Cdca7 enhances stat3 transcriptional activity to regulate aerobic glycolysis and promote pancreatic cancer progression and gemcitabine resistance

Cdca7 enhances stat3 transcriptional activity to regulate aerobic glycolysis and promote pancreatic cancer progression and gemcitabine resistance

Play all audios:

Loading...

ABSTRACT Cell division cycle associated 7 (CDCA7) plays a role in various malignancies, especially pancreatic cancer (PC). However, its expression pattern and functional significance in PC


require further research. Therefore, this study aimed to investigate CDCA7 expression levels and biological functions in PC using in vitro and in vivo experiments. Western blotting,


immunohistochemistry, and real-time polymerase chain reaction were performed to detect CDCA7 expression in PC cells and tissues. Additionally, the biological functions of CDCA7 were assessed


using cell proliferation, wound healing, and Transwell assays. CDCA7 overexpression promoted PC cell proliferation, migration, and invasion, and increased resistance to the chemotherapy


drug gemcitabine, possibly through enhanced aerobic glycolysis. Additionally, immunoprecipitation assay showed that CDCA7 interacted with STAT3 protein and affected the transcriptional


regulation of hexokinase 2. Conclusively, targeting CDCA7 might be a promising therapeutic strategy to increase gemcitabine sensitivity by inhibiting glycolysis in PC cells. SIMILAR CONTENT


BEING VIEWED BY OTHERS FOXQ1 PROMOTES PANCREATIC CANCER CELL PROLIFERATION, TUMOR STEMNESS, INVASION AND METASTASIS THROUGH REGULATION OF LDHA-MEDIATED AEROBIC GLYCOLYSIS Article Open access


24 October 2023 FOXD1 FACILITATES PANCREATIC CANCER CELL PROLIFERATION, INVASION, AND METASTASIS BY REGULATING GLUT1-MEDIATED AEROBIC GLYCOLYSIS Article Open access 03 September 2022


DELETION OF ENO1 SENSITIZES PANCREATIC CANCER CELLS TO GEMCITABINE VIA MYC/RRM1-MEDIATED GLYCOLYSIS Article Open access 22 March 2025 INTRODUCTION Pancreatic cancer (PC) remains a serious


challenge in oncology due to its high mortality rate, with only a 12% 5-year survival rate [1]. Major contributors to the poor prognosis of PC include difficulties in early detection and


limited effectiveness of drug therapies [2]. Notably, due to the limited treatment options for pancreatic cancer, only approximately 20% of patients are eligible for surgical resection [3].


In addition to surgery, chemotherapy is an important treatment strategy for PC. Currently, gemcitabine is the most commonly used drug for PC chemotherapy. However, the efficacy of


gemcitabine treatment in PC remains modest, with an overall response rate of less than 20%. Importantly, the low efficacy of gemcitabine is primarily due to the rapid onset of drug


resistance in tumor cells, leading to inadequate therapeutic responses [4, 5]. Therefore, it is crucial to investigate the mechanisms underlying PC development and chemotherapy resistance,


as well as develop efficacious targeted therapies. Cell division cycle associated protein 7 (CDCA7) belongs to the family of cell cycle regulatory proteins. It has been identified as a c-Myc


responsive gene, contributing to MYC-mediated tumorigenesis by acting as a transcriptional regulator [6, 7]. As an oncogenic gene with copy number amplification, CDCA7 exhibits high


expression in various tumors and is correlated with unfavorable prognosis [8, 9]. CDCA7 promotes tumor cell proliferation, invasion, and metastasis in various cancers, including esophageal,


gastric, and ovarian cancers, and regulates malignant biological processes, including inflammatory factors [10,11,12]. CDCA7 is a drug resistance-associated cell cycle protein that is highly


expressed in paclitaxel-resistant subgroup of non-small cell lung cancer [13, 14]. However, the precise role of CDCA7 in PC progression remains unclear. Recently, the role of metabolic


reprogramming in the development of drug resistance in PC has attracted considerable attention [15]. Research findings indicate that stroma formation and the complex tumor microenvironment,


including aberrant glycolytic metabolism reprogramming, are crucial factors contributing to chemotherapy resistance in PC [16]. Additionally, an increasing number of preclinical and clinical


trials are being performed to develop metabolic-targeted therapies for PC [15]. Moreover, therapeutic approaches targeting metabolism holds great potential for improving unfavorable


prognosis in patients with PC [15]. Overall, metabolism research not only aids in understanding cancer initiation and progression but also offers novel perspectives for PC treatment. Signal


transducer and activator of transcription 3 (STAT3) is a member of the STAT protein family. STAT3 can be phosphorylated by specific kinases, resulting in the formation of heterodimers that


migrate to the nucleus where they function as transcription factors critical in various cellular processes, including cell growth and apoptosis [17,18,19]. For example, induction of STAT3


signaling increases tumor glycolysis and proliferation, prevents apoptosis, and promotes drug resistance [20, 21]. Additionally, FBP1 binds to STAT3, blocks the binding of STAT3 to


STAT3-mediated gene promoters, inhibits glycolysis in ovarian cancer cells, and improves cisplatin resistance in ovarian cancer [22]. Based on these findings, it could be speculated that


regulating glycolysis-related enzymes via STAT3 inhibition may ameliorate chemotherapy resistance [23, 24]. Therefore, this study aimed to investigate CDCA7 expression levels and functional


significance in PC, focusing on its effect on STAT3. CDCA7 expression level was upregulated in PC and closely correlated with adverse clinical outcomes. Additionally, CDCA7 expression was


related to the glycolysis pathway in PC cells. Moreover, high expression of CDCA7 enhanced the transcriptional activity of STAT3, which promoted hexokinase 2 (HK2) expression. Collectively,


these changes increased aerobic glycolysis, ultimately promoting PC cell proliferation and invasion and gemcitabine resistance. Based on these findings, we believe that targeting CDCA7 may


be a potential chemotherapy sensitization strategies for PC. MATERIALS AND METHODS BIOINFORMATICS ANALYSIS Bioinformatics analysis was performed to verify the transcription levels of CDCA7


in the GTEx and TCGA databases using the Gene Expression Profiling Interactive Analysis (GEPIA). Additionally, we examined CDCA7 expression in cancerous and adjacent non-tumor tissues using


datasets (GSE15471, GSE28735, and GSE16515) from the Gene Expression Omnibus database. PATIENT INFORMATION AND CLINICAL SAMPLES Cancer and para-cancerous specimens from 73 patients with PC


were collected from the Department of Hepatobiliary Surgery, Affiliated Hospital of Guizhou Medical University. This study was approved by the Ethics Committee of the Affiliated Hospital of


