Open Access

TLR9 induces colitis‑associated colorectal carcinogenesis by regulating NF‑κB expression levels

  • Authors:
    • Qingtian Luo
    • Ling Zeng
    • Chaotao Tang
    • Zhendong Zhang
    • Youxiang Chen
    • Chunyan Zeng
  • View Affiliations

  • Published online on: August 12, 2020     https://doi.org/10.3892/ol.2020.11971
  • Article Number: 110
  • Copyright: © Luo et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Chronic colorectal inflammation has been associated with colorectal cancer (CRC); however, its exact molecular mechanisms remain unclear. The present study aimed to investigate the effect of Toll‑like receptor 9 (TLR9) on the development of colitis‑associated CRC (CAC) through its regulation of the NF‑κB signaling pathway. By using a CAC mouse model and immunohistochemistry, the present study discovered that the protein expression levels of TLR9 were gradually upregulated during the development of CRC. In addition, the expression levels of TLR9 were revealed to be positively correlated with NF‑κB and Ki67 expression levels. In vitro, inhibiting TLR9 expression levels using chloroquine decreased the cell viability, proliferation and migration of the CRC cell line HT29, and further experiments indicated that this may occur through downregulating the expression levels of NF‑κB, proliferating cell nuclear antigen and Bcl‑xl. In conclusion, the findings of the present study suggested that TLR9 may serve an important role in the development of CAC by regulating NF‑κB signaling.

Introduction

The number of patients with colorectal cancer (CRC) worldwide is increasing annually, with an incidence rate of 6.1% in 2018 (1). Chronic inflammation is the leading cause of immune cell infiltration and proliferation, and it has been suggested to be a high-risk factor for colitis-associated CRC (CAC) (2). Inflammatory bowel disease (IBD), which encompasses both ulcerative colitis (UC) and Crohn's disease, was established as an important precursor to CRC (3,4). For example, the incidence of IBD-associated CRC in patients with UC was reported to have a cumulative risk rate of 2% at 10 years, 8% at 20 years and 18% at 30 years of disease duration (3). Therefore, further in vivo studies are required for researchers to gain an improved understanding of the molecular mechanisms of CAC, which may provide more exact molecular targets for the diagnosis and treatment of CAC during the early stages.

Toll-like receptor (TLR)9, a member of the TLR family, is located in the cytoplasm and intracellular endosomes, and can be activated by unmethylated bacterial CpG DNA (5). The activation of the TLR9 signaling pathway induces a Type 1 T helper cell immune response and stimulates the proliferation of B cells, thus protecting the host from external microbial invasion (69). Multiple studies have revealed that abnormal TLR9 expression levels were involved in the pathogenesis and progression of UC (10,11). In addition, abnormal expression levels of TLR9 were also identified during the tumorigenesis and development of CRC (1215).

NF-κB is an important transcription factor involved in various biological processes, including inflammatory reactions, immune responses, apoptosis and proliferation (16). In fact, NF-κB is regarded as a molecular hub that links inflammation and cancer (17). It was previously suggested that NF-κB may serve an important role in colorectal carcinogenesis by regulating matrix metalloproteinase-9 expression (1820). Previous studies have revealed that TLR9 was related to the biological characteristics of various types of cancer, including bladder, lung and prostate cancer, such as cell proliferation, invasion, tumor growth and progression (2124). In fact, one previous study reported that TLR9 regulated the expression levels of interleukin (IL)-6 through the myeloid differentiation primary response protein MyD88 (MyD88)/NF-κB signaling pathway in myeloid cells to promote tumor recurrence after irradiation, including in melanoma, bladder carcinoma and colorectal carcinoma (25).

The current study aimed to investigate the effect of TLR9 on the development of CAC through its regulation of the NF-κB signaling pathway. Owing to the synergistic effects of azoxymethane (AOM), a tumor-inducing agent, and dextran sodium sulfate (DSS), a tumor-promoting agent (26), the present study established CAC model mice by co-administering AOM and DSS to analyze the expression levels of TLR9, NF-κB and Ki67 in CAC tissues.

Materials and methods

Reagents and antibodies

AOM (cat. no. A5486) and chloroquine (TLR9 inhibitor; cat. no. C6628-25G) were obtained from Sigma-Aldrich; Merck KGaA. DSS (cat. no. 0216011080-100G) was purchased from MP Biomedicals, LLC. Anti-TLR9 (cat. no. ab134368) and anti-MyD88 (cat. no. ab135693) primary antibodies were obtained from Abcam. Anti-NF-κB (NF-κB p65; cat. no. 8242S), anti-Bcl-xl (cat. no. 2764) and anti-Ki67 (cat. no. 9449S) primary antibodies were obtained from Cell Signaling Technology, Inc. The anti-proliferating cell nuclear antigen (PCNA; cat. no. sc-56) primary antibody was obtained from Santa Cruz Biotechnology, Inc. and the anti-GAPDH (cat. no. TA309157) primary antibody was obtained from OriGene Technologies, Inc. Horseradish peroxidase secondary goat anti-rabbit (cat. no. ZB-2301) and goat anti-mouse (cat. no. ZB-2305) antibodies, used for western blotting, and goat anti-mouse/rabbit antibodies (cat. no. TA130001/TA130015) used for immunohistochemistry (IHC) were obtained from OriGene Technologies, Inc.

Cell lines and culture

The human CRC cell line HT29 was obtained from the American Type Culture Collection and cultured in McCoy's 5A (Modified) medium (Gibco; Thermo Fisher Scientific, Inc.), supplemented with 10% FBS (Gibco; Thermo Fisher Scientific, Inc.), 50 mg/ml penicillin and 50 mg/ml streptomycin, maintained in a humidified atmosphere of 5% CO2 at 37°C.

