International Journal of Molecular Medicine is an international journal devoted to molecular mechanisms of human disease.
International Journal of Oncology is an international journal devoted to oncology research and cancer treatment.
Covers molecular medicine topics such as pharmacology, pathology, genetics, neuroscience, infectious diseases, molecular cardiology, and molecular surgery.
Oncology Reports is an international journal devoted to fundamental and applied research in Oncology.
Experimental and Therapeutic Medicine is an international journal devoted to laboratory and clinical medicine.
Oncology Letters is an international journal devoted to Experimental and Clinical Oncology.
Explores a wide range of biological and medical fields, including pharmacology, genetics, microbiology, neuroscience, and molecular cardiology.
International journal addressing all aspects of oncology research, from tumorigenesis and oncogenes to chemotherapy and metastasis.
Multidisciplinary open-access journal spanning biochemistry, genetics, neuroscience, environmental health, and synthetic biology.
Open-access journal combining biochemistry, pharmacology, immunology, and genetics to advance health through functional nutrition.
Publishes open-access research on using epigenetics to advance understanding and treatment of human disease.
An International Open Access Journal Devoted to General Medicine.
Colorectal cancer (CRC) is the third most common cancer and the second leading cause of cancer-related mortality worldwide (1). Surgery, chemotherapy and radiotherapy are three primary modalities in CRC treatment; however, several alternative agents, including vitexin, nitric oxide and sodium selenite, have been reported to inhibit tumor progression and increase sensitivity to chemoradiotherapy in CRC (2–4). This indicates that alternative therapies could be employed to augment the efficacy of primary treatments and even achieve tumor suppressive purposes independently.
Hydrogen is the most abundant element in the universe. Deuterium and protium are two stable isotopes of hydrogen, which exhibit differences not only in physical and chemical properties but also in biological functions (5). The concentration of deuterium in natural water is ~0.015% [150 parts per million (ppm)] (6). Deuterium-depleted water (DDW) is defined as water with a deuterium concentration of <150 ppm, and it was reported to have an inhibitory effect on xenotransplanted tumor growth in mice for the first time in the 1990s (7). Subsequently, studies reported the effects of DDW in several areas, including in cancer suppression (8), anti-senescence (9), insulin sensitization (10) and neuroprotection (11). However, the functions of DDW in CRC treatment remain unclear.
Oxidative stress has been reported to serve a major role in CRC progression (12). Furthermore, the inhibition of reactive oxygen species (ROS) production is considered a critical signaling mechanism of the DDW anticancer function (13,14). Forkhead box (Fox)M1, a member of the FOX transcription factor family, is recognized as a master regulator of cancer growth, stemness and metastasis in CRC (15). Previous studies have reported crosstalk between ROS production and FoxM1 expression (16,17); however, whether the FoxM1 signaling pathway can be blocked by DDW, and the role of ROS in this process, remain unknown.
Therefore, the objective of this study was to systematically investigate the impact of DDW on malignant biological behaviors and potential molecular mechanisms in CRC cells cultured in vitro, specifically focusing on its effects on cell proliferation, stemness, migration, invasion and regulation of the ROS/FoxM1 signaling pathway. The results of the present study may facilitate the clinical application of DDW for CRC adjuvant therapy in the future.
The DDW (deuterium %=25 ppm) used in the present study was provided by HYD LLC. Regular ddH2O or DDW was used to dissolve RPMI-1640 medium powder (Gibco; Thermo Fisher Scientific, Inc.). A total of two CRC cell lines, HT-29 (cat. no. STCC10801G) and DLD-1 (cat. no. STCC10810G), were purchased from Wuhan Servicebio Technology Co., Ltd. and authenticated by STR profiling. Cells were cultured in conditioned RPMI-1640 medium supplemented with 10% fetal bovine serum (FBS, Gibco; Thermo Fisher Scientific, Inc.) and 1% penicillin/streptomycin solution (Sigma-Aldrich; Merck KGaA) at 37°C in a humidified atmosphere containing 5% CO2. The control group contained cells that were cultured in medium prepared with regular ddH2O, whereas the cells in the DDW group were cultured in medium prepared with DDW.
