Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
Gut ; 2024 Jul 18.
Article in English | MEDLINE | ID: mdl-39025494
2.
J Gastroenterol Hepatol ; 38(10): 1768-1777, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37259282

ABSTRACT

BACKGROUND: Colorectal cancer (CRC) incidence has increased among patients aged <50 years. Exploring high-risk factors and screening high-risk populations may help lower early-onset CRC (EO-CRC) incidence. We developed noninvasive predictive models for EO-CRC and investigated its risk factors. METHODS: This retrospective multicenter study collected information on 1756 patients (811 patients with EO-CRC and 945 healthy controls) from two medical centers in China. Sociodemographic features, clinical symptoms, medical and family history, lifestyle, and dietary factors were measured. Patients from one cohort were randomly assigned (8:2) to two groups for model establishment and internal validation, and another independent cohort was used for external validation. Multivariable logistic regression, random forest, and eXtreme Gradient Boosting (XGBoost) were performed to establish noninvasive predictive models for EO-CRC. Some variables in the model influenced EO-CRC occurrence and were further analyzed. Multivariable logistic regression analysis yielded adjusted odd ratios (ORs) and 95% confidence intervals (CIs). RESULTS: All three models showed good performance, with areas under the receiver operator characteristic curves (AUCs) of 0.82, 0.84, and 0.82 in the internal and 0.78, 0.79, and 0.78 in the external validation cohorts, respectively. Consumption of sweet (OR 2.70, 95% CI 1.89-3.86, P < 0.001) and fried (OR 2.16, 95% CI 1.29-3.62, P < 0.001) foods ≥3 times per week was significantly associated with EO-CRC occurrence. CONCLUSION: We established noninvasive predictive models for EO-CRC and identified multiple nongenetic risk factors, especially sweet and fried foods. The model has good performance and can help predict the occurrence of EO-CRC in the Chinese population.


Subject(s)
Colorectal Neoplasms , Life Style , Humans , Asian People , Colorectal Neoplasms/diagnosis , Colorectal Neoplasms/epidemiology , Colorectal Neoplasms/etiology , Retrospective Studies , Risk Factors , Random Allocation
3.
Nat Microbiol ; 8(5): 919-933, 2023 05.
Article in English | MEDLINE | ID: mdl-37069401

ABSTRACT

Epidemiological studies have indicated an association between statin use and reduced incidence of colorectal cancer (CRC), and work in preclinical models has demonstrated a potential chemopreventive effect. Statins are also associated with reduced dysbiosis in the gut microbiome, yet the role of the gut microbiome in the protective effect of statins in CRC is unclear. Here we validated the chemopreventive role of statins by retrospectively analysing a cohort of patients who underwent colonoscopies. This was confirmed in preclinical models and patient cohorts, and we found that reduced tumour burden was partly due to statin modulation of the gut microbiota. Specifically, the gut commensal Lactobacillus reuteri was increased as a result of increased microbial tryptophan availability in the gut after atorvastatin treatment. Our in vivo studies further revealed that L. reuteri administration suppressed colorectal tumorigenesis via the tryptophan catabolite, indole-3-lactic acid (ILA). ILA exerted anti-tumorigenic effects by downregulating the IL-17 signalling pathway. This microbial metabolite inhibited T helper 17 cell differentiation by targeting the nuclear receptor, RAR-related orphan receptor γt (RORγt). Together, our study provides insights into an anti-cancer mechanism driven by statin use and suggests that interventions with L. reuteri or ILA could complement chemoprevention strategies for CRC.


Subject(s)
Colorectal Neoplasms , Hydroxymethylglutaryl-CoA Reductase Inhibitors , Limosilactobacillus reuteri , Microbiota , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Tryptophan , Retrospective Studies , Colorectal Neoplasms/prevention & control
4.
Br J Cancer ; 128(2): 363-374, 2023 01.
Article in English | MEDLINE | ID: mdl-36396820

ABSTRACT

BACKGROUND: Chemotherapy resistance is the major cause of recurrence in patients with colorectal cancer (CRC). A previous study found that Fusobacterium (F.) nucleatum promoted CRC chemoresistance. Additionally, metformin rescued F. nucleatum-induced tumorigenicity of CRC. Here, we aimed to investigate whether metformin could revert F. nucleatum-induced chemoresistance and explore the mechanism. METHODS: The role of metformin in F. nucleatum-infected CRC cells was confirmed using cell counting kit 8 assays and CRC xenograft mice. Stemness was identified by tumorsphere formation. Bioinformatic analyses were used to explore the regulatory molecules involved in metformin and F. nucleatum-mediated regulation of the sonic hedgehog pathway. RESULTS: We found that metformin abrogated F. nucleatum-promoted CRC resistance to chemotherapy. Furthermore, metformin attenuated F. nucleatum-stimulated stemness by inhibiting sonic hedgehog signaling. Mechanistically, metformin diminished sonic hedgehog signaling proteins by targeting the MYC/miR-361-5p cascade to reverse F. nucleatum-induced stemness, thereby rescuing F. nucleatum-triggered chemoresistance in CRC. CONCLUSIONS: Metformin acts on F. nucleatum-infected CRC via the MYC/miR-361-5p/sonic hedgehog pathway cascade, subsequently reversing stemness and abolishing F. nucleatum-triggered chemoresistance. Our results identified metformin intervention as a potential clinical treatment for patients with chemoresistant CRC with high amounts of F. nucleatum.


