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.
DEAH box helicase 15 (DHX15), which is a member of the DEAH box RNA helicase family (1), is involved in several cellular processes, such as splicing and ribosome biogenesis and facilitates remodeling of large RNA-protein complexes (2–4). DHX15 was shown to carry out a role in antiviral immune response in vivo (5). In RNA virus-induced intestinal inflammation, DHX15 carries out a pivotal role in controlling inflammation by sensing IFN-β and other cytokines produced by RNA viruses (6). Moreover, DHX15 is reported to be involved in tumorigenesis as a tumor-promoting factor in acute lymphoblastic leukemia and cancer of the lung, prostate or breast (7–10), but it also acts as a tumor suppressor in hepatocellular carcinoma and gastric cancer (11,12). Using gain- and loss-of-function analyses, we previously revealed that DHX15 suppressed the proliferation of glioma cell lines (13).
Colorectal cancer (CRC) is the third most common type of cancer, with ~2 million cases diagnosed worldwide in 2020, and the second most common cause of cancer-related mortality, according to the International Agency for Research on Cancer of the World Health Organization (14). Repeat analyses of public single-cell sequence and bulk transcriptome data revealed consistent upregulation of DHX15 expression in patients with CRC (15–17). Analysis of the clinical pathological features of CRC samples from the Cancer Gene Atlas and the Clinical Proteomic Tumor Analysis Consortium revealed that reduced expression of DHX15 mRNA and DHX15 protein associated with poor CRC prognosis in terms of survival, metastasis and recurrence (16). Furthermore, the overall survival positively associated with DHX15 expression levels in CRC (18). Conversely, in a case report of a patient with CRC harboring a KRAS p.G12D mutation, DHX15 was implicated as a critical mediator of microbiota-driven pathogenesis of CRC. Specifically, DHX15 was found to be highly expressed and functioned as a receptor on tumor cells, facilitating the invasion of Fusobacterium nucleatum into the nuclei of intestinal epithelial cells (IECs), thereby promoting tumorigenesis (19). The present study examined the effects of DHX15 overexpression using CRC cell lines with different characteristics, including KRAS mutations (19).
Human CRC samples and adjacent normal tissues were obtained by surgical resection from ten patients with CRC; five male, average age 60 years (51–65 years), five female, average age 75.6 years (60–91 years) at Juntendo University Hospital (Tokyo, Japan) between April 2022-August 2025 and embedded in paraffin. The diagnoses of the ten patients were based on clinical and pathological examinations (Table I). Cancer staging was carried out in accordance with the Union for International Cancer Control TNM classification (20). All human tissues were obtained with written informed consent and approval from the medical ethical committee of Juntendo University Hospital (Tokyo, Japan; approval no. E22-0079).
Table I.DHX15 expression in human CRC tissues and clinicopathological factors of the patients with CRC. |
Porcupine inhibitor C59 (cat. no. S7037) was obtained from Selleck Chemicals and the NF-κB inhibitor IMD-0354 (cat. no. HY-10172) was obtained from MedChemExpress. The human CRC cell lines HCT116 (21), SW480 (22) and Caco2 (23) (provided by Dr Akira Orimo, Juntendo University, Tokyo, Japan) were cultured in low glucose DMEM (Nacalai Tesque, Inc.) and DLD1 cells (24) (provided by Dr Akira Orimo, Juntendo University, Tokyo, Japan) were cultured in RPMI1640 (Nacalai Tesque, Inc.); both culture media were supplemented with 10% FBS (MilliporeSigma) and penicillin-streptomycin (MilliporeSigma). The characteristics of the four CRC cell lines are summarized in (Table SI) (25).
HCT116, SW480, Caco2 and DLD1 cells (1.5×106 cells) were seeded on 10-cm plates (Nippon Gene Co., Ltd.) and transfected using GeneJuice Transfection Reagent (Cat. no. 70967-3CN; Merck KGaA) with a combination of the plasmids pMXs-IP (control vector) or pMX-DHX15 and EGFP expression (pCAG-EGFP), at DNA amounts of 0.03 µg for pCAG-EGFP and 0.21 µg for pMXs-IP or pMX-DHX15 per well. The cells were harvested at 37°C after 12 h of culture and EGFP-positive cells were collected using a cell sorter (FACS ARIA III, BD Biosciences). Total RNA was purified using Trizol® reagent (Thermo Fisher Scientific, Inc.).