The EdU assay was performed using a commercial kit (cat. no. C10310-1; Guangzhou RiboBio Co., Ltd.). HT-29 and DLD-1 cells were seeded into 6-well plates at a density of 1×105 cells per well. After culturing in serum-free medium for 72 h, cells were incubated with 0.1% EdU solution for 3 h at 37°C, then fixed with 4% paraformaldehyde (PFA) for 30 min at room temperature. Cells were then incubated with appollo staining complex for 30 min at room temperature to react with EdU, followed by incubation with Hoechst 33342 staining reagent for 30 min at room temperature to label the cell nuclei. Fluorescent staining was recorded using the Operetta CLS high-content analysis system (PerkinElmer, Inc.). A total of five visual fields were randomly selected and the number of EdU positive cells was assessed using ImageJ software (version 1.53 K; National Institutes of Health). The following formula was used to determine the percentage of EdU positive cells: EdU positive cells (%)=(EdU positive cells/total cells) ×100%.
HT-29 and DLD-1 cells were seeded into 6-well plates at a density of 1×103 cells per well and cultured with conditioned medium. The medium was changed every 3 days. After 10 days, the cells were fixed with 4% PFA for 30 min at room temperature and then stained with 0.1% crystal violet for 20 min at room temperature. The colonies consisting of at least 40 cells were counted under an inverted light microscope (ECLIPSE Ts2; Nikon Corporation) using ImageJ software.
DMEM powder and F-12 medium powder (both from Gibco; Thermo Fisher Scientific, Inc.) were dissolved with regular ddH2O or DDW. Both media were mixed at a ratio of 1:1 to prepare the DMEM/F-12 medium. HT-29 and DLD-1 cells were seeded into ultra-low attachment 24-well plates (Corning, Inc.) at a density of 1×103 cells per well and cultured in DMEM/F12 medium supplemented with 1% B27, 20 ng/ml human FGF and 20 ng/ml human EGF (Invitrogen™; Thermo Fisher Scientific, Inc). Fresh medium was added every 3 days. After 2 weeks, cell spheres were counted under an inverted light microscope (ECLIPSE Ts2; Nikon Corporation), and the sphere formation efficiency (SFE) was calculated using the following formula: SFE (%)=number of spheres/1,000 ×100%.
HT-29 and DLD-1 cells were cultured in 6-well plates until they formed confluent monolayers. A gap was created across the middle of the well using a 200 µl sterile tip. The cells were then cultured in serum-free medium for an additional 48 h. An inverted light microscope (ECLIPSE Ts2; Nikon Corporation) was used to capture images both before and after the 48-h incubation period to evaluate the migration rate. Experiments were repeated in quintuplicate. A visual field was randomly selected, and the same field was imaged for every time point. The area of blank space was measured using ImageJ software. The following formula was used to assess the wound healed ratio: Wound healed ratio (%)=[1-(area48 h/area0 h)] ×100%.
HT-29 and DLD-1 cells were suspended in serum-free medium. The inner membranes of Transwell inserts (cat. no. 3464; Corning, Inc.) were coated with 10% Matrigel (cat. no. 354234; Corning, Inc.), followed by the seeding of 2×104 cells with 200 µl serum-free DMEM into each upper chamber. A total of 750 µl DMEM supplemented with 10% FBS was added to each lower chamber of the 24-well plate. After incubating for 24 h at 37°C, the cells were fixed with 4% PFA for 30 min at room temperature and then stained with 0.1% crystal violet for 20 min at room temperature. Cells that successfully invaded the bottom side of membrane were visualized using an inverted light microscope (ECLIPSE Ts2; Nikon Corporation).
Intracellular ROS levels were assessed using the 2′,7′-dichlorodihydrofluorescein diacetate (DCFH-DA) method. HT-29 and DLD-1 cells were incubated with the DCFH-DA probes (Sigma-Aldrich; Merck KGaA) for 30 min at 37°C and then treated with H2O2 (50 µM) for 30 min at 37°C. Cells were then fixed with 4% PFA for 30 min at room temperature. The fluorescence intensity of DCF was recorded using the Operetta CLS High-Content Analysis System (PerkinElmer, Inc.).