Subject(s)
Colorectal Neoplasms , MicroRNAs , Humans , Animals , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , Hedgehog Proteins/genetics , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Fusobacterium nucleatum , Drug Resistance, Neoplasm/genetics
5.
Kaohsiung J Med Sci ; 37(1): 47-54, 2021 Jan.
Article in English | MEDLINE | ID: mdl-32916039

ABSTRACT

Hepatocellular carcinoma (HCC), a primary liver tumor, is the third leading cause of cancer-related mortality worldwide. The proteasome system is overactivated in the majority of tumors, including HCC. However, targeting the proteasome system in HCC is not as effective as in other types of cancer. Therefore, a new target of HCC therapy needs to be identified, and the potential mechanism must be studied. Using the The Cancer Gene Genome Atlas and GEO datasets, the present investigation demonstrated for the first time that ADRM1 is overexpressed in HCC, and the high level of its expression predicts poor overall survival in HCC patients. The high expression of ADRM1 in HCC was verified using tumor tissue arrays. By comparing paired tumor and nontumor tissues, it was shown that the majority of HCC patients (76.25%) exhibited higher ADRM1 expression in the tumor than in normal tissues. in vitro experiments demonstrated that targeting ADRM1 with shRNAs significantly suppressed the proliferation of HCC cells. RA190, a specific inhibitor of ADRM1, suppressed cell proliferation and colony formation by HCC cells in a concentration-dependent manner. The study of the mechanism of the effects of RA190 revealed that targeting ADRM1 blocked the G2/M transition in the cell cycle and induced apoptosis of HCC cells. Together, the obtained results indicate that ADRM1 is a promising target for HCC therapy and suggest that ADRM1 inhibitors, such as RA190, have the potential for clinical application in the treatment of HCC.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Liver Neoplasms/metabolism , Molecular Targeted Therapy , Apoptosis/drug effects , Benzylidene Compounds/pharmacology , Carcinoma, Hepatocellular/pathology , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Gene Knockdown Techniques , Humans , Liver Neoplasms/pathology , Tumor Stem Cell Assay
6.
Clin Lab ; 66(12)2020 Dec 01.
Article in English | MEDLINE | ID: mdl-33337839

ABSTRACT

BACKGROUND: PD-L1 expression on tumor-infiltrating lymphocytes (TILs) has recently been reported as a biomarker for colorectal cancer (CRC). However, the prognostic and clinical significance of PD-L1 on TILs in CRC remains controversial. We performed this meta-analysis to evaluate the association between the PD-L1 expression on TILs and clinicopathological features and prognosis of CRC patients. METHODS: A comprehensive literature search for relevant studies published up to Feb 2020 was performed using Medline, Embase, and Web of Science. Odds ratio (OR) with 95% CI was selected to appraise the correlation between PD-L1 expression on TILs with prognostic and clinicopathological characteristics of CRC patients. Begg's and Egger's test were used to assess publication bias. The statistical analysis was conducted using Stata software. RESULTS: A total of 19 studies including 5,213 CRC cases were included in this meta-analysis. The pooled results showed that PD-L1 overexpression on TILs was relevant to longer OS (OR = 1.36, 95% CI = 1.19 - 1.55, p < 0.01) and longer DFS/RFS (OR = 1.22, 95% CI = 1.03 - 1.44, p = 0.02). Moreover, CRC patients with high expression of PD-L1 on TILS was associated with lower T stage (OR = 2.30, 95% CI = 1.85 - 2.87, p < 0.01), less lymph node in-vasion (OR = 1.48, 95% CI = 1.03 - 2.13, p = 0.03), less distant metastasis (OR = 2.56, 95% CI = 1.81 - 3.64, p < 0.01), earlier TNM stage (OR = 1.93, 95% CI = 1.34 - 2.66, p < 0.01), later tumor grade (OR = 0.38, 95% CI = 0.23 - 0.62, p < 0.01) and high MSI status (OR = 0.36, 95% CI = 0.25 - 0.52, p < 0.01). But it is not related to tumor size, tumor differentiation, MMR status, BRAF mutant, and KRAS mutant. CONCLUSIONS: This meta-analysis revealed that PD-L1 expression on TILs can serve as a significant biomarker for positive prognosis and clinicopathological features of CRC. Our results may provide some useful information when using PD-L1 expression to predict the survival of CRC patients and to select the beneficial CRC patients from PD-1/PD-L1 antibody treatment.


