°C in a humidified atmosphere of 5% CO2.

Clonogenic potency assay

The clonogenic potency of HT1080 cells was estimated with the colony formation assay. Appropriate numbers of cells were seeded and incubated for 2 h, then subjected to X-ray irradiation at 1–6 Gy in the presence of 500 µM 4-MU followed by incubation for 24 h. After 7–10 days of culture with regular changes of medium cells were fixed with methanol (Wako Pure Chemical Industries, Ltd.) stained by Giemsa (Wako Pure Chemical Industries, Ltd.), and quantified.

Irradiation

Ionizing radiation (IR) was delivered using an X-ray generator (MBR-1520R-3; Hitachi Medical, Co., Tokyo, Japan) with 0.5-mm aluminum and 0.3-mm copper filters at a distance of 45 cm between the focus and target (150 kV, 20 mA, 1.0 Gy/min). During X-ray exposure, the total dose and dose rate were monitored with a thimble ionization chamber placed next to the sample.

Flow cytometry analysis

To evaluate the expression of the IL-6 receptors CD126 and CD130, and the IL-1α/β receptor type I IL-1R, cells were resuspended in 100 µl Dulbecco's phosphate-buffered saline without CaCl2 and MgCl (Takara Bio, Inc., Otsu, Japan) containing 5% FBS and PE-conjugated anti-human CD126 antibody (3 µl/106 cells), APC-conjugated anti-human CD130 antibody (3 µl/106 cells), and PE-conjugated anti-human type 1 IL-1R antibody for 15 min at 4°C in the dark. To detect cell death, cells labeled with FITC-annexin V (3 µl/106 cells) were resuspended in annexin V binding buffer (cat. no. 422201; BioLegend) and incubated for 15 min at room temperature in dark with PI (6 µl/106 cells), followed by flow cytometry analysis on FACS Aria instrument (BD Biosciences, Tokyo, Japan).

RNA extraction and analysis

Total RNA was extracted from HT1080 cells 24 h after irradiation and/or 4-MU administration using the RNeasy Mini kit (Qiagen GmbH, Hilden, Germany), and RNA quality was confirmed with an Agilent 2100 Bioanalyzer (Agilent Technologies, Inc., Santa Clara, CA, USA). Cyanine (Cy)3-labeled cRNA samples were synthesized from total RNA and hybridized to 8×60K format SurePrint G3 Human GE v2 microarray slides (eArray Design ID=039494) according to the manufacturer's instructions (Agilent Technologies, Inc.). Cy3 fluorescence was detected with a DNA microarray scanner (G2600A SureScan) and processed using Feature Extraction software (both from Agilent Technologies, Inc.).

Reverse transcription-quantitative polymerase chain reaction (RT-qPCR)

cDNA was synthesized using a RT kits (Applied Biosystems; Thermo Fisher Scientific, Inc.) and used as a template for PCR in a 20-µl reaction mixture containing 2X SYBR Premix Ex Taq (Takara Bio, Inc.) and 0.5 µM forward and reverse primers (Table I). The reaction was carried out on a real-time PCR system (StepOne Plus; Life Technologies) under the following conditions: 30 sec at 95°C, followed by 40 cycles of 95°C for 5 sec, and 54°C for 30 sec. Target gene expression levels were calculated relative to that of glyceraldehyde 3-phosphate dehydrogenase mRNA (internal control) with the comparative ΔΔCq method (23).

Table I.

Primer sequences of the target genes.

Table I.

Primer sequences of the target genes.

PrimersSequences (5′-3′)
IL-1α forward GGTTGAGTTTAAGCCAATCCA
IL-1α reverse TGCTGACCTAGGCTTGATGA
IL-1β forward TACCTGTCCTGCGTGTTGAA
IL-1β reverse TCTTTGGGTAATTTTTGGGATCT
IL-6 forward CACTGGGCACAGAACTTATGTTG
IL-6 reverse AAAATAATTAAAATAGTGTCCTAA CGCTCAT
GAPDH forward GTGAAGGTCGGAGTCAACG
GAPDH reverse TGAGGTCAATGAAGGGGTC

[i] IL, interleukin

Enzyme-linked immunosorbent assay (ELISA)

The levels of IL-1α, −1β, and −6 secreted by cells were measured using the Human IL-1α, IL-1β, and IL-6 DuoSet ELISA kits (all from R&D Systems), respectively, according to the manufacturer's protocols. Cells were treated with 500 µM 4-MU and irradiated with 2 Gy X ray, followed by incubation in serum-free medium. Culture medium conditioned for 24 h was used for the ELISA assay. Cytokine concentration was determined per million cells.