Western blotting

Cells were lysed in RIPA lysis buffer (Beijing Solarbio Science & Technology Co., Ltd.) at 4°C for 30 min. The protein concentration was measured following centrifugation at 12,000 × g for 15 min at 4°C and quantified using a bicinchoninic acid protein assay kit (Beijing Solarbio Science & Technology Co., Ltd.). An equal amount of protein (50 µg/lane) was separated via 10% SDS-PAGE and electrophoretically transferred onto polyvinylidene difluoride membranes (EMD Millipore). The membranes were blocked with 5% skimmed milk for 2 h at room temperature and were subsequently incubated overnight at 4°C with the following primary antibodies: Anti-TLR9 (1:1,000), anti-NF-κB (1:1,000), anti-Bcl-xl (1:1,000), anti-PCNA (1:1,000), anti-MyD88 (1:500) and anti-GAPDH (1:1,000). Following the primary antibody incubation, the membranes were washed 3 times with PBS for 10 min each and incubated with the corresponding goat anti-rabbit or goat anti-mouse secondary antibodies at 4°C for 4 h. Protein bands were visualized using an ECL reagent (Thermo Fisher Scientific, Inc.) on a Gel Doc XR+ system (Bio-Rad Laboratories, Inc.) and analyzed using Image Lab version 2.0 software (Bio-Rad Laboratories, Inc.).

Wound healing assay

A wound healing assay was performed to analyze the cell migratory ability. Briefly, HT29 cells (3×105/well) were seeded into six-well plates and cultured to 90–100% confluence. Subsequently, a 200-µl pipette tip was used to scratch a wound in the cell monolayer. Fresh serum-free McCoy's 5A (Modified) medium containing different concentrations of chloroquine (0, 15 or 25 µg/ml) was added to each well and cultured for 48 h in a humidified atmosphere of 5% CO2 at 37°C. Images of each well were captured at 0 and 48 h using a light phase contrast microscope (Olympus, ZKX-41; Olympus Corporation) with a magnification of ×100. The wound-healing areas were assessed using ImageJ 1.52a software (National Institutes of Health). The migratory rate of cells = (wound area at 0 h-wound area at 48 h)/area at 0 h.

Colony formation assay

HT29 cells (1.2×105/well) were cultured in 6-well plates for 48 h in medium containing different concentrations of chloroquine (0, 15 or 25 µg/ml) at 37°C, and then seeded into 6-cm cell culture dishes (500 cells/dish) and incubated in complete medium at 37°C. After 14 days, the cells were fixed with 4% paraformaldehyde for 15 min and stained with 0.1% crystal violet for 30 min, both at room temperature. The number of colonies, defined as >50 cells/colony, was counted manually using a light microscope with a magnification of ×100. Relative colony number = number of colonies in observed group/number of colonies in control group. All of the experiments were repeated ≥3 times.

Cell viability assay

The viability of HT29 cells was analyzed using an MTT assay. Briefly, HT29 cells were seeded at a density of 5,000 cells/well in a 96-well plate and treated for 24, 48 or 72 h with chloroquine (0, 100, 125 or 150 µg/ml) at 37°C and then analyzed using an MTT assay as previously described (27).

Animal studies

All animal experiments were approved by the ethics committee of The First Affiliated Hospital of Nanchang University (Nanchang, China). A total of 144 female BALB/c mice (weight, 20–24 g; age, 8–10 weeks) were obtained from Beijing Vital River Laboratory Animal Technology Co. Ltd. (Laboratory Animal License no. SCXK 2017-0012). The animals were maintained with a normal diet and tap water ad libitum at a temperature of 23±2°C and a relative humidity of 40–60%, and artificially illuminated on an approximate 12 h light/dark cycle at the Animal Care Facility in the Medical College of Nanchang University. All of the mice experiments were approved by the Animal Care and Use Committee of Nanchang University. The total experiment lasted for 23 weeks. The mice were divided into four groups (36 mice/group): i) Group A (AOM + DSS), which was intraperitoneally injected without anesthesia with 10 mg/kg AOM once on the first day, followed by 3% DSS given in the drinking water for 1 week and then 2 weeks of distilled water (one cycle), which was repeated for two additional cycles; ii) Group B (AOM), which was intraperitoneally injected with 10 mg/kg AOM once on the first day and provided with distilled drinking water during weeks 0–9; iii) Group C (DSS), which was treated with DSS as described for group A but without the AOM treatment; and iv) Group D (blank control), which received neither DSS nor AOM treatment and was provided with distilled drinking water for the first 9 weeks. All the mice were provided with a normal diet and tap water during weeks 10–23 (Fig. 1A).

The disease activity index (DAI) was evaluated at the end of the experiment using the numerical system described by Tian et al (28). The DAI parameters included total body weight loss (0, none; 1, 1–5%; 2, 5–10%; 3, 10–20%; 4, >20%), stool consistency (0, well-formed pellets; 2, loose stool; 4, diarrhoea) and the presence of fecal occult blood (0, negative; 2, positive; 4, gross bleeding).

Several mice were randomly sacrificed at certain times points following AOM injection (the 1st, 2nd, 3rd, 6th, 9th, 12th, 18th and 23rd weeks). Six mice were sacrificed at weeks 1, 2, 3 and 6, one mouse was sacrificed at week 9 and four mice were sacrificed at weeks 12 and 18 in each group. Additionally, six mice in group D were randomly sacrificed at week 0 as control. All remaining mice were sacrificed at week 23. After the large bowels were resected and washed with PBS, they were carefully examined, photographed and fixed in 10% formalin at room temperature for 24 h. Further histological examinations were subsequently performed.