RT-qPCR was carried out as previously described (26). The expression levels of the transcripts of interest were normalized using GAPDH and ACTB values. The respective forward and reverse primer sequences were as follows: human ACTB: 5′-GAAGGAGATCACTGCCCTGG-3′ and 5′-ACTCCTGCTTGCTGATCCAC-3′; GAPDH: 5′-ATTGCCCTCAACGACCACTT-3′ and 5′-TGGTCCAGGGGTCTTACTCC-3′; DHX15: 5′-TGCTGAACGTCTACCATGCTT-3′ and 5′-ATTCGAGATAGCTGCTGGCG-3′; IRF1: 5′-AAGGGGTGTGGCCTTTTTAGA-3′ and 5′-TGTCCCTGTTCACCCCAAAG-3′; IRF3: 5′-CCTGCACATTTCCAACAGCC-3′ and 5′-AATCCATGCCCTCCACCAAG-3′: IRF7: 5′-GCTCCCCACGCTATACCATC-3′ and 5′-CAGGGAAGACACACCCTCAC-3′; NFKB1: 5′-GTGAAGACCACCTCTCAGGC-3′ and 5′-CTGTCGCAGACACTGTCACT-3′; NFKB2: 5′-ACGCCTCTTGACCTCACTTG-3′ and 5′-GTGGCTCCATGGTGTTCTGA-3′; RELA: 5′-GGACATGGACTTCTCAGCCC-3′ and 5′-AAAGTTGGGGGCAGTTGGAA-3′; and REL: 5′-TCCTTAGCCCAGCCATCTCT-3′ and 5′-GGCAGTCTCCGCTCATCTTT-3′.
DHX15 protein expression in human clinical samples was examined by immunohistochemistry using an anti-DDX15 antibody (H-4, 1:500; Santa Cruz Biotechnology; cat. no. sc-271686). Paraffin-embedded 3-µm sections were first deparaffinized with EZ Prep (cat. no. 950-102) and subjected to antigen retrieval using Cell Conditioner #2 (pH 9.0; cat. no. 950-223) on a BenchMark Ultra automated stainer (Roche Tissue Diagnostics). After antigen retrieval, the sections were incubated with the primary anti-DDX15 antibody at 37°C for 1 h.
Primary antibody signals were then detected using the UltraView Universal DAB Detection Kit (Roche Tissue Diagnostics, cat. no. 760-500) under standard BenchMark Ultra settings, including an 8-min incubation with the HRP-conjugated multimer reagent, followed by DAB chromogen visualization. Counterstaining was performed with Hematoxylin II for 8 min and Bluing Reagent for 4 min at 37°C. Stained sections were observed using a Zeiss Axio Imager M2 fluorescence microscope (Zeiss GmbH), and images were acquired using ZEN software (version 3.12; Zeiss GmbH).
Stained samples were observed using Zeiss Axio Imager M2 fluorescence microscope (Zeiss GmbH) and images were acquired using ZEN software (ZEN3.12; Zeiss GmbH). For quantification of DHX15 positive signals, we used ImageJ (version 1.54p; National Institutes of Health). The images were imported to ImageJ and DAB signals were isolated using the color deconvolution command of ImageJ. DAB images were converted to gray scale binary images, and doublet cells were separated by the Watershed command and positive and negative cells were counted. To quantify intensity of the signals, the log10 value of mean of sample ROI vs. mean of background ROI by using 8-bit index color was calculated.
Immunohistochemistry of the cultured cells was carried out as described previously (27). The primary antibodies used were mouse monoclonal antibodies against Ki67 (1:250; cat. no. 55060; BD); LC3 (1:2,000; cat. no. M152-3; MEDICAL & BIOLOGICAL LABORATORIES CO., LTD.); rabbit polyclonal antibody against active Caspase-3 (AC3; 1:250; cat. no. 9664S; Promega Corporation); and chicken polyclonal antibody against GFP (1:2,000; cat. no. ab13970; Abcam). Appropriate secondary antibodies conjugated with Alexa Fluor 488 (1:500; cat. no. A11039; Thermo Fisher Scientific, Inc.) or Alexa Fluor 594 (1:500; cat. no. C10639; Thermo Fisher Scientific, Inc.) were used. The nuclei were visualized using DAPI. Stained cells were observed using Zeiss Axio Imager I fluorescence microscope (Zeiss GmbH) and images were acquired using ZEN software (version, 3.12; Zeiss GmbH).