Statistical analysis

Statistical analysis of microarray data was performed using GeneSpring (Agilent Technologies, Inc.). Up- and downregulated mRNAs transcripts were selected based on fold change (>2-fold) of irradiated and/or 4-MU-administered samples relative to control samples. Ingenuity Pathway Analysis (IPA; Qiagen Silicon Valley, Redwood City, CA, USA) was used for functional analysis of each transcript. The significance of differences between control and experimental cultures was evaluated with one-way analysis of variance and the Tukey-Kramer test. Statistical analyses were performed using Microsoft Excel 2010 (Microsoft Corporation, Redmond, WA, USA) with the add-on software Statcel v3 (OMS Publishing, Saitama, Japan). P<0.05 was considered to indicate a statistically significant difference.

Results

Radiosensitization by 4-MU

To investigate whether the anti-inflammatory effect of 4-MU enhances radiosensitization, we evaluated the clonogenic potency of HT1080 cells exposed to 4-MU and/or X-ray radiation with the colony formation assay. The survival of cells treated with 4-MU combined with X-ray irradiation was decreased in a radiation dose-dependent manner compared to X-ray irradiation alone (Fig. 1A). This was confirmed by annexin V and PI staining. Representative dot plots and quantified data are shown in Fig. 1B and C. The number of cells positive for both annexin V and PI was increased 6 h after X-ray irradiation (9.12±0.52%) (Fig. 1C). A comparable increase was observed upon treatment with 4-MU alone for 6 h (10.13±0.03%). On the other hand, there were more annexin V and PI positive cells in the group exposed to both 4-MU and X-ray radiation as compared to either treatment alone (12.58±0.23%). These results suggest that 4-MU enhances the sensitivity of HT1080 cells to the lethal effects of X-ray radiation.

mRNA profiling and IPA

We performed a microarray analysis to determine the signaling pathways affected by 4-MU treatment combined with X-ray irradiation. A total of 2873, 4085, and 3179 genes were differentially expressed in cells treated with 4-MU alone, X-ray alone, and 4-MU + X-ray, respectively. The IPA z-scores revealed that signaling pathways associated with inflammation including IL-8 and −6 and Toll-like receptor signaling were altered by treatment with 4-MU or/and 2 Gy X-ray radiation (Fig. 2A). Moreover, IL-1α and −1β were inactivated in cells treated with 4-MU only (Fig. 2B).

IL-1α, IL-1β, and IL-6 mRNA levels in cells and IL-1α, IL-1β, and IL-6 concentrations in the cell culture supernatant were analyzed by RT-qPCR and ELISA, respectively. IL-1α and −1β transcript levels in cells exposed to X-ray radiation alone were increased about 2-fold relative to the control, whereas the levels in cells treated with 4-MU showed the opposite trend (Fig. 3A). However, IL-1α concentration in the supernatant was 3-fold higher in 4-MU treated as compared to control cultures (1.73±0.21 vs. 5.09±0.48 pg/ml), whereas IL-1α concentration (per 106 cells) in the culture supernatant of cells exposed to both 4-MU and X-ray radiation was higher than that in the radiation-only group (5.71±1.73 vs. 2.20±0.31) (Fig. 3B). On the other hand, the IL-1β concentration in the culture supernatant was correlated with the mRNA expression. X-ray irradiation increased IL-6 mRNA level 1.7-fold relative to the control (Fig. 3A); however, the level was decreased 0.8-fold by treatment with 4-MU combined with X-ray irradiation. IL-6 level in the culture supernatant of cells exposed to radiation alone was 2-fold higher than that in the control group (2.84±0.21 vs. 5.95±0.11 pg/ml) (Fig. 3B), whereas concentrations for cells treated with 4-MU with or without X-ray irradiation were similar to that of control cultures (2.05±0.36 and 2.39±0.21 pg/ml, respectively). This suggests that the increase in IL-6 expression and release induced by X-ray irradiation was reversed by 4-MU treatment which suppressed inflammation by inhibiting IL-1β production.