Histopathological analysis and immunohistochemistry (IHC)

Paraffin-embedded colorectal sections (5-µm-thick) were stained with hematoxylin for 2 min and eosin for 5 min at room temperature to analyze the degree of inflammation using a light microscope with magnifications of ×40 and ×100. Briefly, the severity of inflammation, the thickness of inflammation the severity of epithelial damage and the extent of the lesions were each graded from 0 to 3 by two investigators who were blinded to the treatment groups, as previously described (29,30). The severity of inflammation was adapted from the grading system developed by Truelove and Richards (31,32) as follows: i) Grade 0, no neutrophil infiltration in the lamina propria; ii) Grade I, infiltration of a small number of neutrophils [<10 cells/high power field (HPF)] in the lamina propria, involving a few crypts; iii) Grade II, obvious neutrophil infiltration in the lamina propria (10–50 cells/HPF), involving >50% of the crypts; iv) Grade III, infiltration of neutrophils (>50 cells/HPF) in the lamina propria with crypt abscess; and v) Grade IV, obvious acute inflammation in the lamina propria with ulcer formation. Grade I was classified as mild, grade II was classified as moderate, and grades III and IV were classified as severe. The severity of inflammation ranged from 0 to 3 (0, no inflammation; 1, mild; 2, moderate; 3, severe), the thickness of inflammation ranged from 0 to 3 (0, no inflammation; 1, mucosa; 2, mucosa plus submucosa; 3, transmural), the severity of epithelial damage ranged from 0 to 3 (0, intact epithelium; 1, disruption of architectural structure; 2, erosion; 3, ulceration) and the extent of lesions ranged from 0 to 3 (0, no lesions; 1, punctuate; 2, multifocal; 3, diffuse).

IHC was performed as described in our previous study (33)

Colorectal tissues were fixed in 10% formalin for 24 h at room temperature. Formalin-fixed and paraffin-embedded tissue blocks were cut into 5-µm-thick sections and mounted on glass slides. Slides were heated in an oven at 70°C for 90 min and deparaffined in xylene twice for 10 min each at room temperature, rehydrated in a descending ethanol series (100, 95 and 85% ethanol for 5 min each at room temperature) and incubated in 3% H2O2 for 8 min at room temperature to block endogenous peroxidase. Antigen retrieval was performed by heating in a microwave at 100°C in sodium citrate buffer (3 mM, pH 6.0) for 15 min. Slides were blocked with 5% bovine serum albumin (Beijing Solarbio Science & Technology Co., Ltd.) for 1 h at room temperature to block non-specific antibody binding and incubated overnight at 4°C with the following primary antibodies: Anti-TLR9 (1:400), anti-NF-κB (1:200) and anti-Ki67 (1:100). Following the primary antibody incubation, the sections were washed three times with PBS and incubated with horseradish peroxidase secondary goat anti-mouse/rabbit antibodies (ready to use) at 37°C for 30 min. The sections were stained with 3,3′-diaminobenzidine at room temperature; the duration of staining was based on the staining observed under a light microscope with a magnification of ×100, and the reaction was terminated when the staining was yellowish-brown. The sections were then counterstained with hematoxylin for 1 min at room temperature. The slides were observed under a light microscope with a magnification of ×100. The widely accepted German semi-quantitative scoring system (34,35) was used to determine the staining intensity and area of staining, according to the recommendations of Remmele and Stegner (36). Each specimen was assigned a score according to the intensity of the nucleic, cytoplasmic and/or membrane staining (no staining, not detected=0; weak staining, light yellow=1; moderate staining, yellowish brown=2; strong staining, brown=3) and the extent of stained cells (no staining, 0; 1–24%, 1; 25–49%, 2; 50–74%, 3; 75–100%, 4). The final immunoreactive score was determined by multiplying the intensity score with the extent of stained cells score, ranging from 0 (the minimum) to 12 (the maximum).

Co-immunoprecipitation assay

HT29 cells were lysed in RIPA lysis buffer (Beijin Solarbio Science & Technology Co., Ltd.) at 4°C for 30 min. Whole-cell lysates were pelleted via centrifugation at 10,000 × g for 10 min at 4°C. The supernatant was incubated with an anti-TLR9 antibody (1:200) or goat anti-mouse IgG (1:200; cat. no. ZB-2305; OriGene Technologies, Inc.), together with protein A/G PLUS-Agarose beads (Santa Cruz Biotechnology) at 4°C overnight. The beads were washed three times with the non-lubrol lysis buffer at 2,500 × g centrifugation for 5 min at 4°C, then subjected to SDS-PAGE and subsequent western blotting analysis as aforementioned. Whole cell lysate was used as a control.

Statistical analysis

Statistical analysis was performed using SPSS 20.0 software (IBM Corp.). The data are presented as the mean ± SD. All experiments were performed at least in triplicate. A one-way ANOVA followed by a Tukey's post hoc test was used to determine the statistical differences between >2 groups, whereas an unpaired Student's t-test were used to determine the statistical differences between 2 groups. A Spearman's rank correlation test was used to determine the correlation between the expression levels of TLR9, NF-κB and Ki67 in CRC tissues. P<0.05 was considered to indicate a statistically significant difference.

Results

Construction of the CAC model mice

In groups A (AOM + DSS) and C (DSS), the body weights of the mice decreased with time compared with group D (blank control group; Fig. 1B). The average weight of the mice in group C declined from 21.6±1.3 g at the beginning of the experiment to 20.8±0.8 g by week 23, while the average weight of the mice in group A decreased from 21.6±1.23 g at the beginning of the experiment to 19.5±0.7 g by week 23; the differences between groups A and D, and groups C and D were statistically significant at week 23 (P<0.05). The DAI, which reflects the severity of colitis, was also markedly increased in groups A and C after DSS administration (on the 1st, 3rd and the 6th week; Fig. 1C). No weight loss or any signs of inflammation, such as loose stool or diarrhea, positive fecal occult blood or gross bleeding, were observed in groups B (AOM) and D (blank control). The DAI of groups B and D was zero throughout the modeling process and is therefore not shown in Fig. 1C. The lengths of the large bowels in groups A and C were decreased compared with groups B and D (Fig. 2A). Notably, the lengths of the large bowels were significantly decreased in groups A and C from week 3 compared with the control group (mice sacrificed in group D at week 0) or with group D(P<0.05; Fig. 2B). However, the severity of inflammation was significantly increased in group A compared with in group C at week 23, and the extent of inflammation was significantly increased in group A compared with in group C after week 18 (P<0.05), while there was no significant differences in the thickness of inflammation and the severity of epithelial damage between groups A and C (Fig. 2C).

