1 Department of Gastroenterology Surgery, Yichang Central People’s Hospital, The First College of Clinical Medical Science, China Three Gorges University, 443000 Yichang, Hubei, China
2 Department of Chemistry and Biochemistry, University of California, Santa Barbara, CA 93106, USA
3 Department of Chemistry, University of Warwick Coventry, CV4 7AL Coventry, UK
4 Center for Clinical Research and Translational Medicine, Yangpu Hospital, School of Medicine, Tongji University, 200438 Shanghai, China
Abstract
Dysregulated metabolic pathways, including glycolysis and a compromised DNA damage response (DDR), are linked to the progression of colorectal cancer (CRC). The mitotic arrest deficient-like 2 (MAD2L2) and aurora kinase B (AURKB) genes play roles in cell cycle regulation and the DDR, making them potential targets for CRC therapy.
Differential expression analysis was performed using The Cancer Genome Atlas-Colon Adenocarcinoma (TCGA-COAD) and GSE47074 datasets. A predictive model was established, and gene expression levels were further analyzed. The Gene Expression Profiling Interaction Analysis database and co-immunoprecipitation experiments assessed the correlation between AURKB and MAD2L2. Knockdown experiments in CRC cell lines further investigated the role of AURKB, followed by analyses of cell behavior, oxidative stress, glycolysis, DDR, and interaction with MAD2L2.
The risk model identified six prognostic genes (BUB1 mitotic checkpoint serine/threonine kinase B (BUB1B), AURKB, aurora kinase A (AURKA), exonuclease 1 (EXO1), topoisomerase II alpha (TOP2A), cyclin A2 (CCNA2)) associated with CRC, which were significantly expressed in tumor samples from the TCGA-COAD and GSE47074 datasets. In vitro assays confirmed that AURKB knockdown inhibited CRC cell behavior, induced G1 cell cycle arrest, and increased oxidative stress and apoptosis. AURKB knockdown also impaired glycolysis, reducing lactate production, glucose uptake, and ATP levels. Overexpression of MAD2L2 partially reversed these effects, restored glycolytic activity, and mitigated the cell cycle arrest and DDR caused by AURKB knockdown.
AURKB regulates CRC progression by modulating glycolysis and DDR pathways. Targeting the AURKB-MAD2L2 axis offers a promising therapeutic strategy for disrupting fundamental metabolic and DNA repair mechanisms in CRC.
Keywords
- colorectal cancer
- AURKB
- MAD2L2
- DNA damage response
- glycolysis
Colorectal cancer (CRC) is considered one of the most prevalent and lethal cancers globally, imposing a substantial health burden due to its high incidence and mortality [1]. According to global cancer statistics, approximately 1.9 million new cases of CRC are diagnosed each year, accounting for about 10% of all new cancer cases. About 935,000 people die of CRC each year, accounting for 9.2% of all cancer deaths [2]. The incidence of CRC significantly increases with age, especially in individuals over 50 years of age. However, in recent years, the incidence of this disease has also gradually increased in people younger than 50. Globally, the incidence of CRC is slightly higher in men than in women. The incidence of CRC is higher in North America, Europe, Australia, and New Zealand, whereas in some parts of Asia and Africa, the incidence is relatively low. However, it is worth noting that the incidence of CRC is increasing in East Asia. Risk factors for CRC include a family history of bowel cancer, unhealthy eating habits (e.g., high-fat, low-fiber diets), physical inactivity, smoking, and alcohol consumption [3]. Despite advancements in treatment approaches such as radiotherapy, chemotherapy, and surgical resection, the prognosis for advanced-stage CRC remains poor, highlighting the urgent need for more effective therapeutic strategies [4]. Recent research has increasingly recognized the role of metabolic reprogramming, particularly aerobic glycolysis, in cancer progression [5]. In several malignancies, such as breast and lung cancers, enhanced glycolysis is closely associated with aggressive tumor behavior and poor clinical outcomes. For example, in breast cancer, heightened glycolysis is correlated with increased tumor invasiveness [6], whereas in lung cancer, the overexpression of key glycolytic enzymes such as hexokinase 2 (HK2) drives tumor growth and metastasis [7]. The above findings highlight the critical necessity of targeting aerobic glycolysis during cancer treatment.
The serine/threonine kinase aurora kinase B (AURKB) is essential for chromosome segregation and cytokinesis, making it a significant focus in cancer research [8]. Abnormal AURKB activity during cell division can lead to chromosomal instability and uncontrolled cell proliferation [9], both of which are critical drivers of tumorigenesis. Research on CRC has revealed that inhibiting AURKB could elevate the susceptibility of CRC cells to fluorouracil, delay the development of chemoresistance, and improve chemotherapy outcomes [10]. Furthermore, in other malignancies such as thyroid cancer, AURKB has been tied to cancer development and inhibition of apoptosis [11], underscoring its significance in aggressive tumor behavior.
Mitotic arrest deficient-like 2 (MAD2L2) is another key regulator involved in mitotic checkpoint control and the DNA damage response (DDR) [12]. Its involvement in cancer has been extensively documented, particularly for its contribution to tumorigenesis and treatment resistance [13]. In ovarian cancer, high levels of MAD2L2 correlate with a poor prognosis, partly due to its role in repairing DNA double-strand breaks (DSBs) via the non-homologous end-joining pathway [14]. Similarly, in glioblastoma, MAD2L2 overexpression promotes tumor progression by enhancing cell survival and proliferation [13]. Emerging evidence suggests a functional connection between MAD2L2 and AURKB. In bladder cancer, AURKB has been shown to activate MAD2L2 expression while downregulating the p53 DDR pathway, which promotes cancer development [15]. Moreover, recent study indicates that MAD2L2 may influence aerobic glycolysis by regulating vital glycolytic enzymes, contributing to the metabolic reprogramming essential for cancer progression [16].