Expression of inflammatory cytokine receptors

To investigate whether IL-1α, −1β, and −6 receptors are suppressed by 4-MU, we examined the expression of the cognate surface receptors [type 1 IL-1R (CD121a), CD126, and CD130, respectively] by flow cytometry. The mean fluorescence intensity of CD121a on cells exposed to X-ray radiation alone was higher than that of control cells (59.88±18.3 vs. 74.29±10.3, respectively) (Fig. 4A and D). On the other hand, the mean fluorescence intensity of CD121a in the 4-MU only (55.22±4.38) and 4-MU/X-ray radiation combination (62.79±3.72) groups did not differ significantly from that of the control group (P=0.429, 0.268, respectively), suggesting that the increase in CD121a expression induced by X-ray irradiation was marginally suppressed.

The mean fluorescence intensity of CD126, the ligand binding protein for IL-6, was unaltered by X-ray irradiation relative to the control (74.48±5.74 vs. 96.17±14.1). 4-MU treatment reduced the signal intensity to about half that of the control group (4-MU alone: 56.54±6.12; 4-MU combined with X-ray: 52.11±3.71) (Fig. 4B and D). On the other hand, the mean fluorescence intensity of CD130, the signal transducer for IL-6 receptor, was downregulated by 2 Gy X-ray irradiation as compared to the control (260.40±11.4 vs. 295.76±1.46) and was further decreased by 4-MU administration (4-MU alone: 239.42±9.99; 4-MU combined: 249.57±13.5) (Fig. 4C and D). These results indicate that expression of IL-6 receptors were suppressed by 4-MU treatment.

Discussion

Recent some studies have described the anti-tumor and -invasion effects of 4-MU and the role of HAS in various malignancies (2426). HA is closely related to cancer cell proliferation, invasion, and metastasis (27). 4-MU was reported to inhibit activation of Akt signaling by modulating the interaction between HA, CD44, and the receptor for hyaluronan-mediated motility complex (15). On the other hand, angiogenesis was suppressed by downregulation the proinflammatory cytokines IL-6 and −8 and the chemokine C-X-C motif chemokine ligand 12 (17). In accordance with these findings, we found that the increase in IL-1β and −6 expression caused by X-ray irradiation was abolished by 4-MU treatment. However, whether this effect leads to radiosensitization effects, needs to be verified using neutralizing antibodies and gene knockdown experiments. IL-1α has been reported to consolidate cellular scaffolds in HT1080 cells (28). Liberation of cells by processes such as epithelial-mesenchymal transition is known to be the initiation of invasion into the blood vessel (29). Therefore, it was suggested that 4-MU prevents liberation of cells by promoting the release of IL-1α. However, conflicting IL-1α between mRNA and protein levels indicate that 4-MU might have the effect of stabilizing IL-1α. Since 4-MU has multiple functions, further verification is necessary.

Inflammatory cytokines have been linked to radioresistance in cancer stem cells. In particular, IL-6, an activator of the Janus kinase/signal transducer and activator of transcription 3 (STAT)3 signaling pathway, has been shown to inhibit the oxidative stress response (30) and DNA repair in non-small cell lung cancer cells (31,32) and promote cancer cell resistance to radio- and chemotherapy. On the other hand, IL-1β promotes angiogenesis and migration and is implicated in the tumorigenicity of fibrosarcoma cells (28). Blockade of IL-1 signaling overcame erlotinib resistance in head and neck squamous cell carcinoma (33). Activation of NF-κB, plays an important role in the release of IL-1β and −6 and TNFα, which triggers a positive feedback loop in the inflammatory response (34). NF-κB activation induces anti-apoptotic genes such as B cell lymphoma (Bcl)-2 and Bcl-extra-large leading to radio- and chemotherapeutic resistance (35). Thus, controlling the inflammatory cascade can potentially improve the outcome of radiotherapy. The expression of CD121a was not significantly among treatments indicating that 4-MU has no effect on the IL-1 receptor but suppresses only release of IL-1β. However, we found that expression of CD126 and CD130 on cell surface was suppressed in the presence of 4-MU, suggesting that the exchange of inflammatory response between adjacent cells causing radioresistance was indirectly suppressed through inhibition of IL-1β and IL-6 release, and the receptors of IL-6.