Luciferase reporter assays for the Wnt/β-catenin and NF-κB signaling pathways were performed using TCF-luc (IFN minimal promoter with 7× TCF-binding sites driving firefly luciferase) and NFκB-luc (SV40 early promoter with 8× NF-κB-binding sites driving firefly luciferase) (28). These plasmids were kindly provided by Dr. K. Shimotono (Kyoto University, Kyoto, Japan) and Dr. R. Tsuruta (University of Tokyo, Tokyo, Japan), respectively. No 3′ UTR reporter constructs and no miRNA mimics or inhibitors were used in this study. Cells were transfected with the reporter plasmids using GeneJuice Transfection Reagent (cat. no. 70967-3CN; MilliporeSigma) according to the manufacturer's instructions. Luciferase activity was measured 18 h after transfection using a luminometer (Lumat LB9507; Titertek-Berthold) and the Luciferase Assay System (Promega, cat. no. E1500). Because only firefly luciferase reporters were used, luciferase activity was analyzed without Renilla luciferase normalization.
SW480 (1×104cell/well), Caco2 (1×104 cell/well), HCT116 (2×104 cells) and DLD1 (2×104 cells) cells were plated on 48-well plates (Corning, Inc.) and then transfected with a combination of plasmids pMXs-IP (control vector) or pMX-DHX15 and pCAG-EGFP using GeneJuice (cat. no. 70967-3CN; MilliporeSigma). After 1 h of incubation, the plate was set on IncuCyte ZOOM (Sartorius AG). Live-cell images were captured every 8 h and analyzed using IncuCyte Live-Cell Imaging Software (v2024B).
To evaluate the time-dependent changes in the number of GFP-positive cells after transfection, cells were monitored using an Incucyte live-cell imaging system under standard culture conditions (37°C; 5% CO2) at 8-h intervals immediately after plating.
Cell proliferation was examined by incorporating 5-ethynyl 2′-deoxyuridine (EdU) using Click-iT EdU Alexa Fluor 594 Imaging kit (Thermo Fisher Scientific, Inc.). After transfecting the cells as aforementioned, EdU was added to the culture at a final concentration of 10 µM and incubated for 2 h at 37°C. For inhibitor experiments, cells were treated with the Wnt inhibitor C59 (10 µM) or the NF-κB inhibitor IMD-0354 (5 µM) at 37°C for 2 h prior to EdU addition. The incorporated EdU signals were visualized by immunostaining with a chicken polyclonal antibody against GFP (cat. no. ab13970; 1:2,000; Abcam) at room temperature overnight and a secondary antibody conjugated with Alexa Fluor 488 at room temperature for 30 min (1:500; Thermo Fisher Scientific, Inc.). EGFP was immunostained with an anti-GFP antibody (1:2,000; cat. no. ab13970; Abcam). Nuclei were stained with DAPI. Stained cells were observed using Zeiss Axio Imager I (Zeiss GmbH) and images were acquired using ZEN software (Zeiss GmbH).
Cell lysates were collected from cell lines (2×106 cells) and analyzed by western blotting, as previously described (29). Protein was extracted using RIPA buffer (Nacalai Tesque, Inc.), containing 50 mM Tris-HCl (pH 7.4), 150 mM NaCl, 1% NP-40, 0.5% sodium deoxycholate and 0.1% SDS, supplemented with protease inhibitor cocktail (Nacalai Tesque, Inc.).
Protein concentration was determined using the BCA assay (Nacalai Tesque, Inc..) according to the manufacturer's protocol. Equal amounts of protein (3 µg per lane) were mixed with 4× Laemmli sample buffer (Nacalai Tesque, Inc.) and boiled at 98°C for 5 min before electrophoresis. Proteins were separated on 10% SDS-PAGE gels. Proteins were transferred onto PVDF membranes. Membranes were blocked in 5% non-fat dry milk (Bio-Rad) in TBS-Tween (0.1%) at room temperature for 1 h. Membranes were incubated with primary antibodies at 4°C overnight, followed by incubation with HRP-conjugated secondary antibodies for 1 h at room temperature. Signals were detected using Chemi-Lumi One Super (Nacalai Tesque, Inc.). The primary antibodies used were mouse monoclonal antibodies against ACTIN (1:1,000; cat. no. A4700; MilliporeSigma) and DDX15 (1:1,000; cat. no. sc-271686; Santa Cruz Biotechnology, Inc.). The secondary antibody used was HRP-linked secondary antibodies (1:5,000; cat. no. NA931; Cytiva).
