Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 154
1.
Chin J Integr Med ; 30(2): 135-142, 2024 Feb.
Article En | MEDLINE | ID: mdl-37434030

OBJECTIVE: To investigate the effect of Huangqin Decoction (HQD) on nuclear factor erythroid 2 related-factor 2 (Nrf2)/heme oxygenase (HO-1) signaling pathway by inducing the colitis-associated carcinogenesis (CAC) model mice with azoxymethane (AOM)/dextran sodium sulfate (DSS). METHODS: The chemical components of HQD were analyzed by liquid chromatography-quadrupole-time-of-flight mass spectrometry (LC-Q-TOF-MS/MS) to determine the molecular constituents of HQD. Totally 48 C57BL/6J mice were randomly divided into 6 groups by a random number table, including control, model (AOM/DSS), mesalazine (MS), low-, medium-, and high-dose HQD (HQD-L, HQD-M, and HQD-H) groups, 8 mice in each group. Except for the control group, the mice in the other groups were intraperitoneally injected with AOM (10 mg/kg) and administrated with 2.5% DSS orally for 1 week every two weeks (totally 3 rounds of DSS) to construct a colitis-associated carcinogenesis mouse model. The mice in the HQD-L, HQD-M and HQD-H groups were given HQD by gavage at doses of 2.925, 5.85, and 11.7 g/kg, respectively; the mice in the MS group was given a suspension of MS at a dose of 0.043 g/kg (totally 11 weeks). The serum levels of malondialdehyde (MDA) and superoxide dismutase (SOD) were measured by enzyme-linked immunosorbent assay. The mRNA and protein expression levels of Nrf2, HO-1, and inhibitory KELCH like ECH-related protein 1 (Keap1) in colon tissue were detected by quantitative real-time PCR, immunohistochemistry, and Western blot, respectively. RESULTS: LC-Q-TOF-MS/MS analysis revealed that the chemical constituents of HQD include baicalin, paeoniflorin, and glycyrrhizic acid. Compared to the control group, significantly higher MDA levels and lower SOD levels were observed in the model group (P<0.05), whereas the expressions of Nrf2 and HO-1 were significantly decreased, and the expression of Keap1 increased (P<0.01). Compared with the model group, serum MDA level was decreased and SOD level was increased in the HQD-M, HQD-H and MS groups (P<0.05). Higher expressions of Nrf2 and HO-1 were observed in the HQD groups. CONCLUSION: HQD may regulate the expression of Nrf2 and HO-1 in colon tissue, reduce the expression of MDA and increase the expression of SOD in serum, thus delaying the progress of CAC in AOM/DSS mice.


Antioxidants , Colitis , Mice , Animals , Antioxidants/pharmacology , Kelch-Like ECH-Associated Protein 1/metabolism , Scutellaria baicalensis/chemistry , Scutellaria baicalensis/metabolism , NF-E2-Related Factor 2/metabolism , Tandem Mass Spectrometry , Mice, Inbred C57BL , Colitis/complications , Colitis/drug therapy , Colitis/metabolism , Signal Transduction , Carcinogenesis , Azoxymethane/pharmacology , Superoxide Dismutase/metabolism
2.
Am J Physiol Gastrointest Liver Physiol ; 325(4): G318-G333, 2023 10 01.
Article En | MEDLINE | ID: mdl-37489869

Currently available colorectal cancer (CRC) therapies have limited efficacy and severe adverse effects that may be overcome with the alternative use of natural compounds. We previously reported that panaxynol (PA), a bioactive component in American ginseng, possesses anticancer properties in vitro and suppresses murine colitis through its proapoptotic and anti-inflammatory properties. Because colitis is a predisposing factor of CRC and inflammation is a major driver of CRC, we sought to evaluate the therapeutic potential of PA in CRC. Azoxymethane-dextran sodium sulfate (AOM/DSS) mice (C57BL/6) were administered 2.5 mg/kg PA or vehicle 3 times/wk via oral gavage over 12 wk. PA improved clinical symptoms (P ≤ 0.05) and reduced tumorigenesis (P ≤ 0.05). This improvement may be reflective of PA's restorative effect on intestinal barrier function; PA upregulated the expression of essential tight junction and mucin genes (P ≤ 0.05) and increased the abundance of mucin-producing goblet cells (P ≤ 0.05). Given that macrophages play a substantial role in the pathogenesis of CRC and that we previously demonstrated that PA targets macrophages in colitis, we next assessed macrophages. We show that PA reduces the relative abundance of colonic macrophages within the lamina propria (P ≤ 0.05), and this was consistent with a reduction in the expression of important markers of macrophages and inflammation (P ≤ 0.05). We further confirmed PA's inhibitory effects on macrophages in vitro under CRC conditions (P ≤ 0.05). These results suggest that PA is a promising therapeutic compound to treat CRC and improve clinical symptoms given its ability to inhibit macrophages and modulate the inflammatory environment in the colon.NEW & NOTEWORTHY We report that panaxynol (PA) reduces colorectal cancer (CRC) by improving the colonic and tumor environment. Specifically, we demonstrate that PA improves crypt morphology, upregulates crucial tight junction and mucin genes, and promotes the abundance of mucin-producing goblet cells. Furthermore, PA reduces macrophages and associated inflammation, important drivers of CRC, in the colonic environment. This present study provides novel insights into the potential of PA as a therapeutic agent to ameliorate CRC tumorigenesis.