HA is synthesized by HAS and is cleaved by hyaluronidase or oxidative stress, yielding a lower molecular weight form (36) that activates macrophages and stimulates the production of proinflammatory cytokines such as IL-1β through the HA/CD44 interaction thereby enhancing proliferation and angiogenesis (37). IL-1β and TNFα induced the human manganese superoxide dismutase gene such as SOD2, which is an enzyme that degrades active oxygen generated in cells (38). X-ray irradiation has been shown to produce reactive oxygen species (ROS) or free radicals that indirectly and/or directly induce DNA strand breakage and exert cytotoxic effects (39,40). Radiosensitizing effect of 4-MU was suggested to be due to the accumulation of ROS.

In conclusion, 4-MU increased the sensitivity of HT1080 cells to X-ray radiation by inhibiting the production of the proinflammatory cytokines IL-1β and −6. Thus, the efficacy of radiotherapy can be enhanced by co-administration of 4-MU. Additional research is needed to determine whether 4-MU can also prevent distant metastasis of cancer cells; however, our results highlight the clinical potential of 4-MU as a radiosensitizing agent that can improve treatment outcome.

Acknowledgements

Not applicable.

Funding

The present study was supported by KAKENHI, Young Scientists (B) (grant no. 17K16413).

Availability of data and materials

The datasets used and/or analyzed during the present study are available from the corresponding author on reasonable request.

Authors' contributions

RS, TN, KO, ET and YH conceived the study, participated in its design and coordination. RS and KH drafted the manuscript. RS, KH, KM and MC performed the experiments, and analyzed and interpreted the data. TN, KO, ET and YH critically revised the manuscript for important intellectual content. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Lee SY, Jeong EK, Ju MK, Jeon HM, Kim MY, Kim CH, Park HG, Han SI and Kang HS: Induction of metastasis, cancer stem cell phenotype, and oncogenic metabolism in cancer cells by ionizing radiation. Mol Cancer. 16:102017. View Article : Google Scholar : PubMed/NCBI

2 

Hara T, Iwadate M, Tachibana K, Waguri S, Takenoshita S and Hamada N: Metastasis of breast cancer cells to the bone, lung, and lymph nodes promotes resistance to ionizing radiation. Strahlenther Onkol. 193:848–855. 2017. View Article : Google Scholar : PubMed/NCBI

3 

Kuwahara Y, Mori M, Kitahara S and Fukumoto M, Ezaki T, Mori S, Echigo S, Ohkubo Y and Fukumoto M: Targeting of tumor endothelial cells combining 2 Gy/day of X-ray with Everolimus is the effective modality for overcoming clinically relevant radioresistant tumors. Cancer Med. 3:310–321. 2014. View Article : Google Scholar : PubMed/NCBI

4 

Kim JS, Chang JW, Yun HS, Yang KM, Hong EH, Kim DH, Um HD, Lee KH, Lee SJ and Hwang SG: Chloride intracellular channel 1 identified using proteomic analysis plays an important role in the radiosensitivity of HEp-2 cells via reactive oxygen species production. Proteomics. 10:2589–2604. 2010. View Article : Google Scholar : PubMed/NCBI

5 

Hazawa M, Hosokawa Y, Monzen S, Yoshino H and Kashiwakura I: Regulation of DNA damage response and cell cycle in radiation-resistant HL60 myeloid leukemia cells. Oncol Rep. 28:55–61. 2012.PubMed/NCBI

6 

Arechaga-Ocampo E, Lopez-Camarillo C, Villegas-Sepulveda N, Gonzalez-De la Rosa CH, Perez-Añorve IX, Roldan-Perez R, Flores-Perez A, Peña-Curiel O, Angeles-Zaragoza O, Rangel Corona R, et al: Tumor suppressor miR-29c regulates radioresistance in lung cancer cells. Tumor Biol. 39:10104283176950102017. View Article : Google Scholar