The DDR is essential for maintaining genomic integrity by detecting and repairing DNA damage [17]. In CRC, dysregulation of the DDR pathway contributes to genomic instability and tumor progression [18]. Alterations in homologous recombination repair (HRR) are increasingly recognized as potential therapeutic targets in CRC [18]. For example, DSBs are crucial for inducing cell death during radiotherapy [19]. However, CRC stem cells can enhance radio resistance by activating cell cycle checkpoints and DDR pathways, facilitating efficient DNA repair and promoting tumor cell survival [20]. This enhanced repair capacity complicates treatment and underscores the potential of targeting DDR mechanisms. The development of DDR-targeted therapies, such as poly (Adenosine diphosphate (ADP)-ribose) polymerase inhibitors, shows promise, especially in treating cancers with HRR deficiencies [21]. Given the complexity and heterogeneity of CRC, understanding the interplay among AURKB, MAD2L2, and the DDR can unveil new therapeutic strategies and provide valuable insights into CRC treatment and prognosis.
Despite advances in CRC research, the exact mechanisms of CRC development have not been fully elucidated. The goal of this study was to clarify the roles of AURKB and MAD2L2 in CRC progression, focusing on their contributions to glycolysis and DDR. Given the importance of these pathways in tumor growth and survival, understanding the connection between AURKB and MAD2L2 may reveal novel therapeutic strategies.
The Gene Expression Omnibus database (https://www.ncbi.nlm.nih.gov/gds/?term=GSE47074) provided the GSE47074 dataset, which included four primary colon cancer samples (tumor group) and four standard samples (control group). In addition, The Cancer Genome Atlas (TCGA) (https://tcga-data.nci.nih.gov/tcga/) database provided 414 colon adenocarcinoma (COAD) samples (tumor group) and 41 standard samples (control group). Differentially expressed genes (DEGs) in the two datasets were obtained separately utilizing the limma package in R studio (version 4.0, http://www.bioconductor.org/packages/release/bioc/html/limma.html). Genes with fold change (FC)
Next, protein–protein interaction (PPI) network analysis was conducted on overlapping genes utilizing the Search Tool for the Retrieval of Interacting Genes (STRING) (https://string-db.org/) database. Subsequently, the Cytohubba plugin in Cytoscape (version 3.8.2, IBM, Armonk, NY, USA) was used to apply the maximum clique centrality (MCC), maximum neighborhood component (MNC), and degree algorithms to identify the top 20 genes of three key modules. Lastly, we performed intersection analysis of the genes from these three network modules to pinpoint the essential intersection genes critical for further investigation.
To investigate the 15 essential intersection genes in the TCGA-COAD dataset, we implemented the least absolute shrinkage and selection operator (LASSO) Cox regression algorithm using the glmnet package in R (version 4.0, http://cran.r-project.org/web/packages/glmnet/index.html). A LASSO Cox regression model was built with 10-fold cross-validation, plotting the relationship between the logarithm of lambda (
The association of AURKB and MAD2L2 expression levels was investigated utilizing the Gene Expression Profiling Interactive Analysis (GEPIA) database (http://gepia.cancer-pku.cn/) to learn more about the link between the two. The correlation coefficient across the two genes was obtained. p
CRC cell lines (LoVo, HCT-116, and SW480) and the NCM460 normal human colon mucosal epithelial cell line were purchased from the American Type Culture Collection (ATCC, Shanghai, China). Cells were cultured in Dulbecco’s Modified Eagle Medium supplemented with 10% fetal bovine serum, 100 U/mL penicillin, and streptomycin (all from Sangon, Shanghai, China) at 37 °C in a humidified incubator with 5% CO2. All cell lines were validated by STR profiling and tested negative for mycoplasma. Detailed validation results are provided in Supplementary Fig. 1.
CRC cells were plated in 6-well dishes at a density of 5
Total RNA was extracted from CRC cells using TRIzol reagent (9109, Takara, Dalian, China), and complementary DNA (cDNA) was synthesized using the PrimeScript™ RT Reagent Kit (RR047A, Takara, Dalian, China). SYBR Green (RR820A, Takara, Dalian, China)-based quantitative PCR (qPCR) was conducted on the CFX96 Real-Time PCR Machine (1855195, Bio-Rad, Hercules, CA, USA) according to standard qPCR procedures. The 2-ΔΔCt method was employed to quantify the gene expression levels, with GAPDH serving as an internal control. The range of primer sequences utilized in qPCR is summarized in Table 1.