Colitis , Colorectal Neoplasms , Mice , Animals , Disease Models, Animal , Mice, Inbred C57BL , Inflammation/metabolism , Colitis/chemically induced , Colitis/drug therapy , Colitis/metabolism , Carcinogenesis/metabolism , Cell Transformation, Neoplastic/metabolism , Azoxymethane/metabolism , Azoxymethane/pharmacology , Azoxymethane/therapeutic use , Macrophages/metabolism , Colorectal Neoplasms/metabolism , Mucins/metabolism , Dextran Sulfate/pharmacology
3.
Comp Med ; 72(5): 330-335, 2022 10 01.
Article En | MEDLINE | ID: mdl-36123012

Multiple animal models have been developed to investigate the pathogenesis of colorectal cancer and to evaluate potential treatments. One model system uses azoxymethane, a metabolite of cycasin, alone and in conjunction with dextran sodium sulfate to induce colon cancer in rodents. Azoxymethane is metabolized by hepatic P450 enzymes and can also be eliminated through the kidneys. In this study, C57BL/6J mice were fed either standard or high-fat diet and then all mice received azoxymethane at 10 mg/kg body weight twice a week for 6 wk. Shortly after the end of treatment, high mortality occurred in mice in the high-fat diet group. Postmortem examination revealed hepatic and renal pathology in mice on both diets. Histologic changes in liver included hepatocytomegaly with nuclear pleomorphism and bile duct hyperplasia accompanied by mixed inflammatory-cell infiltrates. Changes in the kidneys ranged from basophilia of tubular epithelium to tubular atrophy. The results indicate that further optimization of this model is needed when feeding a high-fat diet and giving multiple azoxymethane doses to induce colon cancer in C57BL/6J mice.


Colonic Neoplasms , Diet, High-Fat , Mice , Animals , Azoxymethane/metabolism , Azoxymethane/pharmacology , Diet, High-Fat/adverse effects , Mice, Inbred C57BL , Cycasin , Dextrans , Colonic Neoplasms/chemically induced , Liver/pathology , Kidney/pathology , Diet , Colon
4.
Food Funct ; 13(13): 6947-6961, 2022 Jul 04.
Article En | MEDLINE | ID: mdl-35575226

This study aimed to investigate the alleviative effects of Lactobacillus kefiranofaciens JKSP109 (LK) and Saccharomyces cerevisiae JKSP39 (SC) isolated from Tibetan kefir grain on colon inflammation and colorectal carcinogenesis. Azoxymethane (AOM) and dextran sulfate sodium (DSS) were used to establish a mouse model of colorectal cancer (CRC). The treatment group mice were administered with LK, SC, or the combination of LK and SC for five days per week from the day of receiving AOM. The composition of the gut microbiota was assessed using internal transcribed spacer 2 and 16S rRNA gene high-throughput sequencing. Furthermore, the biomarkers associated with gut barrier integrity, inflammation, regulators of cell proliferation, and apoptosis were evaluated. The results showed that the administration of LK, SC, and their combination increased the body weights and decreased the disease activity index (DAI) score and tumor multiplicity. As compared to the CRC model group, the three treatment groups positively regulated the gut microbiota. Meanwhile, the three treatments also enhanced the gut barrier, decreased the expression of proinflammatory cytokines and oncocyte proliferation indicators, and increased the expression of terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL)-positive tumor epithelial cells and content of short chain fatty acids in fecal samples. All these results indicated that the LK and SC alleviated the inflammation and colorectal carcinogenesis in AOM/DSS-induced CRC mouse models, and the majority of tested indexes in the combination group were superior to single strain groups.


Colitis , Colorectal Neoplasms , Kefir , Animals , Azoxymethane/pharmacology , Carcinogenesis , Colitis/metabolism , Colorectal Neoplasms/metabolism , Dextran Sulfate/adverse effects , Disease Models, Animal , Inflammation/metabolism , Lactobacillus , Mice , RNA, Ribosomal, 16S , Saccharomyces cerevisiae , Tibet
5.
Biochim Biophys Acta Mol Basis Dis ; 1867(10): 166179, 2021 10 01.
Article En | MEDLINE | ID: mdl-34082069

Emerging data show a rise in colorectal cancer (CRC) incidence in young men and women that is often chemoresistant. One potential risk factor is an alteration in the microbiome. Here, we investigated the role of TGF-ß signaling on the intestinal microbiome and the efficacy of chemotherapy for CRC induced by azoxymethane and dextran sodium sulfate in mice. We used two genotypes of TGF-ß-signaling-deficient mice (Smad4+/- and Smad4+/-Sptbn1+/-), which developed CRC with similar phenotypes and had similar alterations in the intestinal microbiome. Using these mice, we evaluated the intestinal microbiome and determined the effect of dysfunctional TGF-ß signaling on the response to the chemotherapeutic agent 5-Fluoro-uracil (5FU) after induction of CRC. Using shotgun metagenomic sequencing, we determined gut microbiota composition in mice with CRC and found reduced amounts of beneficial species of Bacteroides and Parabacteroides in the mutants compared to the wild-type (WT) mice. Furthermore, the mutant mice with CRC were resistant to 5FU. Whereas the abundances of E. boltae, B.dorei, Lachnoclostridium sp., and Mordavella sp. were significantly reduced in mice with CRC, these species only recovered to basal amounts after 5FU treatment in WT mice, suggesting that the alterations in the intestinal microbiome resulting from compromised TGF-ß signaling impaired the response to 5FU. These findings could have implications for inhibiting the TGF-ß pathway in the treatment of CRC or other cancers.


Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/metabolism , Fluorouracil/pharmacology , Gastrointestinal Microbiome/physiology , Signal Transduction/physiology , Transforming Growth Factor beta/metabolism , Animals , Antineoplastic Agents/pharmacology , Azoxymethane/pharmacology , Colon/drug effects , Colon/metabolism , Colon/microbiology , Colorectal Neoplasms/microbiology , Dextran Sulfate/pharmacology , Female , Male , Mice , Mice, Inbred C57BL , Signal Transduction/drug effects , Smad4 Protein/metabolism
6.
Int J Mol Sci ; 22(5)2021 Mar 05.
Article En | MEDLINE | ID: mdl-33807620

Glycyrrhizin (GL), an important active ingredient of licorice root, which weakens the proinflammatory effects of high-mobility group box 1 (HMGB1) by blocking HMGB1 signaling. In this study, we investigated whether GL could suppress inflammation and carcinogenesis in an azoxymethane (AOM)/dextran sodium sulfate (DSS)-induced murine model of colorectal cancer. ICR mice were divided into four groups (n = 5, each)-control group, GL group, colon cancer (CC) group, and GL-treated CC (CC + GL) group, and sacrificed after 20 weeks. Plasma levels of interleukin (IL)-6 and tumor necrosis factor (TNF)-α were measured using an enzyme-linked immunosorbent assay. The colonic tissue samples were immunohistochemically stained with DNA damage markers (8-nitroguanine and 8-oxo-7,8-dihydro-2'-deoxy-guanosine), inflammatory markers (COX-2 and HMGB1), and stem cell markers (YAP1 and SOX9). The average number of colonic tumors and the levels of IL-6 and TNF-α in the CC + GL group were significantly lower than those in the CC group. The levels of all inflammatory and cancer markers were significantly reduced in the CC + GL group. These results suggest that GL inhibits the inflammatory response by binding HMGB1, thereby inhibiting DNA damage and cancer stem cell proliferation and dedifferentiation. In conclusion, GL significantly attenuates the pathogenesis of AOM/DSS-induced colorectal cancer by inhibiting HMGB1-TLR4-NF-κB signaling.


Carcinogenesis/drug effects , Colorectal Neoplasms/drug therapy , Glycyrrhizic Acid/pharmacology , Inflammation/drug therapy , Animals , Azoxymethane/pharmacology , Colon/drug effects , Colon/metabolism , Colorectal Neoplasms/metabolism , Disease Models, Animal , Female , HMGB1 Protein/metabolism , Inflammation/metabolism , Interleukin-6/metabolism , Mice , Mice, Inbred ICR , NF-kappa B/metabolism , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/metabolism
7.
Int J Biol Macromol ; 167: 1349-1360, 2021 Jan 15.
Article En | MEDLINE | ID: mdl-33202274

Large intestine cancer is one of the most relevant chronic diseases taking place at present. Despite therapies have evolved very positively, this pathology is still under deep investigation. One of the recent approaches is the prevention by natural compounds such as pectin. In this paper, we have assessed the impact of citrus pectin and modified citrus pectin on colorectal cancer in rats (Rattus norvegicus F344) to which azoxymethane and DSS were supplied. The lowest intake of food and body weight were detected in animals fed with citrus pectin, together with an increase in the caecum weight, probably due to the viscosity, water retention capacity and bulking properties of pectin. The most striking feature was that, neither citrus pectin nor modified citrus pectin gave rise to a tumorigenesis prevention. Moreover, in both, more than 50% of rats with cancer died, probably ascribed to a severe dysbiosis state in the gut, as shown by the metabolism and metagenomics studies carried out. This was related to a decrease of pH in caecum lumen and increase in acetate and lactic acid levels together with the absence of propionic and butyric acids. A relevant increase in Proteobacteria (Enterobacteriaceae) were thought to be one of the reasons for enteric infection that could have provoked the death of rats and the lack of cancer prevention. However, a reduction of blood glucose and triacylglycerides level and an increase of Bifidobacterium and Lactobacillaceae were found in animals that intake pectin, as compared to universal and modified citrus pectin feeding.


Azoxymethane/toxicity , Carcinogenesis/drug effects , Colorectal Neoplasms/diet therapy , Gastrointestinal Microbiome/drug effects , Pectins/therapeutic use , Acetates/metabolism , Animals , Azoxymethane/pharmacology , Bifidobacterium/isolation & purification , Blood Glucose/drug effects , Body Weight/drug effects , Butyrates/metabolism , Carcinogenesis/metabolism , Carcinogenesis/pathology , Chromatography, High Pressure Liquid , Citrus/chemistry , Colorectal Neoplasms/chemically induced , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/mortality , Dextran Sulfate/pharmacology , Disease Models, Animal , Hydrogen-Ion Concentration , Lactic Acid/metabolism , Lactobacillaceae/isolation & purification , Male , Metagenomics , Pectins/analysis , Phylogeny , Propionates/metabolism , Proteobacteria/isolation & purification , Rats , Rats, Inbred F344 , Triglycerides/blood
8.
Mediators Inflamm ; 2020: 7141970, 2020.
Article En | MEDLINE | ID: mdl-33082711

The flavonoid apigenin is common to many plants. Although the responsible mechanisms have yet to be elucidated, apigenin demonstrates tumor suppression in vitro and in vivo. This study uses an azoxymethane (AOM)/dextran sodium sulfate- (DSS-) induced colon cancer mouse model to investigate apigenin's potential mechanism of action exerted through its effects upon gut microbiota. The size and quantity of tumors were reduced significantly in the apigenin treatment group. Using 16S rRNA high-throughput sequencing of fecal samples, the composition of gut microbiota was significantly affected by apigenin. Further experiments in which gut microbiota were reduced and feces were transplanted provided further evidence of apigenin-modulated gut microbiota exerting antitumor effects. Apigenin was unable to reduce the number or size of tumors when gut microbiota were depleted. Moreover, tumor inhibition effects were initiated following the transplant of feces from mice treated with apigenin. Our findings suggest that the effect of apigenin on the composition of gut microbiota can suppress tumors.