7 

Zhou SB, Guo XW, Gu L and Ji SJ: Influential factors on radiotherapy efficacy and prognosis in patients with secondary lymph node metastasis after esophagectomy of thoracic esophageal squamous cell carcinoma. Cancer Manag Res. 10:217–225. 2018. View Article : Google Scholar : PubMed/NCBI

8 

Yang J, Xu H, Guo X, Zhang J, Ye X, Yang Y and Ma X: Pretreatment inflammatory indexes as prognostic predictors for survival in colorectal cancer patients receiving neoadjuvant chemoradiotherapy. Sci Rep. 8:30442018. View Article : Google Scholar : PubMed/NCBI

9 

Matsuda T, Sumi Y, Yamashita K, Hasegawa H, Yamamoto M, Matsuda Y, Kanaji S, Oshikiri T, Nakamura T, Suzuki S and Kakeji Y: Outcomes and prognostic factors of selective lateral pelvic lymph node dissection with preoperative chemoradiotherapy for locally advanced rectal cancer. Int J Colorectal Dis. 33:367–374. 2018. View Article : Google Scholar : PubMed/NCBI

10 

Xia Y, Li YH, Chen Y, Liu Q, Zhang JH, Deng JY, Ai TS, Zhu HT, Badakhshi H and Zhao KL: A phase II trial of concurrent chemoradiotherapy with weekly paclitaxel and carboplatin in advanced oesophageal carcinoma. Int J Clin Oncol. 23:458–465. 2018. View Article : Google Scholar : PubMed/NCBI

11 

Fangzheng W, Chuner J, Quanquan S, Zhimin Y, Tongxin L, Jiping L, Sakamoto M, Peng W, Kaiyuan S, Weifeng Q, et al: Addition of 5-fluorouracil to docetaxel/cisplatin does not improve survival in locoregionally advanced nasopharyngeal carcinoma. Oncotarget. 8:115469–115479. 2017. View Article : Google Scholar : PubMed/NCBI

12 

Kim HJ, Choi GS, Park JS, Park SY, Cho SH, Lee SJ, Kang BW and Kim JG: Optimal treatment strategies for clinically suspicious lateral pelvic lymph node metastasis in rectal cancer. Oncotarget. 8:100724–100733. 2017.PubMed/NCBI

13 

Nakamura T, Takagaki K, Shibata S, Tanaka K, Higuchi T and Endo M: Hyaluronic-acid-deficient extracellular matrix induced by addition of 4-methylumbelliferone to the medium of cultured human skin fibroblasts. Biochem Biophys Res Commun. 208:470–475. 1995. View Article : Google Scholar : PubMed/NCBI

14 

Kuroda Y, Kasai K, Nanashima N, Nozaka H, Nakano M, Chiba M, Yoneda M and Nakamura T: 4-Methylumbelliferone inhibits the phosphorylation of hyaluronan synthase 2 induced by 12-O-tetradecanoyl-phorbol-13-acetate. Biomed Res. 34:97–103. 2013. View Article : Google Scholar : PubMed/NCBI

15 

Yates TJ, Lopez LE, Lokeshwar SD, Ortiz N, Kallifatidis G, Jordan A, Hoye K, Altman N and Lokeshwar VB: Dietary supplement 4-methylumbelliferone: An effective chemopreventive and therapeutic agent for prostate cancer. J Natl Cancer Inst. 107(djv085)2015.PubMed/NCBI

16 

Piccioni F, Fiore E, Bayo J, Atorrasagasti C, Peixoto E, Rizzo M, Malvicini M, Tirado-González I, García MG, Alaniz L and Mazzolini G: 4-methylumbelliferone inhibits hepatocellular carcinoma growth by decreasing IL-6 production and angiogenesis. Glycobiology. 25:825–835. 2015. View Article : Google Scholar : PubMed/NCBI

17 

Li F, Hao P, Liu G, Wang W, Han R, Jiang Z and Li X: Effects of 4-methylumbelliferone and high molecular weight hyaluronic acid on the inflammation of corneal stromal cells induced by LPS. Graefes Arch Clin Exp Ophthalmol. 255:559–566. 2017. View Article : Google Scholar : PubMed/NCBI