Guizhou Medical University, and each patient provided written informed consent. CELL CULTURE Human pancreatic cell lines HPNE, CFPAC-1, BxPC-3, MIA PaCa-2, SW1990, and PANC-1 were obtained


from the American Type Culture Collection. HPNE and BxPC-3 were grown in RPMI-1640 medium supplemented with 10% FBS, whereas CFPAC-1, MIA PaCa-2, SW1990, and PANC-1 were cultivated in DMEM


with 10% FBS. The cultures were maintained in a humidified atmosphere of 5% CO2 at 37 °C. Authentication of the cell lines was performed through STR profiling, and mycoplasma contamination


was ruled out, ensuring the reliability of the experimental outcomes. QUANTITATIVE REAL-TIME POLYMERASE CHAIN REACTION (QRT-PCR) Total RNA was extracted from clinical pancreatic tissues,


adjacent non-tumor tissues, and PC cells subjected to different treatments using the RNA rapid extraction kit (YISHAN, Shanghai, China). Reverse transcription was performed to generate cDNA


using PrimeScript RT (YISHEN, Shanghai, China). qRT-PCR was performed to quantify gene expression levels. The primer sequences are provided in Supplementary Table 1. WESTERN BLOT ASSAY


Proteins were extracted using a lysis buffer, separated using SDS-PAGE, and transferred to Millipore membranes. Thereafter, the membranes were blocked with 5% non-fat milk for a minimum of 1


 h and incubated with the corresponding antibodies overnight at 4 °C. After washing with water, the membranes were incubated with affinity-labeled goat anti-mouse/rabbit antibody at 20 °C


for 2 h. Protein bands were visualized using ECL reagents (Boster Biotechnology Co., Ltd.) and a chemiluminescence imaging system (Tanon). The antibody information is provided in


Supplementary Table 2. CELL PROLIFERATION ASSAYS Cell proliferation activity was detected using cell counting kit-8 (CCK-8), 5-ethyl-2’deoxyuridine (EdU), and plate cloning experiments. In


the CCk8 experiment, cells were seeded in 96-well cells (six replicate wells per group) followed by the addition of CCK-8 reagent and further incubation for 3 h. Finally, the OD value was


measured using a spectrophotometer. In the EdU experiment, cells were seeded into 24-well plates, followed by the addition of EdU reagent (Beyotime, Shanghai, China) to each well and further


incubation. Finally, the ratio of edu-positive nuclei in six microscopic fields (three independent replicates) was analyzed. In the plate cloning experiment, cells were seeded in a 6-well


plate and cultured. After 14 days, cells were fixed with 4% paraformaldehyde (Biosharp, Hefei, Anhui, China) for 30 min and stained with 0.25% crystal violet solution (Biosharp, Hefei,


Anhui, China) for 30 min. Images of the stained cells were captured after washing with PBS. WOUND HEALING ASSAY PC cells were seeded into a 6-well plate and cultured on growth density


reached 90%. Thereafter, a wound was carefully inflicted on the cell layer using a 200 μL pipette tip, followed by the replacement of the culture medium with serum-free medium and incubation


for 48 h. Images of each wound were meticulously captured at both the start (0 h) and end (48 h) of the incubation period using an inverted microscope (Olympus, Tokyo, Japan). TRANSWELL


ASSAY Briefly, PC cells were starved and seeded in the upper chamber, followed by the addition of 600 μL of 20% FBS DMEM medium to the lower chamber. After 24 h, the cells were washed with


PBS, fixed with 4% paraformaldehyde for 30 min, and stained with a 0.25% crystal violet solution for another 30 min. Images were captured using an inverted microscope (Olympus, Tokyo,


Japan). IMMUNOHISTOCHEMISTRY After fixation, embedding, sectioning, and deparaffinization, PC tissue sections were blocked with 3% H2O2 and 5% BSA. Therefore, the sections were incubated


with primary antibodies targeting CDCA7, anti-Ki67, and PCNA overnight at 4 °C, followed by incubation with secondary antibodies for 2 h at room temperature. Positively stained cells and


signal intensity were assessed in three randomly selected areas by two independent observers, who were blinded to the treatments. ANIMAL EXPERIMENT All animal experiments were approved by


the Ethics Committee of Guizhou Medical University. Forty-eight female BALB/c nude mice (6-week-old) were randomly divided into 8 groups (_n_ = 6 mice/group). PC cells (2 × 106) were


injected into the left axilla of each mouse. Tumor volume was assessed every 7 days and calculated using the formula: Tumor volume = (length × width2)/2. Mice were euthanized at 42 days


post-injection, and tumor tissues were harvested, weighed, and subjected to immunohistochemical analysis. DUAL LUCIFERASE REPORTER ASSAY HK2 promoter was cloned into pGL4.10[luc2] vector.


