Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 120
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 170(3): 548-563.e16, 2017 Jul 27.
Article in English | MEDLINE | ID: mdl-28753429

ABSTRACT

Gut microbiota are linked to chronic inflammation and carcinogenesis. Chemotherapy failure is the major cause of recurrence and poor prognosis in colorectal cancer patients. Here, we investigated the contribution of gut microbiota to chemoresistance in patients with colorectal cancer. We found that Fusobacterium (F.) nucleatum was abundant in colorectal cancer tissues in patients with recurrence post chemotherapy, and was associated with patient clinicopathological characterisitcs. Furthermore, our bioinformatic and functional studies demonstrated that F. nucleatum promoted colorectal cancer resistance to chemotherapy. Mechanistically, F. nucleatum targeted TLR4 and MYD88 innate immune signaling and specific microRNAs to activate the autophagy pathway and alter colorectal cancer chemotherapeutic response. Thus, F. nucleatum orchestrates a molecular network of the Toll-like receptor, microRNAs, and autophagy to clinically, biologically, and mechanistically control colorectal cancer chemoresistance. Measuring and targeting F. nucleatum and its associated pathway will yield valuable insight into clinical management and may ameliorate colorectal cancer patient outcomes.


Subject(s)
Autophagy , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Fusobacterium nucleatum/physiology , Gastrointestinal Microbiome , Animals , Antineoplastic Agents/therapeutic use , Capecitabine/therapeutic use , Colorectal Neoplasms/metabolism , Drug Resistance, Neoplasm , Heterografts , Mice , MicroRNAs/metabolism , Neoplasm Transplantation , Platinum Compounds/therapeutic use , Recurrence , Toll-Like Receptors/metabolism , Tumor Microenvironment
2.
Gut ; 73(2): 268-281, 2024 Jan 05.
Article in English | MEDLINE | ID: mdl-37734910

ABSTRACT

BACKGROUND AND AIMS: Deregulation of RNA N6-methyladenosine (m6A) modification in intestinal epithelial cells (IECs) influences intestinal immune cells and leads to intestinal inflammation. We studied the function of fat mass-and obesity-associated protein (FTO), one of the m6A demethylases, in patients with ulcerative colitis (UC). METHODS: We analysed colon tissues of Ftoflox/flox; Villin-cre mice and their Ftoflox/flox littermates with dextran sulfate sodium (DSS) using real-time PCR and 16s rRNA sequencing. RNA and methylated RNA immunoprecipitation sequencing were used to analyse immunocytes and IECs. Macrophages were treated with conditioned medium of FTO-knockdown MODE-K cells or sphingosine-1-phosphate (S1P) and analysed for gene expression. Liquid chromatograph mass spectrometry identified C16-ceramide. RESULTS: FTO downregulation was identified in our in-house cohort and external cohorts of UC patients. Dysbiosis of gut microbiota, increased infiltration of proinflammatory macrophages, and enhanced differentiation of Th17 cells were observed in Ftoflox/flox;Villin-cre mice under DSS treatment. FTO deficiency resulted in an increase in m6A modification and a decrease in mRNA stability of CerS6, the gene encoding ceramide synthetase, leading to the downregulation of CerS6 and the accumulation of S1P in IECs. Subsequentially, the secretion of S1P by IECs triggered proinflammatory macrophages to secrete serum amyloid A protein 1/3, ultimately inducing Th17 cell differentiation. In addition, through bioinformatic analysis and experimental validation, we identified UC patients with lower FTO expression might respond better to vedolizumab treatment. CONCLUSIONS: FTO downregulation promoted UC by decreasing CerS6 expression, leading to increased S1P accumulation in IECs and aggravating colitis via m6A-dependent mechanisms. Lower FTO expression in UC patients may enhance their response to vedolizumab treatment.


Subject(s)
Colitis, Ulcerative , Colitis , Humans , Animals , Mice , Colitis, Ulcerative/metabolism , RNA, Ribosomal, 16S/metabolism , Intestinal Mucosa/metabolism , Colitis/chemically induced , Colitis/genetics , Colon/metabolism , Sphingolipids/metabolism , Dextran Sulfate , Disease Models, Animal , Mice, Inbred C57BL , Alpha-Ketoglutarate-Dependent Dioxygenase FTO/genetics , Alpha-Ketoglutarate-Dependent Dioxygenase FTO/metabolism
3.
Am J Respir Crit Care Med ; 206(12): 1534-1545, 2022 12 15.
Article in English | MEDLINE | ID: mdl-35819321