| Target | Direction | Sequence (5′-3′) |
| AURKB | Forward | AGATCGAAATCCAGGCCCAC |
| AURKB | Reverse | CCTCCATGATCGTGGCTGTT |
| Cyclin D1 | Forward | GATGCCAACCTCCTCAACGA |
| Cyclin D1 | Reverse | ACTTCTGTTCCTCGCAGACC |
| p27 | Forward | CAGCTTGCCCGAGTTCTACT |
| p27 | Reverse | CGACGGATCAGTCTTTGGGT |
| p21 | Forward | AGTCAGTTCCTTGTGGAGCC |
| p21 | Reverse | CATTAGCGCATCACAGTCGC |
| Bcl-2 | Forward | AAAAATACAACATCACAGAGGAAGT |
| Bcl-2 | Reverse | GACGAGGGGGTGTCTTCAAT |
| Bax | Forward | TGATGGACGGGTCCGGG |
| Bax | Reverse | TGAGACACTCGCTCAGCTTC |
| Caspase-3 | Forward | TGTGAGGCGGTTGTAGAAGAGT |
| Caspase-3 | Reverse | CTTTATTAACGAAAACCAGAGCGCC |
| PKM2 | Forward | CGAGCCTCAAGTCACTCCAC |
| PKM2 | Reverse | GACGAGCTGTCTGGGGATTC |
| LDHA | Forward | CTCTGGCAAAGTGGATATCTTGAC |
| LDHA | Reverse | CTCCATACAGGCACACTGGA |
| HK2 | Forward | TGTGAATCGGAGAGGTCCCA |
| HK2 | Reverse | TCCGGAGACGTGATTTTGGC |
| GLUT1 | Forward | GAGCAGCTACCCTGGATGTC |
| GLUT1 | Reverse | GAGGTCCAGTTGGAGAAGCC |
| MAD2L2 | Forward | CAAAGGAGGCAGACAAAGGCG |
| MAD2L2 | Reverse | GTAGACCTCGCGCACGTAGA |
| p53 | Forward | TGACACGCTTCCCTGGATTG |
| p53 | Reverse | TCCGGGGACAGCATCAAATC |
| H2A.X | Forward | GGTGCTTAGCCCAGGACTTT |
| H2A.X | Reverse | CCCAGCGCAGACCTATGAAT |
| GAPDH | Forward | CATGTTGCAACCGGGAAGGA |
| GAPDH | Reverse | ATCACCCGGAGGAGAAATCG |
qPCR, quantitative PCR; AURKB, aurora kinase B; Bcl-2, B-cell lymphoma 2; Bax, Bcl-2-associated X protein; PKM2, pyruvate kinase M2; LDHA, lactate dehydrogenase A; HK2, hexokinase 2; GLUT1, glucose transporter 1; MAD2L2, mitotic arrest deficient-like 2; H2A.X, histone family member X; GAPDH, Glyceraldehyde 3-phosphate dehydrogenase.
Proteins were extracted from cultured cells with Radioimmunoprecipitation Assay (RIPA) buffer (P0013B, Beyotime, Shanghai, China) containing phosphatase and protease inhibitors. Then the cell lysate was centrifuged for 20 min, and proteins were quantified with a bovine serum albumin (BSA) protein assay kit (P0010, Beyotime, Shanghai, China). Equal protein concentrations were resolved by sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE) and electrotransferred to PVDF membranes (Beyotime, shanghai, China). Then the membranes were incubated overnight at 4 °C with the following primary antibodies (all from Abcam, Shanghai, China): AURKB (1:1000, ab2254), cyclin D1 (1:10,000, ab134175), p27 (1:5000, ab32034), p21 (1:1000, ab109520), B-cell lymphoma 2 (Bcl-2; 1:2000, ab182858), Bcl-2-associated X protein (Bax, 1:1000, ab32503), caspase-3 (1:5000, ab32351), pyruvate kinase M2 (PKM2; 1:10,000, 15822-1-AP), lactate dehydrogenase A (LDHA; 1:5000, ab52488), hexokinase 2 (HK2; 1:1000, ab209847), glucose transporter 1 (GLUT1; 1:200, ab150299), MAD2L2 (1:1000, ab180579), p53 (1:1000, ab32049), H2A histone family member X (H2A.X; 1:5000, ab140498), and GAPDH (1:5000, ab8245). After three washes with Tris-Buffered Saline with Tween 20 (TBST), membranes were incubated with horseradish peroxidase conjugated anti-mouse (1:1000, A0216, Beyotime, Shanghai, China) and anti-rabbit (1:1000, A0208, Beyotime, Shanghai, China) antibodies, and placed in an overnight incubation at 4 °C. Proteins were detected using enhanced chemiluminescence (ECL; Beyotime) and quantified with ImageJ software (version 1.8.0, National Institutes of Health, Bethesda, MD, USA).
After centrifugation of HCT-116 and LoVo cells, 500 µg protein from each sample was incubated overnight at 4 °C with 2 µg of either anti-AURKB (1:30; ab45145, Abcam), anti-MAD2L2 (1:40; ab180579, Abcam), or IgG control (1:200; ab200699, Abcam) . Then the samples were incubated for 2 h with Protein A/G agarose beads (Santa Cruz Biotechnology, Dallas, TX, USA). Boiling in SDS-PAGE loading buffer allowed bound proteins to be eluted from the beads after they had been cleaned with RIPA buffer (P0013B, Beyotime, Shanghai, China). Following SDS-PAGE and electrotransfer to PVDF membranes, the proteins were detected by Western blotting using anti-AURKB and anti-MAD2L2 antibodies. Protein detection was performed using an ECL detection system (P0018S, Beyotime, Shanghai, China).
To measure cell viability, CRC cells were trypsinized and plated in 96-well plates at a density of 2
A Transwell assay was conducted to measure the invasive capabilities of CRC cells. The cells were seeded in the upper chamber of Transwell (Corning, Shanghai, China) that were pre-coated with 80 µL Matrigel (BD Biosciences, Shanghai, China). Then 600 µL complete medium was added to the lower chamber, and 200 µL serum-free media containing 1
Flow cytometry was employed to evaluate cell cycle distribution. After washing cells with cold phosphate-buffered saline (PBS), CRC cells were fixed in 70% ethanol overnight at 4 °C. Then the cells were washed, stained with propidium iodide (PI) solution, and incubated for 30 min at room temperature in the dark. The FACSCalibur™ flow cytometer (342973, BD Biosciences, San Jose, CA, USA) was utilized for flow cytometry, and data were evaluated with FlowJo software (version 10.0, BD Biosciences, San Jose, CA, USA) to quantify the cell cycle phases (G1, S, G2).