Subject(s)
B7-H1 Antigen , Colorectal Neoplasms , Lymphocytes, Tumor-Infiltrating , B7-H1 Antigen/genetics , B7-H1 Antigen/metabolism , Colorectal Neoplasms/genetics , Humans , Neoplasm Staging , Prognosis
7.
J Dig Dis ; 21(7): 385-398, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32441482

ABSTRACT

OBJECTIVE: Fusobacterium nucleatum (F. nucleatum) has been reported to be enriched in patients with inflammatory bowel disease (IBD). This study aimed to explore the role of F. nucleatum in IBD and its pathogenic mechanism. METHODS: Several bacteria that have been reported to be associated with IBD or colorectal cancer were measured in the fecal samples of 91 patients with IBD and 43 healthy individuals. Mice with dextran sulfate sodium (DSS)-induced colitis and a Caco-2 cell line were used to explore the pathogenicity of F. nucleatum. Barrier damage was evaluated by a transmission electron microscope, the permeability of fluorescein isothiocyanate-dextran, transepithelial electrical resistance and immunofluorescence. Protein levels of the cell-cell junction and activation of the STAT3 signaling pathway were detected by immunohistochemistry and immunoblot. Cytokine secretion and T-cell differentiation were measured by quantitative real-time polymerase chain reaction and flow cytometry. RESULTS: F. nucleatum was significantly enriched in the feces of patients with IBD and its abundance correlated with disease activity. Administration of F. nucleatum markedly exacerbated colitis in a DSS mouse model. Mechanistically, F. nucleatum damaged epithelial integrity and increased permeability by regulating the expression and distribution of tight junction proteins zonula occludens-1 and occludin. Moreover, F. nucleatum promoted the secretion of cytokines (tumor necrosis factor-α, interferon-γ, interleukin [IL]-1ß, IL-6, and IL-17), activated the STAT3 signaling pathway, and induced CD4+ T cell proliferation and Th1 and Th17 subset differentiations. CONCLUSION: F. nucleatum can damage the intestinal barrier and induce aberrant inflammation, which exacerbates colitis.


Subject(s)
Colitis , Fusobacterium nucleatum , Inflammation , Intestinal Mucosa/pathology , Animals , Caco-2 Cells , Colitis/immunology , Colitis/microbiology , Dextran Sulfate , Disease Models, Animal , Fusobacterium nucleatum/pathogenicity , Humans , Inflammation/microbiology , Intestinal Mucosa/microbiology , Mice , Mice, Inbred C57BL
8.
Cancer Cell Int ; 20: 120, 2020.
Article in English | MEDLINE | ID: mdl-32308565

ABSTRACT

BACKGROUND: Splicing factor SRSF3 is an oncogene and overexpressed in various kinds of cancers, however, the function and mechanism involved in colorectal cancer (CRC) remained unclear. The aim of this study was to explore the relationship between SRSF3 and carcinogenesis and progression of CRC. METHODS: The expression of SRSF3 in CRC tissues was detected by immunohistochemistry. The proliferation and invasion rate was analyzed by CCK-8 assay, colony formation assay, transwell invasion assay and xenograft experiment. The expression of selected genes was detected by western blot or real time PCR. RESULTS: SRSF3 is overexpressed in CRC tissues and its high expression was associated with CRC differentiation, lymph node invasion and AJCC stage. Upregulation of SRSF3 was also associated with shorter overall survival. Knockdown of SRSF3 in CRC cells activated ArhGAP30/Ace-p53 and decreased cell proliferation, migration and survival; while ectopic expression of SRSF3 attenuated ArhGAP30/Ace-p53 and increases cell proliferation, migration and survival. Targeting SRSF3 in xenograft tumors suppressed tumor progression in vivo. CONCLUSIONS: Taken together, our data identify SRSF3 as a regulator for ArhGAP30/Ace-p53 in CRC, and highlight potential prognostic and therapeutic significance of SRSF3 in CRC.