ABSTRACT

Rationale: Previous genetic studies of obstructive sleep apnea (OSA) have limitations in terms of precise case definition, integrated quantitative traits, and interpretation of genetic functions; thus, the heritability of OSA remains poorly explained. Objectives: To identify novel genetic variants associated with OSA and objective sleep-related traits and to explore their functional roles. Methods: A genome-wide association study was performed in 20,590 Han Chinese individuals (5,438 OSA and 15,152 control samples). Human samples and point mutation knockin mice were used for follow-up investigation of gene functions. Measurements and Main Results: Two characteristic study-wide significant loci (P < 2.63 × 10-9) for OSA were identified: the PACRG intronic variant rs6455893 on 6q26 (odds ratio [OR] = 1.62; 95% confidence interval [CI], 1.39-1.89; P = 6.98 × 10-10) and the missense variant rs3746804 (p.Pro267Leu) in the riboflavin transporter SLC52A3 on 20p13 (OR = 0.83; 95% CI, 0.79-0.88; P = 7.57 × 10-10). In addition, 18 genome-wide significant loci associated with quantitative OSA and objective sleep-related traits were identified, 5 of which exceeded the study-wide significance threshold. Rs3746804 was associated with elevated serum riboflavin concentrations, and the corresponding mutation in mice increased riboflavin concentrations, suggesting that this variant may facilitate riboflavin uptake and riboflavin-dependent physiological activity. Conclusions: We identified several novel genome-wide significant loci associated with OSA and objective sleep-related traits. Our findings provide insight into the genetic architecture of OSA and suggest that SLC52A3 might be a therapeutic target, whereas riboflavin might be a therapeutic agent.


Subject(s)
Genome-Wide Association Study , Sleep Apnea, Obstructive , Animals , Humans , Mice , East Asian People , Membrane Transport Proteins/genetics , Microfilament Proteins/genetics , Molecular Chaperones/genetics , Riboflavin , Sleep , Sleep Apnea, Obstructive/genetics
4.
Gastroenterology ; 161(5): 1552-1566.e12, 2021 11.
Article in English | MEDLINE | ID: mdl-34371001

ABSTRACT

BACKGROUND & AIMS: Enterotoxigenic Bacteroides fragilis (ETBF) is strongly associated with the occurrence of inflammatory bowel disease (IBD), colitis-associated colorectal cancer, and colorectal cancer (CRC). However, the mechanism of ETBF-induced intestinal inflammation and tumorigenesis remains unclear. METHODS: microRNA sequencing was used to detect the differentially expressed microRNAs in both ETBF-treated cells and exosomes derived from ETBF-inoculated cells. Cell Counting Kit 8 assays were used to evaluate the effect of ETBF and exosomes on CRC cell proliferation. The biological role and mechanism of ETBF-mediated miR-149-3p in colitis and colon carcinogenesis were determined both in vitro and in vivo. RESULTS: ETBF promoted CRC cell proliferation by down-regulating miR-149-3p both in vitro and in vivo. ETBF-down-regulated miR-149-3p depended on METTL14-mediated N6-methyladenosine methylation. As the target gene of miR-149-3p, PHF5A transactivated SOD2 through regulating KAT2A messenger RNA alternative splicing after ETBF treatment in CRC cells. miR-149-3p could be released in exosomes and mediated intercellular communication by modulating T-helper type 17 cell differentiation. The level of plasma exosomal miR-149-3p was gradually decreased from healthy control individuals to patients with IBD and CRC. miR-149-3p, existing in plasma exosomes, negatively correlated with the abundance of ETBF in patients with IBD and CRC. CONCLUSIONS: Exosomal miR-149-3p derived from ETBF-treated cells facilitated T-helper type 17 cell differentiation. ETBF-induced colorectal carcinogenesis depended on down-regulating miR-149-3p and further promoting PHF5A-mediated RNA alternative splicing of KAT2A in CRC cells. Targeting the ETBF/miR-149-3p pathway presents a promising approach to treat patients with intestinal inflammation and CRC with a high amount of ETBF.


Subject(s)
Bacteroides fragilis/pathogenicity , Colitis, Ulcerative/microbiology , Colon/microbiology , Colorectal Neoplasms/microbiology , Crohn Disease/microbiology , Exosomes/microbiology , MicroRNAs/metabolism , Animals , Cell Differentiation , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Colitis, Ulcerative/genetics , Colitis, Ulcerative/metabolism , Colitis, Ulcerative/pathology , Colon/metabolism , Colon/pathology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Crohn Disease/genetics , Crohn Disease/metabolism , Crohn Disease/pathology , Disease Models, Animal , Exosomes/genetics , Exosomes/metabolism , HCT116 Cells , Histone Acetyltransferases/genetics , Histone Acetyltransferases/metabolism , Host-Pathogen Interactions , Humans , Methyltransferases/genetics , Methyltransferases/metabolism , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , MicroRNAs/genetics , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Th17 Cells/immunology , Th17 Cells/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism
5.
Gut ; 70(11): 2123-2137, 2021 11.
Article in English | MEDLINE | ID: mdl-33318144