The indicated reporter vectors were transfected into PC cells together with pGL4.74[hRluc/TK] vector. After 48 hours cells were harvested. Finally, the fluorescence signals of firefly and


Renilla luciferases were detected using the Dual-Luciferase Reporter Assay System (Promega, Madison, WI, USA). This process was independently repeated three times. PLASMID TRANSFECTION AND


LENTIVIRUS INFECTION The CDCA7-overexpressing plasmid was constructed using the GV341 vector from GeneChem (Shanghai, China). Short hairpin RNA lentiviruses targeting CDCA7, STAT3, and HK2


were also acquired from GeneChem. The lentiviral particles were produced by co-transfecting the plasmids with packaging vectors and Lipo3000 (Invitrogen, MA, USA) into 293 T cells. The


virus-containing supernatant was collected, filtered through a 0.45 μm filter, concentrated using PEG6000 (Sigma, #81253), and then resuspended in PBS before being aliquoted for future


transfections. Infected cells were selected with puromycin for 72 h. The shRNA sequences are listed in Supplementary Table 3. IMMUNOFLUORESCENCE ASSAY After exposure to various treatments,


PC cells were seeded into a 24-well plate, fixed, permeabilized, blocked, and incubated with primary antibodies against CDCA7 and STAT3 at 4 °C for 10 h. Thereafter, the cells were incubated


with secondary antibodies in the dark at room temperature for 2 h. After washing with PBS, cell nuclei were counterstained with 4’,6-diamidino-2-phenylindole, and visualization and


quantification of target protein expression were performed using fluorescence microscopy. IMMUNOPRECIPITATION, SILVER STAINING, AND MASS SPECTROMETRY ANALYSIS Proteins were extracted from PC


cells and incubated overnight at 4 °C with anti-CDCA7 antibody. Thereafter, protein A + G beads (Beyotime, Shanghai, China) were added and incubated for 2 h under constant rotation. The


beads were collected using a magnetic rack and boiled after adding 1× loading buffer. Protein analysis was conducted using western blotting, silver staining, or mass spectrometry (MS).


Silver staining was performed according to the manufacturer’s instructions. GLUCOSE UPTAKE, ATP PRODUCTION RATE, AND LACTIC ACID PRODUCTION ASSAYS PC cells at the log-phase were plated in


96-well plates. Glucose uptake, ATP levels, and lactate production were measured using the Glucose Assay Kit (Absin, abs580025), ATP Assay Kit (Sigma, MAK190), and Lactate Assay Kit (Sigma,


MAK064), respectively, according to the manufacturers’ protocols. CONSTRUCTION OF ORTHOTOPIC TUMOR MODELS The orthotopic pancreatic cancer tumor studies were conducted using C57BL/6 J mice.


Female mice were randomly divided into 8 groups, with 5 mice per group. At the age of 8 weeks, the mice underwent orthotopic pancreatic cancer tumor induction. A suspension of 1 × 106


PANC-02-Luc cells was harvested and injected into the pancreatic tail via a minimal laparotomy. Twenty-one days following the injection, high-resolution ultrasound imaging was conducted on


each mouse to verify tumor establishment. EXTRACELLULAR ACIDIFICATION RATE To assess PC cell metabolism, cells were seeded in Seahorse XF96 plates (Seahorse, Cat No. 101085-004) at a density


of 3.5 × 104 cells/well until cell confluence reached 80%. After rinsing with PBS, glycolysis and extracellular acidification rate (ECAR) were measured using Seahorse Glycolytic Stress Test


Kit (Seahorse, Cat No. 103020-100) and Seahorse XFe96 Analyzer, respectively. The analysis was performed on three independent replicates. STATISTICAL ANALYSIS All statistical analyses were


performed using SPSS (version 23.0; IBM Corp., Armonk, NY, USA). Data are expressed as mean ± standard deviation. Significant differences were determined using Student’s _t_-test for two


group comparison or one-way analysis of variance for multi-group comparison. Overall survival was determined using the Kaplan-Meier method. Statistical significance was set at _p_ < 0.05.


RESULT CDCA7 IS HIGHLY EXPRESSED IN PC TISSUES AND CELLS Expression profile analysis of datasets (GSE16515, GSE28735, and GSE15471) downloaded from the Gene Expression Omnibus database


showed that CDCA7 mRNA expression was higher in PC tissues than in adjacent para-cancerous tissues (Fig. 1A). Similarly, CDCA7 expression was higher in 179 PC samples than in 171 normal


pancreatic tissues in the Cancer Genome Atlas (TCGA) database (Fig. 1B). Additionally, we examined CDCA7 mRNA and protein expression in clinical tissue samples using qRT-PCR and western


blotting, respectively. Notably, CDCA7 expression was markedly higher in PC tissues than in adjacent tissues at both the mRNA and protein levels (Fig. 1C, D). Moreover, we examined CDCA7


mRNA and protein levels in human pancreatic normal ductal epithelial (HPNE) cells and five human PC cell lines (CFPAC-1, BxPC-3, MIA PaCa-2, SW1990, and PANC-1). CDCA7 mRNA and protein


levels were higher in all PC cell lines (except in SW1990) than in HPNE cells (Fig. 1E, F). Similarly, immunohistochemistry (IHC) showed that CDCA7 expression was higher in PC tissues than


in adjacent tissues (Fig. 1G). Moreover, Kaplan-Meier survival analysis showed that patients with elevated levels of CDCA7 had markedly lower survival rate than those with low CDCA7


expression (Fig. 1H). CDCA7 PROMOTES THE PROLIFERATION, MIGRATION, AND INVASION OF PC CELLS To elucidate the functional roles of CDCA7, the PC cell lines PANC-1 and MIA PaCa-2 were


transfected with various plasmids to generate CDCA7 overexpression and knockdown models. CDCA7 overexpression or knockdown efficiency was confirmed using western blotting and qRT-PCR


(Supplementary Fig. 1A-B). CCK-8, colony formation, and EdU assays showed that CDCA7 overexpression significantly promoted the proliferation of PC cells, whereas CDCA7 knockdown had the


effect t (Fig. 2A–C). Similarly, CDCA7 overexpression significantly upregulated the proliferation of SW1990 cells (Supplementary Fig. 1C−E). Moreover, wound healing (Fig. 2D) and Transwell


