IMR Press / FBL / Volume 29 / Issue 11 / DOI: 10.31083/j.fbl2911400
Open Access Original Research
DLX5 Promotes Radioresistance in Renal Cell Carcinoma by Upregulating c-Myc Expression
Show Less
Affiliation
1 Department of Radiation Oncology, Ningbo Medical Center Lihuili Hospital, 315000,Ningbo, Zhejiang, China
2 Department of Urology, Ningbo Urology and Nephrology Hospital, 315100 Ningbo, Zhejiang, China
*Correspondence: chenxiaodong30505@163.com (Xiaodong Chen)
Front. Biosci. (Landmark Ed) 2024, 29(11), 400; https://doi.org/10.31083/j.fbl2911400
Submitted: 11 July 2024 | Revised: 13 August 2024 | Accepted: 23 August 2024 | Published: 26 November 2024
Copyright: © 2024 The Author(s). Published by IMR Press.
This is an open access article under the CC BY 4.0 license.
Abstract
Background:

Renal cell carcinoma (RCC) is a prevalent and aggressive kidney cancer with notable metastatic potential. While radiotherapy is effective for treating metastatic RCC, the emergence of radioresistance presents a major challenge. This study explores the role of DLX5, previously identified as an oncogene in various cancers, in the development of radioresistance in RCC.

Methods:

Distal-less homeobox 5 (DLX5) expression was measured using western blot analysis. To study the effects of DLX5, its expression was knocked down in 786-O and Caki-1 RCC cell lines through si-DLX5 transfection, and the impact of DLX5 on RCC cell proliferation and radioresistance was assessed using cell counting kit-8 (CCK-8), 5-Ethynyl-2′-deoxyuridine (EdU) incorporation assay, flow cytometry, colony formation, immunofluorescence, and western blot assays. The underlying mechanisms were explored through western blot, colony formation, and CCK-8 assays. In vivo effects were examined using a xenograft mouse model.

Results:

In silico results showed increased DLX5 levels in RCC tissues. Similarly, DLX5 expression was elevated in RCC cell lines. Silencing DLX5 reduced RCC cell proliferation and induced apoptosis in vitro. Additionally, DLX5 knockdown decreased radioresistance and increased DNA damage in RCC cells. Mechanistic studies revealed that DLX5 promotes radioresistance through the upregulation of c-Myc. In vivo, DLX5 silencing impeded tumor growth and reduced radioresistance.

Conclusion:

DLX5 contributes to RCC cell growth and radioresistance by upregulating c-Myc expression, highlighting its potential as a target for overcoming radioresistance in RCC.

Keywords
renal cell carcinoma
DLX5
radioresistance
c-Myc
1. Introduction

Renal cell carcinoma (RCC) represents the most prevalent type of renal tumor, comprising nearly 85% of all kidney tumors and 3% of all adult malignancies, and its incidence and mortality rates have been rising in recent years [1, 2]. Current clinical treatment options, including surgery, targeted therapy, and immunotherapy, have improved the 5-year overall survival rate to 70%–80% [3, 4]. However, 30% of patients eventually develop metastatic RCC following surgical intervention [5]. Therefore, elucidating the underlying mechanisms of RCC and identifying potential therapeutic targets is essential.

Radiotherapy is designed to damage the genome of tumor cells, leading to cell death through the generation of irreparable DNA double-strand breaks (DSBs) [6]. Enhanced repair of DNA DSBs can contribute to resistance to ionizing radiation (IR) [7]. c-Myc plays a significant role in various biological processes in kidney cancer, including the regulation of metabolism [8]. c-Myc has been shown to interact with and regulate the promoters of genes involved in DSB repair in prostate cell lines [9]. In this regard, Wang et al. [10] demonstrated that c-Myc binds to the promoters of CHK1 and CHK2, essential regulators of the DNA damage response (DDR), thereby enhancing radioresistance in stem cell-like populations of nasopharyngeal carcinoma cells. Despite these findings, little is known about the effects of c-Myc in RCC radioresistance.