9.
Clin Lab ; 64(7): 1163-1170, 2018 Jul 01.
Article in English | MEDLINE | ID: mdl-30146828

ABSTRACT

BACKGROUND: H. pylori infection has been reported as a risk factor for colorectal adenoma (CRA); however, the clinical results were controversial. Therefore, we performed a meta-analysis to evaluate the association of H. pylori infection and CRA risk. METHODS: A comprehensive literature search for relevant studies published up to November 2017 was performed using Medline and Embase, and the statistical analysis was conducted using Stata software. RESULTS: A total of twenty-five studies including 8,675 cases and 15,275 controls were included in the analysis. The pooled analysis showed that H. pylori infection was associated with an increased risk of CRA (OR = 1.86, 95% CI = 1.55 - 2.23). Subgroup analyses according to the ethnicity, study type, and H. pylori detection method were further conducted. The results showed that H. pylori infection was associated with an increased risk of CRA both in Caucasian (OR = 2.23, 95% CI = 1.36 - 3.66) and Asian population (OR = 1.58, 95% CI = 1.36 - 1.82). Both the case-control studies and cross sectional studies suggested the H. pylori infection could promote the risk of CRA (case control: OR was 2.00, 95% CI = 1.22 - 3.28; cross-sectional: OR was 1.68, 95% CI = 1.43 - 1.99). For H. pylori infection detection methods, there is significant association between H. pylori infection and CRA risk using the serum IgG method and RUT, but not with the UBT and IHC method. CONCLUSIONS: This analysis suggests that H. pylori infection may be a risk factor for CRA.


Subject(s)
Adenoma/diagnosis , Colorectal Neoplasms/diagnosis , Helicobacter Infections/complications , Helicobacter pylori/isolation & purification , Adenoma/chemically induced , Adenoma/ethnology , Asian People/statistics & numerical data , Case-Control Studies , Colorectal Neoplasms/complications , Colorectal Neoplasms/ethnology , Cross-Sectional Studies , Helicobacter Infections/ethnology , Helicobacter Infections/virology , Helicobacter pylori/physiology , Humans , Risk Factors , White People/statistics & numerical data
10.
Clin Lab ; 64(4): 497-505, 2018 Apr 01.
Article in English | MEDLINE | ID: mdl-29739071

ABSTRACT

BACKGROUND: Cluster of differentiation 24 (CD24) has recently been reported as a biomarker for colorectal cancer. However, the clinical and prognostic significance of CD24 in colorectal cancer remains controversial. Therefore, we performed a meta-analysis to clarify this issue. METHODS: A comprehensive literature search was performed using Medline, Embase, Web of Science, and CNKI, and the statistical analysis was conducted using Stata software. RESULTS: A total of thirteen studies including 2,180 cases were included in this meta-analysis. The pooled analysis indicated that CD24 expression was associated with lymph node invasion (RR = 0.71 (negative versus positive), 95% CI = 0.52 - 0.96, p = 0.02, Figure 3), differentiation (RR = 0.81 (well versus poor), 95% CI = 0.67 - 0.99, p = 0.04), and T stage (RR = 0.74 (T1 + T2 versus T3 + T4), 95% CI = 0.65 - 0.85, p = 0.00). The prognosis analysis also suggested CD24 overexpression indicating poorer 5-year OS rate (RR = 0.74, 95% CI = 0.58 - 0.93, p = 0.01) However, CD24 was not associated with other clinicopathological features such as tumor size, tumor grade, distant metastasis, TNM stage and Dukes stage. CONCLUSIONS: Taken together, this meta-analysis suggested that CD24 is an efficient prognostic factor in colorectal cancer.


Subject(s)
Biomarkers, Tumor/genetics , CD24 Antigen/genetics , Colorectal Neoplasms/genetics , Lymph Nodes/metabolism , Biomarkers, Tumor/metabolism , CD24 Antigen/metabolism , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Humans , Lymph Nodes/pathology , Lymphatic Metastasis , Neoplasm Grading , Neoplasm Invasiveness , Neoplasm Staging , Prognosis
11.
Nat Commun ; 9(1): 545, 2018 02 07.
Article in English | MEDLINE | ID: mdl-29416026

ABSTRACT

Reversible post-translational modifications represent a mechanism to control tumor metabolism. Here we show that mitochondrial Sirtuin5 (SIRT5), which mediates lysine desuccinylation, deglutarylation, and demalonylation, plays a role in colorectal cancer (CRC) glutamine metabolic rewiring. Metabolic profiling identifies that deletion of SIRT5 causes a marked decrease in 13C-glutamine incorporation into tricarboxylic-acid (TCA) cycle intermediates and glutamine-derived non-essential amino acids. This reduces the building blocks required for rapid growth. Mechanistically, the direct interaction between SIRT5 and glutamate dehydrogenase 1 (GLUD1) causes deglutarylation and functional activation of GLUD1, a critical regulator of cellular glutaminolysis. Consistently, GLUD1 knockdown diminishes SIRT5-induced proliferation, both in vivo and in vitro. Clinically, overexpression of SIRT5 is significantly correlated with poor prognosis in CRC. Thus, SIRT5 supports the anaplerotic entry of glutamine into the TCA cycle in malignant phenotypes of CRC via activating GLUD1.


