Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
J Cancer Res Clin Oncol ; 150(5): 243, 2024 May 08.
Article in English | MEDLINE | ID: mdl-38717677

ABSTRACT

Colitis-associated colorectal cancer has been a hot topic in public health issues worldwide. Numerous studies have demonstrated the significance of myeloid-derived suppressor cells (MDSCs) in the progression of this ailment, but the specific mechanism of their role in the transformation of inflammation to cancer is unclear, and potential therapies targeting MDSC are also unclear. This paper outlines the possible involvement of MDSC to the development of colitis-associated colorectal cancer. It also explores the immune and other relevant roles played by MDSC, and collates relevant targeted therapies against MDSC. In addition, current targeted therapies for colorectal cancer are analyzed and summarized.


Subject(s)
Colitis-Associated Neoplasms , Colorectal Neoplasms , Myeloid-Derived Suppressor Cells , Humans , Myeloid-Derived Suppressor Cells/immunology , Colitis-Associated Neoplasms/pathology , Colitis-Associated Neoplasms/etiology , Colitis-Associated Neoplasms/immunology , Colorectal Neoplasms/immunology , Colorectal Neoplasms/etiology , Colorectal Neoplasms/pathology , Colorectal Neoplasms/therapy , Animals , Colitis/complications , Colitis/immunology
2.
J Gastroenterol Hepatol ; 39(5): 893-901, 2024 May.
Article in English | MEDLINE | ID: mdl-38273469

ABSTRACT

BACKGROUND AND AIM: Colitis-associated intestinal cancer (CAC) can develop in patients with inflammatory bowel disease; however, the malignant grade of CAC may differ from that of sporadic colorectal cancer (CRC). Therefore, we compared histological findings distinct from cancer stage between CAC and sporadic CRC to evaluate the features of CAC. METHODS: We reviewed the clinical and histological data collected from a nationwide database in Japan between 1983 and 2020. Patient characteristics were compared to distinguish ulcerative colitis (UC), Crohn's disease (CD), and sporadic CRC. Comparisons were performed by using all collected data and propensity score-matched data. RESULTS: A total of 1077 patients with UC-CAC, 297 with CD-CAC, and 136 927 with sporadic CRC were included. Although the prevalence of well or moderately differentiated adenocarcinoma (Tub1 and Tub2) decreased according to tumor progression for all diseases (P < 0.01), the prevalence of other histological findings, including signet ring cell carcinoma, mucinous carcinoma, poorly differentiated adenocarcinoma, or squamous cell carcinoma, was significantly higher in CAC than in sporadic CRC. Based on propensity score-matched data for 982 patients with UC and 268 with CD, the prevalence of histological findings other than Tub1 and Tub2 was also significantly higher in those with CAC. At pT4, mucinous carcinoma occurred at a significantly higher rate in patients with CD (45/86 [52.3%]) than in those with sporadic CRC (13/88 [14.8%]) (P < 0.01). CONCLUSION: CAC, including early-stage CAC, has a higher malignant grade than sporadic CRC, and this difference increases in significance with tumor progression.


Subject(s)
Colitis, Ulcerative , Propensity Score , Humans , Male , Female , Middle Aged , Colitis, Ulcerative/pathology , Colitis, Ulcerative/complications , Colitis, Ulcerative/epidemiology , Aged , Japan/epidemiology , Crohn Disease/pathology , Crohn Disease/epidemiology , Crohn Disease/complications , Colitis-Associated Neoplasms/pathology , Colitis-Associated Neoplasms/etiology , Colitis-Associated Neoplasms/epidemiology , Colorectal Neoplasms/pathology , Colorectal Neoplasms/epidemiology , Colorectal Neoplasms/etiology , Adult , Adenocarcinoma/pathology , Adenocarcinoma/epidemiology , Adenocarcinoma/etiology , Neoplasm Staging , Neoplasm Grading , Adenocarcinoma, Mucinous/pathology , Adenocarcinoma, Mucinous/epidemiology , Adenocarcinoma, Mucinous/etiology , Carcinoma, Signet Ring Cell/pathology , Carcinoma, Signet Ring Cell/epidemiology , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/epidemiology , Carcinoma, Squamous Cell/etiology , Diagnosis, Differential , Prevalence
3.
Front Immunol ; 14: 1103617, 2023.
Article in English | MEDLINE | ID: mdl-37006260

ABSTRACT

Colitis-associated colorectal cancer is the most serious complication of ulcerative colitis. Long-term chronic inflammation increases the incidence of CAC in UC patients. Compared with sporadic colorectal cancer, CAC means multiple lesions, worse pathological type and worse prognosis. Macrophage is a kind of innate immune cell, which play an important role both in inflammatory response and tumor immunity. Macrophages are polarized into two phenotypes under different conditions: M1 and M2. In UC, enhanced macrophage infiltration produces a large number of inflammatory cytokines, which promote tumorigenesis of UC. M1 polarization has an anti-tumor effect after CAC formation, whereas M2 polarization promotes tumor growth. M2 polarization plays a tumor-promoting role. Some drugs have been shown to that prevent and treat CAC effectively by targeting macrophages.