Statistical significance was calculated by an unpaired Student's t test (two tailed). P<0.05 was considered to indicate a statistically significant difference; *P<0.05, **P<0.01. Experiments were conducted in triplicate. Data are shown as mean ± standard deviation)
On examination of the clinical samples, DHX15 protein was detectable in the cell nuclei in both tumor and adjacent normal tissues of all 10 patients (Fig. 1A). The evaluation values of DHX15 expression strength for all 10 samples are summarized (Table I). The average positive nuclei staining of DHX15 in the ten tumor samples was increased compared with that of adjacent tissue (Fig. 1B). Among the CRC tissues, four showed >60% of DHX15 positive signals in tumor tissues, but there was no correlation between clinicopathological factors and DHX15 staining intensity (Table I). Heat-map (Fig. 1C) represents intensity of DHX15 immunostaining of adjacent normal tissue and tumor tissue (Fig. 1D). Again, clear association between pathological observation and signal intensity was not found, but the average DHX15 expression intensity in the tumor region was increased compared with that in adjacent tissue (Fig. 1E).
DHX15 protein expression levels were first examined in the CRC cell lines HCT116, SW480, Caco2 and DLD1 using western blotting of whole protein. All cell lines express DHX15 with expected size (Fig. S1A; upper bands). Subsequently, the cells were co-transfected with DHX15-expression vector or empty control vector together with EGFP-expression plasmid and EGFP positive cells were purified by a cell sorter after 12 h of culture. Overexpression of DHX15 transcripts was confirmed by RT-qPCR in all four cell lines (Fig. S1B).
To examine the effects of DHX15 overexpression on proliferation, the cells were cotransfected with DHX15-expressing vector or empty control vector with EGFP-expression plasmid. The transfected cells were cultured for 2 days, and the number of EGFP-positive cells was counted over time using IncuCyte live-cell imaging. The number of EGFP-positive cells is shown on Fig. 2A and B. Compared with the control samples, samples with DHX15 overexpression had a significantly reduced cell number in all the examined CRC cell lines (Fig. 2B). These results suggested that DHX15 suppressed cell proliferation.
The number of Ki67-positive proliferating HCT116, SW480 and DLD1 cells was significantly reduced after transfection with DHX15 and EGFP-expression plasmid (Fig. 2C and D). The EdU incorporation assay revealed that DHX15 overexpression significantly reduced the number of EdU-positive cells in all four CRC cell lines (Fig. S2A and B). These results suggested that DHX15 suppressed the proliferation of CRC cell lines.
Next, the DHX15-overexpressing CRC cell lines were investigated using the immunohistochemistry apoptosis marker AC3. The cells were cotransfected with either the empty control or DHX15-expressing vector with EGFP-expression plasmid and immunostaining was carried out after 2 days of culture. The number of AC3-positive apoptotic cells was comparable between the control and DHX15-transfected cells in the four CRC cell lines, except HCT116, which showed slight decrease in AC3-positive cells in the DHX15 overexpression group, although this was not significant (Fig. 3A and B). These results indicated that DHX15 overexpression may not affect apoptosis in the CRC cell lines.
To investigate the potential mechanisms underlying this finding, the involvement of the Wnt and NF-κB signaling pathways were examined, as well as autophagy, in DHX15-overexpressing cells. These pathways have previously been reported to play key roles in the regulation of cell proliferation (11,30,31). Analysis revealed that DHX15 overexpression significantly induced luciferase activities that were dependent on the TCF-binding motif in all examined CRC cells (Fig. 4A). The effects of the Wnt signaling inhibitor C59 on the proliferation of CRC cell lines with overexpressed DHX15 were investigated using EdU incorporation assay. Analysis revealed a tendency for higher EdU incorporation in HCT116 and SW480 cells when compared with control cells (Fig. S3A and B).
On examination of NF-κB signaling using a NF-κB target site-dependent luciferase assay, DHX15 overexpression enhanced luciferase activity in HCT116 and SW480 cells; however, in Caco2 and DLD1 cells, luciferase activity was similar between the control and DHX15-overexpressing cells (Fig. 4B). On examination of NF-κB-related gene expression by RT-qPCR in HCT116 and SW480 cells, the control and DHX15-overexpressing cells revealed similar transcript levels of genes, except for IRF3 and REL in SW480 cells (Fig. S4). Examination of the effects of the NF-κB inhibitor IMD-0354 revealed that the number of EdU-positive cells was not significantly different between DHX15-transfected and IMD-0354 treated cells in all four CRC cell lines (Fig. S5A and B).
The expression pattern of LC3 puncta, which is an autophagy marker (32), was subsequently examined. Upon counting the LC3-positive puncta in the nuclei of control and DHX15-overexpressing cells, DHX15 overexpression led to a reduced number of LC3-positive signals in HCT116 and DLD1 cells but not in SW480 and Caco2 cells (Figs. 4C and S6).