Subject(s)
Carcinogenesis/metabolism , Colorectal Neoplasms/metabolism , Gene Expression Regulation, Neoplastic/physiology , Glutamate Dehydrogenase/metabolism , Glutamine/metabolism , Sirtuins/metabolism , Cell Proliferation , Citric Acid Cycle/physiology , Gene Expression Regulation, Enzymologic/physiology , Glutamate Dehydrogenase/genetics , HCT116 Cells , Humans , RNA Interference , Sirtuins/genetics
12.
Oncotarget ; 8(8): 12866-12876, 2017 Feb 21.
Article in English | MEDLINE | ID: mdl-28030817

ABSTRACT

CD44v6 has recently been reported as a biomarker for colorectal cancer. However, the clinical and prognostic significance of CD44v6 in colorectal cancer remains controversial. Therefore, we performed a meta-analysis to clarify this issue. A comprehensive literature search was performed using Medline, Embase and Web of Science, and the statistical analysis was conducted using Stata software. A total of twenty-one studies including 3918 colorectal cancer cases were included. The pooled analysis showed that CD44v6 overexpression in colorectal cancer was an independent prognostic marker correlating with lower 5-year overall survival rate (OR=0.78, 95%CI =0.67-0.91, p=0.001). CD44v6 overexpression was also associated with more lymph node invasion (OR=1.48, 95%CI= 1.02-2.15, p=0.04), and advanced Dukes stage (OR=2.47, 95%CI= 1.29-4.73, p=0.01). In addition, while excluding Zolbec's study, CD44v6 overexpression was associated with distance metastasis (OR=1.65, 95%CI =1.13-2.40, p=0.01). Taken together, this meta-analysis suggested that CD44v6 is an efficient prognostic factor in colorectal cancer.


Subject(s)
Biomarkers, Tumor/analysis , Colorectal Neoplasms/pathology , Hyaluronan Receptors/biosynthesis , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/mortality , Humans , Neoplasm Invasiveness/pathology , Prognosis , Up-Regulation
13.
Future Microbiol ; 10(9): 1433-45, 2015.
Article in English | MEDLINE | ID: mdl-26346930

ABSTRACT

AIMS: To investigate the antitumor effects of probiotics Clostridium butyricum and Bacillus subtilis on colorectal cancer (CRC) progression. MATERIALS & METHODS: The effects of C. butyricum and B. subtilis on CRC cells were studied. Male C57BL/6 mice with 1,2-dimethylhydrazine dihydrochloride (DMH)-induced CRC were intervened by these two probiotics and the antitumor effects were examined by comparing the tumor incidence and detecting the inflammatory and immune-related markers. RESULTS & CONCLUSIONS: C. butyricum and B. subtilis inhibited the proliferation of CRC cells, caused cell cycle arrest and promoted apoptosis. In vivo, these two probiotics inhibited the development of DMH-induced CRC. The molecular mechanism involved reduced inflammation and improved immune homeostasis. This work establishes a basis for the protective role of probiotics B. subtilis and C. butyricum in intestinal tumorigenesis.


Subject(s)
Bacillus subtilis/physiology , Carcinogenesis , Clostridium butyricum/physiology , Colorectal Neoplasms/prevention & control , Probiotics , 1,2-Dimethylhydrazine , Administration, Oral , Animals , Apoptosis , Cell Cycle Checkpoints , Cell Line, Tumor , Colorectal Neoplasms/chemically induced , Culture Media, Conditioned , Disease Progression , Homeostasis , Inflammation/therapy , Male , Mice, Inbred C57BL
14.
Oncotarget ; 5(8): 2230-42, 2014 Apr 30.
Article in English | MEDLINE | ID: mdl-24809982

ABSTRACT

Increasing evidence suggests long non-coding RNAs (lncRNAs) are frequently aberrantly expressed in cancers, however, few related lncRNA signatures have been established for prediction of cancer prognosis. We aimed to develop a lncRNA signature to improve prognosis prediction of colorectal cancer (CRC). Using a lncRNA-mining approach, we performed lncRNA expression profiling in large CRC cohorts from Gene Expression Ominus (GEO), including GSE39582 test series(N=436), internal validation series (N=117); and two independent validation series GSE14333 (N=197) and GSE17536(N=145). We established a set of six lncRNAs that were significantly correlated with the disease free survival (DFS) in the test series. Based on this six-lncRNA signature, the test series patients could be classified into high-risk and low-risk subgroups with significantly different DFS (HR=2.670; P<0.0001). The prognostic value of this six-lncRNA signature was confirmed in the internal validation series and another two independent CRC sets. Gene set enrichment analysis (GSEA) analysis suggested that risk score positively correlated with several cancer metastasis related pathways. Functional experiments demonstrated three dysregulated lncRNAs, AK123657, BX648207 and BX649059 were required for efficient invasion and proliferation suppression in CRC cell lines. Our results might provide an efficient classification tool for clinical prognosis evaluation of CRC.


