Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 66
Filter
2.
BMC Gastroenterol ; 23(1): 424, 2023 Dec 01.
Article in English | MEDLINE | ID: mdl-38041073

ABSTRACT

BACKGROUND: Intraductal papillary mucinous neoplasm (IPMN) is a cystic tumor of the pancreas arising from abnormal papillary proliferation of ductal epithelial cells, and is a precancerous lesion of pancreatic malignancy. This study aimed to evaluate associations between acute pancreatitis (AP) and histologic subtypes of IPMN. METHODS: In the clinical study, patients with IPMN confirmed by surgical resection specimens at our institute between 2009 and 2021 were eligible for inclusion. Associations and predictive accuracy of AP on the presence of HGD were determined by logistic regressions. In addition, a systematic review and meta-analysis was conducted through literatures upon search in PubMed, Embase, CENTRAL, China National Knowledge Infrastructure (CKNI), and Wanfang database, up to June, 2023. Pooled effects of the associations between AP and HGD and intestinal epithelial subtype subtype, shown as odds ratios (ORs) with 95% confidence intervals (CIs), were calculated using random effects model. RESULTS: The retrospective cohort study included 47 patients (32 males, 15 females) diagnosed with IPMN at our center between 2009 and 2021, including 11 cases with AP (median 62 years) and 36 cases (median 64.5 years) without. Accuracy, sensitivity, specificity, positive predictive value (PPV), and negative predictive value (NPV) of AP in predicting HGD were 78.7%, 57.1%, 82.5%, 36.4%, and 91.7%, respectively. Univariate logistic regression analysis showed that AP group had greater odds of presence of HGD (OR: 6.29,95% CI: 1.14-34.57) than non-AP group. Meta-analysis of five case-control studies in the literature included 930 patients and showed that AP-IPMN patients had higher odds for HGD (OR: 2.13, 95% CI 1.38-3.29) and intestinal epithelial subtype (OR: 5.38, 95% CI: 3.50-8.27) compared to non-AP IPMN. CONCLUSIONS: AP is predictive of malignancy in patients with IPMN.


Subject(s)
Adenocarcinoma, Mucinous , Carcinoma, Pancreatic Ductal , Pancreatic Intraductal Neoplasms , Pancreatic Neoplasms , Pancreatitis , Male , Female , Humans , Carcinoma, Pancreatic Ductal/pathology , Pancreatitis/complications , Pancreatitis/pathology , Retrospective Studies , Acute Disease , Adenocarcinoma, Mucinous/complications , Adenocarcinoma, Mucinous/pathology , Pancreatic Neoplasms/pathology
3.
Article in English | MEDLINE | ID: mdl-37711116

ABSTRACT

BACKGROUND: Early detection and resection of colorectal polyps by routine colonoscopy screening can be effective in reducing the risk of colorectal cancer (CRC). OBJECTIVE: This study aimed to determine the association between diabetes mellitus (DM) and different types of colorectal polyps in the Chinese population. METHODS: A retrospective analysis was performed on inpatients admitted to the Gastroenterology Department of our hospital from January to December 2019. Clinical data, and colonoscopy and pathology findings of the subjects were collected. Bivariate analysis was used to assess factors associated with colorectal polyps. Significant variables from the bivariate evaluation were included in a stepwise multivariate logistic regression analysis to recognize independent predictors of neoplastic polyps and high-risk adenomas. RESULTS: The proportion of patients with DM was significantly higher in patients with neoplastic polyps and high-risk adenomas than in patients without polyps. Age ≥ 50 years, male gender, and a first-degree relative with a history of CRC were independent risk factors for neoplastic polyps and high-risk adenomas, even in non-smokers. An independent risk factor analysis that did not include a family history of CRC showed that age, gender, and alcohol consumption were independent risk factors for neoplastic polyps and high-risk adenomas. DM was an independent risk factor for high-risk adenomas (OR=2.902, 95% CI=1.221-6.899; p=0.016) after adjusting for age, gender, alcohol consumption, and body mass index. Thus, a history of DM significantly increases the risk of high-risk adenomas. CONCLUSION: This study demonstrated that patients with DM, age ≥ 50 years, male gender, alcohol consumption, and a first-degree relative with a history of CRC should undergo regular endoscopic screening and colonic polypectomy.

4.
Front Microbiol ; 14: 1096471, 2023.
Article in English | MEDLINE | ID: mdl-36825092

ABSTRACT

Background and objective: Impaired gut barrier contributes to the progression of type 2 diabetes mellitus (T2DM), and the gut microbiota and metabolome play an important role in it. Hemicellulose, a potential prebiotics, how its supplementation impacted the glucose level, the impaired gut barrier, and the gut microbiota and metabolome in T2DM remained unclear. Methods: In this study, some mice were arranged randomly into four groups: db/db mice fed by a compositionally defined diet (CDD), db/db mice fed by a CDD with 10% and 20% hemicellulose supplementation, and control mice fed by a CDD. Body weight and fasting blood glucose levels were monitored weekly. The gut barrier was evaluated. Fresh stool samples were analyzed using metagenomic sequencing and liquid chromatography-mass spectrometry to detect gut microbiota and metabolome changes. Systemic and colonic inflammation were evaluated. Results: Better glycemic control, restoration of the impaired gut barrier, and lowered systemic inflammation levels were observed in db/db mice with the supplementation of 10 or 20% hemicellulose. The gut microbiota showed significant differences in beta diversity among the four groups. Fifteen genera with differential relative abundances and 59 significantly different metabolites were found. In the db/db group, hemicellulose eliminated the redundant Faecalibaculum and Enterorhabdus. The increased succinate and ursodeoxycholic acid (UDCA) after hemicellulose treatment were negatively correlated with Bifidobacterium, Erysipelatoclostridium, and Faecalibaculum. In addition, hemicellulose reduced the colonic expressions of TLR2/4 and TNF-α in db/db mice. Conclusion: Hemicellulose may serve as a potential therapeutic intervention for T2DM by improving impaired intestinal mucosal barrier integrity, modulating gut microbiota composition, and altering the metabolic profile.