The distal-less homeobox (DLX) family (DLXs), which includes DLX1 through DLX6, is related to the Drosophila distal-less gene and is represented by three clusters: DLX1/DLX2, DLX3/DLX4 and DLX5/DLX6 [11]. DLXs are involved in cell differentiation and proliferation, primarily during embryonic development, including in the nervous system, appendages, hematopoiesis, and branchial arches [11, 12]. In addition, there is increasing evidence indicating that DLXs are deregulated in various tumors, suggesting their potential as therapeutic targets [13, 14]. DLX5, in particular, has demonstrated potent oncogenic activity in oral squamous cell carcinoma (OSCC) [15], ovarian cancer [16], and lung cancer [17]. However, the role of DLX5 in RCC is not yet understood. Notably, DLX5 has been shown to directly bind the c-Myc promoter in lung tumor cells [17]. This raises the hypothesis that DLX5 may influence radioresistance in RCC through interaction with c-Myc.

Therefore, this study aims to investigate the role of DLX5 in RCC development and radioresistance both in vitro and in vivo. The results may provide a theoretical foundation for advancing RCC treatment strategies.

2. Materials and Methods
2.1 Expression Profile of DLX5 in RCC

DLX5 expression levels in kidney renal clear cell carcinoma (KIRC) and normal tissues were evaluated using the UALCAN (http://ualcan.path.uab.edu/index.html), TIMER (https://cistrome.shinyapps.io/timer/), and GEPIA (http://gepia2.cancer-pku.cn) databases.

2.2 Cell Culture

RCC cell lines 786-O (CL-0010), A-498 (CL-0254) and Caki-1 (CL-0052), and HK-2 (CL-0109) cells were purchased from Procell (Wuhan, China). A-498 and HK-2 cells were cultured in Minimum Essential Medium (MEM, PM150410, Procell), while 786-O and Caki-1 cells were grown in RPMI-1640 (PM150110, Procell) and McCoy’s 5A media (PM150710, Procell), respectively, with 10% fetal bovine serum (FBS, 164210-50, Procell) and 1% penicillin/streptomycin (PB180120, Procell). Cells were authenticated by short tandem repeat (STR) profiling, tested negative for mycoplasma, and maintained in a 37 °C, 5% CO2 incubator.

2.3 Cell Transfection

Two small interfering RNAs (siRNAs) targeting DLX5 (si-DLX5#1 and si-DLX5#2) and corresponding negative controls (si-NC) were synthesized by GenePharma (Shanghai, China). The sequences of si-DLX5#1, si-DLX5#2, and si-NC were 5-AGCUAUAGUCGGCAUAAGCUU-3, 5-GUGCAGCCAGCUCAAUCAA-3, and 5-UUCUCCGAACGUGUCACGU-3, respectively. Overexpression of c-Myc was achieved by transfecting pcDNA vector plasmids containing c-Myc (NM_005221.5) sequences (OE-c-Myc). The following transfections were performed: si-DLX5#1, si-DLX5#2, si-NC, si-NC+empty pcDNA vector plasmids (vector), si-DLX5#1+vector, and si-DLX5#1+OE-c-Myc into 786-O and Caki-1 cell lines using Lipofectamine 3000 (Invitrogen, Carlsbad, CA, USA), and the cells were then collected for subsequent analyses 48 hours later.

2.4 Western Blot

Total proteins were extracted using RIPA buffer (R0010, Solarbio, Beijing, China) and quantified with a BCA protein assay (ab102536, Abcam, Cambridge, UK). Proteins (20 µg) were separated by SDS-PAGE and transferred to PVDF membranes (EMD Millipore, Burlington, MA, USA). Membranes were blocked with 5% skim milk (D8340, Solarbio) for 1 hour at room temperature, then incubated overnight at 4 °C with primary antibodies against DLX5 (1:1000, ab64827), gamma histone H2AX (γH2AX) (1:1000, ab2893), c-Myc (1:20,000, ab152146), and β-actin (1:5000, ab8227) (all Abcam). Following this, membranes were incubated with horseradish peroxidase (HRP)-conjugated Anti-Rabbit IgG (1:50,000, ab288151, Abcam) for 2 hours and visualized using an ECL assay (P0018S, Beyotime, Shanghai, China).

2.5 CCK-8 Assay

Cells (3000 per well) were plated in 96-well plates and cultured overnight at 37 °C with 5% CO₂. After treatments, 10 µL of CCK-8 reagent (C0037, Beyotime) was added to each well, the plates were incubated for 2 hours at 37 °C, and optical density (450 nm) was measured using a microplate reader (Thermo Fisher Scientific, Waltham, MA, USA).