Total RNA was isolated using TRIzol™ reagent (Invitrogen; Thermo Fisher Scientific, Inc.) following the manufacturer's instructions. RT-qPCR was performed using the SuperScript™ III Platinum™ One-Step qRT-PCR Kit (cat. no. 11732020; Invitrogen; Thermo Fisher Scientific, Inc.). The sequences of the primers used for qPCR were as follows: FoxM1 forward, 5′-ACGTCCCCAAGCCAGGCTC-3′ and reverse, 5′-CTACTGTAGCTCAGGAATAA-3′; cyclin D1 (CCND1) forward, 5′-CGCTTCCTGTCGCTGGAGCC-3′ and reverse, 5′-CTTCTCGGCCGTCAGGGGGA-3′; matrix metalloproteinase 9 (MMP9) forward, 5′-TTCTGCCCGGACCAAGGATA-3′ and reverse, 5′-ACATAGGGTACATGAGCGCC-3′; and β-actin (reference gene) forward, 5′-CATGTACGTTGCTATCCAGGC-3′ and reverse, 5′-CTCCTTAATGTCACGCACGAT-3′. The thermocycling conditions were as follows: 50°C for 15 min and 95°C for 2 min, and then 40 cycles of 95°C for 15 sec and 60°C for 30 sec. The relative gene expression was calculated using the 2−∆∆Cq method (18).
FoxM1-overexpressing (OE-FoxM1) and negative control (OE-NC) lentiviral particles (1×108 TU/ml) were purchased from Shanghai GeneChem Co., Ltd., both originating from the same plasmid backbone (Ubi-MCS-3FLAG-SV40-puromycin). HT-29 and DLD-1 cells were seeded into 6-well plates at a density of 1×105 cells per well and incubated overnight. HT-29 and DLD-1 cells were transduced with the specific lentivirus at a multiplicity of infection of 10. After a 24-h incubation, fresh medium was changed to terminate the transduction. Stable infected cells were selected by puromycin (5 µg/ml) incubation for 48 h, and the surviving cells were continuously maintained in fresh DMEM with 10% FBS and 1.25 µg/ml puromycin for passaging. FoxM1 expression in the recombinant cells was validated by RT-qPCR and western blotting before subsequent experiments.
Total proteins from HT-29 and DLD-1 cells were extracted using RIPA lysis buffer (cat. no. P0013B; Beyotime Institute of Biotechnology) and quantified using the BCA Protein Assay Kit (cat. no. P0012; Beyotime Institute of Biotechnology). Proteins (40 µg per lane) were separated by 4–20% SDS-PAGE (cat. no. P0468S; Beyotime Institute of Biotechnology) and subsequently transferred onto PVDF membranes (MilliporeSigma). The membranes were blocked with 5% fat-free milk (Bio-Rad Laboratories, Inc.) for 1 h at room temperature and incubated with primary antibodies (1:1,000 dilution by PBS) against β-actin (cat. no. 66009-1-Ig), octamer-binding transcription factor-4 (OCT-4; cat. no. 11263-1-AP), Nanog (cat. no. 14295-1-AP), MMP-9 (cat. no. 10375-2-AP), CCND1 (cat. no. 26939-1-AP) and FoxM1 (cat. no. 13147-1-AP) (all Proteintech Group, Inc.) at 4°C overnight. Protein signals were labeled with HRP-conjugated secondary antibodies (1:5,000 dilution by PBS; cat. nos. SA00001-1 or SA00001-2; Proteintech Group, Inc.) for 1 h at room temperature and visualized using an ECL kit (cat. no. WBKLS0500; MilliporeSigma). The densitometry was measured using ImageJ software.
Continuous data are expressed as mean ± standard deviation. Differences between two groups were analyzed using the unpaired Student's t-test or Mann-Whitney U test as appropriate. One-way ANOVA was used to compare the mean differences among three or more groups, and the least significant difference test was used as a post-hoc test. The data analysis for this paper was generated using GraphPad Prism 10 software (Dotmatics). P<0.05 was considered to indicate a statistically significant difference.