Apoptosis was assessed using an Annexin V-FITC/PI staining kit (556547, BD Biosciences, San Jose, CA, USA). After washing with cold PBS, CRC cells (1
To measure the levels of malondialdehyde (MDA) (cat.no. S0131S), LDH (cat.no. C0016), and superoxide dismutase (SOD) (cat. no. S0101S), detection assay kits were used (Beyotime, Shanghai, China). Briefly, CRC cells (5
The levels of intracellular reactive oxygen species (ROS) of CRC cells were measured using a ROS detection kit (E004-1-1, Nanjing Jiancheng Bioengineering Institute, Nanjing, China). Briefly, HCT-116 and LoVo cells were plated at a density of 2
Lactate levels and glucose uptake levels in CRC cells were assessed using lactate (cat.no. A019-2-1) and glucose assay kits (cat.no. F006-1-1), respectively, according to the manufacturer’s instructions (Nanjing Jiancheng Bioengineering Institute, Nanjing, China). Briefly, 200 µL culture medium per well of 96-well plates were used to seed 2
Intracellular ATP levels were measured using an ATP assay kit (S0026, Beyotime, shanghai, China). Briefly, after collecting the cells, they were lysed with 200 µL lysis buffer and centrifuged at 12,000 rpm at 4 °C; the supernatant was removed as the test sample. Subsequently, 100 µL ATP detection reagent and 10 µL test sample were mixed following the manufacturer’s instructions, and the luminescence was measured using a NanoDrop spectrophotometer (ND-2000, Thermo Fisher Scientific, Waltham, MA, USA). A standard curve was used to calculate the relative ATP levels, which then were normalized to cell number or protein concentration to ensure accurate comparisons.
R was used as the programming language for data analyses. Two groups were compared using the unpaired Student’s t-test. One-way analysis of variance and Tukey’s post hoc test were used for multiple group comparisons. p
Initially, we conducted differential expression analysis on the GSE47074 and TCGA-COAD datasets. Fig. 1A shows 1521 upregulated and 1171 downregulated DEGs in the TCGA-COAD dataset, whereas Fig. 1B shows 568 upregulated and 343 downregulated DEGs in the GSE47074 dataset. Subsequently, the DEGs in the two datasets were analyzed, identifying 319 intersecting upregulated DEGs and 171 downregulated DEGs (Fig. 1C). Subsequently, the intersection genes were further explored by PPI network analysis. Cytoscape illustrated the top 20 genes per MCC, MNC, and Degree algorithms. The MCC network contained 20 points and 190 edges (Fig. 1D), and the MNC network and Degree network contained 20 points and 189 edges (Fig. 1E,F). The top 20 genes of the three algorithms were analyzed again, and 15 essential CRC genes were obtained (Fig. 1G).
Fig. 1. Selection of differentially expressed genes and PPI network analysis based on TCGA-COAD and GSE47074 datasets to screen essential intersection genes. (A) Volcano plot of DEGs in the TCGA-COAD dataset, showing 1521 upregulated and 1171 downregulated DEGs. (B) Volcano plot of DEGs in the GSE47074 dataset, with 568 upregulated and 343 downregulated DEGs. (C) Cross-analysis Venn diagram of 1521 upregulated DEGs of TCGA-COAD, 1171 downregulated DEGs of TCGA-COAD, 568 upregulated DEGs of GSE47074, and 343 downregulated DEGs of GSE47074. (D–F) PPI network analysis of intersecting genes using the MCC (D), MNC (E), and Degree (F) algorithms, visualizing the top 20 genes in each network. (G) Venn diagram showing the overlapping regions of the three different network centers of MCC, MNC, and Degree, showing 15 essential intersection genes. TCGA-COAD, The Cancer Genome Atlas-Colon Adenocarcinoma; DEGs, differentially expressed genes; PPI, protein-protein interaction; MCC, maximum correlation criterion; MNC, maximum neighborhood component.
We performed LASSO Cox regression analysis for 15 intersecting genes to identify the critical genes connected with COAD prognosis. The LASSO coefficient spectrum of the candidate genes is shown in Fig. 2A. The optimal value of the regularization parameter (
Fig. 2. Construct a prognostic-related risk model through LASSO regression analysis. (A) Processes of LASSO Cox model fitting. Each curve represents a CRC-related gene. (B) Selection of the optimal
TCGA-COAD and GSE47074 datasets were used to examine the expression of these six genes (BUB1B, AURKB, AURKA, EXO1, TOP2A, and CCNA2), and it was discovered that all of the genes had greater expression levels in the tumor groups of both datasets (Fig. 3A,B). Among these, AURKB was an interesting gene due to its function in controlling mitosis, a critical stage of the cell cycle. Disruption of mitosis by DNA damage can severely impair cell division, and aerobic glycolysis is known to supply the energy necessary for the DDR [23]. Given its potential involvement in the aerobic glycolysis of CRC cells, AURKB was determined as the core gene in this investigation, warranting further investigation into its role in CRC progression.
Fig. 3. Expression of six prognosis-related genes. (A) Box plot of the expression levels of six prognostic genes (BUB1B, AURKB, AURKA, EXO1, TOP2A, and CCNA2) in COAD versus normal tissues in TCGA-COAD dataset. (B) Box plot of the expression levels of six prognostic genes (BUB1B, AURKB, AURKA, EXO1, TOP2A, and CCNA2) in normal and tumor samples in the GSE47074 dataset. *p
To investigate the function of the hub gene AURKB in CRC, qPCR was carried out to assess AURKB expression in NCM460 cells and three CRC cell lines. The findings revealed that the AURKB mRNA levels in the three CRC cells were significantly higher than those in normal colon epithelial cells, of which the expression of AURKB in LoVo and HCT-116 cells was the highest (Fig. 4A). This finding was corroborated by Western blot analysis, as illustrated in Fig. 4B,C. Subsequently, the expression of AURKB was knocked down in CRC cells, and the findings demonstrated a significant reduction in expression levels following AURKB knockdown compared to the control group (Fig. 4D–F). CCK-8 assays showed that the proliferation of CRC cells was reduced following AURKB silencing (Fig. 4G,H). A transwell assay was carried out to assess the invasion and migration of LoVo and HCT-116 cells following AURKB knockdown. The results showed that knockdown of AURKB significantly inhibited the metastasis of CRC cells (Fig. 4I,J).