2.6 5-Ethynyl-2-Deoxyuridine (EdU) Incorporation Assay

Cells (6 × 105 per well) were plated in 6-well plates and incubated at 37 °C with 5% CO2. After 12 hours, cells were exposed to 1 mL of 20 µM EdU solution (C0081, Beyotime) for 2 hours. Cells were then fixed, permeabilized, and treated with an anti-EdU Click solution (C0081, Beyotime). Nuclei were stained with Hoechst 33342 (C1025, Beyotime). Images were captured using fluorescence microscopy (IX71, Olympus, Tokyo, Japan), and the percentage of EdU-positive cells was calculated from five random fields.

2.7 Flow Cytometry

Cells were stained with propidium iodide (PI) and fluoresceinisothiocyanate (FITC)-Annexin V (C1062S, Beyotime), and then washed with cold PBS (C0221A, Beyotime). Apoptosis was assessed using a FACScan flow cytometer (BD Biosciences, NJ, USA), and data was analyzed using BD CellQuest Pro software (version 5.1, BD Biosciences).

2.8 Irradiation

Cells were irradiated in ambient air with total doses of 0, 2, 4, 6, and 8 Gray (Gy) using a JL Shepherd Mark 1–68 137Cs irradiator. Mice with subcutaneous tumors were irradiated with 10 Gy using an XRad 320 irradiator (Precision X-Ray, 250 kV/s, 15 mA) following anesthesia with isoflurane (R510-22, RWD LIFE SCIENCE, Shenzhen, China).

2.9 Colony Formation Assay

Cells (800 per well) were plated in 6-well plates and incubated at 37 °C for 14 days. After fixation with 4% paraformaldehyde (P0099, Beyotime), cells were stained with 0.1% crystal violet (C0121, Beyotime) for 30 minutes. Colonies with 50 or more cells were counted manually and photographed. Plating efficiency (PE) was calculated by dividing the number of colonies in the control group by the number of cells plated. The survival fraction was determined by dividing the number of colonies by the product of the number of cells plated and the PE.

2.10 Immunofluorescence (IF)

Cells were plated on glass coverslips and cultured overnight. After irradiation with 4 Gy, cells were collected at 0, 1, 4, and 8 hours. For immunofluorescence, cells were fixed with 4% paraformaldehyde (P0099, Beyotime) for 15 minutes, washed with PBS (C0221A, Beyotime), and permeabilized with 0.5% Triton X-100 (P0096, Beyotime) for 20 minutes. Following overnight incubation with anti-γH2AX antibody (1:5000, ab11174, Abcam) at 4 °C, cells were washed and incubated with Goat anti-rabbit IgG-AlexaFluor 488 (1:1000, ab150077, Abcam) for 1 hour at 37 °C. Cells were mounted with DAPI-containing medium (S2110, Solarbio) and visualized by fluorescence microscopy.

2.11 In Vivo Experiment

Four-week-old BALB/c nude mice (Vital River, Beijing, China) were housed in a temperature-controlled specific pathogen free (SPF) facility with a 12-hour light-dark cycle. Mice were randomly assigned to four groups (n = 6 each): sh-NC, sh-DLX5#1, 10 Gy, and sh-DLX5#1+10 Gy. Each group had six mice throughout the study. The left flank of mice in the sh-NC and sh-DLX5#1 groups were subcutaneously injected with 2 × 106 786-O cells transfected with sh-NC or sh-DLX5#1, respectively. The 10 Gy group received 2 × 106 786-O cells and was irradiated with 10 Gy when tumors became measurable. Mice in the sh-DLX5#1+10 Gy group were injected with 2 × 106 786-O cells transfected with sh-DLX5#1 and then irradiated with 10 Gy when tumors were measurable. Tumor volumes were measured weekly using the formula: 1/2 × length × width2. After four weeks, mice were euthanized with isoflurane (R510-22, RWD LIFE SCIENCE), and tumors were excised, weighed, and stored at –80 °C. All procedures were approved by the Animal Research Ethics Committee of Zhejiang Huitong Test & Evaluation Technology Group Co., Ltd (Approval No. HT-2023-LWFB-0018) .