By the 12th week, colorectal tumors were only observed in the mice of group A (Fig. 2D). Pathological results further revealed that the mice in group A presented with acute inflammation (AIF), chronic inflammation (CIF), adenoma and adenocarcinoma of the colorectum at weeks 3, 6, 12 and 18, respectively (Fig. 2D and E).

Expression levels of TLR9 and NF-κB are simultaneously upregulated during the development of chronic colitis in CRC

IHC staining revealed that TLR9 was located in the cytoplasm of the intestinal epithelial cells (IECs) and inflammatory cells in the lamina propria (Fig. 3A). TLR9 expression levels were significantly upregulated in AIF, CIF, adenoma and adenocarcinoma tissues compared with the corresponding tissues from control mice (group D; P=0.0334, P=0.0379, P=0.0437 and P=0.0008, respectively; Fig. 3B). Furthermore, the protein expression levels of TLR9 was significantly upregulated in the adenocarcinoma tissue compared with the AIF (P=0.0077), CIF (P=0.0278) and adenoma (P=0.0273) tissue (Fig. 3B).

The positive NF-κB region was mainly confined to the cytoplasm of the IECs and inflammatory cells (Fig. 3A). The IHC results revealed that the expression levels of NF-κB were significantly upregulated in the AIF, CIF, adenoma and adenocarcinoma tissues compared with the corresponding tissues from control mice (P=0.0061, P=0.0043, P=0.0019 and P<0.0001, respectively; Fig. 3D). Moreover, NF-κB expression levels were significantly upregulated in the adenocarcinoma tissue compared with the AIF (P<0.0001) and adenoma (P=0.0161) tissues (Fig. 3D).

Ki67 expression levels were observed in the nuclei of IECs and inflammatory cells (Fig. 3A). The IHC results revealed that the Ki67 expression levels were gradually upregulated across the intestinal lesions, including in the AIF, CIF, adenoma and adenocarcinoma tissues (P=0.0331, P=0.0092, P=0.0241 and P=0.0006, respectively) compared with the expression levels in the corresponding tissues from the control mice (Fig. 3C). Interestingly, the expression levels of TLR9 and NF-κB were discovered to be significantly positively correlated with other (rho=0.8236; P<0.0001; Fig. 3E). In addition, a significant positive correlation was also identified between TLR9 and Ki67 expression levels (rho=0.5515; P<0.001; Fig. 3F) and between NF-κB and Ki67 expression levels (rho=0.5103; P<0.01; Fig. 3G).

Downregulated TLR9 expression levels reduces the migration, viability and colony formation of HT29 cells

To further investigate the role of TLR9 in CRC, the human CRC cell line HT29 was treated with chloroquine (an inhibitor of TLR9) in vitro. The results revealed that suppressing TLR9 with chloroquine (at both doses) inhibited the migration, viability and colony formation ability of HT29 cells in a dose-dependent manner (Fig. 4A-E). Additionally, lysates were collected from HT29 cells treated with either different concentrations of chloroquine for 72 h or 25 µg/ml chloroquine for different time periods (Fig. 4F-I) and western blotting was performed. The analysis revealed that the expression levels of TLR9, NF-κB, PCNA, MyD88 and Bcl-xl were gradually downregulated in HT29 cells treated with increasing doses of chloroquine compared with the control group (Fig. 4F and G). A similar trend was observed in the expression levels of these proteins as the duration of chloroquine treatment increased (Fig. 4H and I). Thus, these results indicated that the expression levels of TLR9, NF-κB, PCNA, MyD88 and Bcl-xl in HT29 cells treated with chloroquine may be downregulated in both a dose- and time-dependent manner (Fig. 4F-I). To verify whether TLR9 affected the colorectal carcinogenesis by interacting with NF-κB, co-immunoprecipitation assay was used to detect the interaction between TLR9 and NF-κB in HT-29 cells. The results revealed that there was an interaction between TLR9 and NF-κB (Fig. 4J)

Figure 4.

Chloroquine inhibits the migration, viability and colony formation of HT29 cells by inhibiting TLR9. Chloroquine (15 and 25 µg/ml) inhibited the migration of HT29 cells in a dose-dependent manner at 48 h compared with the control group, which was determined using a wound healing assay. (A) Representative images were photographed (magnification ×100) and (B) migration rates were calculated. Proliferation rate of HT29 cells was reduced by chloroquine treatment (15 and 25 µg/ml), which was determined using a colony formation assay. (C) Representative images were photographed and (D) the relative colony number was analyzed. *P<0.05, **P<0.01, ***P<0.001. (E) Viability of HT29 cells was decreased by chloroquine (100, 125 and 150 µg/ml) at 24, 48 and 72 h compared with the control cells, as determined using an MTT assay. (F) Expression levels of TLR9, NF-κB, PCNA, MyD88 and Bcl-xl in lysates obtained from HT29 cells treated with numerous concentrations of chloroquine (0, 5, 10, 15, 20 or 25 µg/ml) for 72 h were analyzed using western blotting. (G) Semi-quantification of the expression levels of the proteins presented in part (F) was performed using ImageJ software. GAPDH was used as the loading control and for normalization. (H) Expression levels of TLR9, NF-κB, PCNA, MyD88 and Bcl-xl in lysates obtained from HT29 cells treated with 25 µg/ml chloroquine for numerous durations (0, 24, 48 or 72 h) were analyzed using western blotting. (I) Semi-quantification of the expression levels of the proteins presented in part (H) was performed using ImageJ software. GAPDH was used as the loading control and for normalization. (J) TLR9 interacted with the NF-κB protein in HT29 cells, which was determined using a co-immunoprecipitation assay. Goat anti-mouse IgG antibody was used as the negative control. All data are expressed as the mean ± SD of three independent experiments. *P<0.05, **P<0.01, ***P<0.001 vs. control/0 h. TLR9, Toll-like receptor 9; PCNA, proliferating cell nuclear antigen; MyD88, myeloid differentiation primary response protein MyD88; IP, immunoprecipitated; IB, immunoblotting.