ABSTRACT

OBJECTIVE: Microbiota disorder promotes chronic inflammation and carcinogenesis. High glycolysis is associated with poor prognosis in patients with colorectal cancer (CRC). However, the potential correlation between the gut microbiota and glucose metabolism is unknown in CRC. DESIGN: 18F-FDG (18F-fluorodeoxyglucose) PET (positron emission tomography)/CT image scanning data and microbiota PCR analysis were performed to measure the correlation between metabolic alterations and microbiota disorder in 33 patients with CRC. Multiple colorectal cancer models, metabolic analysis and Seahorse assay were established to assess the role of long non-coding RNA (lncRNA) enolase1-intronic transcript 1 (ENO1-IT1) in Fusobacterium (F.) nucleatum-induced glucose metabolism and colorectal carcinogenesis. RNA immunoprecipitation and chromatin immunoprecipitation sequencing were conducted to identify potential targets of lncRNA ENO1-IT1. RESULTS: We have found F. nucleatum abundance correlated with high glucose metabolism in patients with CRC. Furthermore, F. nucleatum supported carcinogenesis via increasing CRC cell glucose metabolism. Mechanistically, F. nucleatum activated lncRNA ENO1-IT1 transcription via upregulating the binding efficiency of transcription factor SP1 to the promoter region of lncRNA ENO1-IT1. Elevated ENO1-IT behaved as a guider modular for KAT7 histone acetyltransferase, specifying the histone modification pattern on its target genes, including ENO1, and consequently altering CRC biological function. CONCLUSION: F. nucleatum and glucose metabolism are mechanistically, biologically and clinically connected to CRC. Targeting ENO1 pathway may be meaningful in treating patients with CRC with elevated F. nucleatum.


Subject(s)
Carcinogenesis/genetics , Colorectal Neoplasms/genetics , Fusobacterium Infections/genetics , Glycolysis/genetics , RNA, Long Noncoding/genetics , Animals , Biomarkers, Tumor , Colorectal Neoplasms/diagnostic imaging , DNA-Binding Proteins , Fluorodeoxyglucose F18/pharmacokinetics , Fusobacterium nucleatum , Gastrointestinal Microbiome , Gene Expression Regulation, Neoplastic , Histone Acetyltransferases , Humans , Mice , Phosphopyruvate Hydratase , Positron Emission Tomography Computed Tomography , Prognosis , Radiopharmaceuticals/pharmacokinetics , Signal Transduction , Tumor Cells, Cultured , Tumor Suppressor Proteins
6.
Mol Cancer ; 19(1): 72, 2020 04 03.
Article in English | MEDLINE | ID: mdl-32245489

ABSTRACT

BACKGROUND: Epigenetic alterations are involved in various aspects of colorectal carcinogenesis. N6-methyladenosine (m6A) modifications of RNAs are emerging as a new layer of epigenetic regulation. As the most abundant chemical modification of eukaryotic mRNA, m6A is essential for the regulation of mRNA stability, splicing, and translation. Alterations of m6A regulatory genes play important roles in the pathogenesis of a variety of human diseases. However, whether this mRNA modification participates in the glucose metabolism of colorectal cancer (CRC) remains uncharacterized. METHODS: Transcriptome-sequencing and liquid chromatography-tandem mass spectrometry (LC-MS) were performed to evaluate the correlation between m6A modifications and glucose metabolism in CRC. Mass spectrometric metabolomics analysis, in vitro and in vivo experiments were conducted to investigate the effects of METTL3 on CRC glycolysis and tumorigenesis. RNA MeRIP-sequencing, immunoprecipitation and RNA stability assay were used to explore the molecular mechanism of METTL3 in CRC. RESULTS: A strong correlation between METTL3 and 18F-FDG uptake was observed in CRC patients from Xuzhou Central Hospital. METTL3 induced-CRC tumorigenesis depends on cell glycolysis in multiple CRC models. Mechanistically, METTL3 directly interacted with the 5'/3'UTR regions of HK2, and the 3'UTR region of SLC2A1 (GLUT1), then further stabilized these two genes and activated the glycolysis pathway. M6A-mediated HK2 and SLC2A1 (GLUT1) stabilization relied on the m6A reader IGF2BP2 or IGF2BP2/3, respectively. CONCLUSIONS: METTL3 is a functional and clinical oncogene in CRC. METTL3 stabilizes HK2 and SLC2A1 (GLUT1) expression in CRC through an m6A-IGF2BP2/3- dependent mechanism. Targeting METTL3 and its pathway offer alternative rational therapeutic targets in CRC patients with high glucose metabolism.


Subject(s)
Adenosine/analogs & derivatives , Colorectal Neoplasms/pathology , Epigenesis, Genetic , Glucose Transporter Type 1/metabolism , Glycolysis , Hexokinase/metabolism , Methyltransferases/metabolism , Adenosine/chemistry , Animals , Apoptosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Proliferation , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , DNA Methylation , Disease Progression , Gene Expression Regulation, Neoplastic , Glucose Transporter Type 1/genetics , Hexokinase/genetics , Humans , Methyltransferases/genetics , Mice , Mice, Nude , Prognosis , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Survival Rate , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
7.
Int J Cancer ; 145(1): 206-220, 2019 07 01.
Article in English | MEDLINE | ID: mdl-30561033

ABSTRACT

The molecular mechanisms that control the development of colorectal cancer (CRC) remain poorly defined. Here we show Synbindin promoted CRC oncogenesis by activating Wnt signaling and altering gut microbiome. Synbindin upregulation in human CRCs was associated with poor patient prognosis. Intestine-specific disruption of Synbindin balanced the disturbed gut microbiota and protected mice against tumor formation in the colitis-associated cancer (CAC) model. The protective role was compromised after gut microbiota depletion. In host, increased goblet cells and mucin2 expression, together with increased intestinal epithelial cells (IECs) apoptosis and decreased epithelial proliferation were observed. Further transcriptomic sequencing identified Wnt signaling a major regulatory node downstream of Synbindin. Combined molecular and cellular characterizations revealed that Synbindin confers Disheveled-3 (DVL3)-based signalosome assembly and acts as a modular scaffold for DVL3 and Axin2 complex, orchestrating the intensity of Wnt signaling. These findings identify a critical role of Synbindin in gut microbiome composition and Wnt signaling activation in colorectal carcinogenesis, and highlight Synbindin as an adaptor protein with multifaceted roles.