(Fig. 2E) assays indicated that CDCA7 overexpression promoted the migratory and invasive abilities of PC cells, whereas CDCA7 knockdown had the opposite effects. Similarly, CDCA7


overexpression increased the migratory and invasive abilities of SW1990 cells (Supplementary Fig. 1F, G). To confirm whether the cellular phenotypic changes were caused by CDCA7, we


established distinct groups, including sh-NC, sh-CDCA7, and sh-CDCA7 + CDCA7_Res groups. Significantly, sh-mediated knockout of CDCA7 genes suppressed PC cell proliferation, migration, and


invasion. However, reintroducing the CDCA7 gene successfully restored cellular functions (Supplementary Fig. 1H-L). To evaluate the role of CDCA7 on PC cells in vivo, mice were injected with


PANC-1 cells to construct a subcutaneous tumor model (Fig. 2F). CDCA7 overexpression promoted tumor growth, whereas CDCA7 knockdown inhibited tumor growth, as evidenced by tumor volume and


weight (Fig. 2G, H). Additionally, IHC showed that CDCA7 level was positively correlated with the expression of the proliferation indicators Ki-67 and PCNA in subcutaneous tumor tissues


derived from mice (Fig. 2I, J). Furthermore, we developed an orthotopic pancreatic tumor model in C57BL/6 mice. Bioluminescence imaging (BLI) showed a significant increase in signal


intensity in the _cdca7_ overexpression group compared with that in the control group (Supplementary Fig. 1M). CDCA7 ENHANCES GLYCOLYSIS IN PC CELLS AND PROMOTES GEMCITABINE RESISTANCE Gene


set enrichment analysis (GSEA) of dataset from the TCGA database indicated that the glycolysis pathway was positively enriched in PC tissues (Fig. 3A, B). To investigate the role of CDCA7 in


metabolism in PC cells, extracellular glucose levels, extracellular lactate levels, and intracellular ATP levels were measured. Notably, the CDCA7 overexpression group showed decreased


glucose content in the culture medium, increased lactic acid production, and elevated intracellular ATP levels. In contrast, the CDCA7 knockdown group exhibited increased glucose content,


decreased lactic acid production, and downregulated ATP levels (Fig. 3C–E). Additionally, we examined ECAR in the two PC cell lines using Seahorse energy metabolism analyzer. Although CDCA7


upregulation promoted glycolysis, its downregulation suppressed glycolysis in both cells (Fig. 3F, G). Research evidence suggests that drug-resistant tumor cells possess higher glycolytic


activity than sensitive cells. Therefore, modulating the glycolytic pathway could be an effective strategy to overcome tumor resistance to chemotherapy [25]. To confirm this hypothesis, PC


cells overexpressing CDCA7 were treated with 2-DG, a critical inhibitor of glycolysis. Importantly, 2-DG treatment significantly enhanced the sensitivity of the cells to gemcitabine (Fig.


3H). Consistent with the in vitro results, in vivo experiments showed CDCA7 overexpression promoted tumor resistance to gemcitabine, a phenomenon that is reversed following 2-DG treatment


(Fig. 3I–K). IHC showed a marked decrease in the expression of Ki-67 and PCNA in CDCA7-overexpressing cells treated with 2-DG (Fig. 3L). Moreover, the BLI of orthotopic pancreatic tumor


showed decreased signal intensity in the _cdca7_ + 2-DG group compared with that in _cdca7_overexpression group (Supplementary Fig. 1N). CDCA7 INTERACTS WITH STAT3 Considering that CDCA7 has


been shown to inhibit glycolysis in PC cells, we examined the specific regulatory mechanism of CDCA7 in glycolysis. A silver staining assay was conducted to discern differential protein


bands within the immunoprecipitated complexes, followed by mass spectrometry (Fig. 4A, B). Figure 4C shows the secondary protein spectrum of CDCA7. Co-immunoprecipitation assay demonstrated


a possible interaction between CDCA7 and STAT3 (Fig. 4D). Immunofluorescence co-localization assay showed that CDCA7 and STAT3 were colocalized in the nucleus (Fig. 4E). To identify the


binding sites of CDCA7 and STAT3, we generated a several shortened CDCA7 and STAT3 mutants (Fig. 4F). Notably, the D1 (1-120aa) domain of CDCA7 and the D1 (1-385aa) domain of STAT3 are


necessary and sufficient for direct interaction (Fig. 4G). STAT3 PROMOTES PANCREATIC CANCER PROGRESSION To assess the impact of STAT3 on the progression of PC cells, we generated STAT3


overexpression and knockdown PC cell lines. Western blotting and qRT-PCR confirmed successful STAT3 knockdown and overexpression (Supplementary Fig. 2A, B). Cellular functional assays


revealed that STAT3 knockdown significantly inhibited PC cell proliferation (Supplementary Fig. 2C–E). To determine if the observed decrease in cell proliferation was attributable to STAT3,


we performed reconstitution experiment. Importantly, reintroducing the full-length STAT3 construct into STAT3 knockdown cells effectively restored the proliferation rate of PC cells


(Supplementary Fig. 2F–H). Wound healing and Transwell assays demonstrated that STAT3 inhibited the migratory and invasive abilities of PC cells (Supplementary Fig. 2I, J). To examine the


effects of STAT3 on metabolism in PC cells, we measured glucose and lactate level in the extracellular environment and ATP levels within the cells. Compared with those in the control group,


STAT3 knockdown elevated glucose levels, reduced lactate production, and downregulated ATP levels (Supplementary Fig. 2K–M). Additionally, ECAR was notably lower in the STAT3 knockdown group


than in the control group (Supplementary Fig. 2N). Moreover, STAT3 knockdown enhanced the sensitivity of PC cells to gemcitabine (Supplementary Fig. 2O). CDCA7 PROMOTES PC CELL PROGRESSION