Discussion

Colorectal carcinogenesis is a multi-step process, starting from normal crypts to aberrant crypt foci, then to polyps, adenoma and eventually adenocarcinoma (37,38). It has been reported that individuals with IBD may have an increased risk of developing CRC, which is directly proportional to the extent and duration of their disease (39,40). However, the exact mechanism and duration required for chronic colitis to develop into adenoma and then adenocarcinoma remains unclear (40). It has been suggested that patients with IBD have an increased risk of CRC following the inflammation-dysplasia-carcinoma model (41), including dysplasia and CRC as primary consequences of chronic inflammation. However, there are currently still no defined molecular biomarkers or existing monitoring protocols for detecting the occurrence of a malignant tumor, except for frequent colonoscopy examinations.

In the present study, an acute colitis-chronic colitis-adenoma-adenocarcinoma model was successfully constructed via AOM/DSS induction. Using this model, TLR9 expression levels were discovered to be upregulated as the severity of the colorectal lesions increased, which indicated that TLR9 protein expression levels may be continuously activated during colitis-CRC development. TLR9 is a critical protein associated with innate and acquired immunity (42), and it has been demonstrated to serve a significant role in the development of colitis (11,43) and sporadic CRC (12,13). However, the mechanism by which TLR9 regulates the development of CRC remains to be elucidated.

Interestingly, IHC analysis revealed that the expression levels of NF-κB and Ki67 were simultaneously upregulated alongside TLR9 expression levels. Notably, inhibiting TLR9 decreased the migration, proliferation and viability of HT29 cells in vitro, and TLR9 expression levels in vivo were identified to be significantly positively correlated with the expression levels of NF-κB and Ki67 (a cell proliferation marker) during the transition from colitis to CRC. The present study further revealed that the inhibition of TLR9 in vitro significantly downregulated the expression levels of NF-κB, MyD88, PCNA and the anti-apoptotic protein Bcl-xl in a dose- and time-dependent manner. Notably, a previous study reported that TLR9 promoted the tumor-propagating potential of prostate cancer cells via NF-κB signaling (22). Thus, the findings of the present study indicated that TLR9 may promote CAC through NF-κB signaling. However, these findings may be controversial because other previous studies have revealed that TLR9 agonists exerted an antitumor effect in CRC (14,15,44,45). The majority of these studies primarily focused on the role of TLR9 in colitis or sporadic CRC, whereas the current study focused on the role of TLR9 in the pathogenesis of CAC to provide novel targets for the treatment of CAC. For example, alterations in microtubule end-binding protein 1 were identified as a characteristic of sporadic, but not UC-associated CRC (46), and in another previous study, the immune profiling patterns of patients with CAC were significantly different compared with the patients with sporadic CRC (47).

Chloroquine, a non-specific inhibitor of TLR9, is an old antimalarial drug (48), which has recently attracted significant interest for its potential antitumor properties; for example, numerous studies have reported that chloroquine directly regulated cancer cells by inducing apoptosis, inhibiting autophagy, interacting with nucleotides, eliminating cancer stem cells and enhancing the growth of cancer cells (4951). Chloroquine also inhibited the expression levels of TLR9 by preventing the acidification and maturation of the endosomes, and the trafficking of TLRs (52). Due to the multiple effects of chloroquine on tumor cells, different concentrations of chloroquine were selected for use in the present study based on the lowest dose according to previous studies (49,53), in order to obtain the best possible results with low toxicity to the cells. In the future, investigations using small interfering RNA targeting TLR9 should be performed to determine the effect on the biological processes of CRC cell lines to further verify the findings of the present study. Thus, our future studies will focus on investigating the precise molecular mechanism by which TLR9 participates in the early occurrence of colorectal carcinogenesis.

In conclusion, the present study developed a CAC animal model. The findings indicated that TLR9 may be closely associated with the process of inflammation-dysplasia-carcinoma and may impact carcinogenesis by regulating the NF-κB signaling pathway. These results may provide promising potential to be developed into an early detection protocol or therapeutic molecular target for CRC.

Acknowledgements

Not applicable.

Funding

The present study was supported by grants from the National Natural Science Foundation of China (grant nos. 81660404 and 81560398), the Foundation of Jiangxi Educational Committee (grant no. GJJ170016) and the Graduate Student Innovation Funding Program of Nanchang University (grant no. CX2019119).

Availability of data and materials

All data generated or analyzed during this study are included in this published article. The original data are available from the corresponding author on reasonable request.

Authors' contributions

CZ and QL designed the study and drafted the manuscript. QL and LZ performed the experiments. QL, CT and ZZ analyzed the data. QL, CZ and YC revised the manuscript for important intellectual content. CZ and YC made substantial contributions to conception, design and coordination of the study and gave final approval of the version to be published. All authors have read and approved the final manuscript.

Ethics approval and consent to participate

All animal experiments were approved by the Ethics Committee of The First Affiliated Hospital of Nanchang University (Nanchang, China; approval no. 2015- 045).