Apigenin/pharmacology , Apigenin/therapeutic use , Gastrointestinal Microbiome/drug effects , Animals , Azoxymethane/pharmacology , Carcinogenesis/drug effects , Carcinogenesis/metabolism , Dextran Sulfate/pharmacology , Disease Models, Animal , Female , Mice , Mice, Inbred BALB C , RNA, Ribosomal, 16S/genetics
9.
J Microbiol Biotechnol ; 30(11): 1640-1650, 2020 Nov 28.
Article En | MEDLINE | ID: mdl-32958727

Colorectal cancer (CRC) is the leading cause of common malignant neoplasm worldwide. Many studies have analyzed compositions of gut microbiota associated with various diseases such as inflammatory bowel diseases (IBD) and colon cancer. One of the most representative bacteria involved in CRC is enterotoxigenic Bacteroides fragilis (ETBF), a species belonging to phylum Bacteroidetes. We used ETBF colonized mice with azoxymethane (AOM)/dextran sulphate sodium (DSS) and zerumbone, a compound with anti-bacterial effect, to determine whether zerumbone could restore intestinal microbiota composition. Four experimental groups of mice were used: sham, ETBF colonized AOM/DSS group, ETBF colonized AOM/DSS group zerumbone 60 mg kg-1 (ETBF/AOM/ DSS + Z (60)), and only zerumbone (60 mg kg-1)-treated group. We performed reversible dye terminators-based analysis of 16S rRNA gene region V3-V4 for group comparison. Microbiota compositions of ETBF/AOM/DSS + Z (60) group and ETBF colonized AOM/DSS group not given zerumbone were significantly different. There were more Bacteroides in ETBF/AOM/DSS + Z (60) group than those in ETBF colonized AOM/DSS group, suggesting that B. fragilis could be a normal flora activated by zerumbone. In addition, based on linear discriminant analysis of effect size (LEfSe) analysis, microbial diversity decreased significantly in the ETBF colonized AOM/DSS group. However, after given zerumbone, the taxonomic relative abundance was increased. These findings suggest that zerumbone not only influenced the microbial diversity and richness, but also could be helpful for enhancing the balance of gut microbial composition. In this work, we demonstrate that zerumbone could restore the composition of intestinal microbiota.


Azoxymethane/pharmacology , Bacteroides fragilis/growth & development , Dextran Sulfate/pharmacology , Gastrointestinal Microbiome/drug effects , Sesquiterpenes/pharmacology , Animals , Anti-Bacterial Agents/pharmacology , Bacteria/classification , Bacteria/drug effects , Colon/microbiology , Colon/pathology , Colonic Neoplasms/microbiology , Colonic Neoplasms/pathology , Colorectal Neoplasms/microbiology , Colorectal Neoplasms/pathology , Disease Models, Animal , Gastrointestinal Microbiome/genetics , Mice , RNA, Ribosomal, 16S
10.
FASEB J ; 34(6): 7387-7403, 2020 06.
Article En | MEDLINE | ID: mdl-32283574

Colorectal cancer (CRC) is the third most common solid tumor worldwide and has shown resistance to several immunotherapies, particularly immune checkpoint blockade therapy, which is effective in many other types of cancer. Our previous studies indicated that the active fraction of Garcinia yunnanensis (YTE-17), had potent anticancer activities by regulating multiple signaling pathways. However, knowledge regarding the mechanism and effect of YTE-17 in the prevention of CRC is limited. This study tested the effects of YTE-17 on colon cancer development in vivo by using two murine models: the carcigenic azoxymethane/dextran sulfate sodium (AOM/DSS)-induced CRC model and a genetically induced model using ApcMin/+ mice. Here, the tumor load, tumor number, histology, and even some oncogenes were used to evaluate the effect of YTE-17. The intragastric administration of YTE-17 for 12 weeks significantly decreased CRC incidence, tumor number and size, immunity, and some tumor-associated macrophage (TAM) markers, including CD206, Arg-1, IL-10, and TGF-ß. Importantly, the macrophages depletion by clodronate (CEL) also played a role in reducing the tumor burden and inhibiting tumor development, which were not affected by YTE-17 in the ApcMin/+ mice. Moreover, the YTE-17 treatment attenuated CRC cell growth in a co-culture system in the presence of macrophages. Consistently, YTE-17 effectively reduced the tumor burden and macrophage infiltration and enhanced immunity in the AOM/DSS and ApcMin/+ colon tumor models. Altogether, we demonstrate that macrophages in the microenvironment may contribute to the development and progression of CRC cells and propose YTE-17 as a new potential drug option for the treatment of CRC.