2.12 Immunohistochemistry (IHC)

Tumor samples were fixed in 4% formaldehyde (P0099, Beyotime), dehydrated, and embedded in paraffin (YA0012, Solarbio). Antigen retrieval was done with 10 mM sodium citrate buffer (pH 6.0, P0083, Beyotime) at 94 °C for 15 minutes. Sections were blocked with 1% bovine serum albumin (BSA) (ST023, Beyotime), then incubated with primary antibodies against DLX5 (1:100, ab64827, Abcam), c-Myc (1:100, ab32072, Abcam), and Ki-67 (1:100, ab15580, Abcam). Biotinylated secondary antibodies (1:2000, ab64256, Abcam) were used, and sections were counterstained with hematoxylin (C0105S, Beyotime). The samples were imaged with a light microscope (Olympus).

2.13 Statistical Analysis

Data are presented as mean ± standard deviation (SD). Normality and variance homogeneity tests confirmed a normal distribution. For comparisons between two groups, an unpaired Student’s t-test was used. For comparisons among three or more groups, one-way ANOVA with Dunnett’s post hoc test was applied. Post hoc analyses were conducted with the Bonferroni method in SPSS 26.0 (IBM Corp., Chicago, IL, USA). Statistical significance was defined as p < 0.05.

3. Results
3.1 DLX5 Expression is Elevated in RCC

Pan-cancer analysis revealed a significant increase in DLX5 expression across various cancer types, including KIRC (Fig. 1A). Specifically, DLX5 was markedly upregulated in KIRC samples compared to normal kidney tissue samples (Fig. 1B,C). Additionally, high levels of DLX5 were validated in RCC cell lines (Fig. 1D). Notably, DLX5 expression was higher in 786-O and Caki-1 cells compared to A-498 cells, leading to the selection of 786-O and Caki-1 for further experiments. These findings indicate that DLX5 is highly expressed in RCC.

Fig. 1.

Elevated expression of DLX5 in renal cell carcinoma (RCC). (A) Pan-cancer analysis demonstrated increased DLX5 expression in kidney renal clear cell carcinoma (KIRC). *p < 0.05, **p < 0.01, and ***p < 0.001 vs. Normal. (B) The Gene Expression Profiling Interactive Analysis (GEPIA) analysis showed heightened DLX5 expression in KIRC. ***p < 0.001 vs. Normal. (C) Alignable Tight Genomic Clusters (ATGC) database results confirmed the upregulation of DLX5 in KIRC tumor samples compared to normal samples. *p < 0.05 vs. Normal. (D) DLX5 levels in RCC cell lines. ***p < 0.001 vs. HK-2. n = 3.

3.2 Silencing DLX5 Inhibits RCC Cell Growth

Given the high DLX5 expression in RCC cells, we used two siRNAs targeting DLX5 (si-DLX5#1 and si-DLX5#2) to lower DLX5 levels in 786-O and Caki-1 cells (Fig. 2A). DLX5 silencing significantly reduced cell proliferation, as shown by CCK-8 (Fig. 2B) and EdU assays (Fig. 2C and Supplementary Fig. 1), and increased apoptosis, as indicated by flow cytometry (Fig. 2D). Additionally, DLX5 knockdown enhanced p53 protein expression (Fig. 2E), suggesting DLX5’s role in regulating p53-mediated apoptosis and RCC cell proliferation. Thus, DLX5 downregulation impairs RCC cell growth and promotes apoptosis.

Fig. 2.

Silencing of distal-less homeobox 5 (DLX5) represses cell proliferation and enhances apoptosis in RCC cells. (A) DLX5 levels were decreased in 786-O and Caki-1 cells using si-DLX5#1 and si-DLX5#2, with β-actin as the normalization control. (B,C) Proliferation was assessed by cell counting kit-8 (CCK-8) (B) and, 5-Ethynyl-2-deoxyuridine (EdU) incorporation assay (C) . Scale bars = 100 µm. (D) Apoptosis was evaluated by flow cytometry. (E) p53 protein expression was measured by Western blot, normalized to β-actin. **p < 0.01, and ***p < 0.001 vs. small interfering RNA-negative control (si-NC). n = 3.

3.3 Interference with DLX5 Impairs Radioresistance in RCC Cells

The impact of DLX5 on radioresistance was assessed in 786-O and Caki-1 cells. Following transfection with si-DLX5#1 and si-DLX5#2, both cell lines exhibited a dose-dependent reduction in survival rates (Fig. 3A). Notably, the survival rates of 786-O and Caki-1 cells were significantly diminished following 8 Gy of radiation (RI). Additionally, cell viability was markedly reduced at a total dose of 4 Gy (p < 0.01), with further significant decreases observed upon DLX5 silencing (Fig. 3B). These findings indicate that DLX5 knockdown effectively impairs the radioresistance of RCC cells.