Fig. 4. Knockdown of AURKB inhibits CRC cell proliferation, invasion, and migration. (A) The qPCR data of AURKB mRNA expression in NCM450, HCT-116, LoVo, and SW480 cells. (B,C) Western blot analysis of AURKB protein expression levels in NCM450, HCT-116, LoVo, and SW480 cells. (D) qPCR data of AURKB mRNA expression in HCT-116 and LoVo cells after AURKB knockdown. (E,F) Western blot analysis of AURKB protein expression levels in HCT-116 and LoVo cells after AURKB knockdown. (G,H) CCK-8 assay detected the proliferation of HCT-116 (G) and LoVo (H) cells on days 0–4 after knockdown of AURKB. The X-axis is time (days), and the Y-axis is the optical density value when the absorbance was 450 nm. (I,J) Transwell assay detected the invasion and migration abilities of HCT-116 (I) and LoVo. Scale bar: 50 µm. (J) Cells after AURKB knockdown. Scale bar: 50 µm. *p
After AURKB knockdown, flow cytometry results showed that the proportion of cells in the G1 phase significantly increased, whereas the proportion of cells in the S phase significantly decreased. These results suggest that AURKB knockdown blocked cells in the G1 phase by inhibiting the G1/S transition (Fig. 5A,B). The mRNA expression of key cell cycle regulators, including cyclin D1, p27, and p21, were further examined by qPCR. The results showed that AURKB knockdown significantly downregulated cyclin D1 and upregulated p27 and p21 expression in CRC cells (Fig. 5C,D). Western blot analysis confirmed these findings at the protein level, showing decreased protein expression of cyclin D1 and increased p27 and p21 expression after AURKB knockdown in both cell lines (Fig. 5E–G). These findings indicate that AURKB knockdown causes G1 arrest in CRC cells, possibly through downregulating cyclin D1 and upregulating cell cycle inhibitors p27 and p21.
Fig. 5. Effects of AURKB knockdown on cell cycle distribution and cell cycle regulators in CRC cells. (A,B) Flow cytometry analysis of cell cycle distribution in HCT-116 (A) and LoVo (B) cells transfected with control siRNA (si-NC) or AURKB-specific siRNA (si-AURKB-1 and si-AURKB-2). The bar graph on the right summarizes the percentage of cells in each cell cycle phase (G1, S, G2). (C,D) qPCR analysis of cyclin D1, p27, and p21 mRNA expression levels in HCT-116 (C) and LoVo (D) cells after AURKB knockdown. (E–G) Western blot analysis of protein expression of cycle-related proteins cyclin D1, p27, and p21 in HCT-116 and LoVo cells after AURKB knockdown. *p
The effect of AURKB knockdown on apoptosis of colon cancer cells was analyzed by flow cytometry, and an apoptosis rate of approximately 8% was observed (Fig. 6A,B). In addition, qPCR and western blotting were conducted to evaluate the expression changes of key apoptotic markers in CRC cells. Consistent with the flow cytometry results, knockdown of AURKB enhanced the expression of caspase-3 and Bax and decreased the expression of Bcl-2 (Fig. 6C–G). We observed significant changes in the expression patterns of apoptosis marker genes. This discrepancy between the relatively low apoptosis rate and the significant changes in expression of apoptosis-associated genes may stem from the complexity of the apoptosis pathway. Apoptosis is a complex process that involves multiple signaling pathways and feedback mechanisms. Knockdown of AURKB may have affected multiple apoptosis-associated genes and proteins, but these changes did not all directly lead to cell death. In this process, some genes may play a regulatory role mainly in the apoptotic pathway rather than directly inducing cell death. This complex regulatory network may lead to discrepancies between significant changes in gene expression patterns and the apoptosis rates detected by flow cytometry. Our findings suggest that knockdown of AURKB induces apoptosis in CRC cells by regulating the expression of critical apoptotic regulators. Since si-AURKB-1 showed more pronounced effects in inhibiting cell growth and promoting apoptosis, we selected it for subsequent analyses.
Fig. 6. AURKB knockdown induces apoptosis in CRC cells. (A,B) Flow cytometry analysis of apoptosis in HCT-116 (A) and LoVo (B) cells transfected with control siRNA (si-NC) or AURKB-specific siRNA (si-AURKB-1 and si-AURKB-2). The bar graph on the right summarizes the percentage of apoptotic cells in each group. (C,D) qPCR data of mRNA expression of apoptosis-related markers Bax, Bcl-2, and caspase-3 in HCT-116 and LoVo after AURKB knockdown. (E–G) Western blot analysis of protein expression of apoptosis-related proteins Bax, Bcl-2, and caspase-3 in HCT-116 and LoVo cells after AURKB knockdown. *p
To assess the effect of AURKB knockdown on oxidative stress in CRC cells, we used detection kits to measure LDH, ROS, MDA, and SOD expression levels in LoVo and HCT-116 cells. As shown in Fig. 7A, knockdown of AURKB significantly increased LDH activity in CRC cells, indicating increased cell membrane damage. Similarly, ROS levels were markedly elevated following AURKB knockdown in both cell lines (Fig. 7B), suggesting increased oxidative stress. Additionally, MDA levels, a marker of lipid peroxidation, were significantly higher in AURKB knockdown cells than controls (Fig. 7C). Conversely, SOD activity, which serves as an antioxidant defense mechanism, was significantly reduced in cells with AURKB knockdown (Fig. 7D). These findings indicate that AURKB knockdown in CRC cells leads to increased oxidative stress, cellular damage, and lipid peroxidation, which may be involved in the induced apoptosis.