Cell Polarity/drug effects , Colorectal Neoplasms/drug therapy , Garcinia/chemistry , Macrophages/drug effects , Plant Preparations/pharmacology , Animals , Antineoplastic Agents/pharmacology , Azoxymethane/pharmacology , Cell Line , Cell Line, Tumor , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Colon/drug effects , Colon/metabolism , Colon/pathology , Colonic Neoplasms/drug therapy , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Dextran Sulfate/pharmacology , Disease Progression , Gene Expression Regulation, Neoplastic/drug effects , Macrophage Activation/drug effects , Macrophages/metabolism , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Plant Preparations/chemistry , RAW 264.7 Cells , Signal Transduction/drug effects , Tumor Microenvironment/drug effects
11.
Carcinogenesis ; 41(7): 909-917, 2020 07 14.
Article En | MEDLINE | ID: mdl-32115637

Imbalance of the gut microbial community promotes inflammation and colorectal cancer (CRC). Previously, we demonstrated that freeze-dried Parabacteroides distasonis (Pd) suppressed obesity-driven colorectal tumorigenesis in mice. Here, we investigated if Pd could suppress the development of colon tumors in mice independent of obesity. Six-week-old male A/J mice were assigned to receive: (i) chow diet (CTR); (ii) chow with 0.04% wt/wt freeze-dried Pd (Pd-Early) or (iii) chow diet before switching to 0.04% Pd diet (Pd-Late). Mice remained on diet for 25 weeks with the switch for Pd-Late mice occurring after 19 weeks. All mice received 6 weekly injections of the colon carcinogen azoxymethane (AOM; 10 mg/kg I.P.) starting after 1 week on diet. Colon tumors were observed in 77, 55 and 40% in CTR, Pd-Early and Pd-Late mice, respectively (X2 = 0.047). Colonic expression of toll-like receptor 4, IL-4 and TNF-α was 40% (P < 0.01), 58% (P = 0.05) and 55% (P < 0.001) lower, respectively, in Pd-Early compared with CTR mice. Pd-Late mice displayed a 217% (P = 0.05) and 185% (P < 0.001) increase in colonic IL-10 and TGF-ß expression, respectively, compared with CTR mice and similar increases in protein abundances were detected (47-145%; P < 0.05). Pd-Early and Pd-Late mice both demonstrated increased colonic expression of the tight junction proteins Zonula occludens-1 (P < 0.001) and occludin (P < 0.001) at the transcript (2-3-fold; P < 0.01) and protein level (30-50%; P < 0.05) relative to CTR. Our results support a protective role for Pd in colonic tumorigenesis and maintenance of intestinal epithelial barrier in AOM-treated mice.


Azoxymethane/pharmacology , Bacteroidetes/genetics , Carcinogenesis/genetics , Colonic Neoplasms/microbiology , Animals , Bacteroidetes/metabolism , Colon/pathology , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Humans , Inflammation/genetics , Inflammation/microbiology , Inflammation/pathology , Interleukin-4/genetics , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Mice , Obesity/metabolism , Obesity/microbiology , Obesity/pathology , Signal Transduction/genetics , Toll-Like Receptor 4/genetics , Transforming Growth Factor beta/genetics
12.
Mol Ther ; 28(1): 89-99, 2020 01 08.
Article En | MEDLINE | ID: mdl-31607540

Tumor-associated immune-suppressive neutrophils are prevalent in various cancers, including colorectal cancer. However, mechanisms of immune-suppressive neutrophils are not well understood. We report that a key innate suppressor, IRAK-M (interleukin-1 receptor-associated kinase M), is critically involved in the establishment of immune-suppressive neutrophils. In contrast to the wild-type (WT) neutrophils exhibiting immune-suppressive signatures of CD11bhighPD-L1highCD80low, IRAK-M-deficient neutrophils are rewired with reduced levels of inhibitory molecules PD-L1 and CD11b, as well as enhanced expression of stimulatory molecules CD80 and CD40. The reprogramming of IRAK-M-deficient neutrophils is mediated by reduced activation of STAT1/3 and enhanced activation of STAT5. As a consequence, IRAK-M-deficient neutrophils demonstrate enhanced capability to promote, instead of suppress, the proliferation and activation of effector T cells both in vitro and in vivo. Functionally, we observed that the transfusion of IRAK-M-/- neutrophils can potently render an enhanced anti-tumor immune response in the murine inflammation-induced colorectal cancer model. Collectively, our study defines IRAK-M as an innate suppressor for neutrophil function and reveals IRAK-M as a promising target for rewiring neutrophils in anti-cancer immunotherapy.


Adoptive Transfer/methods , Colorectal Neoplasms/therapy , Immunity, Innate/genetics , Interleukin-1 Receptor-Associated Kinases/metabolism , Neutrophils/immunology , Animals , Azoxymethane/pharmacology , B7-1 Antigen/metabolism , B7-H1 Antigen/metabolism , CD11 Antigens/metabolism , CD40 Antigens/metabolism , Colitis/chemically induced , Colitis/complications , Colorectal Neoplasms/etiology , Disease Models, Animal , Inflammation/chemically induced , Inflammation/metabolism , Interleukin-1 Receptor-Associated Kinases/genetics , Lymphocyte Activation/genetics , Mice , Mice, Knockout , Neutrophils/metabolism , T-Lymphocytes/immunology , Treatment Outcome
13.
PLoS One ; 14(12): e0226907, 2019.
Article En | MEDLINE | ID: mdl-31860645

It has been suggested that manipulation of gut microbiota using antibiotics can inhibit colitis-associated colorectal cancer (CAC) in a mouse model. We investigated whether timing of gut microbial manipulation using antibiotics affects colon tumorigenesis in the azoxymethane (AOM)/dextran sodium sulfate (DSS)-induced CAC model. CAC was induced in C57BL/6 mice by injection of 12.5 mg/kg AOM followed by three rounds of 1.7% DSS exposure. There were six groups based on timing of antibiotic administration. Colonic inflammation, proliferation, and tumorigenesis were evaluated after animal sacrifice. High-throughput sequencing of the mice feces was performed to characterize changes in gut microbiota. Full-time antibiotic treatment significantly decreased the number and size of tumors, histological scores, and expression of pro-inflammatory cytokines compared to the AOM/DSS group without antibiotic treatment. The early and late antibiotic groups, antibiotic administration from the first and second rounds of DSS to the end of the study, showed significantly lower histological scores and tumor burden. In contrast, the pretreatment antibiotic group, antibiotic administration from 3 weeks prior to AOM to the first round of DSS, did not exhibit decreased tumorigenesis. Principal coordinate analysis showed similar gut microbial community structures among the full-time, early, and late antibiotic groups, whereas other groups showed distinct gut microbial profiles. There was a positive correlation between number of tumors and number of operational taxonomic units. Colonic tumorigenesis was attenuated by antibiotic administration, except for that only prior to DSS administration, suggesting that gut microbial changes should be maintained throughout the entire period of inflammation to suppress tumorigenesis.


Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Carcinogenesis/drug effects , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/microbiology , Gastrointestinal Microbiome/drug effects , Animals , Azoxymethane/pharmacology , Cell Transformation, Neoplastic/drug effects , Colitis/chemically induced , Colorectal Neoplasms/chemically induced , Cytokines/metabolism , Dextran Sulfate/pharmacology , Disease Models, Animal , Female , Gastrointestinal Microbiome/genetics , High-Throughput Nucleotide Sequencing , Inflammation/drug therapy , Inflammation/metabolism , Mice , Mice, Inbred C57BL , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, DNA , Tumor Burden/drug effects
14.
Food Funct ; 10(10): 6873-6881, 2019 Oct 16.
Article En | MEDLINE | ID: mdl-31584586

Accumulating evidences have reported that the gut microbiota plays an important role in the occurrence and progression of colorectal cancer. Commonly known as a kind of constituent macromolecules, non-digestible polysaccharides have always been fermented by the intestinal microbiota. Nostoc commune Vaucher (N. commune) has always been appreciated as a healthy food and supplementation worldwide as it is rich in nutrients. Particularly, polysaccharides account for 60% of the dry weight of N. commune. In this study, we examined the protective effects of the polysaccharides isolated from N. commune (NVPS) against colitis-associated colon tumorigenesis in mice treated with azoxymethane (AOM) and dextran sulfate sodium salt (DSS) and the impact of these polysaccharides on the intestinal microbiota. NVPS were administered to mice through an intragastric gavage for 14 weeks. Our results demonstrated that the treatment with NVPS significantly decreased the number and sizes of tumors and reduced the incidence of intestinal tumors. Using 16S rRNA gene sequencing and qPCR, we analyzed the bacterial composition of the fecal samples obtained from mice. The results demonstrated that the alterations in the abundance of Firmicutes and Bacteroidetes caused by the AOM/DSS treatment were significantly reversed in response to the NVPS treatment. Moreover, the short-chain fatty acid (SCFA)-producing genera, including butyric acid-producing genera (Butyricicoccus, Butyrivibrio and Butyricimonas) and acetic acid-producing genera (Lachnospiraceae UCG 001, Lachnospiraceae UCG 006, and Blautia), were drastically enriched following the NVPS intervention. These compositional alterations induced by the NVPS were associated with the suppressed colonic inflammation and carcinogenesis. In conclusion, our results demonstrated an appreciable capability of NVPS to restore the gut microbiota profile altered by AOM/DSS, indicating the potential of NVPS as a promising prebiotic candidate for the prevention and treatment of colorectal cancer.


Carcinogenesis/drug effects , Colitis/drug therapy , Colon/drug effects , Gastrointestinal Microbiome/drug effects , Nostoc commune/metabolism , Polysaccharides/isolation & purification , Polysaccharides/pharmacology , Animals , Azoxymethane/pharmacology , Bacteria/classification , Colon/pathology , Colonic Neoplasms/pathology , Dextran Sulfate/pharmacology , Fatty Acids, Volatile/metabolism , Feces/microbiology , Male , Mice , Mice, Inbred C57BL , RNA, Ribosomal, 16S
15.
Cancer Sci ; 110(6): 1921-1930, 2019 Jun.
Article En | MEDLINE | ID: mdl-30973663

To overcome the problem of overlooking colorectal tumors, a new and highly sensitive modality of colonoscopy is needed. Moreover, it is also important to establish a new modality to evaluate viable tumor volume in primary lesions of colorectal cancer (CRC) during chemotherapy. Therefore, we carried out molecular imaging of colorectal tumors targeting epidermal growth factor receptor (EGFR), which is highly expressed on tumor cells, for evaluating chemotherapeutic efficacy and for endoscopic detection of colorectal adenomas. We first attempted to image five CRC cell lines with various levels of EGFR expression using an Alexa Fluor-labeled anti-EGFR monoclonal antibody (AF-EGFR-Ab). A strong fluorescence signal was observed in the cells depending on the level of EGFR expression. When nude mice xenografted with LIM1215 CRC cells, which highly express EGFR, were i.v. injected with AF-EGFR-Ab, a strong fluorescence signal appeared in the tumor with a high signal to noise ratio, peaking at 48 hours after injection and then gradually decreasing, as shown using an IVIS Spectrum system. When the xenografted mice were treated with 5-fluorouracil, fluorescence intensity in the tumor decreased in proportion to the viable tumor cell volume. Moreover, when the colorectum of azoxymethane-treated rats was observed using a thin fluorescent endoscope with AF-EGFR-Ab, all 10 small colorectal adenomas (≤3 mm) were detected with a clear fluorescence signal. These preliminary results of animal experiments suggest that EGFR-targeted fluorescent molecular imaging may be useful for quantitatively evaluating cell viability in CRC during chemotherapy, and also for detecting small adenomas using a fluorescent endoscope.