Subject(s)
Colitis, Ulcerative , Colitis-Associated Neoplasms , Humans , Colitis-Associated Neoplasms/etiology , Macrophages , Inflammation , Cytokines
4.
J Immunol Res ; 2023: 7040113, 2023.
Article in English | MEDLINE | ID: mdl-36741232

ABSTRACT

Ulcerative colitis (UC) is a complex intestinal inflammation with an increasing risk of colitis-associated colorectal cancer (CAC). However, the pathogenesis is still unclear between active UC and inactive UC. Recently, it has been reported that pyroptosis-related genes (PRGs) are closely associated with inflammatory disease activity. Nevertheless, the specific roles of PRGs in the progression and treatment of UC and CAC remain unclear. In this study, we identified 30 differentially expressed PRGs based on the immune landscape of active and inactive UC samples. Meanwhile, weighted gene coexpression network analysis was applied to explore important genes associated with active UC. By intersecting with the differentially expressed PRGs, CASP5, GBP1, GZMB, IL1B, and IRF1 were selected as key PRGs to construct a pyroptosis-related signature (PR-signature). Then, logistic regression analysis was performed to validate the PR-signature and establish a pyroptosis-related score (PR-Score). We demonstrated that PR-Score had a powerful ability to distinguish active UC from inactive UC in multiple datasets. Besides, PR-Score was positively correlated with immune cell infiltration and inflammatory microenvironment in UC. Lower PR-Score was associated with a better response to anti-TNF therapy for patients with UC. Additionally, high-PR-Score was found to suppress CAC and improve the survival outcomes of patients with colorectal cancer. Finally, the levels of the PR-signature genes were validated both in vitro and in vivo. These findings can improve our understanding of PRGs in UC and provide new markers for predicting the occurrence of active UC or CAC and the treatment of UC.


Subject(s)
Colitis, Ulcerative , Colitis-Associated Neoplasms , Humans , Pyroptosis , Colitis-Associated Neoplasms/etiology , Colitis-Associated Neoplasms/genetics , Colitis, Ulcerative/drug therapy , Colitis, Ulcerative/genetics , Tumor Necrosis Factor Inhibitors , Caspases , Tumor Microenvironment/genetics
5.
Inflamm Bowel Dis ; 29(8): 1285-1296, 2023 08 01.
Article in English | MEDLINE | ID: mdl-36745026

ABSTRACT

BACKGROUND: Myeloid cells are critical for iron and immune homeostasis. Ferritin heavy chain (FTH1) is essential for intracellular iron storage. Myeloid FTH1 is important in the pathogenesis of many inflammatory diseases. However, the role of myeloid FTH1 in colitis and colitis-associated cancer has not been determined. METHODS: Myeloid FTH1 deficient and wild-type mice were treated with dextran sodium sulfate (DSS) or azoxymethane (AOM)-DSS to compare their susceptibility to acute colitis or colitis-associated cancer. RESULTS: Myeloid FTH1-deficient mice fed with a high-iron diet were less susceptible to DSS-induced acute colitis than wild type mice. Mechanistic studies showed that myeloid FTH1 deficiency resulted in lower expression of an iron uptake protein divalent metal transporter 1 (DMT1) and active phosphorylated signal transducer and activator of transcription 3 (STAT3) in the colon tissues. Our studies also showed that pharmacological STAT3 reactivation restored the susceptibility of myeloid FTH1-deficient mice to DSS-induced acute colitis. Consistently, myeloid FTH1-deficient mice fed with a high-iron diet had reduced DMT1, phosphorylated STAT3 and inflammation in their colon tissues, and were less susceptible to colitis-associated colorectal cancer. CONCLUSIONS: Our study demonstrated that myeloid FTH1 is required for colitis and colitis-associated colorectal cancer via maintaining of DMT1-iron-STAT3 signaling activation under excess iron condition.


Subject(s)
Colitis-Associated Neoplasms , Colitis , Colorectal Neoplasms , Animals , Mice , Colitis-Associated Neoplasms/etiology , Colitis-Associated Neoplasms/prevention & control , STAT3 Transcription Factor/metabolism , Iron/metabolism , Colitis/chemically induced , Colitis/complications , Colitis/metabolism , Azoxymethane/toxicity , Dextran Sulfate/toxicity , Mice, Inbred C57BL , Disease Models, Animal , Colorectal Neoplasms/chemically induced , Colorectal Neoplasms/prevention & control
7.
Oncogene ; 40(47): 6540-6546, 2021 11.
Article in English | MEDLINE | ID: mdl-34625710