Subject(s)
Colorectal Neoplasms/metabolism , Colorectal Neoplasms/microbiology , Gastrointestinal Microbiome/physiology , Nerve Tissue Proteins/deficiency , Vesicular Transport Proteins/deficiency , Wnt Signaling Pathway , Animals , Axin Protein/metabolism , Carcinogenesis , Colitis/chemically induced , Colitis/metabolism , Colitis/microbiology , Colitis/pathology , Colorectal Neoplasms/pathology , Dextran Sulfate , Dishevelled Proteins/metabolism , HCT116 Cells , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Microbiota/physiology , Mucin-2/metabolism , Nerve Tissue Proteins/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Vesicular Transport Proteins/metabolism
8.
Gut ; 67(3): 534-541, 2018 03.
Article in English | MEDLINE | ID: mdl-28213609

ABSTRACT

OBJECTIVE: A close relationship between gut microbiota and some chronic liver disorders has recently been described. Herein, we systematically performed a comparative analysis of the gut microbiome in primary biliary cholangitis (PBC) and healthy controls. DESIGN: We first conducted a cross-sectional study of 60 ursodeoxycholic acid (UDCA) treatment-naïve patients with PBC and 80 matched healthy controls. Second, an independent cohort composed of 19 treatment-naïve patients and 34 controls was used to validate the results. Finally, a prospective study was performed in a subgroup of 37 patients with PBC who underwent analysis before and after 6 months of UDCA treatment. Faecal samples were collected, and microbiomes were analysed by 16S ribosomal RNA gene sequencing. RESULTS: A significant reduction of within-individual microbial diversity was noted in PBC (p=0.03). A signature defined by decreased abundance of four genera and increased abundance of eight genera strongly correlated with PBC (area under curve=0.86, 0.84 in exploration and validation data, respectively). Notably, the abundance of six PBC-associated genera was reversed after 6 months of UDCA treatment. In particular, Faecalibacterium, enriched in controls, was further decreased in gp210-positive than gp210-negative patients (p=0.002). Of interest was the finding that the increased capacity for the inferred pathway, bacterial invasion of epithelial cells in PBC, highly correlated with the abundance of bacteria belonging to Enterobacteriaceae. CONCLUSIONS: This study presents a comprehensive landscape of gut microbiota in PBC. Dysbiosis was found in the gut microbiome in PBC and partially relieved by UDCA. Our study suggests that gut microbiota is a potential therapeutic target and diagnostic biomarker for PBC.


Subject(s)
Bacteria , Cholagogues and Choleretics/therapeutic use , Cholangitis/microbiology , Dysbiosis/microbiology , Gastrointestinal Microbiome , Ursodeoxycholic Acid/therapeutic use , Adult , Aged , Antibodies/blood , Biomarkers , Case-Control Studies , Cholangitis/complications , Cholangitis/drug therapy , Cross-Sectional Studies , Dysbiosis/complications , Feces/microbiology , Female , Gastrointestinal Microbiome/drug effects , Humans , Male , Middle Aged , Nuclear Pore Complex Proteins/immunology , Prospective Studies , Young Adult
9.
Int J Cancer ; 143(1): 80-87, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29388190

ABSTRACT

The autophagy lysosome pathway is essential to maintain cell viability and homeostasis in response to many stressful environments, which is reported to play a vital role in cancer development and therapy. However, the association of genetic alterations of this pathway with risk of cancer remains unclear. Based on genome-wide association study data of eight kinds of cancers, we used an adaptive rank truncated product approach to perform a pathway-level and gene-level analysis, and used a logistic model to calculate SNP-level associations to examine whether an altered autophagy lysosome pathway contributes to cancer susceptibility. Among eight kinds of cancers, four of them showed significant statistics in the pathway-level analysis, including breast cancer (p = 0.00705), gastric cancer (p = 0.00880), lung cancer (p = 0.000100) and renal cell carcinoma (p = 0.00190). We also found that some autophagy lysosome genes had signals of association with cancer risk. Our results demonstrated that inherited genetic variants in the overall autophagy lysosome pathway and certain associated genes might contribute to cancer susceptibility, which warrant further evaluation in other independent datasets.