Subject(s)
Colorectal Neoplasms/genetics , Colorectal Neoplasms/mortality , RNA, Long Noncoding/genetics , Transcriptome , Disease-Free Survival , Female , Humans , Kaplan-Meier Estimate , Male , Prognosis , Proportional Hazards Models
15.
Cancer Prev Res (Phila) ; 7(6): 607-16, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24691499

ABSTRACT

microRNAs (miRNA) are promising predictors in colorectal cancer (CRC). We investigated whether miRNAs could predict adenoma recurrence in patients with advanced colorectal adenoma (ACRA) after polypectomy. miRNA expression profiling was performed by miRNA microarray to identify recurrence-related miRNAs. Candidate miRNAs extracted from formalin-fixed paraffin-embedded blocks of patients with ACRA were measured using real-time PCR. Logistic regression analysis was conducted to investigate whether validated miRNA expression profiles were independent from other known adenoma recurrence risk factors. The prognostic values of six miRNAs and three independent risk factors were assessed by the area under the receiver operating characteristic (ROC) curve analysis. The expressions of six candidate miRNAs were significantly decreased from levels in normal colorectal tissue compared with ARCA with adenoma recurrence (RACRA) in this retrospective cohort. However, only miRNA (miR)-194 emerged as a practical predictor. The sensitivity and specificity of miR-194 as a predictor were 71.0% and 78.0%, respectively, at a cutoff value of 0.1311 in the retrospective cohort. Sensitivity and specificity were 76.1% and 77.2%, respectively, in the prospective cohort using the same cutoff value. Low expression levels of miR-194, adenoma size ≥2 cm, and ≥3 adenomas were independent risk factors for adenoma recurrence. Moreover, low expression of miR-194 was a better predictor of adenoma recurrence than the adenoma size and numbers according to ROC curve analysis. miR-194 may be an independent predictor for adenoma recurrence in patients with ACRA after polypectomy.


Subject(s)
Adenoma/diagnosis , Adenoma/surgery , Biomarkers, Tumor/physiology , Colorectal Neoplasms/diagnosis , Colorectal Neoplasms/surgery , MicroRNAs/physiology , Adenoma/pathology , Adult , Aged , Aged, 80 and over , Colorectal Neoplasms/pathology , Digestive System Surgical Procedures , Disease Progression , Female , Humans , Intestinal Polyps/diagnosis , Intestinal Polyps/pathology , Intestinal Polyps/surgery , Male , Middle Aged , Prognosis , Recurrence , Treatment Outcome
16.
Sci Rep ; 4: 3648, 2014 Jan 13.
Article in English | MEDLINE | ID: mdl-24413317

ABSTRACT

MicroRNAs (miRNAs) participate in diverse biological pathways and may act as oncogenes or tumor suppressors. Single nucleotide polymorphisms (SNPs) in miRNAs (MirSNPs) might promote carcinogenesis by affecting miRNA function and/or maturation; however, the association between MirSNPs reported and cancer risk remain inconsistent. Here, we investigated the association between nine common MirSNPs and cancer risk using data from large scale case-control studies. Eight precursor-miRNA (pre-miRNA) SNPs (rs2043556/miR-605, rs3746444/miR-499a/b, rs4919510/miR-608, rs2910164/miR-146a, rs11614913/miR-196a2, rs895819/miR-27a, rs2292832/miR-149, rs6505162/miR-423) and one primary-miRNA (pri-miRNA) SNP (rs1834306/miR-100) were analyzed in 16399 cases and 21779 controls from seven published studies in eight common cancers. With a novel statistic, Cross phenotype meta-analysis (CPMA) of the association of MirSNPs with multiple phenotypes indicated rs2910164 C (P = 1.11E-03), rs2043556 C (P = 0.0165), rs6505162 C (P = 2.05E-03) and rs895819 (P = 0.0284) were associated with a significant overall risk of cancer. In conclusion, MirSNPs might affect an individual's susceptibility to various types of cancer.