ABSTRACT

CCL11, also known as eotaxin-1, is described as an eosinophil chemoattractant, which has been implicated in allergic and Th2 inflammatory diseases. We have reported that CCL11 is significantly increased in the serum of inflammatory bowel disease (IBD) patients, colonic eosinophils are increased and correlate with tissue CCL11 levels in ulcerative colitis patients, and CCL11 is increased in dextran sulfate sodium (DSS)-induced murine colitis. Here, we show that CCL11 is involved in the pathogenesis of DSS-induced colitis and in colon tumorigenesis in the azoxymethane (AOM)-DSS model of colitis-associated carcinogenesis (CAC). Ccl11-/- mice exposed to DSS then allowed to recover had significantly less body weight loss and a decrease in histologic injury versus wild-type (WT) mice. In the AOM-DSS model, Ccl11-/- mice exhibited decreased colonic tumor number and burden, histologic injury, and colonic eosinophil infiltration versus WT mice. Ccl11 is expressed by both colonic epithelial and lamina propria immune cells. Studies in bone marrow chimera mice revealed that hematopoietic- and epithelial-cell-derived CCL11 were both important for tumorigenesis in the AOM-DSS model. These findings indicate that CCL11 is important in the regulation of colitis and associated carcinogenesis and thus anti-CCL11 antibodies may be useful for treatment and cancer chemoprevention in IBD.


Subject(s)
Carcinogenesis/pathology , Chemokine CCL11/physiology , Colitis-Associated Neoplasms/pathology , Colitis/complications , Epithelial Cells/pathology , Animals , Azoxymethane/toxicity , Carcinogenesis/metabolism , Carcinogens/toxicity , Colitis/chemically induced , Colitis-Associated Neoplasms/etiology , Colitis-Associated Neoplasms/metabolism , Epithelial Cells/metabolism , Mice , Mice, Knockout
8.
Cells ; 10(8)2021 08 11.
Article in English | MEDLINE | ID: mdl-34440829

ABSTRACT

Many researchers have argued that Western diet (WD)-induced obesity accelerates inflammation and that inflammation is a link between obesity and colorectal cancer (CRC). This study investigated the effect of WDs on the development and progression of colitis-associated colon cancer (CAC) and the efficacy of the anti-obesity agent orlistat on WD-driven CAC in mice. The results revealed that the WD exacerbated CAC in azoxymethane (AOM)/dextran sulfate sodium (DSS)-induced mice, which showed increased mortality, tumor formation, and aggravation of tumor progression. Furthermore, WD feeding also upregulated inflammation, hyperplasia, and tumorigenicity levels through the activation of STAT3 and NF-κB signaling in an AOM/DSS-induced mouse model. In contrast, treatment with orlistat increased the survival rate and alleviated the symptoms of CAC, including a recovery in colon length and tumor production decreases in WD-driven AOM/DSS-induced mice. Additionally, orlistat inhibited the extent of inflammation, hyperplasia, and tumor progression via the inhibition of STAT3 and NF-κB activation. Treatment with orlistat also suppressed the ß-catenin, slug, XIAP, Cdk4, cyclin D, and Bcl-2 protein levels in WD-driven AOM/DSS-induced mice. The results of this study indicate that orlistat alleviates colon cancer promotion in WD-driven CAC mice by suppressing inflammation, especially by inhibiting STAT3 and NF-κB activation.


Subject(s)
Anti-Obesity Agents/therapeutic use , Colitis-Associated Neoplasms/drug therapy , Diet, Western/adverse effects , NF-kappa B/metabolism , Orlistat/therapeutic use , STAT3 Transcription Factor/metabolism , Animals , Antineoplastic Agents/therapeutic use , Azoxymethane/toxicity , Colitis-Associated Neoplasms/etiology , Colitis-Associated Neoplasms/metabolism , Colitis-Associated Neoplasms/pathology , Dextran Sulfate/toxicity , Inflammation , Mice , Signal Transduction/drug effects , Transcription Factor RelA/metabolism
9.
Cell Mol Gastroenterol Hepatol ; 12(4): 1329-1341, 2021.
Article in English | MEDLINE | ID: mdl-34217896

ABSTRACT

BACKGROUND & AIMS: Identifying new approaches to lessen inflammation, as well as the associated malignant consequences, remains crucial to improving the lives and prognosis of patients diagnosed with inflammatory bowel diseases. Although it previously has been suggested as a suitable biomarker for monitoring disease activity in patients diagnosed with Crohn's disease, the role of the acute-phase protein serum amyloid A (SAA) in inflammatory bowel disease remains unclear. In this study, we aimed to assess the role of SAA in colitis-associated cancer. METHODS: We established a model of colitis-associated cancer in wild-type and SAA double-knockout (Saa1/2-/-) mice by following the azoxymethane/dextran sulfate sodium protocol. Disease activity was monitored throughout the study while colon and tumor tissues were harvested for subsequent use in cytokine analyses, Western blot, and immunohistochemistry +experiments. RESULTS: We observed attenuated disease activity in mice deficient for Saa1/2 as evidenced by decreased weight loss, increased stool consistency, decreased rectal bleeding, and decreased colitis-associated tissue damage. Macrophage infiltration, including CD206+ M2-like macrophages, also was attenuated in SAA knockout mice, while levels of interleukin 4, interleukin 10, and tumor necrosis factor-ɑ were decreased in the distal colon. Mice deficient for SAA also showed a decreased tumor burden, and tumors were found to have increased apoptotic activity coupled with decreased expression for markers of proliferation. CONCLUSION: Based on these findings, we conclude that SAA has an active role in inflammatory bowel disease and that it could serve as a therapeutic target aimed at decreasing chronic inflammation and the associated risk of developing colitis-associated cancer.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Colitis-Associated Neoplasms/etiology , Colitis-Associated Neoplasms/metabolism , Disease Susceptibility , Serum Amyloid A Protein/metabolism , Animals , Biomarkers , Cell Transformation, Neoplastic/genetics , Colitis-Associated Neoplasms/pathology , Disease Models, Animal , Immunohistochemistry , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Macrophages/metabolism , Macrophages/pathology , Mice , Mice, Knockout , Protein Isoforms , Serum Amyloid A Protein/genetics
10.
Int J Mol Sci ; 22(14)2021 Jul 19.
Article in English | MEDLINE | ID: mdl-34299314