Fig. 3.

Interference with DLX5 inhibits radioresistance in RCC cells. (A) Colony formation assays assessed survival fractions of 786-O and Caki-1 cells transfected with si-NC, si-DLX5#1, and si-DLX5#2 following irradiation at 0, 2, 4, 6, and 8 Gray (Gy). **p < 0.01 vs. si-NC. (B) Cell viability was measured by CCK-8 assay after 4 Gy irradiation and 96 hours of culture. **p < 0.01, and ***p < 0.001. n = 3.

3.4 Knockdown of DLX5 Promotes DNA Damage in RCC Cells

To examine DLX5’s impact on DNA damage, we analyzed 786-O and Caki-1 cells. DLX5 downregulation significantly increased γH2AX levels (Fig. 4A and Supplementary Fig. 2). After 4 Gy irradiation for 8 hours, DLX5 silencing further elevated γH2AX expression (Fig. 4B), suggesting that DLX5 knockdown exacerbates DNA damage in RCC cells.

Fig. 4.

Interference with DLX5 increases DNA damage in RCC cells. (A) The relative gamma histone H2AX (γH2AX) levels were detected by immunofluorescence (IF) in 786-O and Caki-1 cells. Scale bars = 50 µm. (B) The relative protein expression of γH2AX was measured by Western blot in both cell lines. Data are normalized to β-actin. *p < 0.05, **p < 0.01, and ***p < 0.001 vs. si-NC. n = 3.

3.5 DLX5 Enhances Radioresistance through c-Myc

c-Myc is known to interact with and modulate the promoters of DNA double-strand break (DSB) repair genes, thereby enhancing radioresistance in tumor cells [9, 10]. Additionally, DLX5 has been shown to directly bind to the c-Myc promoter in lung tumor cells [17], suggesting that DLX5 may influence radioresistance via the regulation of c-Myc. In this study, we found that DLX5 modulated c-Myc expression, as silencing DLX5 significantly reduced the 4 Gy-induced relative protein expression of c-Myc in 786-O cells (Fig. 5A). Further validation was performed by co-transfecting 786-O cells with an overexpression plasmid of c-Myc and si-DLX5#1. Results showed that overexpression of c-Myc significantly rescued the reduced c-Myc expression induced by si-DLX5#1, both with and without radiation treatment (Fig. 5B). Moreover, c-Myc overexpression in 786-O cells restored the decreased survival rate (Fig. 5C) and cell viability (Fig. 5D) caused by DLX5 silencing. Additionally, increased c-Myc expression markedly reduced the γH2AX levels induced by si-DLX5#1 (Fig. 5E). Collectively, these findings demonstrate that DLX5 promotes radioresistance through modulation of c-Myc.

Fig. 5.

DLX5 enhances radioresistance through c-Myc. (A) c-Myc protein levels were measured in 786-O cells transfected with si-NC, si-DLX5#1, or si-DLX5#2, normalized to β-actin. (B) c-Myc expression was assessed in cells transfected with si-NC+vector, si-DLX5#1+vector, or si-DLX5#1+OE-c-Myc, with data normalized to β-actin. (C) Colony formation assays determined survival fractions in the same transfections. (D) Cell viability was assessed by the CCK-8 assay for the same conditions. (E) γH2AX protein expression was analyzed by Western blot, normalized to β-actin. *p < 0.05, **p < 0.01, and ***p < 0.001. n = 3.

3.6 Downregulation of DLX5 Reduces Radioresistance in Vivo

Lastly, we evaluated the role of DLX5 in tumor progression and radioresistance in nude mice. DLX5 knockdown or irradiation with 10 Gy significantly reduced tumor volume and weight. This reduction was further pronounced when DLX5 silencing was combined with 10 Gy irradiation (Fig. 6A). Similar trends were observed in the expression levels of ki-67 and c-Myc, which were significantly decreased under these conditions (Fig. 6B). Additionally, both DLX5 silencing and 10 Gy irradiation led to a marked increase in γH2AX protein levels, with the combination of DLX5 knockdown and 10 Gy irradiation resulting in the most substantial increase (Fig. 6C). These findings demonstrate that downregulation of DLX5 effectively suppresses radioresistance in vivo.