Subject(s)
Gene Regulatory Networks , Genome-Wide Association Study/methods , Lysosomes/genetics , Neoplasms/genetics , Polymorphism, Single Nucleotide , Autophagy , Breast Neoplasms/genetics , Carcinoma, Renal Cell/genetics , Female , Genetic Predisposition to Disease , Humans , Lung Neoplasms/genetics , Male , Stomach Neoplasms/genetics
10.
Mol Carcinog ; 57(7): 911-925, 2018 07.
Article in English | MEDLINE | ID: mdl-29573464

ABSTRACT

Kinesin family member 20B (KIF20B) has been reported to have an oncogenic role in bladder and hepatocellular cancer cells, but its role in colorectal cancer (CRC) progression remains unclear. In this study, we assessed the mRNA and protein levels of KIF20B in CRC tissues using qRT-PCR and immunohistochemistry, respectively. KIF20B was overexpressed in CRC tissues and was associated with cancer invasion and metastasis. Mechanistically, KIF20B overexpression promoted the epithelial-mesenchymal transition (EMT) process mediated by glioma-associated oncogene 1 (Gli1) as well as CRC cell migration and invasion. Interestingly, KIF20B was localized in pseudopod protrusions of CRC cells and influenced the formation of cell protrusions, especially the EMT-related invadopodia. Moreover, intracellular actin dynamic participated in the modulation of the Gli1-mediated EMT and EMT-related cell pseudopod protrusion formation induced by KIF20B. We identified a role for KIF20B in CRC progression and revealed a correlation between KIF20B expression in CRC tissues and patient prognosis. The underlying mechanism was associated with the Gli1-mediated EMT and EMT-related cell protrusion formation modulated by intracellular actin dynamic. Thus, KIF20B may be a potential biomarker and promising treatment target for CRC.


Subject(s)
Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Epithelial-Mesenchymal Transition/genetics , Kinesins/genetics , Zinc Finger Protein GLI1/genetics , Actins/genetics , Aged , Cell Line, Tumor , Cell Movement/genetics , Female , Gene Expression Regulation, Neoplastic/genetics , Humans , Male , Prognosis
11.
Mol Cancer ; 15(1): 60, 2016 09 20.
Article in English | MEDLINE | ID: mdl-27647437

ABSTRACT

BACKGROUND: 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 at developing alncRNA signature to improve prognosis prediction of gastric cancer (GC). METHODS: Using a lncRNA-mining approach, we performed lncRNA expression profiling in large GC cohorts from Gene Expression Ominus (GEO), including GSE62254 data set (N = 300) and GSE15459 data set (N = 192). We established a set of 24-lncRNAs that were significantly associated with the disease free survival (DFS) in the test series. RESULTS: Based on this 24-lncRNA signature, the test series patients could be classified into high-risk or low-risk subgroup with significantly different DFS (HR = 1.19, 95 % CI = 1.13-1.25, P < 0.0001). The prognostic value of this 24-lncRNA signature was confirmed in the internal validation series and another external validation series, respectively. Further analysis revealed that the prognostic value of this signature was independent of lymph node ratio (LNR) and postoperative chemotherapy. Gene set enrichment analysis (GSEA) indicated that high risk score group was associated with several cancer recurrence and metastasis associated pathways. CONCLUSIONS: The identification of the prognostic lncRNAs indicates the potential roles of lncRNAs in GC biogenesis. Our results may provide an efficient classification tool for clinical prognosis evaluation of GC.


Subject(s)
Biomarkers, Tumor/genetics , Gene Expression Profiling/methods , RNA, Long Noncoding/genetics , Stomach Neoplasms/genetics , Disease-Free Survival , Female , Gene Expression Regulation, Neoplastic , Humans , Kaplan-Meier Estimate , Male , Prognosis
12.
Thorax ; 71(4): 347-55, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26883674

ABSTRACT

BACKGROUND: Dyslipidaemia is an intermediary exacerbation factor for various diseases but the impact of obstructive sleep apnoea (OSA) on dyslipidaemia remains unclear. METHODS: A total of 3582 subjects with suspected OSA consecutively admitted to our hospital sleep centre were screened and 2983 (2422 with OSA) were included in the Shanghai Sleep Health Study. OSA severity was quantified using the apnoea-hypopnea index (AHI), the oxygen desaturation index and the arousal index. Biochemical indicators and anthropometric data were also collected. The relationship between OSA severity and the risk of dyslipidaemia was evaluated via ordinal logistic regression, restricted cubic spline (RCS) analysis and multivariate linear regressions. RESULTS: The RCS mapped a nonlinear dose-effect relationship between the risk of dyslipidaemia and OSA severity, and yielded knots of the AHI (9.4, 28.2, 54.4 and 80.2). After integrating the clinical definition and RCS-selected knots, all subjects were regrouped into four AHI severity stages. Following segmented multivariate linear modelling of each stage, distinguishable sets of OSA risk factors were quantified: low-density lipoprotein cholesterol (LDL-C), apolipoprotein E and high-density lipoprotein cholesterol (HDL-C); body mass index and/or waist to hip ratio; and HDL-C, LDL-C and triglycerides were specifically associated with stage I, stages II and III, and stages II-IV with different OSA indices. CONCLUSIONS: Our study revealed the multistage and non-monotonic relationships between OSA and dyslipidaemia and quantified the relationships between OSA severity indexes and distinct risk factors for specific OSA severity stages. Our study suggests that a new interpretive and predictive strategy for dynamic assessment of the risk progression over the clinical course of OSA should be adopted.