ABSTRACT

Signal transducer and activator of transcription 1 (STAT1) acts as a tumor suppressor molecule in colitis-associated colorectal cancer (CAC), particularly during the very early stages, modulating immune responses and controlling mechanisms such as apoptosis and cell proliferation. Previously, using an experimental model of CAC, we reported increased intestinal cell proliferation and faster tumor development, which were consistent with more signs of disease and damage, and reduced survival in STAT1-/- mice, compared with WT counterparts. However, the mechanisms through which STAT1 might prevent colorectal cancer progression preceded by chronic inflammation are still unclear. Here, we demonstrate that increased tumorigenicity related to STAT1 deficiency could be suppressed by IL-17 neutralization. The blockade of IL-17 in STAT1-/- mice reduced the accumulation of CD11b+Ly6ClowLy6G+ cells resembling granulocytic myeloid-derived suppressor cells (MDSCs) in both spleen and circulation. Additionally, IL-17 blockade reduced the recruitment of neutrophils into intestinal tissue, the expression and production of inflammatory cytokines, and the expression of intestinal STAT3. In addition, the anti-IL-17 treatment also reduced the expression of Arginase-1 and inducible nitric oxide synthase (iNOS) in the colon, both associated with the main suppressive activity of MDSCs. Thus, a lack of STAT1 signaling induces a significant change in the colonic microenvironment that supports inflammation and tumor formation. Anti-IL-17 treatment throughout the initial stages of CAC related to STAT1 deficiency abrogates the tumor formation possibly caused by myeloid cells.


Subject(s)
Colitis-Associated Neoplasms/etiology , Granulocytes/pathology , Interleukin-17/physiology , STAT1 Transcription Factor/physiology , Animals , Antibodies, Neutralizing/administration & dosage , Colitis-Associated Neoplasms/pathology , Colitis-Associated Neoplasms/physiopathology , Disease Progression , Female , Granulocytes/immunology , Interleukin-17/antagonists & inhibitors , Interleukin-17/immunology , Mice , Mice, Inbred BALB C , Mice, Knockout , Myeloid-Derived Suppressor Cells/immunology , Myeloid-Derived Suppressor Cells/pathology , Neoplasms, Experimental/etiology , Neoplasms, Experimental/pathology , Neoplasms, Experimental/physiopathology , STAT1 Transcription Factor/deficiency , STAT1 Transcription Factor/genetics , Tumor Microenvironment/immunology
11.
Neoplasia ; 23(6): 561-573, 2021 06.
Article in English | MEDLINE | ID: mdl-34077834

ABSTRACT

Previously, we found that rosmarinic acid (RA) exerted anti-inflammatory activities in a dextran sulfate sodium (DSS)-induced colitis model. Here, we investigated the anti-tumor effects of RA on colitis-associated colon cancer (CAC) and the underlying molecular mechanisms. We established an azoxymethane (AOM)/DSS-induced CAC murine model for in vivo studies and used a conditioned media (CM) culture system in vitro. H&E staining, immunohistochemistry, western blot assay, enzyme-linked immunosorbent assay, molecular docking, co-immunoprecipitation, and immunofluorescence assay were utilized to investigate how RA prevented colorectal cancer. In the AOM/DSS-induced CAC murine model, RA significantly reduced colitis severity, inflammation-related protein expression, tumor incidence, and colorectal adenoma development. It significantly modulated toll-like receptor-4 (TLR4)-mediated nuclear factor-kappa B (NF-κB) and signal transducer and activator of transcription 3 (STAT3) activation, thus attenuating the expression of anti-apoptotic factors, which mediate transcription factor-dependent tumor growth. In vitro, RA inhibited CM-induced TLR4 overexpression and competitively inhibited TLR4-myeloid differentiation factor 2 complex in an inflammatory microenvironment. Thus, RA suppressed NF-κB and STAT3 activation in colon cancer cells in an inflammatory microenvironment. Therefore, RA suppressed colitis-associated tumorigenesis in the AOM/DSS-induced CAC murine model and abrogated human colon cancer progression in an inflammatory microenvironment by propitiating TLR4-mediated NF-κB and STAT3 activation, pleiotropically.