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

AIF

acute inflammation

AOM

azoxymethane

CAC

colitis-associated colorectal cancer

CIF

chronic inflammation

CRC

colorectal cancer

DAI

disease activity index

DSS

dextran sodium sulfate

IBD

inflammatory bowel disease

IECs

intestinal epithelial cells

IHC

immunohistochemistry

PCNA

proliferating cell nuclear antigen

TLR9

Toll-like receptor 9

UC

ulcerative colitis

References

1 

Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA and Jemal A: Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 68:394–424. 2018. View Article : Google Scholar : PubMed/NCBI

2 

Balkwill F and Mantovani A: Inflammation and cancer: Back to virchow? Lancet. 357:539–545. 2001. View Article : Google Scholar : PubMed/NCBI

3 

Eaden JA, Abrams KR and Mayberry JF: The risk of colorectal cancer in ulcerative colitis: A meta-analysis. Gut. 48:526–535. 2001. View Article : Google Scholar : PubMed/NCBI

4 

Moldoveanu AC, Diculescu M and Braticevici CF: Cytokines in inflammatory bowel disease. Rom J Intern Med. 53:118–127. 2015.PubMed/NCBI

5 

Hemmi H, Takeuchi O, Kawai T, Kaisho T, Sato S, Sanjo H, Matsumoto M, Hoshino K, Wagner H, Takeda K and Akira S: A toll-like receptor recognizes bacterial DNA. Nature. 408:740–745. 2000. View Article : Google Scholar : PubMed/NCBI

6 

Krieg AM, Yi AK, Matson S, Waldschmidt TJ, Bishop GA, Teasdale R, Koretzky GA and Klinman DM: CpG motifs in bacterial DNA trigger direct B-cell activation. Nature. 374:546–549. 1995. View Article : Google Scholar : PubMed/NCBI

7 

Kolumam GA, Thomas S, Thompson LJ, Sprent J and Murali-Krishna K: Type I interferons act directly on CD8 T cells to allow clonal expansion and memory formation in response to viral infection. J Exp Med. 202:637–650. 2005. View Article : Google Scholar : PubMed/NCBI

8 

Havenar-Daughton C, Kolumam GA and Murali-Krishna K: Cutting Edge: The direct action of type I IFN on CD4 T cells is critical for sustaining clonal expansion in response to a viral but not a bacterial infection. J Immunol. 176:3315–3319. 2006. View Article : Google Scholar : PubMed/NCBI

9 

Akira S and Takeda K: Toll-Like receptor signaling. Nat Rev Immunol. 4:499–511. 2004. View Article : Google Scholar : PubMed/NCBI

10 

Sanchez-Munoz F, Fonseca-Camarillo G, Villeda-Ramirez MA, Miranda-Pérez E, Mendivil EJ, Barreto-Zúñiga R, Uribe M, Bojalil R, Domínguez-López A and Yamamoto-Furusho JK: Transcript levels of toll-like receptors 5, 8 and 9 correlate with inflammatory activity in ulcerative colitis. BMC Gastroenterol. 11:1382011. View Article : Google Scholar : PubMed/NCBI

11 

Fan Y and Liu B: Expression of Toll-Like receptors in the mucosa of patients with ulcerative colitis. Exp Ther Med. 9:1455–1459. 2015. View Article : Google Scholar : PubMed/NCBI

12 

Eiro N, Gonzalez L, Gonzalez LO, Andicoechea A, Fernández-Díaz M, Altadill A and Vizoso FJ: Study of the expression of toll-like receptors in different histological types of colorectal polyps and their relationship with colorectal cancer. J Clin Immunol. 32:848–854. 2012. View Article : Google Scholar : PubMed/NCBI

13 

Gao C, Qiao T, Zhang B, Yuan S, Zhuang X and Luo Y: TLR9 signaling activation at different stages in colorectal cancer and NF-kappaB expression. Onco Targets Ther. 11:5963–5971. 2018. View Article : Google Scholar : PubMed/NCBI

14 

Nojiri K, Sugimoto K, Shiraki K, Tameda M, Inagaki Y, Kusagawa S, Ogura S, Tanaka J, Yoneda M, Yamamoto N, et al: The expression and function of Toll-like receptors 3 and 9 in human colon carcinoma. Oncol Rep. 29:1737–1743. 2013. View Article : Google Scholar : PubMed/NCBI

15 

Shahriari S, Rezaeifard S, Moghimi HR, Khorramizadeh MR and Faghih Z: Cell membrane and intracellular expression of toll-like receptor 9 (TLR9) in colorectal cancer and breast cancer cell-lines. Cancer Biomark. 18:375–380. 2017. View Article : Google Scholar : PubMed/NCBI

16 

Taniguchi K and Karin M: NF-κB, inflammation, immunity and cancer: Coming of age. Nat Rev Immunol. 18:309–324. 2018. View Article : Google Scholar : PubMed/NCBI

17 

Capece D, Verzella D, Tessitore A, Alesse E, Capalbo C and Zazzeroni F: Cancer secretome and inflammation: The bright and the dark sides of NF-kappaB. Semin Cell Dev Biol. 78:51–61. 2018. View Article : Google Scholar : PubMed/NCBI

18 

Said AH, Raufman JP and Xie G: The role of matrix metalloproteinases in colorectal cancer. Cancers. 6:366–375. 2014. View Article : Google Scholar : PubMed/NCBI

19 

Xue Q, Cao L, Chen XY, Zhao J, Gao L, Li SZ and Fei Z: High expression of MMP9 in glioma affects cell proliferation and is associated with patient survival rates. Oncol Lett. 13:1325–1330. 2017. View Article : Google Scholar : PubMed/NCBI