AND GEMCITABINE RESISTANCE BY TARGETING STAT3 TO REGULATE GLYCOLYSIS LEVELS In the present study, we examined the effect of STAT3 on the proliferation and invasive ability of PC cells


overexpressing CDCA7. CCK8, colony formation, and EDU assays showed that STAT3 knockdown inhibited CDCA7-mediated PC cell proliferation (Fig. 5A–C). Additionally, wound healing and Transwell


assays revealed that STAT3 knockdown attenuated CDCA7 overexpression-induced upregulation of PC cell migration and invasion (Fig. 5D, E). Moreover, STAT3 knockdown increased glucose levels


and decreased both lactate production and intracellular ATP levels in PC cells overexpressing CDCA7 compared with those in the CDCA7 overexpression group (Fig. 5F–H). Similarly, STAT3


knockdown attenuated CDCA7 overexpression-induced gemcitabine resistance in PC cells to a certain degree (Fig. 5I). Collectively, these results indicate that CDCA7 promotes PC progression


and gemcitabine resistance possibly through STAT3-mediated regulation of aerobic glycolysis. CDCA7-STAT3 AXIS ENHANCES HK2 EXPRESSION AND PROMOTES GLYCOLYSIS To elucidate alterations in


glycolysis levels, we conducted CDCA7 overexpression experiments and assessed changes in key glycolytic enzymes. Western blotting showed that HK2 protein levels were significantly


upregulated in the CDCA7 overexpression group (Fig. 6A). Additionally, the GEPIA database showed a positive correlation between CDCA7 and HK2 (Fig. 6B). Moreover, HK2 expression was


upregulated in PC samples in the TCGA database (Fig. 6C). qRT-PCR and western blotting showed that CDCA7 overexpression or knockout influenced the expression levels of HK2 (Fig. 6D, E).


Considering that CDCA7 interacts with STAT3 to regulate glycolysis, we examined changes in HK2 expression in PC cells overexpressing CDCA7 after STAT3 knockdown. qRT-PCR and western blotting


indicated that STAT3 suppression abrogated the regulatory impact of CDCA7 on HK2 expression (Fig. 6F, G). Similarly, the regulation of HK expression by STAT3 was confirmed using qRT-PCR and


western blotting (Fig. 6H, I). CHIP-qPCR showed that the binding of STAT3 to HK2 promoter was significantly enhanced in CDCA7-overexpressing PC cells (Fig. 6J). Moreover, luciferase assay


showed that high expression of CDCA7 upregulated STAT3 transcriptional activity and promoted HK2 transcription (Fig. 6K). To elucidate the underlying mechanism by which CDCA7 enhances the


DNA binding capacity of STAT3, we investigated changes in STAT3 phosphorylation following CDCA7 overexpression. Notably, CDCA7 enhanced STAT3 phosphorylation in PC cells (Fig. 6L).


Additionally, western blotting revealed a significant increase in the levels of STAT3 protein within the nucleus following CDCA7 overexpression in PANC-1 cell (Fig. 6M). In summary, CDCA7


augments STAT3 activity within the HK2 promoter region, thereby stimulating HK2 transcription and intensifying glycolytic activity in PC cell. STAT3 REGULATES HK2 TO PROMOTE THE PROGRESSION


AND DRUG RESISTANCE OF PC Western blot and qRT-PCR confirmed successful HK2 knockdown in PC cells (Supplementary Fig. 3A, B). To determine whether STAT3 facilitates PC progression through


HK2, we knocked down HK2 in STAT3-overexpressing PC cells. Compared with those in the STAT3 overexpression group, co-transfection with OE-STAT3 and sh-HK2 significantly reduced the


proliferation and migration and invasive abilities of PC cells (Fig. 7A–G). Similar trends were observed in glycolysis level and ECAR (Fig. 7H–K). HK2 knockdown partially ameliorated STAT3


overexpression induced gemcitabine resistance (Fig. 7L). Collectively these suggest that STAT3 may promote aerobic glycolysis to enhance PC cell progression and gemcitabine resistance via


HK2. DISCUSSION PC is currently one of the deadliest types of cancer, with poor treatment outcomes. Notably, surgical opportunities are missed because of the insidious onset and late


detection of PC. Moreover, the highly dense nature and metabolic complexity of the tumor tissue result in poor drug treatment. Although combined radiotherapy and immunotherapy has shown poor


outcomes, research evidence suggests that metabolic reprogramming may play a key role in PC carcinogenesis, progression, treatment, and prognosis. Moreover, metabolic reprogramming is


intricately linked to chemotherapy, radiotherapy, and immunotherapy, ultimately contributing to unfavorable prognostic outcomes [15]. Glycolysis reprogramming caused by dense tissue of PC is


one of the metabolic characteristics [26]. Therefore, exploring and analyzing the mechanism of aerobic glycolysis in PC may have important clinical value. CDCA7 is a member of the CDCA


family, and its abnormal upregulation in tumors regulate the malignant biological behavior. CDCA7 promotes TGF-β-induced epithelial-mesenchymal transition to promote tumor progression by


transcriptionally regulating Smad4/Smad7 in esophageal cancer cells [10]. Chen et al. showed that CDCA7 promotes zeste homolog 2 (EZH2) expression to promote ovarian cancer angiogenesis


[12]. Yu et al. found that CDCA7 regulates inflammatory responses through the TLR4/NF-κB signaling pathway in gastric adenocarcinoma [11]. In the present study, analysis of public databases,


IHC, and qRT-PCR showed that CDCA7 expression was significantly higher in human PC tissues than in adjacent non-cancerous tissues, and is significantly negatively correlated with prognosis


and survival. In vivo and in vitro experiments confirmed that CDCA7 overexpression promoted the proliferation of PC cells. Moreover, the reprogramming of glucose, amino acid, and lipid


metabolism, along with metabolic interplay within the tumor microenvironment, plays a pivotal role in driving the progression of pancreatic tumors. Research evidence indicates a correlation


between gemcitabine resistance and metabolic pathways involving glucose, amino acids, and lipids [27]. Zhao et al. showed that gemcitabine-resistant PC cell lines exhibited high aerobic


glycolysis and low ROS levels, which may contribute to chemotherapy resistance [28]. Additionally, Surendra et al. showed that elevated expression of the transmembrane protein MUC1 activated


and stabilized HIF-1α, thereby promoting glycolysis and chemotherapy resistance [29]. In this study, bioinformatics analysis showed that CDCA7 was enriched in the glycolysis pathway. In


vivo and in vitro experiments showed that CDCA7 overexpression increased glycolysis and promoted gemcitabine resistance in PC cells. However, treatment with a key inhibitor of glycolysis


