Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
Ann Epidemiol ; 94: 106-112, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38663524

ABSTRACT

PURPOSE: This study aimed to assess the correlation between glycemic status (prediabetes and type 2 diabetes mellitus) and the risk of gastric cancer according to menopausal status. METHODS: A total of 982,559 pre/peri-menopausal and 1445,419 postmenopausal women aged ≥ 40, who underwent the Korean national health screening in 2009, were included and followed up until 2018. Hazard ratio (HR) and 95% confidence interval (CI) were calculated for development of gastric cancers according to hyperglycemic status in both groups using Cox proportional hazards models. RESULTS: Over a mean follow-up period of 8.3 years, 3259 (0.33%) pre/peri-menopausal women and 13,245 (0.92%) postmenopausal women were diagnosed with gastric cancer. In postmenopausal women, only diabetes mellitus conferred a higher risk of gastric cancer compared to normal glycemic status (HR, 1.15; 95% CI, 1.09-1.20), with an increasing trend of gastric cancers from prediabetes to diabetes (P for trend < 0.001) observed regardless of menopausal status. Obesity, smoking, and heavy alcohol consumption was associated with increased gastric cancer risk mainly in the postmenopausal period. CONCLUSIONS: The risk of gastric cancer escalates with deteriorating glycemic status in a dose-response manner. Diabetes mellitus is linked with an elevated risk of gastric cancer in postmenopausal women.


Subject(s)
Diabetes Mellitus, Type 2 , Postmenopause , Stomach Neoplasms , Humans , Female , Stomach Neoplasms/epidemiology , Middle Aged , Republic of Korea/epidemiology , Diabetes Mellitus, Type 2/epidemiology , Diabetes Mellitus, Type 2/blood , Risk Factors , Adult , Blood Glucose/analysis , Prediabetic State/epidemiology , Prediabetic State/blood , Premenopause , Cohort Studies , Aged , Proportional Hazards Models
2.
Mol Carcinog ; 60(3): 188-200, 2021 03.
Article in English | MEDLINE | ID: mdl-33544929

ABSTRACT

Interaction between a tumor and its microenvironment is important for tumor initiation and progression. Cancer stem cells (CSCs) within the tumor interact with a microenvironmental niche that controls their maintenance and differentiation. We investigated the CSC-promoting effect of factors released from myofibroblasts into the microenvironment of early colorectal cancer tumors and its molecular mechanism. By messenger RNA microarray analysis, expression of HES1, a Notch signaling target, significantly increased in Caco-2 cells cocultured with 18Co cells (pericryptal myofibroblasts), compared to its expression in Caco-2 cells cultured alone. Caco-2 cells cultured in 18Co-conditioned media (CM) showed a significant increase in CD133+CD44+ cells and HES1 expression compared to that in Caco-2 cells cultured in regular media. Significant amounts of interleukin-6 (IL-6) and IL-8 were detected in 18Co-CM compared to levels in regular media. The 18Co-CM-induced increase in CD133+CD44+ cells was attenuated by IL-6- and IL-8-neutralizing antibodies. Furthermore, these neutralizing antibodies and inhibitors of STAT3 and gamma-secretase reduced the expression of HES1 induced in Caco-2 cells cultured in 18Co-CM. Immunohistochemical analysis of human tissues revealed that IL-6, IL-8, and HES1 expression increased from normal to adenoma, and from adenoma to cancer tissues. In addition, IL-6 and HES1 expression was positively correlated in early colorectal cancer tissues. In conclusion, the increase of CSCs by myofibroblasts could be mediated by IL-6/IL-8-induced HES1 activation in the tumor microenvironment. Based on these data, the IL-6/IL-8-mediated Notch/HES1 and STAT3 pathway, through which CSCs interact with their microenvironment, might be a potential target for the prevention and treatment of colorectal tumors.


Subject(s)
Colorectal Neoplasms/pathology , Interleukin-6/metabolism , Interleukin-8/metabolism , Neoplastic Stem Cells/pathology , Transcription Factor HES-1/metabolism , Caco-2 Cells , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Culture Media, Conditioned/pharmacology , Gene Expression Regulation, Neoplastic , Humans , Myofibroblasts/metabolism , Myofibroblasts/pathology , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Organoids/pathology , STAT3 Transcription Factor/antagonists & inhibitors , STAT3 Transcription Factor/metabolism , Transcription Factor HES-1/genetics , Tumor Microenvironment/drug effects
3.
Nat Commun ; 11(1): 5321, 2020 10 21.
Article in English | MEDLINE | ID: mdl-33087710

ABSTRACT

5-Fluorouracil (5-FU) remains the first-line treatment for colorectal cancer (CRC). Although 5-FU initially de-bulks the tumor mass, recurrence after chemotherapy is the barrier to effective clinical outcomes for CRC patients. Here, we demonstrate that p53 promotes WNT3 transcription, leading to activation of the WNT/ß-catenin pathway in ApcMin/+/Lgr5EGFP mice, CRC patient-derived tumor organoids (PDTOs) and patient-derived tumor cells (PDCs). Through this regulation, 5-FU induces activation and enrichment of cancer stem cells (CSCs) in the residual tumors, contributing to recurrence after treatment. Combinatorial treatment of a WNT inhibitor and 5-FU effectively suppresses the CSCs and reduces tumor regrowth after discontinuation of treatment. These findings indicate p53 as a critical mediator of 5-FU-induced CSC activation via the WNT/ß-catenin signaling pathway and highlight the significance of combinatorial treatment of WNT inhibitor and 5-FU as a compelling therapeutic strategy to improve the poor outcomes of current 5-FU-based therapies for CRC patients.


Subject(s)
Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/metabolism , Fluorouracil/pharmacology , Tumor Suppressor Protein p53/metabolism , Wnt Signaling Pathway/drug effects , beta Catenin/metabolism , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Colorectal Neoplasms/pathology , HCT116 Cells , Humans , Mice , Mice, Inbred NOD , Mice, Nude , Mice, SCID , Mice, Transgenic , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Organoids/drug effects , Organoids/metabolism , Organoids/pathology , Pyrazines/administration & dosage , Pyridines/administration & dosage , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Wnt3 Protein/genetics , Wnt3 Protein/metabolism , Xenograft Model Antitumor Assays
4.
Sci Rep ; 9(1): 18440, 2019 12 05.
Article in English | MEDLINE | ID: mdl-31804558

ABSTRACT

WNT signaling activation in colorectal cancers (CRCs) occurs through APC inactivation or ß-catenin mutations. Both processes promote ß-catenin nuclear accumulation, which up-regulates epithelial-to-mesenchymal transition (EMT). We investigated ß-catenin localization, transcriptome, and phenotypic differences of HCT116 cells containing a wild-type (HCT116-WT) or mutant ß-catenin allele (HCT116-MT), or parental cells with both WT and mutant alleles (HCT116-P). We then analyzed ß-catenin expression and associated phenotypes in CRC tissues. Wild-type ß-catenin showed membranous localization, whereas mutant showed nuclear localization; both nuclear and non-nuclear localization were observed in HCT116-P. Microarray analysis revealed down-regulation of Claudin-7 and E-cadherin in HCT116-MT vs. HCT116-WT. Claudin-7 was also down-regulated in HCT116-P vs. HCT116-WT without E-cadherin dysregulation. We found that ZEB1 is a critical EMT factor for mutant ß-catenin-mediated loss of E-cadherin and Claudin-7 in HCT116-P and HCT116-MT cells. We also demonstrated that E-cadherin binds to both WT and mutant ß-catenin, and loss of E-cadherin releases ß-catenin from the cell membrane and leads to its degradation. Alteration of Claudin-7, as well as both Claudin-7 and E-cadherin respectively caused tight junction (TJ) impairment in HCT116-P, and dual loss of TJs and adherens junctions (AJs) in HCT116-MT. TJ loss increased cell motility, and subsequent AJ loss further up-regulated that. Immunohistochemistry analysis of 101 CRCs revealed high (14.9%), low (52.5%), and undetectable (32.6%) ß-catenin nuclear expression, and high ß-catenin nuclear expression was significantly correlated with overall survival of CRC patients (P = 0.009). Our findings suggest that ß-catenin activation induces EMT progression by modifying cell-cell junctions, and thereby contributes to CRC aggressiveness.


Subject(s)
Colorectal Neoplasms/genetics , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic , Wnt Signaling Pathway/genetics , beta Catenin/genetics , Adherens Junctions/metabolism , Adherens Junctions/pathology , Antigens, CD/genetics , Antigens, CD/metabolism , Cadherins/genetics , Cadherins/metabolism , Claudins/genetics , Claudins/metabolism , Colorectal Neoplasms/pathology , Down-Regulation , Gene Expression Profiling , HCT116 Cells , Humans , Mutation , Oligonucleotide Array Sequence Analysis , Tight Junctions/metabolism , Tight Junctions/pathology , Zinc Finger E-box-Binding Homeobox 1/genetics , Zinc Finger E-box-Binding Homeobox 1/metabolism , beta Catenin/metabolism
5.
Exp Mol Med ; 50(11): 1-12, 2018 11 20.
Article in English | MEDLINE | ID: mdl-30459318

ABSTRACT

Drugs targeting the epidermal growth factor receptor (EGFR), such as cetuximab and panitumumab, have been prescribed for metastatic colorectal cancer (CRC), but patients harboring KRAS mutations are insensitive to them and do not have an alternative drug to overcome the problem. The levels of ß-catenin, EGFR, and RAS, especially mutant KRAS, are increased in CRC patient tissues due to mutations of adenomatous polyposis coli (APC), which occur in 90% of human CRCs. The increases in these proteins by APC loss synergistically promote tumorigenesis. Therefore, we tested KYA1797K, a recently identified small molecule that degrades both ß-catenin and Ras via GSK3ß activation, and its capability to suppress the cetuximab resistance of KRAS-mutated CRC cells. KYA1797K suppressed the growth of tumor xenografts induced by CRC cells as well as tumor organoids derived from CRC patients having both APC and KRAS mutations. Lowering the levels of both ß-catenin and RAS as well as EGFR via targeting the Wnt/ß-catenin pathway is a therapeutic strategy for controlling CRC and other types of cancer with aberrantly activated the Wnt/ß-catenin and EGFR-RAS pathways, including those with resistance to EGFR-targeting drugs attributed to KRAS mutations.


Subject(s)
Antineoplastic Agents/therapeutic use , Colorectal Neoplasms/drug therapy , Drug Resistance, Neoplasm , ErbB Receptors/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Animals , Cell Line, Tumor , Cetuximab/therapeutic use , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , Humans , Mice , Mice, Inbred C57BL , Mutation , Proteolysis , Proto-Oncogene Proteins p21(ras)/genetics , Wnt Signaling Pathway , beta Catenin/metabolism
6.
Sci Rep ; 8(1): 13111, 2018 Aug 28.
Article in English | MEDLINE | ID: mdl-30154414

ABSTRACT

A correction to this article has been published and is linked from the HTML and PDF versions of this paper. The error has been fixed in the paper.

7.
Sci Rep ; 8(1): 409, 2018 01 11.
Article in English | MEDLINE | ID: mdl-29323154

ABSTRACT

Metformin has been known to suppress cancer stem cells (CSCs) in some cancers. However, the differential effects of metformin on CSCs and their mechanisms have not been reported. Herein, metformin induced pAMPK activation and pS6 suppression in metformin-sensitive (HT29) cells, but not in metformin-resistant (SW620) cells. The oxygen consumption rate was higher in HT29 cells than in SW620 cells and showed a prominent decrease after metformin treatment in HT29 cells. In glutamine-depleted medium, but not in low-glucose medium, SW620 cells became sensitive to the CSC-suppressing effect of metformin. A combination of metformin and glutaminase C inhibitor (compound 968) suppressed CSCs in SW620 cells and enhanced that effect in HT29 cells. SW620 cells showed higher expression of glutaminase 1 and glutamine transporter (ASCT2) than HT29 cells, especially ASCT2 in CSCs. Knockdown of glutaminase 1, ASCT2, and c-Myc induced significant CSC-suppression and enhanced CSC-suppressing effect of metformin and compound 968. In xenografts and human cancer organoids, combined treatment with metformin and compound 968 showed the same results as those shown in vitro. In conclusion, the effect of metformin on CSCs varies depending on the AMPK-mTOR and glutamine metabolism. The inhibition of glutamine pathway could enhance the CSC-suppressing effect of metformin, overcoming metformin resistance.


Subject(s)
Benzophenanthridines/pharmacology , Colorectal Neoplasms/drug therapy , Glutamine/metabolism , Metformin/pharmacology , Neoplastic Stem Cells/drug effects , Adenylate Kinase/metabolism , Animals , Benzophenanthridines/therapeutic use , Cell Line, Tumor , Colorectal Neoplasms/metabolism , Drug Resistance, Neoplasm/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Glucose/metabolism , HT29 Cells , Humans , Metformin/therapeutic use , Mice , Neoplasm Transplantation , Neoplastic Stem Cells/metabolism , Oxygen Consumption , Ribosomal Protein S6 Kinases/metabolism
8.
Sci Rep ; 7: 39850, 2017 01 03.
Article in English | MEDLINE | ID: mdl-28045058

ABSTRACT

Several recent genome-wide association studies (GWAS) identified susceptibility loci/genes for Behçet's disease (BD). However, no study has specifically investigated the genetic susceptibility loci associated with intestinal involvement in BD. We aimed to identify distinctive genetic susceptibility loci/genes associated with intestinal involvement in BD and determine their roles in intestinal inflammation as well as their interactions with genes involved in inflammatory bowel disease (IBD). GWAS and validation studies showed intestinal BD-specific associations with an NAALADL2 gene locus (rs3914501, P = 3.8 × 10-4) and a YIPF7 gene locus (rs6838327, P = 3.5 × 10-4). Validation, haplotype, and pathway analyses showed distinct genetic architectures between intestinal BD and BD without intestinal involvement. Furthermore, network analysis revealed shared pathogenic pathways between intestinal BD and IBD. Gene functional analyses indicated that down-regulation of NAALADL2 and YIPF7 expression was associated with exacerbating intestinal inflammatory responses both in vitro and in vivo. Our results provide new insights into intestinal BD-specific genetic variations, which represents a distinct pathway from BD without intestinal involvement. Functional consequences of the intestinal BD-specific NAALADL2 and YIPF7 expression patterns proved a suggestive association with intestinal inflammation risk, which warrants further validation.


Subject(s)
Behcet Syndrome/genetics , Glutamate Carboxypeptidase II/genetics , Intestinal Diseases/genetics , Membrane Proteins/genetics , Adult , Behcet Syndrome/pathology , Female , Glutamate Carboxypeptidase II/metabolism , HT29 Cells , Humans , Intestinal Diseases/pathology , Male , Middle Aged
9.
Scand J Gastroenterol ; 43(9): 1116-21, 2008.
Article in English | MEDLINE | ID: mdl-18609172

ABSTRACT

OBJECTIVE: Several reports have suggested that flat colorectal adenomas might exhibit a higher potential for malignancy compared to polypoid adenomas. Although the clinical importance of the shape of polyps is stressed, the controversy surrounding the malignant potential of flat adenomas continues. The aim of this study was to compare the clinicopathologic characteristics, including degree of dysplasia and malignancy, between flat and polypoid adenomas 5 mm in size or larger. MATERIAL AND METHODS: A total of 3263 polyps (254 flat adenomas and 3009 polypoid adenomas), >/=5 mm in size, diagnosed in 1883 patients by colonoscopy were analyzed. RESULTS: Flat adenomas were found in 10% of patients, which represented 7.8% of all adenomas removed. The flat adenomas were larger in diameter than the polypoid adenomas (14.8+/-12.6 mm versus 8.6+/-5.0 mm, p <0.01), had a higher rate of villous components (18.5% versus 11.4%, p <0.01), a higher rate of high-grade dysplasia (9.4% versus 4.2%, p <0.01), and a higher rate of malignancy (10.2% versus 3.6%, p <0.01) than polypoid adenomas. However, there was no difference in the rate of high-grade dysplasia or carcinoma between flat and polypoid adenomas of equal size. It was shown by multivariate analysis that rectosigmoid location, larger size, and presence of a villous component were associated with a higher rate of malignancy, but not with flat morphology. CONCLUSIONS: Flat adenomas, which were of a relatively large size in this study, were not associated with a higher risk for high-grade dysplasia and carcinoma compared with polypoid adenomas.


Subject(s)
Adenoma/pathology , Adenomatous Polyps/pathology , Colonic Polyps/pathology , Colorectal Neoplasms/pathology , Precancerous Conditions/pathology , Aged , Analysis of Variance , Biopsy, Needle , Cohort Studies , Colonoscopy/methods , Diagnosis, Differential , Female , Humans , Immunohistochemistry , Intestinal Mucosa/pathology , Logistic Models , Male , Middle Aged , Multivariate Analysis , Probability , Retrospective Studies , Risk Assessment
10.
Biochem Biophys Res Commun ; 337(2): 457-63, 2005 Nov 18.
Article in English | MEDLINE | ID: mdl-16202385

ABSTRACT

Intestinal epithelial cells not only present a physical barrier to bacteria but also participate actively in immune and inflammatory responses. The migration of epithelial cells from the crypt base to the surface is accompanied by a cellular differentiation that leads to important morphological and functional changes. It has been reported that the differentiation of colonic epithelial cells is associated with reduced interleukin (IL)-8 responses to IL-1beta. Although toll-like receptor 4 (TLR4) has been previously identified to be an important component of mucosal immunity to lipopolysaccharide (LPS) in the colon, little is known about the regulation of TLR4 in colonic epithelial cells during cellular differentiation. We investigated the effects of differentiation on LPS-induced IL-8 secretion and on the expression of TLR4. Differentiation was induced in colon cancer cell line HT-29 cells by butyrate treatment or by post-confluence culture and assessed by measuring alkaline phosphatase (AP) activity. IL-8 secretion was measured by ELISA, and TLR4 protein and mRNA expressions were followed by Western blot and RT-PCR, respectively. HT-29 cells were found to be dose-dependently responsive to LPS. AP activity increased in HT-29 cells by differentiation induced by treatment with butyrate or post-confluence culture. We found that IL-8 secretion induced by LPS was strongly attenuated in differentiated cells versus undifferentiated cells, and that cellular differentiation also attenuated TLR4 mRNA and protein expressions. Pretreating HT-29 cells with tumor necrosis factor (TNF)-alpha or interferon (INF)-gamma augmented LPS-induced IL-8 secretion and TLR4 expression. These TNF-alpha- or INF-gamma-induced augmentations of LPS response and TLR4 expression were all down-regulated by differentiation. Collectively, we conclude that cellular differentiation attenuates IL-8 secretion induced by LPS in HT-29 cells, and this attenuation is related with the down-regulation of TLR4 expression.


Subject(s)
Cell Differentiation/physiology , Gene Expression Regulation/drug effects , Lipopolysaccharides/pharmacology , Toll-Like Receptor 4/metabolism , Alkaline Phosphatase/metabolism , Butyrates/pharmacology , Cells, Cultured , Colon/metabolism , Down-Regulation/drug effects , HT29 Cells , Humans , Interleukin-8/metabolism , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...