Subject(s)
MicroRNAs/genetics , Neoplasms/genetics , Polymorphism, Single Nucleotide , Case-Control Studies , Genetic Association Studies , Genetic Predisposition to Disease , Humans , Odds Ratio , Publication Bias , Risk
17.
Apoptosis ; 19(4): 643-56, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24327257

ABSTRACT

The development of gastric cancer (GC) is a complex multistep process, including numerous genetic and epigenetic changes. CD24 is associated with enhanced invasiveness of GC and a poor prognosis. However, the mechanism by which CD24 induces GC progression remains poorly characterized. Here, we found that the expression of CD24 gradually increased in samples of normal gastric mucosa, non-atrophic chronic gastritis, chronic atrophic gastritis (CAG), CAG with intestinal metaplasia, dysplasia and GC. Moreover, the knockdown of CD24 induced significant levels of apoptosis in GC cells via the mitochondrial apoptotic pathway. CD24 may also promote cellular invasion and regulate the expression of E-cadherin, fibronectin and vitamin D receptor in GC cells. The activation of signal transducer and activator of transcription 3 (STAT3) may mediate CD24-induced GC survival and invasion in vitro. Furthermore, CD24-induced GC progression and STAT3 activation could also be detected in vivo and in clinical GC tissues samples. Taken together, our results indicate that CD24 mediates gastric carcinogenesis and may promote GC progression by suppressing apoptosis and promoting invasion, with the activation of STAT3 playing a critical role.


Subject(s)
CD24 Antigen/metabolism , Carcinogenesis/metabolism , STAT3 Transcription Factor/metabolism , Stomach Neoplasms/metabolism , Adult , Aged , Animals , Apoptosis , CD24 Antigen/genetics , Carcinogenesis/pathology , Caspase 3/metabolism , Cell Line, Tumor , Cell Proliferation , Cell Survival , Chronic Disease , Disease Progression , Female , Gastritis/metabolism , Heterografts , Humans , Intestinal Mucosa/metabolism , Male , Metaplasia , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Mitochondria/metabolism , Neoplasm Transplantation , Stomach Neoplasms/pathology
18.
Int J Cancer ; 134(9): 2030-40, 2014 May 01.
Article in English | MEDLINE | ID: mdl-24127267

ABSTRACT

Persistent infection with Helicobacter pylori (H. pylori) contributes to gastric diseases including chronic gastritis and gastric cancer. However, the pathogenesis of this carcinogenic bacterium has not been completely elucidated. Here, we report that H. pylori rapidly triggers STAT3 signaling and induces STAT3-dependent COX-2 expression both in vitro and in vivo. STAT3 upregulates COX-2 by binding to and increasing the activity of COX-2 promoter. COX-2 in turn regulates IL-6/STAT3 signaling under basal conditions and during H. pylori infection. These findings suggest that a positive feedback loop between STAT3 and COX-2 exists in the basal condition and H. pylori infectious condition. Immunohistochemical staining revealed that H. pylori-positive gastritis tissues exhibited markedly higher levels of pSTAT3(Tyr705) than H. pylori-negative ones. High pSTAT3(Tyr705) levels are correlated with intestinal metaplasia and dysplasia, suggesting pSTAT3(Tyr705) may be useful in the early detection of gastric tumorigenesis. Additionally, a strong positive correlation between STAT3/pSTAT3(Tyr705) levels and COX-2 expression was identified in gastritis and gastric cancer tissues. Together, these findings provide new evidence for a positive feedback loop between STAT3 signaling and COX-2 in H. pylori pathogenesis and may lead to new approaches for early detection and effective therapy of gastric cancer


Subject(s)
Carcinogenesis/metabolism , Cyclooxygenase 2/biosynthesis , Helicobacter Infections/metabolism , STAT3 Transcription Factor/metabolism , Stomach Neoplasms/metabolism , Animals , Blotting, Western , Carcinogenesis/genetics , Cell Line , Chromatin Immunoprecipitation , Cyclooxygenase 2/genetics , Enzyme-Linked Immunosorbent Assay , Feedback, Physiological , Gastritis/genetics , Gastritis/metabolism , Gastritis/pathology , Gene Expression Regulation, Neoplastic/physiology , Gerbillinae , Helicobacter Infections/complications , Helicobacter pylori , Humans , Microscopy, Confocal , Precancerous Conditions/genetics , Precancerous Conditions/metabolism , Precancerous Conditions/pathology , RNA, Small Interfering , Real-Time Polymerase Chain Reaction , Signal Transduction/physiology , Stomach Neoplasms/genetics , Stomach Neoplasms/microbiology
19.
J Natl Cancer Inst ; 105(22): 1738-49, 2013 Nov 20.
Article in English | MEDLINE | ID: mdl-24104608