20 

Ha SH, Kwon KM, Park JY, Abekura F, Lee YC, Chung TW, Ha KT, Chang HW, Cho SH, Kim JK and Kim CH: Esculentoside H inhibits colon cancer cell migration and growth through suppression of MMP-9 gene expression via NF-kB signaling pathway. J Cell Biochem. 120:9810–9819. 2019. View Article : Google Scholar : PubMed/NCBI

21 

Xu L, Wang C, Wen Z, Yao X, Liu Z, Li Q, Wu Z, Xu Z, Liang Y and Ren T: Selective up-regulation of CDK2 is critical for TLR9 signaling stimulated proliferation of human lung cancer cell. Immunol Lett. 127:93–99. 2010. View Article : Google Scholar : PubMed/NCBI

22 

Moreira D, Zhang Q, Hossain DM, Nechaev S, Li H, Kowolik CM, D'Apuzzo M, Forman S, Jones J, Pal SK and Kortylewski M: TLR9 signaling through NF-kappaB/RELA and STAT3 promotes tumor-propagating potential of prostate cancer cells. Oncotarget. 6:17302–17313. 2015. View Article : Google Scholar : PubMed/NCBI

23 

Cai Y, Huang J, Xing H, Li B, Li L, Wang X, Peng D and Chen J: Contribution of FPR and TLR9 to hypoxia-induced chemoresistance of ovarian cancer cells. Onco Targets Ther. 12:291–301. 2019. View Article : Google Scholar : PubMed/NCBI

24 

Olbert PJ, Kesch C, Henrici M, Subtil FS, Honacker A, Hegele A, Hofmann R and Hänze J: TLR4- and TLR9-dependent effects on cytokines, cell viability, and invasion in human bladder cancer cells. Urol Oncol. 33:e119–e127. 2015. View Article : Google Scholar

25 

Gao C, Kozlowska A, Nechaev S, Li H, Zhang Q, Hossain DM, Kowolik CM, Chu P, Swiderski P, Diamond DJ, et al: TLR9 signaling in the tumor microenvironment initiates cancer recurrence after radiotherapy. Cancer Res. 73:7211–7221. 2013. View Article : Google Scholar : PubMed/NCBI

26 

Tanaka T: Colorectal carcinogenesis: Review of human and experimental animal studies. J Carcinog. 8:52009. View Article : Google Scholar : PubMed/NCBI

27 

Zeng CY, Zhan YS, Huang J and Chen YX: MicroRNA7 suppresses human colon cancer invasion and proliferation by targeting the expression of focal adhesion kinase. Mol Med Rep. 13:1297–1303. 2016. View Article : Google Scholar : PubMed/NCBI

28 

Tian Y, Xu Q, Sun L, Ye Y and Ji G: Short-Chain fatty acids administration is protective in colitis-associated colorectal cancer development. J Nutr Biochem. 57:103–109. 2018. View Article : Google Scholar : PubMed/NCBI

29 

Tian Y, Wang K, Wang Z, Li N and Ji G: Chemopreventive effect of dietary glutamine on colitis-associated colon tumorigenesis in mice. Carcinogenesis. 34:1593–1600. 2013. View Article : Google Scholar : PubMed/NCBI

30 

Hudert CA, Weylandt KH, Lu Y, Wang J, Hong S, Dignass A, Serhan CN and Kang JX: Transgenic mice rich in endogenous omega-3 fatty acids are protected from colitis. Proc Natl Acad Sci USA. 103:11276–11281. 2006. View Article : Google Scholar : PubMed/NCBI

31 

Truelove SC and Richards WC: Biopsy studies in ulcerative colitis. Br Med J. 9:1315–1318. 1956. View Article : Google Scholar

32 

Sangfelt P, Carlson M, Thörn M, Lööf L and Raab Y: Neutrophil and eosinophil granule proteins as markers of response to local prednisolone treatment in distal ulcerative colitis and proctitis. Am J Gastroenterol. 96:1085–1090. 2001. View Article : Google Scholar : PubMed/NCBI

33 

Zeng C, Wang Y, Lu Q, Chen J, Zhang J, Liu T, Lv N and Luo S: SPOP suppresses tumorigenesis by regulating Hedgehog/Gli2 signaling pathway in gastric cancer. J Exp Clin Cancer Res. 33:752014. View Article : Google Scholar : PubMed/NCBI

34 

Scharl A, Vierbuchen M, Conradt B, Moll W, Würz H and Bolte A: Immunohistochemical detection of progesterone receptor in formalin-fixed and paraffin-embedded breast cancer tissue using a monoclonal antibody. Arch Gynecol Obstet. 247:63–71. 1990. View Article : Google Scholar : PubMed/NCBI

35 

Pan X, Zhou T, Tai YH, Wang C, Zhao J, Cao Y, Chen Y, Zhang PJ, Yu M, Zhen C, et al: Elevated expression of CUEDC2 protein confers endocrine resistance in breast cancer. Nat Med. 17:708–714. 2011. View Article : Google Scholar : PubMed/NCBI

36 

Remmele W and Stegner HE: Recommendation for uniform definition of an immunoreactive score (IRS) for immunohistochemical estrogen receptor detection (ER-ICA) in breast cancer tissue. Pathologe. 8:138–140. 1987.PubMed/NCBI

37 

Kinzler KW and Vogelstein B: Lessons from hereditary colorectal cancer. Cell. 87:159–170. 1996. View Article : Google Scholar : PubMed/NCBI

38 

Brenner H, Kloor M and Pox CP: Colorectal cancer. Lancet. 383:1490–1502. 2014. View Article : Google Scholar : PubMed/NCBI

39 

Beaugerie L and Itzkowitz SH: Cancers complicating inflammatory bowel disease. N Engl J Med. 372:1441–1452. 2015. View Article : Google Scholar : PubMed/NCBI