To assess the biological functions of DDW in CRC malignancy, HT-29 and DLD-1 cells were cultured in conditioned medium prepared with regular ddH2O or DDW. It was demonstrated that, compared with the control (regular ddH2O) treatment, DDW treatment reduced the proportion of EdU-positive cells (Fig. 1A) and the number of colonies formed (Fig. 1B). These results indicated that DDW could inhibit the proliferation of CRC cells in vitro. Notably, the results also revealed that DDW treatment impaired the sphere formation efficiency of HT-29 and DLD-1 cells (Fig. 1C). This finding implied that DDW might interfere with the stem cell-like phenotype of CRC cells. Therefore, the expression levels of two typical stemness markers were analyzed using western blotting. The findings demonstrated that, compared with the control group, the protein levels of Nanog and OCT-4 were significantly downregulated in the DDW treatment group (Fig. 1D). These results indicated that DDW treatment impaired the self-renewal ability of HT-29 and DLD-1 cells.
Subsequently, wound healing and Transwell assays were performed to evaluate the effect of DDW on cancer cell mobility. Compared with the control treatment, the migrated distances of HT-29 and DLD-1 cells were significantly reduced when treated with DDW (Fig. 2A and C). Additionally, there was a notable reduction in the number of invaded cells for both the HT-29 and DLD-1 cells treated by DDW in the Transwell assays (Fig. 2B and D). These findings suggested that DDW treatment is associated with a significant reduction in CRC cell migration and invasion.
To determine whether the ROS/FoxM1 axis was involved in the aforementioned findings, ROS production in response to DDW treatment was evaluated in the CRC cells. The findings revealed that, compared with the control treatment, DDW treatment significantly reduced the ROS levels in the HT-29 and DLD-1 cell lines (Fig. 3A). Subsequently, RT-qPCR and western blotting were used to evaluate FoxM1 expression. Compared with the control treatment, DDW treatment significantly reduced the mRNA and protein levels of FoxM1 (Fig. 3B and C). Moreover, the expression levels of CCND1 and MMP9, two typical downstream targets of FoxM1 (19,20), were lower in HT-29 and DLD-1 cells treated with DDW, compared with the control treatment (Fig. 3B and C). Furthermore, H2O2 was utilized to induce intracellular ROS production. Compared with the cells treated by DDW alone, additional treatment with H2O2 not only blocked the antioxidant effect of DDW (Fig. 3A) but also rescued the expression of FoxM1 (Fig. 3D). These results indicated that DDW treatment downregulated FoxM1 expression possibly through inhibiting ROS production.
To assess whether the anticancer effects of DDW were dependent on the inhibition of FoxM1, FoxM1 was overexpressed in HT-29 and DLD-1 cells using lentiviral transduction. The mRNA and protein expression levels were significantly upregulated in OE-FoxM1 cells compared with the OE-NC cells (Fig. 4). Furthermore, compared with the OE-NC lentiviral transduction, the inhibitory effects of DDW on the proliferation (Fig. 5A and B), stemness (Fig. 5C and D), migration (Fig. 6A) and invasion (Fig. 6B) of HT-29 and DLD-1 cells were significantly abrogated by OE-FoxM1 lentiviral transduction. In addition, overexpression of FoxM1 also rescued the expression levels of CCND1 and MMP-9 in CRC cells treated with DDW, as demonstrated using RT-qPCR (Fig. 7A and C) and western blotting (Fig. 7B and D). These results validated that DDW may exert its tumor suppressive effects through inhibiting FoxM1 expression.
Water is the most common compound on Earth, but its isotopic composition is heterogeneous due to kinetic isotope fractionation (21). Given that the variation of the levels of deuterium in different terrestrial water sources is minimal, it is difficult to observe significant differences in biological effects among populations consuming natural water with varying deuterium concentrations. Artificial DDW can be prepared by desalination, distillation and catalytic exchange (22). The deuterium-content of DDW used in translational and clinical studies ranges from 25 to 125 ppm; however, Yavari et al (23) reported that the synergistic anticancer effects of DDW combined with 5-fluorouracil gradually faded with the increase of deuterium content from 30 to 150 ppm. This indicated that a dose-dependent inhibition of cancer progression may exist in DDW-treated cells. Therefore, the present study used 25 ppm DDW to treat HT-29 and DLD-1 cells.