Antineoplastic Agents/pharmacology , Colorectal Neoplasms/drug therapy , ErbB Receptors/antagonists & inhibitors , Molecular Targeted Therapy/methods , Xenograft Model Antitumor Assays , Animals , Azoxymethane/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Colorectal Neoplasms/diagnostic imaging , Colorectal Neoplasms/metabolism , ErbB Receptors/metabolism , Female , Fluorouracil/pharmacology , HT29 Cells , Humans , Male , Mice, Inbred BALB C , Mice, Nude , Rats, Inbred F344
16.
Cell Death Dis ; 10(4): 304, 2019 04 03.
Article En | MEDLINE | ID: mdl-30944312

Colitis-associated cancer (CAC), a prototype of inflammation-associated cancer, is one of the most common gastrointestinal tumors. As a potential cancer testis antigen (CT antigen), cancer testis antigen 55 (CT55) is expressed in different tumors and normal testes. However, its role in CAC remains unknown. Here, we identified CT55 as a new potent promoter of CAC. We discovered that Ct55 deficiency alleviated inflammatory responses, decreased cell proliferation and colitis-associated tumorigenesis in an azoxymethane/dextran sulfate sodium (AOM/DSS) mouse model. Mechanistically, CT55 acts as an accelerator of tumor necrosis factor (TNF)-α-induced nuclear factor-κB (NF-κB) signaling. Upon stimulation with TNF-α, CT55 interacts with the IκB kinase (IKK) complex, which increases the phosphorylation of IKKα/ß and activates IKK-p65 signaling, while knockout of CT55 blocks IKK-p65 signaling. Notably, inhibition of IKK abolished the positive effect of CT55 on NF-κB activation. Collectively, our findings strongly indicate that CT55 deficiency suppresses the development of CAC and that the CT55-TNF-α-induced NF-κB axis may represent a promising target for CAC therapy.


Antigens, Neoplasm/metabolism , Colitis/complications , Colorectal Neoplasms/metabolism , NF-kappa B/metabolism , Animals , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Azoxymethane/pharmacology , Carcinogenesis/genetics , Carcinogenesis/metabolism , Cell Proliferation/drug effects , Cell Proliferation/genetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/immunology , Colorectal Neoplasms/pathology , Dextran Sulfate/pharmacology , Disease Models, Animal , HCT116 Cells , HEK293 Cells , Humans , I-kappa B Kinase/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Nude , NF-kappa B/genetics , Phosphorylation/drug effects , Signal Transduction/drug effects , Signal Transduction/genetics , Tumor Necrosis Factor-alpha/metabolism
17.
Int J Cancer ; 144(12): 3086-3098, 2019 06 15.
Article En | MEDLINE | ID: mdl-30515752

Chronic inflammation of the colon (colitis) is a risk factor for colorectal cancer (CRC). Hormone-replacement therapy reduces CRC incidences, and the estrogen receptor beta (ERß/ESR2) has been implicated in this protection. Gut microbiota is altered in both colitis and CRC and may influence the severity of both. Here we test the hypothesis that intestinal ERß impacts the gut microbiota. Mice with and without intestine-specific deletion of ERß (ERßKOVil ) were generated using the Cre-LoxP system. Colitis and CRC were induced with a single intraperitoneal injection of azoxymethane (AOM) followed by administration of three cycles of dextran sulfate sodium (DSS) in drinking water. The microbiota population were characterized by high-throughput 16S rRNA gene sequencing of DNA extracted from fecal samples (N = 39). Differences in the microbiota due to AOM/DSS and absence of ERß were identified through bioinformatic analyses of the 16S-Seq data, and the distribution of bacterial species was corroborated using qPCR. We demonstrate that colitis-induced CRC reduced the gut microbiota diversity and that loss of ERß enhanced this process. Further, the Bacteroidetes genus Prevotellaceae_UCG_001 was overrepresented in AOM/DSS mice compared to untreated controls (3.5-fold, p = 0.004), and this was enhanced in females and in ERßKOVil mice. Overall, AOM/DSS enriched for microbiota impacting immune system diseases and metabolic functions, and lack of ERß in combination with AOM/DSS enriched for microbiota impacting carbohydrate metabolism and cell motility, while reducing those impacting the endocrine system. Our data support that intestinal ERß contributes to a more favorable microbiome that could attenuate CRC development.


Colitis/metabolism , Colitis/microbiology , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/microbiology , Estrogen Receptor beta/metabolism , Gastrointestinal Microbiome/physiology , Animals , Azoxymethane/pharmacology , Dextran Sulfate/pharmacology , Estrogen Receptor beta/deficiency , Female , Gastrointestinal Microbiome/drug effects , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
18.
Asian Pac J Cancer Prev ; 19(10): 2839-2844, 2018 Oct 26.
Article En | MEDLINE | ID: mdl-30362310

Background: The intestinal mucosa is commonly exposed to oxidant nutrients and carcinogens, which can lead to the generation of free radicals. The antioxidants present in the diet assume great importance as possible protective agents, reducing the oxidative damage. In this way, we evaluated the antioxidant action of grape juice on preneoplastic lesions induced by azoxymethane (AOM) in Wistar rats. Methods: The colorectal carcinogenesis was induced by two intraperitoneal injections of 15mg/kg of AOM in Wistar rats. The animals were divided in 7 groups and treated with 1 and 2% concentrations of grape juice before and after carcinogen administration. After euthanasia, the expression of antioxidant enzymes catalase (CAT), copper-zinc superoxide dismutase (Cu/Zn-SOD) and manganese superoxide dismutase (Mn-SOD) CAT, SOD1 and SOD2 were evaluated by immunohistochemistry. Results: AOM decreased the expression of CAT and Mn-SOD enzymes, but not for Cu/Zn-SOD. We observed an increase expression of CAT and Mn-SOD after grape juice administration in some concentrations according to the time of administration of the grape juice before the carcinogen or just after the carcinogen. Conclusion: Our results suggest an independent action of each enzyme and a possible antioxidant action of the grape juice components in the diet being able to balance the body to neutralize the superoxide radicals and not leave them in the cell-damaging form.