(2-DG) reversed the effects of CDCA7 overexpression. Importantly, CDCA7 is a protein related to transcription factors [10]. To verify the molecular mechanism by which CDCA7 regulates


glycolysis and affects drug resistance, we performed mass spectrometry and immunoprecipitation assay. Immunoprecipitation and colocalization assays showed that CDCA7 interacted with STAT3,


with both proteins mainly colocalized in the nucleus. Further analysis using truncated bodies confirmed the interaction between the D1 (1-120aa) domain of CDCA7 and the D1 (1-385aa) domain


of STAT3. STAT3 is a signal transducer and transcriptional activator that is often phosphorylated and activated to function as a transcriptional activator. For example, enhanced STAT3


transcriptional signal can increase glycolysis and promote tumor progression [20]. STAT3 directly regulates the transcription of solute carrier family 2 member 1 (SLC2A1) to promote


glycolysis [30]. Li et al. showed that STAT3 overexpression regulated the expression of key enzymes in glycolysis, leading to a direct increase in glucose consumption and lactate formation


[31]. Consistent with the above findings, western blotting and qRT-PCR showed that CDCA7-STAT3 interaction affected HK2 expression in PC cells. HK2, a key enzyme that regulates the first


step of glycolysis, is overexpressed in several tumors and upregulates glycolysis to promote gemcitabine drug resistance [32,33,34]. Dual-luciferase assay showed that CDCA7 overexpression


increased the transcriptional activity of STAT3. CHIP-qPCR showed a significant increase in the binding of STAT3 to HK2 promoter in CDCA7-overexpressing PC cells, upregulating the


transcription and protein levels of HK2 and promoting the resistance of PC cell lines to gemcitabine. Notably, HK2 downregulation partly ameliorated STAT3 overexpression-induced increase in


aerobic glycolysis and PC cell proliferation, migration, and invasion, and suppressed gemcitabine resistance. Collectively, these results suggest that the CDCA7/STAT3/HK axis may play a key


role in aerobic glycolysis and PC progression and influence the sensitivity of PC to gemcitabine. Conclusively, CDCA7 acts as an oncogene in PC and regulates aerobic glycolysis by enhancing


the transcriptional activity of the transcription factor STAT3 and promoting the transcription of the key glycolysis enzyme HK2, which may contribute to drug resistance in PC cells.


Therefore, targeting CDCA7 could be a potential therapeutic strategy for increasing the sensitivity of PC cells to gemcitabine. DATA AVAILABILITY The datasets produced and examined during


the present investigation can be obtained from the corresponding author upon a reasonable inquiry. REFERENCES * Siegel RL, Giaquinto AN, Jemal A. Cancer statistics, 2024. CA Cancer J Clin.


2024;74:12–49. Article  PubMed  Google Scholar  * Moore A, Donahue T. Pancreatic Cancer. JAMA 2019;322:1426. Article  PubMed  PubMed Central  Google Scholar  * Rahib L, Smith BD, Aizenberg


R, Rosenzweig AB, Fleshman JM, Matrisian LM. Projecting cancer incidence and deaths to 2030: the unexpected burden of thyroid, liver, and pancreas cancers in the United States. Cancer Res.


2014;74:2913–2921. Article  CAS  PubMed  Google Scholar  * Conroy T, Pfeiffer P, Vilgrain V, Lamarca A, Seufferlein T, O’Reilly EM, et al. Pancreatic cancer: ESMO Clinical Practice Guideline


for diagnosis, treatment and follow-up. Ann Oncol. 2023;34:987–1002. Article  CAS  PubMed  Google Scholar  * Tang R, Xu J, Wang W, Meng Q, Shao C, Zhang Y, et al. Targeting neoadjuvant


chemotherapy-induced metabolic reprogramming in pancreatic cancer promotes anti-tumor immunity and chemo-response. Cell Rep Med. 2023;4:101234. Article  CAS  PubMed  PubMed Central  Google


Scholar  * Osthus RC, Karim B, Prescott JE, Smith BD, McDevitt M, Huso DL, et al. The Myc target gene JPO1/CDCA7 is frequently overexpressed in human tumors and has limited transforming


activity in vivo. Cancer Res 2005;65:5620–7. Article  CAS  PubMed  PubMed Central  Google Scholar  * Gill RM, Gabor TV, Couzens AL, Scheid MP. The MYC-associated protein CDCA7 is


phosphorylated by AKT to regulate MYC-dependent apoptosis and transformation. Mol Cell Biol. 2013;33:498–513. Article  CAS  PubMed  PubMed Central  Google Scholar  * Wang Y, Zhao Y, Zhang Z,


Zhang J, Xu Q, Zhou X, et al. High Expression of CDCA7 in the Prognosis of Glioma and Its Relationship with Ferroptosis and Immunity. Genes (Basel). 2023;14:1406. Article  CAS  PubMed 


Google Scholar  * Li S, Huang J, Qin M, Zhang J, Liao C. High expression of CDCA7 predicts tumor progression and poor prognosis in human colorectal cancer. Mol Med Rep. 2020;22:57–66. CAS 


PubMed  PubMed Central  Google Scholar  * Li H, Wang S, Li X, Weng Y, Guo D, Kong P, et al. CDCA7 promotes TGF-beta-induced epithelial-mesenchymal transition via transcriptionally regulating


Smad4/Smad7 in ESCC. Cancer Sci. 2023;114:91–104. Article  CAS  PubMed  Google Scholar  * Guo Y, Zhou K, Zhuang X, Li J, Shen X. CDCA7-regulated inflammatory mechanism through