Subject(s)
Apolipoproteins/blood , Cholesterol, HDL/blood , Cholesterol, LDL/blood , Dyslipidemias/blood , Sleep Apnea, Obstructive/blood , Triglycerides/blood , Adult , Biomarkers/blood , Body Mass Index , Cardiovascular Diseases/complications , Dyslipidemias/complications , Dyslipidemias/diagnosis , Female , Humans , Male , Middle Aged , Obesity/complications , Polysomnography , Retrospective Studies , Risk Factors , Severity of Illness Index , Sleep Apnea, Obstructive/complications , Sleep Apnea, Obstructive/diagnosis , Sleep Apnea, Obstructive/etiology
13.
Proc Natl Acad Sci U S A ; 110(51): 20705-10, 2013 Dec 17.
Article in English | MEDLINE | ID: mdl-24248364

ABSTRACT

Variation in the 3' untranslated region (3'UTR) of the HLA-C locus determines binding of the microRNA Hsa-miR-148a, resulting in lower cell surface expression of alleles that bind miR-148a relative to those alleles that escape its binding. The HLA-C 3'UTR variant was shown to associate with HIV control, but like the vast majority of disease associations in a region dense with causal candidates, a direct effect of HLA-C expression level on HIV control was not proven. We demonstrate that a MIR148A insertion/deletion polymorphism associates with its own expression levels, affecting the extent to which HLA-C is down-regulated, the level of HIV control, and the risk of Crohn disease only among those carrying an intact miR-148a binding site in the HLA-C 3'UTR. These data illustrate a direct effect of HLA-C expression level on HIV control that cannot be attributed to other HLA loci in linkage disequilibrium with HLA-C and highlight the rich complexity of genetic interactions in human disease.


Subject(s)
Base Sequence , Crohn Disease/genetics , HIV Infections/genetics , HLA-C Antigens/genetics , INDEL Mutation , Linkage Disequilibrium , MicroRNAs/genetics , 3' Untranslated Regions/genetics , 3' Untranslated Regions/immunology , Alleles , Crohn Disease/immunology , Crohn Disease/metabolism , Female , Gene Expression Regulation/genetics , Gene Expression Regulation/immunology , HIV Infections/immunology , HIV Infections/metabolism , HLA-C Antigens/biosynthesis , HLA-C Antigens/immunology , Humans , Male , MicroRNAs/biosynthesis , MicroRNAs/immunology
14.
Gut ; 64(1): 37-48, 2015 Jan.
Article in English | MEDLINE | ID: mdl-24717932

ABSTRACT

OBJECTIVE: Octamer transcription factor 1 (OCT1) was found to be expressed in intestinal metaplasia and gastric cancer (GC), but the exact roles of OCT1 in GC remain unclear. The objective of this study was to determine the functional and prognostic implications of OCT1 in GC. DESIGN: Expression of OCT1 was examined in paired normal and cancerous gastric tissues and the prognostic significance of OCT1 was analysed by univariate and multivariate survival analyses. The functions of OCT1 on synbindin expression and extracellular signal-regulated kinase (ERK) phosphorylation were studied in vitro and in xenograft mouse models. RESULTS: The OCT1 gene is recurrently amplified and upregulated in GC. OCT1 overexpression and amplification are associated with poor survival in patients with GC and the prognostic significance was confirmed by independent patient cohorts. Combining OCT1 overexpression with American Joint Committee on Cancer staging improved the prediction of survival in patients with GC. High expression of OCT1 associates with activation of the ERK mitogen-activated protein kinase signalling pathway in GC tissues. OCT1 functions by transactivating synbindin, which binds to ERK DEF domain and facilitates ERK phosphorylation by MEK. OCT1-synbindin signalling results in the activation of ERK substrates ELK1 and RSK, leading to increased cell proliferation and invasion. Immunofluorescent study of human GC tissue samples revealed strong association between OCT1 protein level and synbindin expression/ERK phosphorylation. Upregulation of OCT1 in mouse xenograft models induced synbindin expression and ERK activation, leading to accelerated tumour growth in vivo. CONCLUSIONS: OCT1 is a driver of synbindin-mediated ERK signalling and a promising marker for the prognosis and molecular subtyping of GC.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/physiology , MAP Kinase Signaling System/physiology , Nerve Tissue Proteins/physiology , Organic Cation Transporter 1/physiology , Stomach Neoplasms/physiopathology , Vesicular Transport Proteins/physiology , Animals , Mice , Prognosis
15.
Int J Cancer ; 136(5): 1053-64, 2015 Mar 01.
Article in English | MEDLINE | ID: mdl-24996057