Fig. 7. Knockdown of AURKB affects oxidative stress in CRC cells. (A) Relative LDH activity in HCT-116 and LoVo cells transfected with control siRNA (si-NC) or AURKB-specific siRNA (si-AURKB-1). (B) Relative ROS levels in HCT-116 and LoVo cells following AURKB knockdown. (C) Relative MDA levels in HCT-116 and LoVo cells after AURKB knockdown. (D) Relative SOD levels in HCT-116 and LoVo cells after AURKB knockdown. *p
Through kit testing, it was found that AURKB knockdown significantly reduced ATP production, glucose uptake, and lactate production in CRC cells compared with the control group (Fig. 8A–C). qPCR was conducted to assess the expression levels of glycolysis-related genes (PKM2, LDHA, HK2, GLUT1) in CRC cells. Compared to the control group, the expression of glycolysis-related genes was significantly downregulated after AURKB knockdown (Fig. 8D,E). Western blot analysis confirmed that AURKB knockdown led to decreased protein expression of PKM2, LDHA, HK2, and GLUT1 in CRC cells (Fig. 8F–H). These results indicate that AURKB knockdown impairs glycolysis and energy production in CRC cells by downregulating critical glycolytic enzymes and glucose transporters, thereby decreasing ATP levels.
Fig. 8. Effects of AURKB knockdown on glycolysis and ATP production in CRC cells. (A) Relative lactate production in HCT-116 and LoVo cells transfected with control siRNA (si-NC) or AURKB-specific siRNA (si-AURKB-1). (B) Relative glucose uptake in HCT-116 and LoVo cells following AURKB knockdown. (C) ATP levels in HCT-116 and LoVo cells after AURKB knockdown. (D,E) qPCR data of mRNA expression of glycolysis-related genes PKM2, LDHA, HK2, and GLUT1 in HCT-116 and LoVo cells after AURKB knockdown. (F–H) Western blot analysis of protein expression of glycolysis-related proteins PKM2, LDHA, HK2, and GLUT1 in HCT-116 and LoVo cells after AURKB knockdown. *p
The GEPIA database analyzed the expression correlation between AURKB and MAD2L2. Bioinformatics analysis revealed a significant positive relationship between the expression levels of AURKB and MAD2L2, with a correlation coefficient of 0.415 (Fig. 9A). The connection between AURKB and MAD2L2 was further elucidated by co-immunoprecipitation experiments, which further confirmed that AURKB can interact with MAD2L2 in LoVo and HCT-116 cells (Fig. 9B). Subsequently, qPCR was used to detect the significant overexpression efficiency of MAD2L2 in CRC cells, and Western blotting experimental results also demonstrated effective knockdown (Fig. 9C–E). The CCK-8 assay demonstrated that MAD2L2 overexpression significantly reversed the proliferation of CRC cells inhibited by AURKB knockdown (Fig. 9F,G). In addition, Transwell experiments performed in CRC cells showed that MAD2L2 overexpression rescued the inhibitory effects of AURKB knockdown on cell behavior (Fig. 9H,I).
Fig. 9. MAD2L2 overexpression rescues the inhibitory effects of AURKB knockdown on CRC cell proliferation, migration, and invasion. (A) Correlation analysis between AURKB and MAD2L2 expression in CRC detected by the GEPIA database. (B) Co-IP demonstrated the interaction of AURKB and MAD2L2 in HCT-116 and LoVo cells. (C) qPCR data of MAD2L2 mRNA expression in HCT-116 and LoVo after MAD2L2 overexpression. (D,E) Western blot analysis of MAD2L2 protein expression levels in HCT-116 and LoVo cells after MAD2L2 overexpression. (F,G) CCK-8 assay was used to detect the proliferation of control, si-AURKB-1, and si-AURKB-1+over-MAD2L2 treated HCT-116 and LoVo cells from day 0 to day 4. (H,I) Transwell assay detected the invasion and migration abilities of Control, si-AURKB-1, and si-AURKB-1+over-MAD2L2 treated HCT-116 and LoVo cells. Scale bar: 50 μm. *p
To understand the impact of MAD2L2 overexpression on the CRC cell cycle and DDR in the context of AURKB knockdown, we evaluated the expression of key cell cycle proteins (cyclin D1, p21, and p53) and DNA damage marker H2A.X in CRC cells. qPCR analysis showed that AURKB knockdown significantly reduced cyclin D1 mRNA expression while increasing p53, p21, and H2A.X expression in CRC cells (Fig. 10A,B). However, overexpression of MAD2L2 partially reversed these effects, resulting in the restoration of cyclin D1 levels and decreased expression of p53, p21, and H2A.X. Western blot analysis confirmed these findings at the protein level (Fig. 10C–E). These results suggest that MAD2L2 overexpression can mitigate the impact of AURKB knockdown on DDR and cell cycle progression, highlighting the potential compensatory mechanism in CRC cells. We also observed a decrease in the mRNA and protein expression of MAD2L2 after knockdown of AURKB, suggesting that AURKB may have regulatory effects on the expression level of MAD2L2. However, we also found that overexpression of MAD2L2 did not restore the decrease in AURKB mRNA and protein expression caused by AURKB knockdown. This finding suggests that MAD2L2 may not be directly involved in regulating AURKB expression despite the effect of AURKB on MAD2L2 expression.