TLR4/NF-kappaB signaling pathway in stomach adenocarcinoma. Biofactors 2021;47:865–78. Article  CAS  PubMed  Google Scholar  * Cai C, Peng X, Zhang Y. Downregulation of cell division


cycle-associated protein 7 (CDCA7) suppresses cell proliferation, arrests cell cycle of ovarian cancer, and restrains angiogenesis by modulating enhancer of zeste homolog 2 (EZH2)


expression. Bioengineered. 2021;12:7007–19. Article  CAS  PubMed  PubMed Central  Google Scholar  * Walker MG. Drug target discovery by gene expression analysis: cell cycle genes. Curr


Cancer Drug Targets 2001;1:73–83. Article  CAS  PubMed  Google Scholar  * Hou J, Lambers M, den Hamer B, den Bakker MA, Hoogsteden HC, Grosveld F, et al. Expression profiling-based subtyping


identifies novel non-small cell lung cancer subgroups and implicates putative resistance to pemetrexed therapy. J Thorac Oncol. 2012;7:105–14. Article  CAS  PubMed  Google Scholar  * Qin C,


Yang G, Yang J, Ren B, Wang H, Chen G, et al. Metabolism of pancreatic cancer: paving the way to better anticancer strategies. Mol Cancer 2020;19:50. Article  PubMed  PubMed Central  Google


Scholar  * Neesse A, Algul H, Tuveson DA, Gress TM. Stromal biology and therapy in pancreatic cancer: a changing paradigm. Gut 2015;64:1476–84. Article  CAS  PubMed  Google Scholar  * Jiang


H, Yu J, Guo H, Song H, Chen S. Upregulation of survivin by leptin/STAT3 signaling in MCF-7 cells. Biochem Biophys Res Commun. 2008;368:1–5. Article  CAS  PubMed  Google Scholar  * Gao X,


Wang H, Yang JJ, Liu X, Liu ZR. Pyruvate kinase M2 regulates gene transcription by acting as a protein kinase. Mol Cell 2012;45:598–609. Article  CAS  PubMed  PubMed Central  Google Scholar


  * Shen S, Niso-Santano M, Adjemian S, Takehara T, Malik SA, Minoux H, et al. Cytoplasmic STAT3 represses autophagy by inhibiting PKR activity. Mol Cell 2012;48:667–80. Article  CAS  PubMed


  Google Scholar  * Sadrkhanloo M, Paskeh MDA, Hashemi M, Raesi R, Motahhary M, Saghari S, et al. STAT3 signaling in prostate cancer progression and therapy resistance: An oncogenic pathway


with diverse functions. Biomed Pharmacother. 2023;158:114168. Article  CAS  PubMed  Google Scholar  * Hashemi M, Abbaszadeh S, Rashidi M, Amini N, Talebi Anaraki K, Motahhary M, et al. STAT3


as a newly emerging target in colorectal cancer therapy: Tumorigenesis, therapy response, and pharmacological/nanoplatform strategies. Environ Res. 2023;233:116458. Article  CAS  PubMed 


Google Scholar  * Li H, Qi Z, Niu Y, Yang Y, Li M, Pang Y, et al. FBP1 regulates proliferation, metastasis, and chemoresistance by participating in C-MYC/STAT3 signaling axis in ovarian


cancer. Oncogene 2021;40:5938–49. Article  CAS  PubMed  PubMed Central  Google Scholar  * Zhu W, Sheng D, Shao Y, Zhang Q, Peng Y. STAT3-regulated LncRNA LINC00160 mediates cell


proliferation and cell metabolism of prostate cancer cells by repressing RCAN1 expression. Mol Cell Biochem. 2022;477:865–75. Article  CAS  PubMed  Google Scholar  * Huo N, Cong R, Sun ZJ,


Li WC, Zhu X, Xue CY, et al. STAT3/LINC00671 axis regulates papillary thyroid tumor growth and metastasis via LDHA-mediated glycolysis. Cell Death Dis. 2021;12:799. Article  CAS  PubMed 


PubMed Central  Google Scholar  * Chen F, Chen J, Yang L, Liu J, Zhang X, Zhang Y, et al. Extracellular vesicle-packaged HIF-1alpha-stabilizing lncRNA from tumour-associated macrophages


regulates aerobic glycolysis of breast cancer cells. Nat Cell Biol. 2019;21:498–510. Article  CAS  PubMed  Google Scholar  * Bosello-Travain V, Forman HJ, Roveri A, Toppo S, Ursini F,


Venerando R, et al. Glutathione peroxidase 8 is transcriptionally regulated by HIFalpha and modulates growth factor signaling in HeLa cells. Free Radic Biol Med. 2015;81:58–68. Article  CAS


  PubMed  Google Scholar  * Henderson SE, Makhijani N, Mace TA. Pancreatic Cancer-Induced Cachexia and Relevant Mouse Models. Pancreas 2018;47:937–45. Article  PubMed  PubMed Central  Google


Scholar  * Zhao H, Duan Q, Zhang Z, Li H, Wu H, Shen Q, et al. Up-regulation of glycolysis promotes the stemness and EMT phenotypes in gemcitabine-resistant pancreatic cancer cells. J Cell


Mol Med. 2017;21:2055–67. Article  CAS  PubMed  PubMed Central  Google Scholar  * Shukla SK, Purohit V, Mehla K, Gunda V, Chaika NV, Vernucci E, et al. MUC1 and HIF-1alpha Signaling


Crosstalk Induces Anabolic Glucose Metabolism to Impart Gemcitabine Resistance to Pancreatic Cancer. Cancer Cell 2017;32:71–87.e7. Article  CAS  PubMed  PubMed Central  Google Scholar  * Li


Y, Song Z, Han Q, Zhao H, Pan Z, Lei Z, et al. Targeted inhibition of STAT3 induces immunogenic cell death of hepatocellular carcinoma cells via glycolysis. Mol Oncol. 2022;16:2861–80.