To the best of our knowledge, the present study is the first to demonstrate that DDW exerts anticancer effects through inhibiting the ROS/FoxM1 signaling pathway in CRC cells. This finding was demonstrated by the following results: i) DDW treatment inhibited cell proliferation, stemness, migration and invasion in vitro; ii) DDW treatment decreased ROS production as well as the expression of FoxM1 and its downstream targets, CCND1 and MMP-9; iii) the induction of ROS partly rescued FoxM1 expression in DDW-treated CRC cells; and iv) overexpression of FoxM1 abrogated DDW-mediated tumor suppressive effects.
DDW has been shown to suppress the proliferation of breast cancer cells by arresting them at the G0 or G1 phases (23). Notably, the results of the present study revealed that DDW treatment decreased the expression of CCND1 in CRC cells. As a critical mediator of cell cycle progression, CCND1 is responsible for transiting cells from the G1 to S phase (24). The results of the present study also demonstrated that DDW inhibited cell migration, cell invasion and MMP-9 expression, which is in accordance with the results in nasopharyngeal carcinoma cells reported by Wang et al (25). Moreover, a growing body of research has reported promising outcomes regarding the clinical application of DDW. Concomitant administration of DDW with conventional therapies has been demonstrated to achieve improved overall survival rates compared with the use of chemoradiotherapy alone in patients with advanced-stage glioblastoma multiforme (26) and lung cancer (27). Furthermore, ~30% of patients with CRC who receive initial curative treatment typically develop tumor relapse (28). However, a retrospective analysis reported that consumption of DDW markedly reduced this rate to 18.5% (8), supporting the strong tumor-prevention effect of DDW.
Although overproduction of ROS induced by chemoradiotherapy leads to the death of cancer cells, slightly elevated ROS levels resulting from intracellular hypermetabolism facilitate tumor progression by activating pro-survival transcription factors including hypoxia-inducible factor 1α (HIF-1α), activator protein-1 and NF-κB (29). The present study demonstrated that manipulation of the degree of ROS production could disrupt the inhibitory efficacy of DDW toward the expression of FoxM1, and the overexpression of FoxM1 counteracted the tumor-inhibiting effects of DDW. These results indicate that ROS-induced FoxM1 could be the target of DDW in CRC treatment. However, the molecular mechanisms by which ROS upregulates FOXM1 are complex. Park et al (30) reported that endogenous ROS were constantly generated when cancer cells were cultured in favorable conditions with sufficient energy metabolism and oxygen supply, causing the upregulation of FoxM1 to enhance the rates of proliferation and metastasis. This viewpoint was corroborated by the finding that ROS-induced upregulation of FoxM1 promotes aerobic glycolysis in glioblastoma (31). Moreover, ROS have also been demonstrated to activate the FoxM1 promoter and induce its expression in a HIF-1α-dependent manner (32).
However, some limitations of the present study should be noted. First, the detailed mechanisms by which DDW inhibits ROS production remain unclear. Future research will focus on mitochondrial-related biological changes to explore whether DDW inhibits the production of ROS by disrupting mitochondrial function. Second, the in vivo anticancer effects of DDW were not evaluated in the present study. Animal experiments and population-based randomized controlled clinical trials will be introduced to further demonstrate the therapeutic effects of DDW in colorectal cancer.
In summary, the findings of the present study suggest that DDW has multiple anticancer effects in CRC cells, and DDW-mediated ROS/FoxM1 signaling inactivation could be a possible pathway in these effects. Future studies should focus on the clinical application of DDW as a promising alternative therapeutic agent in CRC.
Not applicable.
This study was supported by grants from the Beijing Hospitals Authority Youth Programme (grant no. QML20210906) and the Natural Science Basic Research Program of Shaanxi (grant no. 2021JQ-397).
The data generated in the present study may be requested from the corresponding author.
CL and YJ conceptualized the present study. CL wrote the original draft of the manuscript. CL and XC contributed to the experiments. CL, XC and YJ analyzed the data. All authors read and approved the final version of the manuscript. CL and YJ confirm the authenticity of all the raw data.
Not applicable.
Not applicable.