Fig. 10. MAD2L2 overexpression mitigates the effects of AURKB knockdown on cell cycle regulators and the p53 DDR pathway. (A,B) qPCR was used to detect the mRNA expression levels of AURKB, MAD2L2, cyclin D1, p53, p21, and H2A.X in the control group, si-AURKB-1, si-AURKB-1+over-MAD2L2 treated HCT-116 and LoVo cells. (C–E) Western blot analysis was used to detect the protein expression levels of AURKB, MAD2L2, cyclin D1, p53, p21, and H2A.X in the control group, si-AURKB-1, si-AURKB-1+over-MAD2L2 treated HCT-116 and LoVo cells. * p
Next, ATP levels, glucose uptake, and lactate production in CRC cells were assessed. In contrast to the control, AURKB knockdown inhibited ATP levels, lactate production, and glucose uptake, while MAD2L2 overexpression reversed this inhibitory phenomenon (Fig. 11A–C). The expression of key glycolytic enzymes at the mRNA and protein levels was further evaluated. qPCR analysis showed that AURKB knockdown significantly reduced the mRNA levels of these glycolysis-related genes in CRC cells (Fig. 11D,E). Western blot analysis showed that PKM2, LDHA, HK2, and GLUT1 protein levels were significantly reduced after AURKB knockdown (Fig. 11F). Notably, MAD2L2 overexpression reversed these effects and restored PKM2, LDHA, HK2, and GLUT1 levels in AURKB knockdown cells (Fig. 11G,H). These results indicate that MAD2L2overexpression can restore glycolytic activity and ATP production impaired by AURKB knockdown, highlighting its role in maintaining CRC cell metabolism.
Fig. 11. Effects of MAD2L2 overexpression on glycolysis and ATP production in AURKB knockdown CRC cells. (A) Lactate production levels in control, si-AURKB-1, si-AURKB-1+over-MAD2L2 treated HCT-116 and LoVo cells. (B) Glucose uptake levels in control, si-AURKB-1, si-AURKB-1+over-MAD2L2 treated HCT-116 and LoVo cells. (C) ATP levels in control, si-AURKB-1, si-AURKB-1+over-MAD2L2 treated HCT-116 and LoVo cells. (D,E) qPCR was used to detect the mRNA expression levels of glycolysis-related genes PKM2, LDHA, HK2, and GLUT1 in the control group, si-AURKB-1, si-AURKB-1+over-MAD2L2 treated HCT-116 and LoVo cells. (F–H) Western blot analysis was used to detect the protein expression levels of glycolysis-related proteins PKM2, LDHA, HK2, and GLUT1 in the control group, si-AURKB-1, si-AURKB-1+over-MAD2L2 treated HCT-116 and LoVo cells. *p
CRC is a major worldwide health challenge because of its high morbidity and death rates. Identifying critical genes involved in CRC progression has been a central focus of recent research. Notable genes such as APC regulator of WNT signaling pathway (APC), tumor protein p53 (TP53), and KRAS proto-oncogene, GTPase (KRAS) have been extensively studied for their roles in CRC. APC mutations are commonly linked to early-stage CRC [24], while TP53 mutations are associated with advanced disease and poor prognosis [25]. KRAS mutations, on the other hand, are often related to chemotherapy resistance, influencing treatment outcomes [26]. In this research, we obtained six CRC prognosis-related genes through bioinformatics analysis. AURKA is linked to a poor prognosis in various cancers and regulates the wingless/integrated (Wnt) and RAS-mitogen-activated protein kinase (Ras-MAPK) signaling pathways, promoting CRC development. BUB1B, critical for mitotic checkpoints and genomic stability, is closely linked to CRC development [27]. CCNA2, a crucial cell cycle regulator, drives CRC cell proliferation [28]. EXO1 participates in DNA damage repair and is essential for maintaining genomic stability, with its dysfunction leading to instability [29]. TOP2A is crucial for DNA topology regulation and cancer cell proliferation [30]. AURKB, essential for cell division and mitosis, was identified as a critical gene in this study, with its role in glycolysis and DDR in CRC cells warranting further investigation.
AURKB is a crucial regulator of mitosis, chromosome segregation, and cytokinesis, with significant implications for cancer cell behavior. Studies have demonstrated that AURKB overexpression promotes proliferation and invasion while inhibiting apoptosis in various cancers, including breast cancer and hepatocellular carcinoma [31, 32]. In prostate cancer, AURKB overexpression is associated with a poor prognosis and increased tumor aggressiveness [33]. Tumor cell cycle regulation is tightly controlled by proteins such as p27 and p21, which, when downregulated, are linked to accelerated tumor growth and poor outcomes in CRC [34]. Apoptosis-related proteins such as Bcl-2, known for their anti-apoptotic properties, are also upregulated in CRC, contributing to tumor progression [35]. Building upon this knowledge, our study focused on the function of AURKB in CRC. We discovered that AURKB expression was significantly higher in CRC cells than in normal cells. Knockdown of AURKB in CRC cells decreased invasion and proliferation, caused cell cycle arrest, and increased apoptosis. These outcomes offer essential perspectives regarding the molecular processes by which AURKB contributes to CRC progression.
Oxidative stress, because it damages cells and tissuesand is characterized by an imbalance between the production of ROS and antioxidant defenses, plays an essential role in cancer pathogenesis. Critical factors in this process include LDH, ROS, MDA, and SOD [36]. In CRC, elevated ROS levels inhibit cell growth and migration, and increased MDA levels indicate heightened oxidative damage in CRC tissues compared to normal tissues [37]. Concurrently, aerobic glycolysis, the phenomenon where tumor cells preferentially produce ATP by glycolysis even under aerobic circumstances, is a characteristic of cancer metabolism [38]. Greater lactate generation and better glucose absorption are the hallmarks of this metabolic change, contributing to an acidic tumor microenvironment that promotes invasion and immune evasion in CRC cells [39]. Elevated ATP levels support the invasive behavior of these cells [40], and overexpression of glycolysis-related genes such as HK2 and GLUT1 enhances glucose metabolism in CRC [41, 42]. We explored the impact of AURKB knockdown on oxidative stress and glycolysis in CRC cells. Our results revealed that AURKB knockdown disrupted cellular oxidative balance and inhibited aerobic glycolysis, demonstrating that AURKB is essential for maintaining oxidative stress and glycolytic pathways in CRC cells. Therefore, targeting AURKB could impair the metabolism and viability of CRC cells by disrupting their oxidative stress response and glycolytic activity.