ABSTRACT

The transmembrane protein with epidermal growth factor and two follistatin motifs 2 (TMEFF2) is a single-pass transmembrane protein, and it is downregulated in human gastric cancer and levels correlate with tumor progression and time of survival. However, the mechanism of its dysregulation in gastric cancer is little known. Here we investigate its regulatory mechanism and the bidirectional regulation between TMEFF2 and STAT3 in gastric carcinogenesis. TMEFF2 expression was decreased after Helicobacter pylori (H. pylori) infection in vivo and in vitro. STAT3 directly binds to the promoter of TMEFF2 and regulates H. pylori-induced TMEFF2 downregulation in normal gastric GES-1 cells and gastric cancer AGS cells. Conversely, TMEFF2 may suppress the phosphorylation of STAT3 and TMEFF2-induced downregulation of STAT3 phosphorylation may depend on SHP-1. A highly inverse correlation between the expression of TMEFF2 and pSTAT3 was also revealed in gastric tissues. We now show the deregulation mechanism of TMEFF2 in gastric carcinogenesis and identify TMEFF2 as a new target gene of STAT3. The phosphorylation of STAT3 may be negatively regulated by TMEFF2, and the bidirectional regulation between TMEFF2 and STAT3 may contribute to H. pylori-associated gastric carcinogenesis.


Subject(s)
Cell Transformation, Neoplastic/pathology , Gastric Mucosa/pathology , Gene Expression Regulation, Neoplastic , Helicobacter Infections/pathology , Membrane Proteins/metabolism , Neoplasm Proteins/metabolism , STAT3 Transcription Factor/metabolism , Stomach Neoplasms/pathology , Animals , Cell Transformation, Neoplastic/metabolism , Chromatin Immunoprecipitation , Gastric Mucosa/metabolism , Gastric Mucosa/microbiology , Gerbillinae , Helicobacter Infections/metabolism , Helicobacter Infections/microbiology , Helicobacter pylori/pathogenicity , Humans , Immunoenzyme Techniques , Male , Membrane Proteins/genetics , Neoplasm Proteins/genetics , Phosphorylation , Protein Tyrosine Phosphatase, Non-Receptor Type 6/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , STAT3 Transcription Factor/genetics , Signal Transduction , Stomach Neoplasms/metabolism , Stomach Neoplasms/microbiology , Tumor Cells, Cultured
16.
Int J Colorectal Dis ; 30(3): 353-61, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25518811

ABSTRACT

PURPOSE: Since observational data in the urban residents are required to better assess the risk factors of colorectal neoplasm occurrence and the effectiveness of colonoscopy screening and surveillance, we conducted a case-control study at multicenters in China to identify patient characteristics and neoplasm features of colorectal adenoma (CRA) and colorectal carcinoma (CRC). METHODS: A total of 4089 patients who had undergone a colonoscopy from 19 hospitals were enrolled, of which 1106 had CRA and 466 had CRC. They were compared with controls. The analysis provides features and risk factors of colorectal neoplasm using multivariate logistic regression. RESULTS: Increasing age, a family history of colorectal cancer or previous cases of colorectal adenoma or hypertension disease, gastrointestinal surgery, regular intake of pickled food (adjusted odds ratio [aOR] 1.42, 95 % confidence interval [CI], 1.048-1.924), consumption of alcohol, and a positive result of fecal occult blood testing (FOBT; aOR 2.509, 95 % CI 1.485-4.237) were associated with an increased risk of CRA. In the CRC group, increasing age, regular intake of pickled foods, and a positive FOBT result were risk factors. In addition, a positive abdominal computed tomography (CT) before a colonoscopy and physical signs of emaciation were also significantly associated with an increasing risk of colorectal carcinoma. Regular intake of vegetables decreased the risk of both CRA and CRC. CONCLUSIONS: Age, pickled foods, and a positive FOBT are risk factors for colorectal neoplasm. Vegetable intake was associated with a decreased risk of CRA and CRC.


Subject(s)
Adenoma/epidemiology , Adenoma/pathology , Carcinoma/epidemiology , Carcinoma/pathology , Colonoscopy , Colorectal Neoplasms/epidemiology , Colorectal Neoplasms/pathology , Age Factors , Aged , Case-Control Studies , China/epidemiology , Cross-Sectional Studies , Early Detection of Cancer , Feeding Behavior , Humans , Mass Screening , Middle Aged , Occult Blood , Risk Factors
17.
PLoS Genet ; 8(2): e1002514, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22577363

ABSTRACT

An important paradigm in evolutionary genetics is that of a delicate balance between genetic variants that favorably boost host control of infection but which may unfavorably increase susceptibility to autoimmune disease. Here, we investigated whether patients with psoriasis, a common immune-mediated disease of the skin, are enriched for genetic variants that limit the ability of HIV-1 virus to replicate after infection. We analyzed the HLA class I and class II alleles of 1,727 Caucasian psoriasis cases and 3,581 controls and found that psoriasis patients are significantly more likely than controls to have gene variants that are protective against HIV-1 disease. This includes several HLA class I alleles associated with HIV-1 control; amino acid residues at HLA-B positions 67, 70, and 97 that mediate HIV-1 peptide binding; and the deletion polymorphism rs67384697 associated with high surface expression of HLA-C. We also found that the compound genotype KIR3DS1 plus HLA-B Bw4-80I, which respectively encode a natural killer cell activating receptor and its putative ligand, significantly increased psoriasis susceptibility. This compound genotype has also been associated with delay of progression to AIDS. Together, our results suggest that genetic variants that contribute to anti-viral immunity may predispose to the development of psoriasis.