The authors declare that they have no competing interests.
|
Bray F, Laversanne M, Sung H, Ferlay J, Siegel RL, Soerjomataram I and Jemal A: Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 74:229–263. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Chen Y, Liang J, Chen S, Lin N, Xu S, Miao J, Zhang J, Chen C, Yuan X, Xie Z, et al: Discovery of vitexin as a novel VDR agonist that mitigates the transition from chronic intestinal inflammation to colorectal cancer. Mol Cancer. 23:1962024. View Article : Google Scholar : PubMed/NCBI | |
|
Zou Y, He Y, Tan L, Xu X, Qi C and Zhang Y: Discovery of cytotoxic nitric oxide-releasing piperlongumine derivatives targeting Wnt/β-catenin in colon cancer cells. J Nat Prod. 87:1893–1902. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Wang H, Yang X, Zhang Z and Xu H: Both calcium and ROS as common signals mediate Na(2)SeO(3)-induced apoptosis in SW480 human colonic carcinoma cells. J Inorg Biochem. 97:221–230. 2003. View Article : Google Scholar : PubMed/NCBI | |
|
Dunlap MK, Kringle L, Kay BD and Kimmel GA: Proton diffusion and hydrogen/deuterium exchange in amorphous solid water at temperatures from 114 to 134 K. J Chem Phys. 161:2445042024. View Article : Google Scholar : PubMed/NCBI | |
|
Korchinsky N, Davis AM and Boros LG: Nutritional deuterium depletion and health: A scoping review. Metabolomics. 20:1172024. View Article : Google Scholar : PubMed/NCBI | |
|
Somlyai G, Jancsó G, Jákli G, Vass K, Barna B, Lakics V and Gaál T: Naturally occurring deuterium is essential for the normal growth rate of cells. FEBS Lett. 317:1–4. 1993. View Article : Google Scholar : PubMed/NCBI | |
|
Kovács BZ, Puskás LG, Nagy LI, Papp A, Gyöngyi Z, Fórizs I, Czuppon G, Somlyai I and Somlyai G: Blocking the increase of intracellular deuterium concentration prevents the expression of cancer-related genes, tumor development, and tumor recurrence in cancer patients. Cancer Control. 29:107327482110689632022. View Article : Google Scholar : PubMed/NCBI | |
|
Avila DS, Somlyai G, Somlyai I and Aschner M: Anti-aging effects of deuterium depletion on Mn-induced toxicity in a C. elegans model. Toxicol Lett. 211:319–324. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Kondo M, Sawada K, Matsuda Y, Abe M, Sanechika N, Takanashi Y, Mori Y, Kimura M and Toyoda M: Study of the effects of deuterium-depleted water on the expression of GLUT4 and insulin resistance in the muscle cell line C2C12. Biomedicines. 12:17712024. View Article : Google Scholar : PubMed/NCBI | |
|
Wu Y, Qin D, Yang H, Wang W, Xiao J, Zhou L and Fu H: Neuroprotective effects of deuterium-depleted water (DDW) against H2O2-induced oxidative stress in differentiated pc12 cells through the PI3K/Akt signaling pathway. Neurochem Res. 45:1034–1044. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Lin S, Li Y, Zamyatnin AA, Werner J and Bazhin AV: Reactive oxygen species and colorectal cancer. J Cell Physiol. 233:5119–5132. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Bayrak BB, Kulak GY, Yanardag R and Yarat A: Short term deuterium depletion in drinking water reduced tumor induced oxidative stress in mice liver. Pathol Res Pract. 240:1541862022. View Article : Google Scholar : PubMed/NCBI | |
|
Fatemi F, Golbodagh A, Hojihosseini R, Dadkhah A, Akbarzadeh K, Dini S and Malayeri MRM: Anti-inflammatory effects of deuterium-depleted water plus Rosa Damascena Mill. essential oil via cyclooxygenase-2 pathway in rats. Turk J Pharm Sci. 17:99–107. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Laissue P: The forkhead-box family of transcription factors: Key molecular players in colorectal cancer pathogenesis. Mol Cancer. 18:52019. View Article : Google Scholar : PubMed/NCBI | |
|