18 

Piao L, Canguo Z, Wenjie L, Xiaoli C, Wenli S and Li L: Lipopolysaccharides-stimulated macrophage products enhance Withaferin A-induced apoptosis via activation of caspases and inhibition of NF-κB pathway in human cancer cells. Mol Immunol. 81:92–101. 2017. View Article : Google Scholar : PubMed/NCBI

19 

Yu H, Aravindan N, Xu J and Natarajan M: Inter- and intra-cellular mechanism of NF-kB-dependent survival advantage and clonal expansion of radio-resistant cancer cells. Cell Signal. 31:105–111. 2017. View Article : Google Scholar : PubMed/NCBI

20 

Mochizuki D, Adams A, Warner KA, Zhang Z, Pearson AT, Misawa K, McLean SA, Wolf GT and Nör JE: Anti-tumor effect of inhibition of IL-6 signaling in mucoepidermoid carcinoma. Oncotarget. 6:22822–22835. 2015. View Article : Google Scholar : PubMed/NCBI

21 

Lee SO, Yang X, Duan S, Tsai Y, Strojny LR, Keng P and Chen Y: IL-6 promotes growth and epithelial-mesenchymal transition of CD133+ cells of non-small cell lung cancer. Oncotarget. 7:6626–6638. 2016.PubMed/NCBI

22 

Saga R, Monzen S, Chiba M, Yoshino H, Nakamura T and Hosokawa Y: Anti-tumor and anti-invasion effects of a combination of 4-methylumbelliferone and ionizing radiation in human fibrosarcoma cells. Oncol Lett. 13:410–416. 2017. View Article : Google Scholar : PubMed/NCBI

23 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

24 

Lokeshwar VB, Lopez LE, Munoz D, Chi A, Shirodkar SP, Lokeshwar SD, Escudero DO, Dhir N and Altman N: Antitumor activity of hyaluronic acid synthesis inhibitor 4-methylumbelliferone in prostate cancer cells. Cancer Res. 70:2613–2623. 2010. View Article : Google Scholar : PubMed/NCBI

25 

Arai E, Nishida Y, Wasa J, Urakawa H, Zhuo L, Kimata K, Kozawa E, Futamura N and Ishiguro N: Inhibition of hyaluronan retention by 4-methylumbelliferone suppresses osteosarcoma cells in vitro and lung metastasis in vivo. Br J Cancer. 105:1839–1849. 2011. View Article : Google Scholar : PubMed/NCBI

26 

Urakawa H, Nishida Y, Wasa J, Arai E, Zhuo L, Kimata K, Kozawa E, Futamura N and Ishiguro N: Inhibition of hyaluronan synthesis in breast cancer cells by 4-methylumbelliferone suppresses tumorigenicity in vitro and metastatic lesions of bone in vivo. Int J Cancer. 130:454–466. 2012. View Article : Google Scholar : PubMed/NCBI

27 

Li Y, Li L, Brown TJ and Heldin P: Silencing of hyaluronan synthase 2 suppresses the malignant phenotype of invasion breast cancer cells. Int J Cancer. 120:2557–2567. 2007. View Article : Google Scholar : PubMed/NCBI

28 

Nazarenko I, Marhaba R, Reich E, Voronov E, Vitacolonna M, Hildebrand D, Elter E, Rajasagi M, Apte RN and Zöller M: Tumorigenicity of IL-1alpha- and IL-1beta-deficient fibrosarcoma cells. Neoplasia. 10:549–562. 2008. View Article : Google Scholar : PubMed/NCBI

29 

Sun Y and Ma L: The emerging molecular machinery and therapeutic targets of metastasis. Trends Pharmacol Sci. 36:349–359. 2015. View Article : Google Scholar : PubMed/NCBI

30 

Tamari Y, Kashino G and Mori H: Acquisition of radioresistance by IL-6 treatment is caused by suppression of oxidative stress derived from mitochondria after γ-irradiation. J Radiat Res. 58:412–420. 2017. View Article : Google Scholar : PubMed/NCBI

31 

Chen Y, Zhang F, Tsai Y, Yang X, Yang L, Duan S, Wang X, Keng P and Lee SO: IL-6 signaling promotes DNA repair and prevents apoptosis in CD133+ stem-like cells of lung cancer after radiation. Radiat Oncol. 10:2272015. View Article : Google Scholar : PubMed/NCBI