Subject(s)
Cinnamates/pharmacology , Colitis-Associated Neoplasms/etiology , Colitis-Associated Neoplasms/metabolism , Depsides/pharmacology , NF-kappa B/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Toll-Like Receptor 4/metabolism , Animals , Biomarkers , Cell Line, Tumor , Cinnamates/chemistry , Colitis-Associated Neoplasms/drug therapy , Colitis-Associated Neoplasms/pathology , Colonic Neoplasms/drug therapy , Colonic Neoplasms/etiology , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Depsides/chemistry , Disease Models, Animal , Humans , Immunohistochemistry , Male , Mice , Models, Molecular , Structure-Activity Relationship , Xenograft Model Antitumor Assays , Rosmarinic Acid
12.
PLoS One ; 16(1): e0245292, 2021.
Article in English | MEDLINE | ID: mdl-33503019

ABSTRACT

Early diagnosis of colorectal cancer is needed to reduce the mortal consequence by cancer. Lipid mediators play critical role in progression of colitis and colitis-associated colon cancer (CAC) and some of their metabolites are excreted in urine. Here, we attempted to find novel biomarkers in urinary lipid metabolite of a murine model of CAC. Mice were received single administration of azoxymethane (AOM) and repeated administration of dextran sulfate sodium (DSS). Lipid metabolites in their urine was measured by liquid chromatography mass spectrometry and their colon was collected to perform morphological study. AOM and DSS caused inflammation and tumor formation in mouse colon. Liquid chromatography mass spectrometry-based comprehensive analysis of lipid metabolites showed that cyclooxygenase-mediated arachidonic acid (AA) metabolites, prostaglandins, and reactive oxygen species (ROS)-mediated AA metabolites, isoprostanes, were predominantly increased in the urine of tumor-bearing mice. Among that, urinary prostaglandin (PG)E2 metabolite tetranor-PGEM and PGD2 metabolite tetranor-PGDM were significantly increased in both of urine collected at the acute phase of colitis and the carcinogenesis phase. On the other hand, two F2 isoprostanes (F2-IsoPs), 8-iso PGF2α and 2,3-dinor-8-iso PGF2α, were significantly increased only in the carcinogenesis phase. Morphological study showed that infiltrated monocytes into tumor mass strongly expressed ROS generator NADPH (p22phox). These observations suggest that urinary 8-iso PGF2α and 2,3-dinor-8-iso PGF2α can be indexes of CAC.


Subject(s)
Colitis-Associated Neoplasms/pathology , Colitis/pathology , Dinoprost/analogs & derivatives , F2-Isoprostanes/urine , Animals , Biomarkers/urine , Chromatography, High Pressure Liquid , Colitis/chemically induced , Colitis/complications , Colitis-Associated Neoplasms/etiology , Colitis-Associated Neoplasms/urine , Cyclooxygenase 2/metabolism , Cytochrome b Group/metabolism , Dextran Sulfate/toxicity , Dinoprost/urine , Disease Models, Animal , Female , Lipid Metabolism , Mass Spectrometry , Mice , Mice, Inbred C57BL , Monocytes/immunology , NADPH Oxidases/metabolism , Reactive Oxygen Species/metabolism
13.
Adv Med Sci ; 66(1): 89-97, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33461100

ABSTRACT

PURPOSE: Ulcerative colitis (UC) carries a high risk of developing colorectal cancer (CRC). FK506-binding protein 51 (FKBP51) is a key regulator of glucocorticoid resistance and inflammatory tumor microenvironment. This study aimed to investigate the role of FKBP51 in UC-CRC prognosis. MATERIALS AND METHODS: The FKBP51 expression was measured by immunohistochemistry, qRT-PCR and western blot in control and tumor-containing tissues from UC-CRC patients. H&E staining was used to analyze the inflammatory status of each sample. The relationship between FKBP51 expression and UC-CRC prognosis was assessed by Kaplan-Meier curves and Mann-Whitney U test, and receiver-operating characteristic curves were generated to clarify the role of FKBP51 in predicting survival period and recurrence of UC-CRC patients. RESULTS: The FKBP51 expression was significantly (p â€‹< â€‹0.01) increased by 36.3% in tumor-containing tissues compared to control tissues in UC-CRC patients. Nuclear enrichment of FKBP51 in tumor-containing tissues was significantly (p â€‹< â€‹0.001) increased by 78.5%. The UC-CRC patients with higher levels of FKBP51 expression ratio between tumor-containing tissues and control tissues had shorter survival periods, but greater neutrophil invasion and neutrophils to lymphocytes ratio (NLR) in peripheral blood. Moreover, the FKBP51 expression ratio was more helpful in predicting the survival periods and recurrence in the UC-CRC patients than the NLR in peripheral blood. CONCLUSIONS: The FKBP51 expression ratio between tumor-containing tissue and control tissue may be an important biomarker of inflammatory tumor microenvironment and more helpful for the UC-CRC prognosis.