The DDR is critical for maintaining genomic stability by detecting and repairing DNA damage. Dysregulation of the DDR pathway in CRC leads to uncontrolled cell proliferation and tumorigenesis [43]. Defects in DDR mechanisms result in the accumulation of mutations, driving CRC progression and resistance to therapy [44]. Similarly, p53, a critical tumor suppressor involved in DDR, is often mutated in CRC, leading to impaired cell cycle control and resistance to apoptosis [25]. These disruptions in DDR promote genomic instability and contribute to the aggressive behavior of CRC cells. MAD2L2, a mitotic spindle assembly checkpoint component, is essential for cell cycle regulation and proper chromosome segregation. Dysregulation of MAD2L2 has been noted in various cancers, such as CRC [45]. In CRC, MAD2L2 overexpression is linked to poor prognosis and enhanced tumor invasiveness. According to a study, high levels of MAD2L2 are related to increased proliferation, resistance to apoptosis, and the promotion of metastasis, underscoring its role in enhancing the invasive and migratory capabilities of CRC cells [46]. Our study further revealed a direct functional interaction between AURKB and MAD2L2 in CRC progression, which provides a basis for understanding their synergistic effects. In addition, after AURKB knockdown, we observed a significant decrease in the expression MAD2L2, and the overexpression of MAD2L2 did not restore the reduction in AURKB mRNA and protein expression caused by AURKB knockdown, suggesting that AURKB may positively regulate the expression of MAD2L2. This regulatory relationship suggests that AURKB may be critical in maintaining MAD2L2 function. Moreover, overexpression of MAD2L2 partially reversed the effects of AURKB knockdown on CRC cells, restoring cell proliferation, migration, and invasion abilities and attenuating the effects of cell cycle arrest and the DDR. These findings suggest that MAD2L2 may play a compensatory role in maintaining cell metabolism and cell cycle progression after AURKB knockdown. Finally, AURKB and MAD2L2 jointly regulate the p53 DDR pathway and aerobic glycolysis, critical pathways for CRC progression. The effects of AURKB knockdown and MAD2L2 overexpression on these pathways suggest that they may synergistically regulate metabolism and DNA repair mechanisms in CRC cells. The findings of this study underscore the pivotal role of the AURKB-MAD2L2 axis in modulating p53 DDR and metabolic pathways, highlighting its potential as a therapeutic target of CRC.
In this study, we revealed the potential role of AURKB and MAD2L2 in CRC progression by in vitro experiments. However, we must recognize certain limitations in translating the results from in vitro experiments to clinical applications. First, in vitro, experimental conditions cannot fully mimic the complex tumor microenvironment in the human body. Factors such as tumor-host immune system interactions, tumor heterogeneity, and dynamic changes during tumor development are difficult to adequately capture in in vitro models. These factors may impact the expression and function of AURKB and MAD2L2, thus limiting the generalizability of our results. Second, in vitro experiments usually cannot assess long-term therapeutic and side effects, which are very important considerations in clinical applications. Assessing these factors in an in vivo model is essential to determine the feasibility of AURKB and MAD2L2 as therapeutic targets. In addition, there are significant individual differences and tumor heterogeneity among CRC patients, which may affect the expression and function of AURKB and MAD2L2. Our in vitro study failed to fully capture this heterogeneity, which may limit the generalizability of our results. Furthermore, CRC frequently develops resistance to therapy, and in vitro experiments may not be able to model this complex biological phenomenon fully. Therefore, investigating the roles of AURKB and MAD2L2 in treatment resistance in an in vivo model is essential for developing effective therapeutic strategies. To overcome these limitations, we plan to incorporate in vivo models in future studies to validate the roles of AURKB and MAD2L2 in CRC progression and to assess the in vivo effects of potential therapeutic strategies. We believe that with these additional studies, we can provide a stronger scientific foundation for AURKB and MAD2L2 as therapeutic targets for CRC.
In conclusion, the research revealed that AURKB is a critical regulator of CRC cell proliferation, invasion, oxidative stress, and glycolysis. Our findings demonstrated that AURKB knockdown significantly inhibited CRC cell growth and metastasis while inducing apoptosis and disrupting the balance of oxidative stress and glycolysis. Furthermore, we discovered a novel interaction between AURKB and MAD2L2, elucidating their combined role in modulating the p53 DDR pathway and promoting aerobic glycolysis. These results suggest that targeting the AURKB-MAD2L2 axis could be a promising therapeutic strategy for CRC. Our study provides new insights into the molecular underpinnings of CRC while discovering potential targets for future cancer research and treatment.
While preparing this research, the authors used artificial intelligence technology, ChatGPT, to assist in proofreading spelling and grammar. This tool was focused on the initial draft stage of the manuscript to help us identify and correct spelling and grammatical errors in the text. After initial language editing using ChatGPT, the authorsthoroughly reviewed the manuscript content and edited it as necessary to ensure scientific accuracy, clarity of expression, and originality of the article. We solemnly declare that the authors take full responsibility for the content of the final submitted publication despite using artificial intelligence aids. We ensure that all opinions, data, and conclusions in the manuscript are derived from our research and analysis and do not infringe copyright or intellectual property rights.
The datasets used and/or analyzed during the current study are available from the corresponding author upon reasonable request.
Conception and design of the research: BS, SL, ZY and JX. Acquisition of data: SL. Analysis and interpretation of data: SL, JY, KY, CX, ZQ, YX, LY, and TZ. Statistical analysis: JY, KY, CX, ZQ, YX, LY, and TZ. Drafting the manuscript: SL and ZY. Revision of manuscript for important intellectual content: BS and JX. All authors contributed to editorial changes in 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.
Not applicable.
Not applicable.
This research received no external funding.
The authors declare no conflict of interest.
Supplementary material associated with this article can be found, in the online version, at https://doi.org/10.31083/FBL26532.
References
Publisher’s Note: IMR Press stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.