Fig. 6.

Silencing of DLX5 inhibits radioresistance in vivo. Nude mice were inoculated subcutaneously with 2 × 106 786-O cells transfected with sh-NC or sh-DLX5#1, and irradiated with 10 Gy when tumors became measurable. (A) Tumor volume and weight were monitored. (B) c-Myc and Ki-67 expression levels were assessed by immunohistochemistry (IHC). Scale bars = 200 µm. (C) γH2AX protein levels were measured by Western blot, normalized to β-actin. *p < 0.05, and ***p < 0.001. n = 6.

4. Discussion

RCC is a malignant kidney tumor characterized by a rising incidence and mortality rate. The prognosis of RCC is further compromised by metastasis, which exacerbates the disease outcome [5]. Despite the potential of radiotherapy for managing metastatic RCC, radioresistance has emerged as a significant clinical challenge [18]. DLX5, an oncogene implicated in various cancers, has been shown to play a critical role in tumorigenesis [15, 16, 17]. Our study demonstrates that DLX5 expression is significantly elevated in RCC, and its downregulation inhibits cell proliferation while promoting apoptosis. Additionally, DLX5 silencing reduces radioresistance and enhances DNA damage in RCC cells. Mechanistically, DLX5 appears to facilitate radioresistance through the upregulation of c-Myc. Moreover, the suppression of DLX5 impedes tumor growth and radioresistance in vivo. Collectively, DLX5 can lead to RCC progression and radioresistance by modulating c-Myc expression.

DLX5 expression is elevated in various tumors, including OSCC [15], ovarian cancer [16] and lung cancer [17]. Elevated DLX5 levels are generally associated with adverse clinical outcomes. For instance, high DLX5 expression was found to be correlated with increased tumor growth both in vitro and in vivo, advanced TNM stages, lymph node metastasis, poor cellular differentiation, and worse prognosis [15]. In ovarian cancer, DLX5 upregulation has been linked to enhanced cell proliferation and tumor size [16]. Consistent with these observations, our study shows that high DLX5 expression in RCC cells promotes growth and radioresistance and that DLX5 downregulation increases apoptosis and reduces tumor growth in vitro and in vivo, highlighting its role in RCC progression.

In addition, our study reveals that DLX5 interference impedes radioresistance in RCC cells. The primary mechanism of radiation damage in radiotherapy involves DNA damage, which disrupts the cell cycle and activates various signaling pathways to repair the damaged DNA [19]. The effectiveness of radiation therapy in treating malignancies can be enhanced by modulating DNA damage repair processes, which are influenced by the interplay between DNA damage response (DDR) activation and repair mechanisms [20, 21]. A key feature of DDR activation is the accumulation of γH2AX at sites of DNA damage, making γH2AX a reliable marker for DNA damage [22, 23]. The phosphorylation of γH2AX is a hallmark of the recognition and repair of DNA double-strand breaks (DSBs) [24]. Ionizing radiation (IR) commonly induces cell death by generating irreparable DNA DSBs [6]. Here, our findings demonstrate that DLX5 downregulation significantly increased the relative level of γH2AX in 786-O and Caki-1 cells, indicating that silencing DLX5 promotes DNA damage in RCC cells. Thus, DLX5 silencing inhibits radioresistance by enhancing DNA damage. Additionally, DLX5 downregulation notably reduced the relative protein expression of c-Myc in 786-O cells subjected to radiation, a result further validated by c-Myc overexpression experiments. This suggests that DLX5 regulates c-Myc expression, consistent with previous study showing that DLX5 directly binds to the c-Myc promoter in lung tumor cells [17]. Overexpression of c-Myc in 786-O cells significantly restored survival rates, cell viability, and reduced γH2AX levels that were elevated due to DLX5 silencing. Therefore, DLX5 enhances radioresistance in RCC through c-Myc. However, the precise mechanisms through which DLX5 promotes radioresistance via c-Myc warrant further investigation. The transcription factor c-Myc has been shown to bind to the promoters of checkpoint kinase 1 (CHK1) and checkpoint kinase 2 (CHK2), enhancing radioresistance in stem cell-like populations of nasopharyngeal carcinoma cells [10]. It is plausible that DLX5 enhances radioresistance through a c-Myc/CHK axis. Moreover, the ataxia-telangiectasia mutated and Rad3 related (ATR)-CHK1 pathway is crucial for the cellular response to DNA damage. ATR is a central regulator of the cellular DNA damage response [25, 26]. During the G2 phase of the cell cycle, activation of the ATR/CHK1 pathway prevents mitosis in cells with damaged DNA [27, 28]. Future studies will explore the role of the ATR-CHK1 pathway in DLX5/c-Myc-mediated radioresistance in RCC.