Subject(s)
Biomarkers, Tumor/analysis , Colitis, Ulcerative/complications , Colitis-Associated Neoplasms/pathology , Colorectal Neoplasms/pathology , Lymphocytes/pathology , Neutrophils/pathology , Tacrolimus Binding Proteins/metabolism , Adolescent , Adult , Case-Control Studies , Child , Colitis-Associated Neoplasms/etiology , Colitis-Associated Neoplasms/metabolism , Colorectal Neoplasms/etiology , Colorectal Neoplasms/metabolism , Female , Follow-Up Studies , Humans , Male , Prognosis , ROC Curve , Risk Factors , Survival Rate , Tacrolimus Binding Proteins/genetics , Tumor Microenvironment , Young Adult
14.
Cell Mol Gastroenterol Hepatol ; 11(4): 1177-1197, 2021.
Article in English | MEDLINE | ID: mdl-33418102

ABSTRACT

Inflammatory bowel disease (IBD) patients have an increased risk of developing colitis-associated colon cancer (CAC); however, the basis for inflammation-induced genetic damage requisite for neoplasia is unclear. Several studies have shown that IBD patients have signs of increased oxidative damage, which could be a result of genetic and environmental factors such as an excess in oxidant molecules released during chronic inflammation, mitochondrial dysfunction, a failure in antioxidant capacity, or oxidant promoting diets. It has been suggested that chronic oxidative environment in the intestine leads to the DNA lesions that precipitate colon carcinogenesis in IBD patients. Indeed, several preclinical and clinical studies show that different endogenous and exogenous antioxidant molecules are effective at reducing oxidation in the intestine. However, most clinical studies have focused on the short-term effects of antioxidants in IBD patients but not in CAC. This review article examines the role of oxidative DNA damage as a possible precipitating event in CAC in the context of chronic intestinal inflammation and the potential role of exogenous antioxidants to prevent these cancers.


Subject(s)
Antioxidants/pharmacology , Colitis-Associated Neoplasms/prevention & control , Colitis/complications , Animals , Colitis-Associated Neoplasms/etiology , Colitis-Associated Neoplasms/pathology , Humans
15.
Dig Dis Sci ; 66(5): 1488-1498, 2021 05.
Article in English | MEDLINE | ID: mdl-32445051

ABSTRACT

BACKGROUND: Heparanase (HPSE) is considered to play an important role in the occurrence, development and carcinogenesis of ulcerative colitis (UC). There are no reports about the detection of HPSE mRNA in feces to predict UC activity and cancerization risk. AIMS: To explore the feasibility and effectiveness of fecal epithelial HPSE mRNA in monitoring patients' UC activity and predicting cancer risk. METHODS: The clinical part of the study enrolled 20 patients with UC and 20 controls. Meanwhile, a UC-induced carcinogenesis mouse model was established using a combination treatment of dimethylhydrazine and dextran sulfate sodium. Tissue expression of HPSE protein was detected by immunohistochemistry. RT-qPCR was used to detect the expression of HPSE mRNA in colonic mucosa and feces. RESULTS: In the human study, the relative expressions of HPSE mRNA in colonic mucosa and feces were positively correlated with the Mayo score (P < 0.05), and with a significant correlation between feces and colonic mucosa (P < 0.05). In the mouse model, the relative expressions of HPSE mRNA in colonic mucosa and feces in the ulcerative colitis-associated colorectal cancer group was significantly higher than that of the UC group and the normal control group (P < 0.05), and with a significant correlation between feces and colonic mucosa (P < 0.05). CONCLUSIONS: The relative level of HPSE mRNA was positively correlated with UC activity and cancerization. The relative level of HPSE mRNA in feces was correlated with that in colonic mucosa. The detection of HPSE mRNA in feces can be used as a new marker for disease monitoring and cancer risk prediction of UC.


Subject(s)
Colitis, Ulcerative/genetics , Colitis-Associated Neoplasms/etiology , Feces/enzymology , Glucuronidase/genetics , Intestinal Mucosa/enzymology , RNA, Messenger/genetics , Animals , Case-Control Studies , Colitis, Ulcerative/complications , Colitis, Ulcerative/diagnosis , Colitis, Ulcerative/enzymology , Disease Models, Animal , Feasibility Studies , Genetic Markers , Humans , Immunohistochemistry , Male , Mice, Inbred C57BL , Polymerase Chain Reaction , Predictive Value of Tests , Risk Assessment , Risk Factors
16.
Mucosal Immunol ; 14(1): 187-198, 2021 01.
Article in English | MEDLINE | ID: mdl-32467604

ABSTRACT

Inflammation is a critical player in the development and progression of colon cancer. Basic leucine zipper transcription factor ATF-like 3 (BATF3) plays an important role in infection and tumor immunity through regulating the development of conventional type 1 dendritic cells (cDC1s). However, the function of BATF3 in colitis and colitis-associated colon cancer (CAC) remains unclear. Here, BATF3 wild-type and knockout mice were used to construct an AOM/DSS-induced CAC model. In addition, DSS-induced chronic colitis, bone marrow cross-transfusion (BMT), neutrophil knockout, and other animal models were used for in-depth research. We found that BATF3 deficiency in intestinal epithelial cells rather than in cDC1s inhibited CAC, which was depended on inflammatory stimulation. Mechanistically, BATF3 directly promoted transcription of CXCL5 by forming a heterodimer with JunD, and accelerated the recruitment of neutrophils through the CXCL5-CXCR2 axis, ultimately increasing the occurrence and development of CAC. Tissue microarray and TCGA data also indicated that high expression of BATF3 was positively correlated with poor prognosis of colorectal cancer and other inflammation-related tumors. In summary, our results demonstrate that intestinal epithelial-derived BATF3 relies on inflammatory stimulation to promote CAC, and BATF3 is expected to be a novel diagnostic indicator for colitis and CAC.


Subject(s)
Basic-Leucine Zipper Transcription Factors/metabolism , Chemokine CXCL5/metabolism , Colitis-Associated Neoplasms/etiology , Colitis-Associated Neoplasms/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Neutrophil Infiltration/immunology , Repressor Proteins/metabolism , Animals , Chemokine CXCL5/genetics , Colitis , Colitis-Associated Neoplasms/pathology , Colonic Neoplasms/etiology , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Dextran Sulfate/adverse effects , Disease Models, Animal , Intestinal Mucosa/pathology , Mice , Mice, Knockout , Neutrophils/immunology , Neutrophils/metabolism
17.
Cell Mol Immunol ; 18(2): 350-362, 2021 02.
Article in English | MEDLINE | ID: mdl-32094504

ABSTRACT

Loss of the colonic inner mucus layer leads to spontaneously severe colitis and colorectal cancer. However, key host factors that may control the generation of the inner mucus layer are rarely reported. Here, we identify a novel function of TRIM34 in goblet cells (GCs) in controlling inner mucus layer generation. Upon DSS treatment, TRIM34 deficiency led to a reduction in Muc2 secretion by GCs and subsequent defects in the inner mucus layer. This outcome rendered TRIM34-deficient mice more susceptible to DSS-induced colitis and colitis-associated colorectal cancer. Mechanistic experiments demonstrated that TRIM34 controlled TLR signaling-induced Nox/Duox-dependent ROS synthesis, thereby promoting the compound exocytosis of Muc2 by colonic GCs that were exposed to bacterial TLR ligands. Clinical analysis revealed that TRIM34 levels in patient samples were correlated with the outcome of ulcerative colitis (UC) and the prognosis of rectal adenocarcinoma. This study indicates that TRIM34 expression in GCs plays an essential role in generating the inner mucus layer and preventing excessive colon inflammation and tumorigenesis.


Subject(s)
Carrier Proteins/physiology , Colitis-Associated Neoplasms/prevention & control , Colitis/prevention & control , Colon/pathology , Goblet Cells/pathology , Mucus/physiology , Animals , Carcinogenesis , Carrier Proteins/genetics , Carrier Proteins/metabolism , Colitis/etiology , Colitis-Associated Neoplasms/etiology , Colitis-Associated Neoplasms/pathology , Colon/immunology , Colon/metabolism , Goblet Cells/immunology , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Mucin-2/metabolism
18.
Cell Mol Gastroenterol Hepatol ; 11(5): 1505-1518, 2021.
Article in English | MEDLINE | ID: mdl-33316454

ABSTRACT

BACKGROUND & AIMS: Inflammatory bowel disease is commonly treated by administration of glucocorticoids. While the importance of intestinal epithelial cells for the pathogenesis of this disorder is widely accepted, their role as target cells for glucocorticoids has not been explored. To address this issue, we induced colonic inflammation in GRvillin mice, which carry an inducible deletion of the glucocorticoid receptor in intestinal epithelial cells. METHODS: Colitis and colitis-associated colorectal cancer were induced by administration of dextran sulfate sodium and azoxymethane in mice. Clinical parameters, epithelial permeability and tumor development were monitored during disease progression. Colon tissue, lamina propria cells and intestinal epithelial cells were examined by gene expression analyses, flow cytometry, histopathology, and immunohistochemistry. RESULTS: The absence of the intestinal epithelial glucocorticoid receptor aggravated clinical symptoms and tissue damage, and compromised epithelial barrier integrity during colitis. Gene expression of chemokines, pattern recognition receptors and molecules controlling epithelial permeability was dysregulated in intestinal epithelial cells of GRvillin mice, leading to a reduced recruitment and a hyperactivation of leukocytes in the lamina propria of the colon. Importantly, the exaggerated inflammatory response in GRvillin mice also enhanced associated tumorigenesis, resulting in a higher number and larger size of tumors in the colon. CONCLUSIONS: Our results reveal an important role of intestinal epithelial cells as targets of glucocorticoid action in inflammatory bowel disease and suggest that the efficacy with which colitis is kept at bay directly affects the progression of colorectal cancer.