The present study investigated the role of DHX15 in CRC by examining DHX15 expression levels in clinical samples and changes in DHX15 levels in CRC cell lines. In the clinical samples from 10 patients with CRC, DHX15 expression levels varied. The absence of a clear association between clinicopathological factors and intensity of DHX15 expression was likely attributable to the small sample size (n=10 patients). Further analysis of DHX15 expression using additional clinical samples may help clarify its relationship with clinicopathological factors. Although the relationship between DHX15 expression levels and tumor malignancy remains inconclusive, the quantification of its expression may provide a potential prognostic indicator, such as recurrence or reduced survival in cases with low DHX15 expression. At present, to the best of our knowledge, no association has been observed between DHX15 expression and tumor stage. In addition, we hypothesize that there is a possibility that the DHX15 expression patterns and levels are associated with disease progression which may be revealed by monitoring the condition of patients continuously and carefully. Conversely, analysis revealed that DHX15 upregulation hampered the proliferation activity of all examined CRC cell lines. This result is consistent with a recent study which revealed that DHX15 inhibits CRC progression, invasion and metastasis (18). In the present study, all CRC cell lines examined exhibited endogenous DHX15 expression. Therefore, further forced overexpression of DHX15 may have exceeded the physiological threshold, leading to cytotoxicity. Such supraphysiological expression levels could disrupt essential cellular processes, ultimately resulting in growth suppression. Mutation in the well-known protooncogene KRAS is found in HCT116, SW480 and DLD1 cell lines but not in Caco2 cells; therefore, the DHX15 effects observed in the present study were likely not associated with KRAS mutation. However, evaluation of DHX15 protein expression patterns and their relationship with clinicopathological factors in hepatocellular carcinoma revealed that DHX15 was markedly upregulated, and its high expression associated with poor prognosis (33). Taken together with the reports of other types of cancer, we hypothesize that the effects on DHX15 expression levels to cancer prognosis depend on the types of cancer.
Among the various mechanisms of DHX15 activity, Wnt involvement has been reported in different model systems. In one study that analyzed the role of DHX15 in antibacterial responses in IECs, mice with DHX15 deletion in IECs were susceptible to infection with enteric bacteria because of low levels of α defensin, which is an antimicrobial peptide; considering that α defensin is induced by the TLR/Wnt pathways this study suggested that DHX15 regulates TLR/Wnt signals (34). Another study revealed that DHX15 regulates zebrafish intestinal development through the Wnt signaling pathway (35). The present study revealed that DHX15 overexpression significantly induced Wnt signaling in the four CRC cell lines; however, inhibition of Wnt signaling did not prevent the suppressed proliferation of DHX15-overexpressing cells. Furthermore, given its well-known contribution to the development and progression of various tumors (36,37), Wnt signaling was less likely to participate in the suppression of cell proliferation by DHX15. However, other physiological processes, such as metastasis, were not analyzed in the present study and requires in vivo clarification in future research.
DHX15 acts as an immune modulator through regulation of NF-κB. Human DHX15 contributes to activation of the NF-κB, JNK and p38 MAPK pathways in HeLa cells in response to the synthetic double-stranded RNA analog poly(I:C) (5). In acute myeloid leukemia, DHX15 is downregulated in disease remission or cell line differentiation (7). Furthermore, knockdown of DHX15 inhibits nuclear translocation and activation of the NF-κB subunit P65 in leukemia cells (7). Positive feedback of NF-κB and DHX15 was reported in breast cancer cells (10). The present study revealed that DHX15 overexpression induced NF-κB signaling in HCT116 and SW480 cells, but the expression levels of known downstream target genes of the NF-κB pathway were not upregulated. Furthermore, NF-κB inhibition by the IMD-0354 did not affect cell proliferation, suggesting the need to clarify the signaling pathways and biological roles of DHX15 using cell lines in the future.
Involvement of autophagy in cancer progression has been reported in various types of cancer (38–41). DHX15 negatively regulates autophagy in association with mTORC1 activation in hepatoma cells (11). There have been several reports on KRAS-activating mutations and autophagy. KRAS-activating mutations increase autophagy and contribute to the survival of CRC cells under starvation conditions (42). In addition, autophagy was induced to different degrees, depending on mutations of the KRAS allele (42). In the present study, given the decreased number of LC3 puncta in HCT116 and DLD1 cells, which have a KRASG13D/− mutation, a relationship between KRAS mutation and autophagy was suspected. However, the contribution of this pathway to cell proliferation remains unclear. The effects of other signaling pathway inhibitors were also explored using this experimental model (data not shown). Preliminary experiments were performed with various inhibitor concentrations, and cell proliferation was assessed by EdU immunostaining. No significant differences were observed between the groups. However, because the efficacy of the inhibitor itself was not fully confirmed, these data are not included in the present study. Experiments in which knockdown of DHX15 is achieved by siRNA oligonucleotide are planned as future work.