32 

Duan S, Tsai Y, Keng P and Chen Y, Lee SO and Chen Y: IL-6 signaling contributes to cisplatin resistance in non-small cell lung cancer via the up-regulation of anti-apoptotic and DNA repair associated molecules. Oncotarget. 6:27651–27660. 2015. View Article : Google Scholar : PubMed/NCBI

33 

Stanam A, Gibson-Corley KN, Love-Homan L, Ihejirika N and Simons AL: Interleukin-1 blockade overcomes erlotinib resistance in head and neck squamous cell carcinoma. Oncotarget. 7:76087–76100. 2016. View Article : Google Scholar : PubMed/NCBI

34 

Grivennikov SI and Karin M: Dangerous liaisons: STAT3 and NF-kappaB collaboration and crosstalk in cancer. Cytokine Growth Factor Rev. 21:11–19. 2010. View Article : Google Scholar : PubMed/NCBI

35 

Hei TK, Zhou H, Chai Y, Ponnaiya B and Ivanov VN: Radiation induced non-targeted response: Mechanism and potential clinical implications. Curr Mol Pharmacol. 4:96–105. 2011. View Article : Google Scholar : PubMed/NCBI

36 

Nagy N, Kuipers HF, Frymoyer AR, Ishak HD, Bollyky JB, Wight TN and Bollyky PL: 4-methylumbelliferone treatment and hyaluronan inhibition as a therapeutic strategy in inflammation, autoimmunity, and cancer. Front Immunol. 6:1232015. View Article : Google Scholar : PubMed/NCBI

37 

Hauser-Kawaguchi A, Luyt LG and Turley E: Design of peptide mimetics to block pro-inflammatory functions of HA fragments. Matrix Biol. S0945(053X): 30444–30454. 2018.

38 

Xu Y, Kiningham KK, Devalaraja MN, Yeh CC, Majima H, Kasarskis EJ and St Clair DK: An intronic NF-kappaB element is essential for induction of the human manganese superoxide dismutase gene by tumor necrosis factor-alpha and interleukin-1beta. DNA Cell Biol. 18:709–722. 1999. View Article : Google Scholar : PubMed/NCBI

39 

Bajinskis A, Natarajan AT, Erixon K and Harms-Ringdahl M: DNA double strand breaks induced by the indirect effect of radiation are more efficiently repaired by non-homologous end joining compared to homologous recombination repair. Mutat Res. 756:21–29. 2013. View Article : Google Scholar : PubMed/NCBI

40 

Vignard J, Mirey G and Salles B: Ionizing-radiation induced DNA double-strand breaks: A direct and indirect lighting up. Radiother Oncol. 108:362–369. 2013. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

March-2019
Volume 17 Issue 3

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Saga R, Hasegawa K, Murata K, Chiba M, Nakamura T, Okumura K, Tsuruga E and Hosokawa Y: Regulation of radiosensitivity by 4‑methylumbelliferone via the suppression of interleukin‑1 in fibrosarcoma cells. Oncol Lett 17: 3555-3561, 2019
APA
Saga, R., Hasegawa, K., Murata, K., Chiba, M., Nakamura, T., Okumura, K. ... Hosokawa, Y. (2019). Regulation of radiosensitivity by 4‑methylumbelliferone via the suppression of interleukin‑1 in fibrosarcoma cells. Oncology Letters, 17, 3555-3561. https://doi.org/10.3892/ol.2019.9990
MLA
Saga, R., Hasegawa, K., Murata, K., Chiba, M., Nakamura, T., Okumura, K., Tsuruga, E., Hosokawa, Y."Regulation of radiosensitivity by 4‑methylumbelliferone via the suppression of interleukin‑1 in fibrosarcoma cells". Oncology Letters 17.3 (2019): 3555-3561.
Chicago
Saga, R., Hasegawa, K., Murata, K., Chiba, M., Nakamura, T., Okumura, K., Tsuruga, E., Hosokawa, Y."Regulation of radiosensitivity by 4‑methylumbelliferone via the suppression of interleukin‑1 in fibrosarcoma cells". Oncology Letters 17, no. 3 (2019): 3555-3561. https://doi.org/10.3892/ol.2019.9990