Carcinogenesis/drug effects , Catalase/metabolism , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/prevention & control , Superoxide Dismutase/metabolism , Vitis/chemistry , Animals , Antioxidants/pharmacology , Azoxymethane/pharmacology , Carcinogenesis/chemically induced , Carcinogens/pharmacology , Colorectal Neoplasms/chemically induced , Fruit and Vegetable Juices , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Male , Oxidants/metabolism , Oxidation-Reduction/drug effects , Oxidative Stress/drug effects , Rats , Rats, Wistar
19.
Gene ; 677: 299-307, 2018 Nov 30.
Article En | MEDLINE | ID: mdl-30121380

Chronic intestinal inflammation predisposes patients with Inflammatory Bowel Disease (IBD) to Colitis-Associated Cancer (CAC). In the setting of chronic inflammation, microsatellite instability (MSI) results from early loss of DNA damage response (DDR) genes, ultimately leading to tumor formation. Despite continued efforts to improve early detection of high risk, pre-dysplastic regions in IBD patients, current macroscopic and genetic surveillance modalities remain limited. Therefore, understanding the regulation of key DDR genes in the progression from colitis to cancer may improve molecular surveillance of CAC. To evaluate DDR gene regulation in the transition from colitis to tumorigenesis, we utilized the well-established Azoxymethane/Dextran Sodium Sulfate (AOM/DSS) pre-clinical murine model of CAC in C57BL/6 mice. In order to assess colonic tumor burden in the setting of mutagen and intestinal irritation, tumors were visualized and graded in real time through high-resolution murine colonoscopy. Upon sacrifice, colons were opened and assessed for macroscopic tumor via high magnification surgical lenses (HMSL). Tissues were then sectioned and separated into groups based on the presence or absence of macroscopically visible tumor. Critical DDR genes were evaluated by semi-quantitative RT-PCR. Interestingly, colon tissue with macroscopically visible tumor (MVT) and colon tissue prior to observable tumor (the non-macroscopically visible tumor-developing group, NMVT) were identical in reduced mRNA expression of mlh1, anapc1, and ercc4 relative to colitic mice without mutagen, or those receiving mutagen alone. Colitis alone was sufficient to reduce colonic ercc4 expression when compared to NMVT mice. Therefore, reduced ercc4 expression may mark the early transition to CAC in a pre-clinical model, with expression reduced prior to the onset of observable tumor. Moreover, the expression of select DDR genes inversely correlated with chronicity of inflammatory disease. These data suggest ercc4 expression may define early stages in the progression to CAC.


Carcinogenesis/genetics , Colitis/genetics , Colitis/pathology , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , DNA Damage/genetics , Animals , Azoxymethane/pharmacology , Carcinogenesis/pathology , Colon/drug effects , Colon/pathology , DNA Damage/drug effects , Dextran Sulfate/pharmacology , Disease Models, Animal , Disease Progression , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Inflammation/genetics , Inflammation/pathology , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/pathology , Intestines/drug effects , Intestines/pathology , Male , Mice , Mice, Inbred C57BL , Microsatellite Instability/drug effects , Signal Transduction/drug effects , Signal Transduction/genetics
20.
Anticancer Res ; 38(8): 4485-4491, 2018 Aug.
Article En | MEDLINE | ID: mdl-30061214

BACKGROUND/AIM: The expression of cannabinoid receptor-1 (CB1-R) seems to be modulated by bioactive natural components such as the flavonoid quercetin. The aim of this study was to determine in an animal model of induced-colon cancer, whether quercetin inhibits colon carcinogenesis through changes in the expression of CB1-R. MATERIALS AND METHODS: C57BL/6J male mice were randomly assigned to standard diet or experimental diet supplemented with 0.5% quercetin. Azoxymethane (AOM) (10 mg/kg body weight) or saline solution (PBS) was intraperitoneally injected, once weekly for 6 weeks. RESULTS: The diet supplemented with quercetin induced CB1-R gene expression and protein, inhibiting the protein levels of STAT3 and p-STAT3 (both mediators of cell proliferation). Dietary quercetin also caused a significant increase in Bax/Bcl2 ratio protein expression. CONCLUSION: The anti-proliferative and pro-apoptotic effects of quercetin in AOM-treated mice are mediated by induction of the protein and gene expression levels of CB1-R.


Azoxymethane/pharmacology , Quercetin/pharmacology , Receptor, Cannabinoid, CB1/metabolism , Up-Regulation/drug effects , Animals , Apoptosis/drug effects , Carcinogenesis/drug effects , Cell Proliferation/drug effects , Colonic Neoplasms/drug therapy , Colonic Neoplasms/metabolism , Diet , Dietary Supplements , Flavonoids/pharmacology , Gene Expression/drug effects , Male , Mice , Mice, Inbred C57BL , Proto-Oncogene Proteins c-bcl-2/metabolism , bcl-2-Associated X Protein/metabolism
...