Wu G, Yang L, Xu Y, Jiang X, Jiang X, Huang L, Mao L and Cai S: FABP4 induces asthmatic airway epithelial barrier dysfunction via ROS-activated FoxM1. Biochem Biophys Res Commun. 495:1432–1439. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Sun L, Wang Y, Wang L, Yao B, Chen T, Li Q, Liu Z, Liu R, Niu Y, Song T, et al: Resolvin D1 prevents epithelial-mesenchymal transition and reduces the stemness features of hepatocellular carcinoma by inhibiting paracrine of cancer-associated fibroblast-derived COMP. J Exp Clin Cancer Res. 38:1702019. View Article : Google Scholar : PubMed/NCBI | |
|
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 | |
|
Zhao L, Liu L, Dong Z and Xiong J: miR-149 suppresses human non-small cell lung cancer growth and metastasis by inhibiting the FOXM1/cyclin D1/MMP2 axis. Oncol Rep. 38:3522–3530. 2017.PubMed/NCBI | |
|
Jin H, Li XJ, Park MH and Kim SM: FOXM1-mediated downregulation of uPA and MMP9 by 3,3′-diindolylmethane inhibits migration and invasion of human colorectal cancer cells. Oncol Rep. 33:3171–3177. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
de Szoeke SP, Sarkar M, Quiñones Meléndez E, Blossey PN and Noone D: A simple model for the evaporation of hydrometeors and their isotopes. J Geophys Res Atmos. 129:e2024JD0411262024. View Article : Google Scholar : PubMed/NCBI | |
|
Huang F and Meng C: Method for the production of deuterium-depleted potable water. Ind Eng Chem Res. 50:378–381. 2011. View Article : Google Scholar | |
|
Yavari K and Kooshesh L: Deuterium depleted water inhibits the proliferation of human MCF7 breast cancer cell lines by inducing cell cycle arrest. Nutr Cancer. 71:1019–1029. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Wang W and Medeiros LJ: Utility of cyclin D1 in the diagnostic workup of hematopoietic neoplasms: What can cyclin D1 Do for Us? Adv Anat Pathol. 26:281–291. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Wang H, Zhu B, He Z, Fu H, Dai Z, Huang G, Li B, Qin D, Zhang X, Tian L, et al: Deuterium-depleted water (DDW) inhibits the proliferation and migration of nasopharyngeal carcinoma cells in vitro. Biomed Pharmacother. 67:489–496. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Somlyai G, Kovács BZ, Papp A and Somlyai I: A preliminary study indicating improvement in the median survival time of glioblastoma multiforme patients by the application of deuterium depletion in combination with conventional therapy. Biomedicines. 11:19892023. View Article : Google Scholar : PubMed/NCBI | |
|
Gyöngyi Z, Budán F, Szabó I, Ember I, Kiss I, Krempels K, Somlyai I and Somlyai G: Deuterium depleted water effects on survival of lung cancer patients and expression of Kras, Bcl2, and Myc genes in mouse lung. Nutr Cancer. 65:240–246. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Nikolouzakis TK, Vassilopoulou L, Fragkiadaki P, Mariolis Sapsakos T, Papadakis GZ, Spandidos DA, Tsatsakis AM and Tsiaoussis J: Improving diagnosis, prognosis and prediction by using biomarkers in CRC patients (Review). Oncol Rep. 39:2455–2472. 2018.PubMed/NCBI | |
|
Averill-Bates D: Reactive oxygen species and cell signaling. Review. Biochim Biophys Acta Mol Cell Res. 1871:1195732024. View Article : Google Scholar : PubMed/NCBI | |
|
Park HJ, Carr JR, Wang Z, Nogueira V, Hay N, Tyner AL, Lau LF, Costa RH and Raychaudhuri P: FoxM1, a critical regulator of oxidative stress during oncogenesis. EMBO J. 28:2908–2918. 2009. View Article : Google Scholar : PubMed/NCBI | |
|
Su X, Yang Y, Yang Q, Pang B, Sun S, Wang Y, Qiao Q, Guo C, Liu H and Pang Q: NOX4-derived ROS-induced overexpression of FOXM1 regulates aerobic glycolysis in glioblastoma. BMC Cancer. 21:11812021. View Article : Google Scholar : PubMed/NCBI | |
|
Xia L, Mo P, Huang W, Zhang L, Wang Y, Zhu H, Tian D, Liu J, Chen Z, Zhang Y, et al: The TNF-α/ROS/HIF-1-induced upregulation of FoxMI expression promotes HCC proliferation and resistance to apoptosis. Carcinogenesis. 33:2250–2259. 2012. View Article : Google Scholar : PubMed/NCBI |