Subject(s)
Genes, MHC Class II , Genes, MHC Class I , Psoriasis/genetics , Psoriasis/immunology , Genes, MHC Class I/immunology , Genes, MHC Class II/immunology , Genetic Association Studies , Genetic Predisposition to Disease , HIV Infections/genetics , HIV Infections/immunology , HIV-1/genetics , HIV-1/pathogenicity , HLA-B Antigens/genetics , HLA-C Antigens/genetics , Humans , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Killer Cells, Natural/virology , Polymorphism, Genetic , Protein Binding , Receptors, KIR3DS1/genetics
18.
Carcinogenesis ; 35(6): 1389-98, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24608043

ABSTRACT

C9orf140 is a newly identified and characterized gene which is associated with cell proliferation and tumorigenicity. Expression of C9orf140 is upregulated in human gastric cancer and colorectal cancer (CRC); however, little is known about its role in CRC progression. We have investigated the clinical significance, biological effects and mechanisms of C9orf140 signaling. We found that the expression of C9orf140 is dramatically increased in a subset of CRC and correlates significantly with vascular invasion and lymph node metastasis. Our finding showed that knockdown of C9orf140 significantly reduced cell proliferation and invasion in vitro and dramatically increased overall survival and decreased lung metastasis in vivo. Conversely, overexpression of C9orf140 significantly increased lung metastasis and shortened overall survival when compared with control tumors. C9orf140-induced CRC cell invasion may depend on promoting the epithelial-mesenchymal transition progression. STAT5 may directly interact with the enhancer of zeste homolog 2 (EZH2) and ß-catenin to enhance C9orf140 gene transactivation. Furthermore, C9orf140 may participate in cell invasion which is induced by STAT5, EZH2 or ß-catenin activation. We describe the role of C9orf140 in CRC progression and find that C9orf140 overexpression may be regulated by STAT5, EZH2 and ß-catenin interaction.


Subject(s)
Cell Cycle Proteins/genetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Polycomb Repressive Complex 2/metabolism , STAT5 Transcription Factor/metabolism , beta Catenin/metabolism , Adult , Aged , Animals , Base Sequence , Binding Sites , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Colorectal Neoplasms/mortality , Colorectal Neoplasms/pathology , Disease Models, Animal , Disease Progression , Enhancer of Zeste Homolog 2 Protein , Female , Gene Expression , Gene Knockdown Techniques , Heterografts , Humans , Immunohistochemistry , Male , Mice , Middle Aged , Molecular Sequence Data , Neoplasm Metastasis , Neoplasm Staging , Nuclear Proteins , Protein Binding , Signal Transduction
19.
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
20.
J Pathol ; 230(3): 277-90, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23424038

ABSTRACT

The polycomb group protein enhancer of zeste homologue 2 (EZH2), which has histone methyltransferase (HMT) activity, is overexpressed in malignant tumours. However, the role of EZH2 in colorectal cancer (CRC) invasion is little known. Here we investigated the clinical significance, biological effects, and mechanisms of EZH2 signalling. Knockdown of EZH2 significantly reduced cell invasion and secretion of matrix metalloproteinases 2/9 (MMP2/9) in in vitro studies. Knockdown of EZH2 dramatically increased overall survival and decreased metastasis of lung in in vivo studies. Conversely, overexpression of EZH2 significantly increased lung metastasis and shortened overall survival when compared with control tumours. EZH2-induced CRC cell invasion may depend on down-regulation of vitamin D receptor (VDR), which is considered to be a marker of CRC invasion. EZH2 regulates the histone trimethylation of lysine 27 (H3K27me3) in the VDR promoter. Moreover, we found that STAT3 directly binds to the EZH2 promoter and regulates VDR down-regulation in CRC cells. Significant inverse correlations were observed between the expression of EZH2 and pSTAT3 and that of VDR in CRC tissues compared with normal tissue in patients. We show the role of EZH2 in CRC metastasis and identify VDR as a target gene of EZH2. EZH2 expression may be directly regulated by STAT3, and STAT3 may play an important role in EZH2-mediated VDR down-regulation in CRC. This pathway may provide potential targets in aggressive CRC.


Subject(s)
Colorectal Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Polycomb Repressive Complex 2/metabolism , Receptors, Calcitriol/metabolism , STAT3 Transcription Factor/metabolism , Animals , Base Sequence , Cell Line, Tumor , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Down-Regulation , Enhancer of Zeste Homolog 2 Protein , Histone Methyltransferases , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Histones/genetics , Histones/metabolism , Humans , Lysine/metabolism , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Methylation , Mice , Mice, Inbred BALB C , Mice, Nude , Molecular Sequence Data , Neoplasm Invasiveness , Polycomb Repressive Complex 2/genetics , RNA Interference , Receptors, Calcitriol/genetics , Recombinant Fusion Proteins , STAT3 Transcription Factor/genetics , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL