Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Front Oncol ; 12: 929092, 2022.
Article in English | MEDLINE | ID: mdl-35847876

ABSTRACT

Background: Accumulating data support that regular physical activity potentially inhibits chronic colitis, a risk factor for colitis-associated cancer (CAC). However, possible effects of physical activity on CAC and the underlying mechanisms remain poorly understood. Methods: A pretreatment of swimming on azoxymethane/dextran sodium sulfate (AOM/DSS)-induced CAC mice was implemented to determine its protective effect. Inflammation and tumorigenesis were assessed using colorectums from C57BL/6 mice. In order to determine how swimming alters colonic lipid metabolism and gene expression, a comparative analysis was conducted. Meanwhile, alterations in intestinal microbiota and short-chain fatty acids (SCFAs) were detected and analyzed. Finally, an integration analysis of colonic lipid metabolism with gene expression and intestinal microbiota was performed respectively. Result: Swimming pretreatment relieved bowel inflammation and minimized tumor formation. We demonstrated that prostaglandin E2 (PGE2)/PGE2 receptor 2 subtype (EP2) signaling as a potential regulatory target for swimming induces colonic lipid metabolites. Swimming-induced genera, Erysipelatoclostridium, Parabacteroides, Bacteroides, and Rikenellaceae_RC9_gut_group, induced intestinal SCFAs and affected the function of colonic lipid metabolites enriched in glycerophospholipid metabolism and choline metabolism in cancer. Conclusion: According to our experiments, swimming pretreatment can protect mice from CAC by intervention in the possible link between colonic lipid metabolites and PGE2/EP2 signaling. Further, swimming-induced genera and probiotics promoted glycerophospholipid metabolism and choline metabolism in cancer, the major constituents of colonic lipid metabolites, and increased SCFAs, which were also important mechanisms for the anti-inflammatory and anti-tumorigenic effects of swimming.

2.
Ann Transl Med ; 10(4): 169, 2022 Feb.
Article in English | MEDLINE | ID: mdl-35280417

ABSTRACT

Background: Microsatellite instability-high (MSI-H) is a form of genomic instability present in 15% of colorectal cancer (CRC) cases. Several differential gene analyses have been conducted on CRC; however, none have specifically explored the differentially expressed genes in MSI-H CRC. Research on the different gene expressions between MSI-H CRC and microsatellite stable (MSS) CRC, and their different patterns of metastasis will provide invaluable insights for diagnosis, prognosis, and treatment. Methods: In this study, the differential expression of 46,602 genes were analyzed across 613 different tissue samples from The Cancer Genome Atlas (TCGA)-colon adenocarcinoma (COAD) and TCGA-rectum adenocarcinoma (READ) as part of a gene association analysis. R package TCGAbiolinks (version 2.18.0) was used to download the data set, and DESeq2 (version 1.30.1) was used for the differential gene analysis. The resulting genes were then analyzed for shared pathways with R package clusterProfiler (version 3.0.4). Results: A total of 237 significantly differentially expressed genes (Padj<0.05) were found between MSI-H and MSS CRC. Differentially expressed genes include insulin like growth factor 2 (IGF2) and fibroblast growth factor 3 (FGF3), and the enriched pathways mostly involve hearing, digestive regulation, and neurogenesis.463 differentially expressed genes were found between metastatic and non-metastatic CRC. Notably differentially expressed genes in metastatic CRC include DEAD-box helicase 53 (DDX53) and adiponectin, C1Q and collagen domain containing (ADIPOQ), and enriched pathways include the immune system, cell adhesion, and cell signaling. For MSI-H CRC, a total of 34 genes were significantly differently expressed between metastatic and non-metastatic CRC. These include notum, palmitoleoyl-protein carboxylesterase (NOTUM), serpin family B member 2 (SERPINB2), and several keratin (KRT) genes, and the pathway analysis showed the major enrichment of the hormonal and secretion and regulation pathways. Of the differentially expressed genes in metastatic CRC, 25 were immunity related and include fatty acid binding protein 4 (FABP4), and the pathway analysis showed the enrichment of humoral immunity and lymphocyte regulation. Conclusions: Of the biologically plausible differentially expressed genes, the most notable were NOTUM, KRT6A, KRT14, SERPINB2, and serum amyloid A1 (SAA1). NOTUM, KRT6A, and KRT14 are active in the Wnt pathway. All five are also involved in various inflammation pathways.

3.
Neoplasma ; 68(5): 1015-1022, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34374296

ABSTRACT

Colorectal cancer (CRC) is the most common malignancy worldwide, and its underlying molecular mechanisms remain largely unexplored. Accumulating evidences indicate cancer-associated fibroblasts (CAFs), abundant stromal cell population in the tumor microenvironment, play a key role in tumor development. Herein, we have successfully isolated CAFs and paired normal fibroblasts (NFs) from colorectal cancer tissues (n=10). By using a multiplex cytokine profiling assay, we have identified IL-6 as a major cytokine released by CAFs. Co-culturing of CAFs with CRC cell lines HCT116 or SW480 increases IL-6 release, and the secretion by CAFs can be further enhanced under hypoxia. By using the CCK-8 assay, we have found that HCT116 or SW480 cells treated with culture medium from CAFs, IL-6, or hypoxia showed a significant cell growth compared to control cells (p<0.01). Mechanistically, we have found that hypoxia could enhance the effect of the IL-6/STAT3 signaling on CRC cells, in part, through HIF-1a targeting PKM2. In conclusion, our data clearly proposes the interconnected mechanisms for constitutive activation of STAT3 signal by CAFs-derived IL-6 under hypoxia in colorectal cancer. The pharmacological inhibition of STAT3, PKM2, or HIF-1α can significantly reduce the oncogenic effect of IL-6, providing a potential therapeutic target for CRC patients.


Subject(s)
Cancer-Associated Fibroblasts , Colorectal Neoplasms , Interleukin-6/metabolism , Cell Line, Tumor , Cell Proliferation , Humans , Hypoxia , Tumor Microenvironment
4.
J Cell Mol Med ; 25(16): 8006-8014, 2021 08.
Article in English | MEDLINE | ID: mdl-34173718

ABSTRACT

Ajuba is a multiple LIM domain-containing protein and functions as a transcriptional coregulator to modulate many gene expressions in various cellular processes. Here, we describe that the LIM domain of Ajuba interacts with Twist, and the Twist box is a pivotal motif for the interaction. Biologically, Ajuba enhances transcription of target gene N-cadherin as an obligate coactivator of Twist. The enhancement is achieved by binding to the E-box element within N-cadherin promoter as revealed by luciferase reporter and chromatin immunoprecipitation assays. Mechanistic investigation demonstrates that Ajuba recruits CBP and Twist to form a ternary complex at the Twist target promoter region and concomitantly enhances histone acetylation at these sites. These findings identify that Twist is a new interacting protein of Ajuba and Ajuba/Twist/CBP ternary complex may be a potential treatment strategy for Twist-related tumour metastasis.


Subject(s)
Antigens, CD/metabolism , Cadherins/metabolism , Colorectal Neoplasms/pathology , Gene Expression Regulation , LIM Domain Proteins/metabolism , Nuclear Proteins/metabolism , Peptide Fragments/metabolism , Promoter Regions, Genetic , Sialoglycoproteins/metabolism , Twist-Related Protein 1/metabolism , Cell Line, Tumor , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Humans , Protein Interaction Domains and Motifs
5.
J Dig Dis ; 18(3): 169-178, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28296190

ABSTRACT

OBJECTIVE: MicroRNA-1254 (miR-1254) has not been studied in colorectal cancer (CRC) to date. This study aimed to investigate the inhibitory mechanism of miR-1254 in CRC tumorigenesis. METHODS: MiR-1254 expression was examined using real-time polymerase chain reaction in CRC and adjacent non-tumorous tissues. The correlation between miR-1254 expressions and proliferation and migration of cancer cells was determined using the CCK-8 and transwell assays. RNA sequencing was used to identify differentially expressed genes downstream from miR-1254. A luciferase reporter assay was used to confirm the direct interaction between miR-1254 and its predicted target gene, PSMD10. Moreover, PSMD10 was either overexpressed or silenced in colon carcinoma cells overexpressing miR-1254 to determine whether their interaction contributed to CRC migration and epithelial-mesenchymal transition (EMT). RESULTS: Significantly lower miR-1254 expressions were observed in CRC tissues than in adjacent non-tumorous tissues. Exogenous miR-1254 expression suppressed the migration of colon carcinoma cell lines SW1116 and HCT116. RNA sequencing and luciferase assays revealed that miR-1254 directly binded to the 3'-untranslated region of PSMD10, an important regulator of EMT and cell migration. PSMD10 knockdown inhibited EMT and colon cancer cell migration, whereas PSMD10 overexpression reversed the inhibition of EMT and cell migration caused by miR-1254. CONCLUSION: MiR-1254 may act as a tumor suppressor in CRC and may inhibit CRC migration by directly targeting PSMD10 to suppress the EMT process.


Subject(s)
Adenocarcinoma/genetics , Colorectal Neoplasms/genetics , MicroRNAs/genetics , Proteasome Endopeptidase Complex/genetics , Proto-Oncogene Proteins/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Cell Movement/genetics , Cell Proliferation/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Epithelial-Mesenchymal Transition/genetics , Epithelial-Mesenchymal Transition/physiology , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Gene Targeting/methods , Humans , Proteasome Endopeptidase Complex/biosynthesis , Proto-Oncogene Proteins/biosynthesis , RNA, Neoplasm/genetics , Real-Time Polymerase Chain Reaction , Tumor Cells, Cultured
6.
Int J Clin Exp Pathol ; 8(9): 10512-22, 2015.
Article in English | MEDLINE | ID: mdl-26617761

ABSTRACT

OBJECTIVE: Perfluorooctanoic acid (PFOA) is widely used in consumer products and detected in human serum. Our study meant to elucidate the uncovered molecular mechanisms underlying the PFOA induced colorectal cancer cell DLD-1 invasion and matrix metalloproteinases (MMP) expression. METHODS AND RESULTS: Trans-well filter assay appeared that PFOA treatment stimulated DLD-1 cells invasion significantly. Meanwhile, the results of luciferase reporter, quantitative real-time PCR, western blotting, and gelatin zymography showed that PFOA induced MMP-2/-9 expression and enzyme activation levels consistently (P < 0.05 each). Subsequently, western blotting and immunofluorescence assay demonstrated that PFOA could enhance nuclear factor kappaB (NF-κB) activity by stimulating NF-κB translocation into nuclear in DLD-1 cells. Furthermore, JSH-23, a well-known NF-κB inhibitor, could reverse the PFOA induced colorectal cancer cell invasion and MMP-2/-9 expression. CONCLUSIONS: Our study confirmed that PFOA could induce colorectal cancer cell DLD-1 invasive ability and MMP-2/-9 expression through activating NF-κB, which deserves more concerns on environmental pollutant-resulted public health risk.


Subject(s)
Caprylates/toxicity , Colorectal Neoplasms/pathology , Fluorocarbons/toxicity , Gene Expression/drug effects , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , NF-kappa B/metabolism , Blotting, Western , Cell Line, Tumor , Environmental Pollutants/toxicity , Fluorescent Antibody Technique , Humans , Neoplasm Invasiveness/pathology , Real-Time Polymerase Chain Reaction
7.
Biomed Res Int ; 2013: 181384, 2013.
Article in English | MEDLINE | ID: mdl-23509688

ABSTRACT

PURPOSE: DLC-1 is a tumor suppressor gene frequently silenced in human cancers. However, the pathogenicity of DLC-1 epigenetic silencing in the mucosa-adenoma-carcinoma transformation process of colorectal cancer (CRC) has not been studied. METHODS: Promoter methylation status of DLC-1 was evaluated in 4 human CRC cell lines, 48 normal mucosa, 57 adenomas, and 80 CRC tissues with methylation-sensitive high-resolution melting analysis (MS-HRMA), while the mRNA expression was examined by qPCR. HRMA was utilized to detect the KRAS codon 12, 13 and BRAF V600E mutations. RESULTS: Partial (1%-10%) and extensive (10%-100%) DLC-1 promoter methylations were observed in 10% and 0% of normal mucosa, 46% and 14% of adenomas, and 60% and 36% of CRCs, respectively. The promoter methylation of DLC-1 was related with the reduction of gene expression and the advanced Duke's stages (Stage C and D). DLC-1 promoter methylation and KRAS mutations are common concurrent pathological alternations. CONCLUSIONS: Epigenetic alternation plays a key role in the transcriptional silencing of DLC-1. It is also an independent risk factor related to the carcinogenesis of colorectal tumors and spans over its pathogenesis process. Therefore, DLC-1 promoter methylation quantitation may have a promising significance in the evaluation and management of CRC patients.


Subject(s)
Adenoma/genetics , Colorectal Neoplasms/genetics , DNA Methylation , GTPase-Activating Proteins/metabolism , Gene Expression Regulation, Neoplastic , Promoter Regions, Genetic , Tumor Suppressor Proteins/metabolism , Adenoma/metabolism , Base Sequence , Cell Line, Tumor , Colorectal Neoplasms/metabolism , CpG Islands , Disease Progression , Epigenesis, Genetic , GTPase-Activating Proteins/genetics , Gene Expression Profiling , Gene Silencing , Humans , Molecular Sequence Data , Mutation , Risk Factors , Sequence Analysis, DNA , Sequence Homology, Nucleic Acid , Tumor Suppressor Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...