Subject(s)
Carcinogenesis/pathology , Colitis-Associated Neoplasms/pathology , Colitis/complications , Inflammation/pathology , Intestinal Mucosa/pathology , Receptors, Glucocorticoid/physiology , Animals , Azoxymethane/toxicity , Carcinogenesis/metabolism , Carcinogens/toxicity , Colitis/chemically induced , Colitis/pathology , Colitis-Associated Neoplasms/etiology , Colitis-Associated Neoplasms/metabolism , Dextran Sulfate/toxicity , Female , Gene Expression Profiling , Inflammation/etiology , Inflammation/metabolism , Intestinal Mucosa/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Permeability , Receptors, Glucocorticoid/deficiency
19.
Asian Pac J Cancer Prev ; 21(12): 3579-3586, 2020 Dec 01.
Article in English | MEDLINE | ID: mdl-33369455

ABSTRACT

OBJECTIVE: Eleutherine palmifolia (L.) Merr. extract (EPE) containing isoliquiritigenin and oxyresveratrol is believed to be an anticancer agent. This study evaluates colon histopathology, TNF-α, TGF-ß, and hepatotoxicity on BALB/c mice colitis-associated colon cancer (CAC) model treated with EPE. METHODS: In vivo study was performed on BALB/c mice CAC model induced by 10 mg/kgBW AOM on the first day followed by administration that each cycle consisted of 5% DSS in water for seven days and regular water for seven days. The indicators of the formation of CAC were observed by a fecal occult blood test (FOBT) and serum amyloid α (SAA) test. The treatment was conducted once a week started from the seventh week up to the twentieth week with six treatment groups: I was administrated by regular water only (negative control), II was administrated by AOM and DSS only (positive control), III was administrated by doxorubicin,  IV-VI were treated by EPE (0.25 mg/kg BW, 0.50 mg/kg BW, and 1.00 mg/kg BW) respectively. The colon and liver's histopathology was observed using hematoxylin-eosin (HE) staining, TNF-α with immunohistochemistry (IHC), and level measurement of TGF-ß colon with ELISA reader. The data were used one-way ANOVA followed by post hoc as statistical analysis. RESULTS: The administration of EPE increased the expression of TNF-α, the total of goblet cells of the colon, and decreased the level of TGF-ß. Administration of EPE 0.50 mg/20g BW decreased a liver histopathological score but induced a histopathological alteration of the liver at a dose of 1.00 mg/20g BW. CONCLUSION: This study indicate that EPE could be recommended as a colon anticancer through increase the goblet cells, induce apoptosis through increase TNF-α, and decrease TGF-ß.


Subject(s)
Colitis-Associated Neoplasms/drug therapy , Colitis/complications , Iridaceae/chemistry , Plant Extracts/pharmacology , Animals , Colitis/chemically induced , Colitis-Associated Neoplasms/etiology , Colitis-Associated Neoplasms/pathology , Dextran Sulfate/toxicity , Female , Goblet Cells/drug effects , Mice , Mice, Inbred BALB C , Toxicity Tests , Transforming Growth Factor beta/drug effects , Transforming Growth Factor beta/metabolism , Tumor Necrosis Factor-alpha/drug effects , Tumor Necrosis Factor-alpha/metabolism
20.
Int J Mol Sci ; 21(21)2020 Oct 24.
Article in English | MEDLINE | ID: mdl-33114313

ABSTRACT

Inflammatory bowel disease (IBD) includes Crohn's disease (CD) and ulcerative colitis (UC). These are chronic autoimmune diseases of unknown etiology affecting the gastrointestinal tract. The IBD population includes a heterogeneous group of patients with varying disease courses requiring personalized treatment protocols. The complexity of the disease often delays the diagnosis and the initiation of appropriate treatments. In a subset of patients, IBD leads to colitis-associated cancer (CAC). MicroRNAs are single-stranded regulatory noncoding RNAs of 18 to 22 nucleotides with putative roles in the pathogenesis of IBD and colorectal cancer. They have been explored as biomarkers and therapeutic targets. Both tissue-derived and circulating microRNAs have emerged as promising biomarkers in the differential diagnosis and in the prognosis of disease severity of IBD as well as predictive biomarkers in drug resistance. In addition, knowledge of the cellular localization of differentially expressed microRNAs is a prerequisite for deciphering the biological role of these important epigenetic regulators and the cellular localization may even contribute to an alternative repertoire of biomarkers. In this review, we discuss findings based on RT-qPCR, microarray profiling, next generation sequencing and in situ hybridization of microRNA biomarkers identified in the circulation and in tissue biopsies.


Subject(s)
Colitis-Associated Neoplasms/genetics , Genetic Markers , Inflammatory Bowel Diseases/genetics , MicroRNAs/genetics , Colitis-Associated Neoplasms/etiology , Diagnosis, Differential , Early Diagnosis , Epigenesis, Genetic , Female , Gene Expression Regulation , Humans , Inflammatory Bowel Diseases/complications , Male
SELECTION OF CITATIONS
SEARCH DETAIL
...