Article  CAS  PubMed  PubMed Central  Google Scholar  * Li M, Jin R, Wang W, Zhang T, Sang J, Li N, et al. STAT3 regulates glycolysis via targeting hexokinase 2 in hepatocellular carcinoma


cells. Oncotarget 2017;8:24777–84. Article  PubMed  PubMed Central  Google Scholar  * He H, Guo J, Hu Y, Zhang H, Li X, Zhang J, et al. Saikosaponin D reverses epinephrine- and


norepinephrine-induced gemcitabine resistance in intrahepatic cholangiocarcinoma by downregulating ADRB2/glycolysis signaling. Acta Biochim Biophys Sin. (Shanghai) 2023;55:1404–14. CAS 


PubMed  Google Scholar  * Jiang SH, Dong FY, Da LT, Yang XM, Wang XX, Weng JY, et al. Ikarugamycin inhibits pancreatic cancer cell glycolysis by targeting hexokinase 2. FASEB J.


2020;34:3943–55. Article  CAS  PubMed  Google Scholar  * Fan K, Fan Z, Cheng H, Huang Q, Yang C, Jin K, et al. Hexokinase 2 dimerization and interaction with voltage-dependent anion channel


promoted resistance to cell apoptosis induced by gemcitabine in pancreatic cancer. Cancer Med. 2019;8:5903–15. Article  CAS  PubMed  PubMed Central  Google Scholar  Download references


ACKNOWLEDGEMENTS We thank the Department of Hepatobiliary Surgery, the Affiliated Hospital of Guizhou Medical University for providing the pancreatic cancer tissue samples and related


anonymous clinical data. FUNDING This work was supported by grants from: 1. The Project of Science and Technology of Guizhou Province, Qian Ke He Zhi Cheng [2021] normal 080. 2. National


Natural Science Foundation of China (No. 82360519); 3. Eighth Batch of Hundred Multiplier Level Talent Program, Qiankehe Talent (No. GCC [2023]082); 4. 2023 Discipline Leaders (No.


gyfyxkyc-2023-03). AUTHOR INFORMATION Author notes * These authors contributed equally: Dijie Zheng, Yazhu Deng, Lu Deng. AUTHORS AND AFFILIATIONS * School of Basic Medical Sciences, Guizhou


Medical University, Guiyang, Guizhou Province, 550025, China Dijie Zheng, Yazhu Deng & Deqin Lu * Department of Hepatobiliary Surgery, the Affiliated Hospital of Guizhou Medical


University, Guiyang, Guizhou Province, 550001, China Dijie Zheng, Lu Deng, Zhiwei He, Xinghao Sun, Yanyu Gong, Binbin Shi & Chao Yu * Guizhou Provincial Institute of Hepatobiliary,


Pancreatic and Splenic Diseases, Guiyang, Guizhou Province, 550001, China Dijie Zheng, Lu Deng, Zhiwei He, Xinghao Sun, Yanyu Gong, Binbin Shi & Chao Yu * Key Laboratory of Liver,


Gallbladder, Pancreas and Spleen of Guizhou Medical University, Guiyang, Guizhou Province, 550001, China Dijie Zheng, Lu Deng, Zhiwei He, Xinghao Sun, Yanyu Gong, Binbin Shi & Chao Yu *


Department of Vascular Surgery, the Affiliated Hospital of Guizhou Medical University, Guizhou Medical University, 550001, Guiyang, China Yazhu Deng * School of Clinical Medicine, Guizhou


Medical University, Guiyang, Guizhou Province, 550025, China Lu Deng, Zhiwei He, Xinghao Sun, Yanyu Gong, Binbin Shi & Chao Yu Authors * Dijie Zheng View author publications You can also


search for this author inPubMed Google Scholar * Yazhu Deng View author publications You can also search for this author inPubMed Google Scholar * Lu Deng View author publications You can


also search for this author inPubMed Google Scholar * Zhiwei He View author publications You can also search for this author inPubMed Google Scholar * Xinghao Sun View author publications


You can also search for this author inPubMed Google Scholar * Yanyu Gong View author publications You can also search for this author inPubMed Google Scholar * Binbin Shi View author


publications You can also search for this author inPubMed Google Scholar * Deqin Lu View author publications You can also search for this author inPubMed Google Scholar * Chao Yu View author


publications You can also search for this author inPubMed Google Scholar CONTRIBUTIONS DZ, YD, and LD contributed to the experimental implementation. XS and ZH were responsible for


writing-review and editing. Data analysis was performed by YG and BS. DL and CY contributed to the experimental design. All authors participated in the preparation of the manuscript and


approved the final versions submitted and published. CORRESPONDING AUTHORS Correspondence to Deqin Lu or Chao Yu. ETHICS DECLARATIONS COMPETING INTERESTS The authors declare no competing


interests. ETHICS APPROVAL AND CONSENT TO PARTICIPATE All procedures followed were in accordance with the ethical standards of the Ethical Committee of the Affiliated Hospital of Guizhou


Medical University. All institutional and national guidelines for the care and use of laboratory animals were followed. CONSENT TO PUBLICATION All authors have agreed to publish this


manuscript. ADDITIONAL INFORMATION PUBLISHER’S NOTE Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations. Edited by Stephen


Tait SUPPLEMENTARY INFORMATION SUPPLEMENTARY INFORMATION ORIGINAL DATA1 ORIGINAL DATA2 RIGHTS AND PERMISSIONS OPEN ACCESS This article is licensed under a Creative Commons Attribution 4.0


International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the


source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative


Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by


statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit


http://creativecommons.org/licenses/by/4.0/. Reprints and permissions ABOUT THIS ARTICLE CITE THIS ARTICLE Zheng, D., Deng, Y., Deng, L. _et al._ CDCA7 enhances STAT3 transcriptional


activity to regulate aerobic glycolysis and promote pancreatic cancer progression and gemcitabine resistance. _Cell Death Dis_ 16, 68 (2025). https://doi.org/10.1038/s41419-025-07399-1


Download citation * Received: 15 May 2024 * Revised: 10 December 2024 * Accepted: 27 January 2025 * Published: 04 February 2025 * DOI: https://doi.org/10.1038/s41419-025-07399-1 SHARE THIS


ARTICLE Anyone you share the following link with will be able to read this content: Get shareable link Sorry, a shareable link is not currently available for this article. Copy to clipboard


Provided by the Springer Nature SharedIt content-sharing initiative