40 

Terzić J, Grivennikov S, Karin E and Karin M: Inflammation and colon cancer. Gastroenterology. 138:2101–2114. 2010. View Article : Google Scholar : PubMed/NCBI

41 

Itzkowitz SH and Yio X: Inflammation and cancer IV. Colorectal cancer in inflammatory bowel disease: The role of inflammation. Am J Physiol Gastrointest Liver Physiol. 287:G7–G17. 2004. View Article : Google Scholar : PubMed/NCBI

42 

Akira S, Takeda K and Kaisho T: Toll-Like receptors: Critical proteins linking innate and acquired immunity. Nat Immunol. 2:675–680. 2001. View Article : Google Scholar : PubMed/NCBI

43 

Atreya R, Bloom S, Scaldaferri F, Gerardi V, Admyre C, Karlsson Å, Knittel T, Kowalski J, Lukas M, Löfberg R, et al: Clinical effects of a topically applied toll-like receptor 9 agonist in active moderate-to-severe ulcerative colitis. J Crohn's Colitis. 10:1294–1302. 2016. View Article : Google Scholar

44 

Schmoll HJ, Wittig B, Arnold D, Riera-Knorrenschild J, Nitsche D, Kroening H, Mayer F, Andel J, Ziebermayr R and Scheithauer W: Maintenance treatment with the immunomodulator MGN1703, a Toll-like receptor 9 (TLR9) agonist, in patients with metastatic colorectal carcinoma and disease control after chemotherapy: A randomised, double-blind, placebo-controlled trial. J Cancer Res Clin Oncol. 140:1615–1624. 2014. View Article : Google Scholar : PubMed/NCBI

45 

Dong T, Yi T, Yang M, Lin S, Li W, Xu X, Hu J, Jia L, Hong X and Niu W: Co-Operation of alpha-galactosylceramide-loaded tumour cells and TLR9 agonists induce potent anti-tumour responses in a murine colon cancer model. Biochem J. 473:7–19. 2016. View Article : Google Scholar : PubMed/NCBI

46 

Gemoll T, Kollbeck SL, Karstens KF, Hò GG, Hartwig S, Strohkamp S, Schillo K, Thorns C, Oberländer M, Kalies K, et al: EB1 protein alteration characterizes sporadic but not ulcerative colitis associated colorectal cancer. Oncotarget. 8:54939–54950. 2017. View Article : Google Scholar : PubMed/NCBI

47 

Soh JS, Jo SI, Lee H, Do EJ, Hwang SW, Park SH, Ye BD, Byeon JS, Yang SK, Kim JH, et al: Immunoprofiling of colitis-associated and sporadic colorectal cancer and its clinical significance. Sci Rep. 9:68332019. View Article : Google Scholar : PubMed/NCBI

48 

Kuznik A, Bencina M, Svajger U, Jeras M, Rozman B and Jerala R: Mechanism of endosomal TLR inhibition by antimalarial drugs and imidazoquinolines. J Immunol. 186:4794–4804. 2011. View Article : Google Scholar : PubMed/NCBI

49 

Pascolo S: Time to use a dose of chloroquine as an adjuvant to anti-cancer chemotherapies. Eur J Pharmacol. 771:139–144. 2016. View Article : Google Scholar : PubMed/NCBI

50 

Maes H, Kuchnio A, Carmeliet P and Agostinis P: Chloroquine anticancer activity is mediated by autophagy-independent effects on the tumor vasculature. Mol Cell Oncol. 3:e9700972016. View Article : Google Scholar : PubMed/NCBI

51 

Lin YC, Lin JF, Wen SI, Yang SC, Tsai TF, Chen HE, Chou KY and Hwang TI: Chloroquine and hydroxychloroquine inhibit bladder cancer cell growth by targeting basal autophagy and enhancing apoptosis. Kaohsiung J Med Sci. 33:215–223. 2017. View Article : Google Scholar : PubMed/NCBI

52 

Mohamed FE, Al-Jehani RM, Minogue SS, Andreola F, Winstanley A, Damink SW, Habtesion A, Malagó M, Davies N, Luong TV, et al: Effect of toll-like receptor 7 and 9 targeted therapy to prevent the development of hepatocellular carcinoma. Liver Int. 35:1063–1076. 2015. View Article : Google Scholar : PubMed/NCBI

53 

Zhang Y, Li Y, Li Y, Ma Y, Wang H and Wang Y: Chloroquine inhibits MGC803 gastric cancer cell migration via the Toll-like receptor 9/nuclear factor kappa B signaling pathway. Mol Med Rep. 11:1366–1371. 2015. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

October-2020
Volume 20 Issue 4

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Luo Q, Zeng L, Tang C, Zhang Z, Chen Y and Zeng C: TLR9 induces colitis‑associated colorectal carcinogenesis by regulating NF‑&kappa;B expression levels. Oncol Lett 20: 110, 2020
APA
Luo, Q., Zeng, L., Tang, C., Zhang, Z., Chen, Y., & Zeng, C. (2020). TLR9 induces colitis‑associated colorectal carcinogenesis by regulating NF‑&kappa;B expression levels. Oncology Letters, 20, 110. https://doi.org/10.3892/ol.2020.11971
MLA
Luo, Q., Zeng, L., Tang, C., Zhang, Z., Chen, Y., Zeng, C."TLR9 induces colitis‑associated colorectal carcinogenesis by regulating NF‑&kappa;B expression levels". Oncology Letters 20.4 (2020): 110.
Chicago
Luo, Q., Zeng, L., Tang, C., Zhang, Z., Chen, Y., Zeng, C."TLR9 induces colitis‑associated colorectal carcinogenesis by regulating NF‑&kappa;B expression levels". Oncology Letters 20, no. 4 (2020): 110. https://doi.org/10.3892/ol.2020.11971