Although the present study did not yield fully conclusive mechanistic results, it provides novel evidence that DHX15 overexpression suppresses colorectal cancer cell proliferation and highlights DHX15 as a previously under-recognized regulator of tumor biology. These findings offer a foundation for future mechanistic studies and suggest that DHX15 may serve as a potential biomarker or therapeutic target in colorectal cancer.
The authors would like to thank Dr. Kanji Uchida (Department of Anesthesiology, University of Tokyo Hospital, Tokyo, Japan) for discussions and allowing the use of IncuCyte in their department.
Funding: No funding was received.
The data generated in the present study are included in the figures and/or tables of this article.
YI and KoS performed the experiments, TI, SI, KoS, KiS, KaS and SW designed the research, YI and SW wrote the manuscript. SW contributed to the study conception, experimental design, data interpretation, and provided critical revisions of the manuscript. YI and SW confirm the authenticity of all the raw data. All authors read and approved the final manuscript.
All human tissues were obtained with written informed consent and approval from the medical ethical committee of Juntendo University Hospital, Tokyo, Japan (approval no. E22-0079).
Not applicable.
The authors declare that they have no competing interests.
|
DHX15 |
DEAH (Asp-Glu-Ala-His) box helicase 15 |
|
CRC |
colorectal cancer |
|
IEC |
epithelial cell |
|
RT-qPCR |
reverse transcription-quantitative PCR |
|
TCF-luc |
TCF-binding site luciferase |
|
NFκB-luc |
NF-κB binding sites luciferase |
|
Linder P: Dead-box proteins: A family affair-active and passive players in RNP-remodeling. Nucleic Acids Res. 34:4168–4180. 2006. View Article : Google Scholar : PubMed/NCBI | |
|
Arenas JE and Abelson JN: Prp43: An RNA helicase-like factor involved in spliceosome disassembly. Proc Natl Acad Sci USA. 94:11798–11802. 1997. View Article : Google Scholar : PubMed/NCBI | |
|
Combs DJ, Nagel RJ, Ares M Jr and Stevens SW: Prp43p is a DEAH-box spliceosome disassembly factor essential for ribosome biogenesis. Mol Cell Biol. 26:523–534. 2006. View Article : Google Scholar : PubMed/NCBI | |
|
Tanaka N, Aronova A and Schwer B: Ntr1 activates the Prp43 helicase to trigger release of lariat-intron from the spliceosome. Genes Dev. 21:2312–2325. 2007. View Article : Google Scholar : PubMed/NCBI | |
|
Mosallanejad K, Sekine Y, Ishikura-Kinoshita S, Kumagai K, Nagano T, Matsuzawa A, Takeda K, Naguro I and Ichijo H: The DEAH-box RNA helicase DHX15 activates NF-κB and MAPK signaling downstream of MAVS during antiviral responses. Sci Signal. 7:ra402014. View Article : Google Scholar : PubMed/NCBI | |
|
Xing J, Zhou X, Fang M, Zhang E, Minze LJ and Zhang Z: DHX15 is required to control RNA virus-induced intestinal inflammation. Cell Rep. 35:1092052021. View Article : Google Scholar : PubMed/NCBI | |
|
Pan L, Li Y, Zhang HY, Zheng Y, Liu XL, Hu Z, Wang Y, Wang J, Cai YH, Liu Q, et al: DHX15 is associated with poor prognosis in acute myeloid leukemia (AML) and regulates cell apoptosis via the NF-kB signaling pathway. Oncotarget. 8:89643–89654. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Yao G, Chen K, Qin Y, Niu Y, Zhang X, Xu S, Zhang C, Feng M and Wang K: Long non-coding RNA JHDM1D-AS1 interacts with DHX15 protein to enhance non-small-cell lung cancer growth and metastasis. Mol Ther Nucleic Acids. 18:831–840. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Jing Y, Nguyen MM, Wang D, Pascal LE, Guo W, Xu Y, Ai J, Deng FM, Masoodi KZ, Yu X, et al: DHX15 promotes prostate cancer progression by stimulating Siah2-mediated ubiquitination of androgen receptor. Oncogene. 37:638–650. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Zheng W, Wang X, Yu Y, Ji C and Fang L: CircRNF10-DHX15 interaction suppressed breast cancer progression by antagonizing DHX15-NF-κB p65 positive feedback loop. Cell Mol Biol Lett. 28:342023. View Article : Google Scholar : PubMed/NCBI | |