This study acknowledges several limitations. Firstly, the clinical parameters associated with DLX5 will be explored in future research to further validate the clinical relevance of our findings. Additionally, while our in vitro results are promising, further validation through in vivo experiments involving the overexpression of c-Myc is necessary to confirm the role of DLX5 in RCC radioresistance.

5. Conclusion

In summary, the findings demonstrate that DLX5 expression is significantly increased in RCC. Loss-of-function assays revealed that DLX5 enhances RCC cell growth and radioresistance, with mechanistic insights indicating that DLX5 promotes these effects through the upregulation of c-Myc. These results support DLX5 as an essential player for RCC progression and radioresistance, identifying it as a potential marker for diagnosis and therapeutic targeting. Furthermore, targeting DLX5 or its downstream pathways may offer new strategies for overcoming radioresistance in RCC.

Availability of Data and Materials

All data containing in the study are available upon requirement by contacting the corresponding author on reasonable request.

Author Contributions

DH, ML and XC contributed to the study conception and design. Material preparation and the experiments were performed by DH. Data collection and analysis were performed by ML. The first draft of the manuscript was written by XC and all authors commented on previous versions of the manuscript. All authors read and approved the final manuscript. All authors have participated sufficiently in the work and agreed to be accountable for all aspects of the work.

Ethics Approval and Consent to Participate

All animal experiments were approved by the Ethics Committee of Zhejiang Huitong Test & Evaluation Technology Group Co., Ltd (Approval No. HT-2023-LWFB-0018) for the use of animals and conducted in accordance with the National Institutes of Health Laboratory Animal Care and Use Guidelines. The animal experiment complies with the ARRIVE guidelines and in accordance with the National Institutes of Health guide for the care and use of Laboratory animals (NIH Publications No. 8023, revised 1978).

Acknowledgment

Not applicable.

Funding

This study was supported by the Medical Science and Technology Project of Zhejiang Provincial Health Commission (Grant No.2024KY1605).

Conflict of Interest

The authors declare no conflict of interest.

Supplementary Material

Supplementary material associated with this article can be found, in the online version, at https://doi.org/10.31083/j.fbl2911400.