ABSTRACT

BACKGROUND: The molecular mechanisms that control the aggressiveness of gastric cancer (GC) remain poorly defined. Here we show that synbindin contributes to the aggressiveness of GC by activating extracellular signal-regulated protein kinase (ERK) signaling on the Golgi apparatus. METHODS: Expression of synbindin was examined in normal gastric mucosa (n = 44), intestinal metaplastic gastric mucosa (n = 66), and GC tissues (n=52), and the biological effects of synbindin on tumor growth and ERK signaling were detected in cultured cells, nude mice, and human tissue samples. The interaction between synbindin and mitogen-activated protein kinase kinase (MEK1)/ERK was determined by immunofluorescence and fluorescence resonance energy transfer assays. The transactivation of synbindin by nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) was detected using luciferase reporter assay and chromatin immunoprecipitation. RESULTS: High expression of synbindin was associated with larger tumor size (120.8 vs 44.8 cm(3); P = .01), advanced tumor node metastasis (TNM) stage (P = .003), and shorter patient survival (hazard ratio = 1.51; 95% confidence interval [CI] = 1.01 to 2.27; P = .046). Synbindin promotes cell proliferation and invasion by activating ERK2 on the Golgi apparatus, and synbindin is directly transactivated by NF-κB. Synbindin expression level was statistically significantly higher in human GCs with activated ERK2 than those with low ERK2 activity (intensity score of 11.5, 95% CI = 10.4 to 12.4 vs intensity score of 4.6, 95% CI 3.9 to 5.3; P < .001). Targeting synbindin in xenograft tumors decreased ERK2 phosphorylation and statistically significantly reduced tumor volume (451.2mm(3), 95% CI = 328.3 to 574.1 vs 726.1mm(3), 95% CI = 544.2 to 908.2; P = .01). CONCLUSIONS: Synbindin contributes to malignant phenotypes of GC by activating ERK on the Golgi, and synbindin is a potential biomarker and therapeutic target for GC.


Subject(s)
Golgi Apparatus/metabolism , MAP Kinase Kinase 1/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , NF-kappa B/metabolism , Nerve Tissue Proteins/metabolism , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Vesicular Transport Proteins/metabolism , Animals , Cell Line, Tumor , Chromatin Immunoprecipitation , Flow Cytometry , Fluorescence Resonance Energy Transfer , Fluorescent Antibody Technique , Gastric Mucosa/metabolism , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Golgi Apparatus/enzymology , Heterografts , Humans , Kaplan-Meier Estimate , Luciferases , Luminescent Agents , MAP Kinase Signaling System , Mice , Mice, Nude , Nerve Tissue Proteins/genetics , Odds Ratio , Phosphorylation , Protein Array Analysis , Retrospective Studies , Stomach Neoplasms/enzymology , Stomach Neoplasms/mortality , Transcriptional Activation , Up-Regulation , Vesicular Transport Proteins/genetics
20.
Nutr Cancer ; 65(8): 1171-83, 2013.
Article in English | MEDLINE | ID: mdl-24099273

ABSTRACT

The mechanism by which butyrate prevents colorectal cancer (CRC) is unclear. The objective of this study was to identify potential target genes of butyrate in 1,2-dimethylhydrazine (DMH)-induced CRC in mice. Nontumor colorectal tissues of mice from DMH + butyrate, DMH, and control groups were hybridized on Agilent Mouse Whole Genome 44K Oligo Microarrays. Selected genes were validated by qRT-PCR. Data was further analyzed by KEGG, gene ontology (GO), and pathway studio software. The tumor incidence in the DMH + butyrate and DMH groups was 30% and 90%, respectively (P < 0.05). There were 355 genes downregulated due to DMH treatment while upregulated by butyrate, and 475 genes upregulated by DMH while downregulated by butyrate. The results revealed that most of the tumor-related signaling pathways (e.g., MAPK pathway, Wnt pathway, insulin pathway, and VEGF pathway) were downregulated by butyrate. The GO terms related to cell differentiation, cell cycle, cell proliferation, cell death, cell adhesion, and cell migration were significantly affected. The chemopreventive effects of butyrate were confirmed in the DMH-induced CRC mice model. And mechanisms encompassing multiple pathways and GO terms are involved in the regulation of gene expression.


Subject(s)
Butyrates/pharmacology , Colorectal Neoplasms/genetics , Gene Expression Regulation, Neoplastic , 1,2-Dimethylhydrazine/toxicity , Animals , Cell Adhesion/drug effects , Cell Differentiation/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Colorectal Neoplasms/chemically induced , Disease Models, Animal , Down-Regulation/drug effects , Female , Gene Expression Profiling , Genes, Neoplasm , Mice , Mice, Inbred ICR , Microarray Analysis , Signal Transduction/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...