|
Zhao M, Ying L, Wang R, Yao J, Zhu L, Zheng M, Chen Z and Yang Z: DHX15 inhibits autophagy and the proliferation of hepatoma cells. Front Med (Lausanne). 7:5917362020. View Article : Google Scholar : PubMed/NCBI | |
|
Zong Z, Li H, Ning Z, Hu C, Tang F, Zhu X, Tian H, Zhou T and Wang H: Integrative bioinformatics analysis of prognostic alternative splicing signatures in gastric cancer. J Gastrointest Oncol. 11:685–694. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Ito S, Koso H, Sakamoto K and Watanabe S: RNA helicase DHX15 acts as a tumour suppressor in glioma. Br J Cancer. 117:1349–1359. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A and Bray F: Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 71:209–249. 2021.PubMed/NCBI | |
|
Lv X, Ma W, Miao X, Hu S and Xie H: Navigating colorectal cancer prognosis: A Treg-related signature discovered through single-cell and bulk transcriptomic approaches. Environ Toxicol. 39:3512–3522. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Lu P, Zhang Y, Cui Y, Liao Y, Liu Z, Cao ZJ, Liu JE, Wen L, Zhou X, Fu W and Tang F: Systematic characterization of full-length RNA isoforms in human colorectal cancer at single-cell resolution. Protein Cell. 16:873–895. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Tao Y, Li J, Pan J, Wang Q, Ke RW, Yuan D, Wu H, Cao Y and Zhao L: Integration of scRNA-Seq and bulk RNA-Seq identifies circadian rhythm disruption-related genes associated with prognosis and drug resistance in colorectal cancer patients. Immunotargets Ther. 14:475–489. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Fan L, Guo X, Zhang J, Wang Y, Wang J and Li Y: Relationship between DHX15 expression and survival in colorectal cancer. Rev Esp Enferm Dig. 115:234–240. 2023.PubMed/NCBI | |
|
Zhu H, Li M, Bi D, Yang H, Gao Y, Song F, Zheng J, Xie R, Zhang Y, Liu H, et al: Fusobacterium nucleatum promotes tumor progression in KRAS p.G12D-mutant colorectal cancer by binding to DHX15. Nat Commun. 15:16882024. View Article : Google Scholar : PubMed/NCBI | |
|
Huang SH and O'Sullivan B: Overview of the 8th edition TNM classification for head and neck cancer. Curr Treat Options Oncol. 18:402017. View Article : Google Scholar : PubMed/NCBI | |
|
Rajput A, Martin ID, Rose R, Beko A, Levea C, Sharratt E, Mazurchuk R, Hoffman RM, Brattain MG and Wang J: Characterization of HCT116 human colon cancer cells in an orthotopic model. J Surg Res. 147:276–281. 2008. View Article : Google Scholar : PubMed/NCBI | |
|
Verhagen MP, Xu T, Stabile R, Joosten R, Tucci FA, van Royen M, Trerotola M, Alberti S, Sacchetti A and Fodde R: The SW480 cell line as a model of resident and migrating colon cancer stem cells. iScience. 27:1106582024. View Article : Google Scholar : PubMed/NCBI | |
|
Sambuy Y, De Angelis I, Ranaldi G, Scarino ML, Stammati A and Zucco F: The Caco-2 cell line as a model of the intestinal barrier: Influence of cell and culture-related factors on Caco-2 cell functional characteristics. Cell Biol Toxicol. 21:1–26. 2005. View Article : Google Scholar : PubMed/NCBI | |
|
Dexter DL, Spremulli EN, Fligiel Z, Barbosa JA, Vogel R, VanVoorhees A and Calabresi P: Heterogeneity of cancer cells from a single human colon carcinoma. Am J Med. 71:949–956. 1981. View Article : Google Scholar : PubMed/NCBI | |
|
Ahmed D, Eide PW, Eilertsen IA, Danielsen SA, Eknæs M, Hektoen M, Lind GE and Lothe RA: Epigenetic and genetic features of 24 colon cancer cell lines. Oncogenesis. 2:e712013. View Article : Google Scholar : PubMed/NCBI | |
|