References
[1]
Siegel RL, Giaquinto AN, Jemal A. Cancer statistics, 2024. CA: a Cancer Journal for Clinicians. 2024; 74: 12–49.
[2]
Young M, Jackson-Spence F, Beltran L, Day E, Suarez C, Bex A, et al. Renal cell carcinoma. Lancet (London, England). 2024; 404: 476–491.
[3]
Singh D. Current updates and future perspectives on the management of renal cell carcinoma. Life Sciences. 2021; 264: 118632.
[4]
Chen YW, Wang L, Panian J, Dhanji S, Derweesh I, Rose B, et al. Treatment Landscape of Renal Cell Carcinoma. Current Treatment Options in Oncology. 2023; 24: 1889–1916.
[5]
De Meerleer G, Khoo V, Escudier B, Joniau S, Bossi A, Ost P, et al. Radiotherapy for renal-cell carcinoma. The Lancet. Oncology. 2014; 15: e170–7.
[6]
Lester-Coll NH, Topol EJ. Radiation oncology 2.0. Lancet (London, England). 2021; 398: 654.
[7]
Tan J, Sun X, Zhao H, Guan H, Gao S, Zhou PK. Double-strand DNA break repair: molecular mechanisms and therapeutic targets. MedComm. 2023; 4: e388.
[8]
Liu Z, Sun T, Piao C, Zhang Z, Kong C. METTL13 inhibits progression of clear cell renal cell carcinoma with repression on PI3K/AKT/mTOR/HIF-1α pathway and c-Myc expression. Journal of Translational Medicine. 2021; 19: 209.
[9]
Luoto KR, Meng AX, Wasylishen AR, Zhao H, Coackley CL, Penn LZ, et al. Tumor cell kill by c-MYC depletion: role of MYC-regulated genes that control DNA double-strand break repair. Cancer Research. 2010; 70: 8748–8759.
[10]
Wang WJ, Wu SP, Liu JB, Shi YS, Huang X, Zhang QB, et al. MYC regulation of CHK1 and CHK2 promotes radioresistance in a stem cell-like population of nasopharyngeal carcinoma cells. Cancer Research. 2013; 73: 1219–1231.
[11]
Tan Y, Testa JR. DLX Genes: Roles in Development and Cancer. Cancers. 2021; 13: 3005.
[12]
Rubenstein JL, Nord AS, Ekker M. DLX genes and proteins in mammalian forebrain development. Development (Cambridge, England). 2024; 151: dev202684.
[13]
Zhang TJ, Xu ZJ, Gu Y, Wen XM, Ma JC, Zhang W, et al. Identification and validation of prognosis-related DLX5 methylation as an epigenetic driver in myeloid neoplasms. Clinical and Translational Medicine. 2020; 10: e29.
[14]
Wang J, Tao L, Liu Y, Liu H, Shen X, Tao L. Identification and validation of DLX4 as a prognostic and diagnostic biomarker for clear cell renal cell carcinoma. Oncology Letters. 2023; 25: 146.
[15]
Zhang J, Wu J, Chen Y, Zhang W. Dlx5 promotes cancer progression through regulation of CCND1 in oral squamous cell carcinoma (OSCC). Biochemistry and Cell Biology. 2021; 99: 424–434.
[16]
Tan Y, Cheung M, Pei J, Menges CW, Godwin AK, Testa JR. Upregulation of DLX5 promotes ovarian cancer cell proliferation by enhancing IRS-2-AKT signaling. Cancer Research. 2010; 70: 9197–9206.
[17]
Xu J, Testa JR. DLX5 (distal-less homeobox 5) promotes tumor cell proliferation by transcriptionally regulating MYC. The Journal of Biological Chemistry. 2009; 284: 20593–20601.
[18]
De Felice F, Tombolini V. Radiation therapy in renal cell carcinoma. Critical Reviews in Oncology/hematology. 2018; 128: 82–87.
[19]
Fabbrizi MR, Parsons JL. Radiotherapy and the cellular DNA damage response: current and future perspectives on head and neck cancer treatment. Cancer Drug Resistance (Alhambra, Calif.). 2020; 3: 775–790.
[20]
Biau J, Chautard E, Verrelle P, Dutreix M. Altering DNA Repair to Improve Radiation Therapy: Specific and Multiple Pathway Targeting. Frontiers in Oncology. 2019; 9: 1009.
[21]
Santivasi WL, Xia F. Ionizing radiation-induced DNA damage, response, and repair. Antioxidants & Redox Signaling. 2014; 21: 251–259.
[22]
Kuo LJ, Yang LX. Gamma-H2AX - a novel biomarker for DNA double-strand breaks. In Vivo (Athens, Greece). 2008; 22: 305–309.
[23]
Maliszewska-Olejniczak K, Dróżdż A, Waluś M, Dorosz M, Gryziński MA. Immunofluorescence Imaging of DNA Damage and Repair Foci in Human Colon Cancer Cells. Journal of Visualized Experiments: JoVE. 2020.
[24]
Rao VA, Agama K, Holbeck S, Pommier Y. Batracylin (NSC 320846), a dual inhibitor of DNA topoisomerases I and II induces histone gamma-H2AX as a biomarker of DNA damage. Cancer Research. 2007; 67: 9971–9979.
[25]
Priya B, Ravi S, Kirubakaran S. Targeting ATM and ATR for cancer therapeutics: Inhibitors in clinic. Drug Discovery Today. 2023; 28: 103662.
[26]
Cheng B, Pan W, Xing Y, Xiao Y, Chen J, Xu Z. Recent advances in DDR (DNA damage response) inhibitors for cancer therapy. European Journal of Medicinal Chemistry. 2022; 230: 114109.
[27]
Rundle S, Bradbury A, Drew Y, Curtin NJ. Targeting the ATR-CHK1 Axis in Cancer Therapy. Cancers. 2017; 9: 41.
[28]
Qiu Z, Oleinick NL, Zhang J. ATR/CHK1 inhibitors and cancer therapy. Radiotherapy and Oncology: Journal of the European Society for Therapeutic Radiology and Oncology. 2018; 126: 450–464.

Publisher’s Note: IMR Press stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share
Back to top