Saita K, Moriuchi Y, Iwagawa T, Aihara M, Takai Y, Uchida K and Watanabe S: Roles of CSF2 as a modulator of inflammation during retinal degeneration. Cytokine. 158:1559962022. View Article : Google Scholar : PubMed/NCBI | |
|
Koso H, Yi H, Sheridan P, Miyano S, Ino Y, Todo T and Watanabe S: Identification of RNA-binding protein LARP4B as a tumor suppressor in glioma. Cancer Res. 76:2254–2264. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Tsuruta L, Lee HJ, Masuda ES, Yokota T, Arai N and Arai K: Regulation of expression of the IL-2 and IL-5 genes and the role of proteins related to nuclear factor of activated T cells. J Allergy Clin Immunol. 96:1126–1135. 1995. View Article : Google Scholar : PubMed/NCBI | |
|
Kuribayashi H, Iwagawa T, Murakami A, Kawamura T, Suzuki Y and Watanabe S: NMNAT1 is essential for human iPS cell differentiation to the retinal lineage. Invest Ophthalmol Vis Sci. 65:372024. View Article : Google Scholar : PubMed/NCBI | |
|
Kabiri Z, Greicius G, Zaribafzadeh H, Hemmerich A, Counter CM and Virshup DM: Wnt signaling suppresses MAPK-driven proliferation of intestinal stem cells. J Clin Invest. 128:3806–3812. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Jiao L, Jiang M, Liu J, Wei L and Wu M: Nuclear factor-kappa B activation inhibits proliferation and promotes apoptosis of vascular smooth muscle cells. Vascular. 26:634–640. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Levine B and Kroemer G: Biological functions of autophagy genes: A disease perspective. Cell. 176:11–42. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Xie C, Liao H, Zhang C and Zhang S: Overexpression and clinical relevance of the RNA helicase DHX15 in hepatocellular carcinoma. Hum Pathol. 84:213–220. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Wang Y, He K, Sheng B, Lei X, Tao W, Zhu X, Wei Z, Fu R, Wang A, Bai S, et al: The RNA helicase Dhx15 mediates Wnt-induced antimicrobial protein expression in Paneth cells. Proc Natl Acad Sci USA. 118:e20174321182021. View Article : Google Scholar : PubMed/NCBI | |
|
Yao J, Cai Y, Chen Z, Wang X, Lai X, Pan L, Li Y and Wang S: Dhx15 regulates zebrafish intestinal development through the Wnt signaling pathway. Genomics. 115:1105782023. View Article : Google Scholar : PubMed/NCBI | |
|
Yu F, Yu C, Li F, Zuo Y, Wang Y, Yao L, Wu C, Wang C and Ye L: Wnt/β-catenin signaling in cancers and targeted therapies. Signal Transduct Target Ther. 6:3072021. View Article : Google Scholar : PubMed/NCBI | |
|
Ge X and Wang X: Role of Wnt canonical pathway in hematological malignancies. J Hematol Oncol. 3:332010. View Article : Google Scholar : PubMed/NCBI | |
|
Chang SH, Huang SW, Wang ST, Chung KC, Hsieh CW, Kao JK, Chen YJ, Wu CY and Shieh JJ: Imiquimod-induced autophagy is regulated by ER stress-mediated PKR activation in cancer cells. J Dermatol Sci. 87:138–148. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Russell RC and Guan KL: The multifaceted role of autophagy in cancer. EMBO J. 41:e1100312022. View Article : Google Scholar : PubMed/NCBI | |
|
Jalali P, Shahmoradi A, Samii A, Mazloomnejad R, Hatamnejad MR, Saeed A, Namdar A and Salehi Z: The role of autophagy in cancer: From molecular mechanism to therapeutic window. Front Immunol. 16:15282302025. View Article : Google Scholar : PubMed/NCBI | |
|
Niu X, You Q, Hou K, Tian Y, Wei P, Zhu Y, Gao B, Ashrafizadeh M, Aref AR, Kalbasi A, et al: Autophagy in cancer development, immune evasion, and drug resistance. Drug Resist Updat. 78:1011702025. View Article : Google Scholar : PubMed/NCBI | |
|
Alves S, Castro L, Fernandes MS, Francisco R, Castro P, Priault M, Chaves SR, Moyer MP, Oliveira C, Seruca R, et al: Colorectal cancer-related mutant KRAS alleles function as positive regulators of autophagy. Oncotarget. 6:30787–30802. 2015. View Article : Google Scholar : PubMed/NCBI |