5.
Cancer Med ; 12(7): 8351-8366, 2023 04.
Article in English | MEDLINE | ID: mdl-36708020

ABSTRACT

BACKGROUND: Colorectal cancer (CRC) is a disease with high morbidity and mortality rates globally. Long noncoding RNAs (lncRNAs) play a fundamental role in tumor progression, and increasing attention has been paid to their role in CRC. This study aimed to determine the function of lncRNA DICER1 antisense RNA 1 (DICER1-AS1) in CRC and confirm its potential regulatory mechanisms in CRC. METHODS: The publicly available dataset was used to assess DICER1-AS1 function and expression in CRC. RT-qPCR or western blot assays were performed to verify DICER1-AS1, miR-650, and mitogen-activated protein kinase 1 (MAPK1) expression in CRC cells or tissues. To determine the function of DICER1-AS1, we performed CCK-8, colony formation, transwell, cell cycle, and in vivo animal assays. Using RNA sequence analysis, luciferase reporter assays, and bioinformatics analysis, the connection between DICER1-AS1, MAPK1, and miR-650 was investigated. RESULTS: DICER1-AS1 was significantly upregulated in CRC tissue compared to normal colon tissue. High DICER1-AS1 expression suggested a poor prognosis in CRC patients. Functionally, upregulation of DICER1-AS1 effectively promoted CRC proliferation, migration, and invasion ex vivo and tumor progression in vivo. Mechanistically, DICER1-AS1 functions as a competitive endogenous RNA (ceRNA) that sponges miR-650 to upregulate MAPK1, promotes ERK1/2 phosphorylation, and sequentially activates the MAPK/ERK signaling pathway. CONCLUSION: Our investigations found that upregulation of DICER1-AS1 activates the MAPK/ERK signaling pathway by sponging miR-650 to promote CRC progression, revealing a possible clinically significant biomarker and therapeutic target.


Subject(s)
Colorectal Neoplasms , MicroRNAs , RNA, Long Noncoding , Animals , Cell Line, Tumor , Cell Movement , Cell Proliferation , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Gene Expression Regulation, Neoplastic , MAP Kinase Signaling System , MicroRNAs/genetics , MicroRNAs/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Signal Transduction
6.
Pancreas ; 51(4): 330-337, 2022 04 01.
Article in English | MEDLINE | ID: mdl-35695761

ABSTRACT

OBJECTIVES: Embryonic stem cells (ESCs)-derived pancreatic precursor cells have great potential for pancreas repair. Expression of pancreatic duodenal homeobox 1 (Pdx1) in definitive endoderm (DE) cells is the premise that DE cells differentiate into pancreatic cells. To achieve the required number of Pdx1-expressing DE cells for cell transplantation therapy, a valid model must be established. Using this model, researchers investigated how Pdx1 regulates ESC differentiation into pancreatic cells. METHODS: Tet-On inducible lentiviral vector encoding Pdx1 or mock vector was transduced into mouse ESC (ES-E14TG2a). The mouse ESCs were divided into 3 groups: control (ESC), mock vector (Pdx1 - -ESC), and vector encoding Pdx1 (Pdx1 + -ESC). All groups were separately cocultured with the DE cells sorted by immune beads containing CXCR-4 + (C-X-C chemokine receptor type-4) antibody. Doxycycline induced the expression of Pdx1 on the Pdx1 + -ESC cells. The markers of cell differentiation and Notch pathway were examined. RESULTS: Significantly increased expression levels of Ptf1a, CK19, and amylase on day (d) 3 and d7, Neuro-D1 on d10 and d14, Pax6 and insulin on d14, as well as Notch1, Notch2, Hes1, and Hes5 on d3 and thereafter declined on d14 were observed in Pdx1 + -ESC group. CONCLUSIONS: Pdx1 + -ESC could differentiate into pancreatic-like cells with involvement of the Notch pathway.


Subject(s)
Endoderm , Homeodomain Proteins , Mouse Embryonic Stem Cells , Pancreas , Trans-Activators , Animals , Cell Differentiation , Endoderm/cytology , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Mice , Mouse Embryonic Stem Cells/cytology , Pancreas/cytology , Receptors, Notch/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism
7.
Front Med (Lausanne) ; 8: 690995, 2021.
Article in English | MEDLINE | ID: mdl-34336894

ABSTRACT

Background: Proton pump inhibitors (PPIs) are validated gastric acid suppressors and have been widely used to treat patients with active duodenal ulcers. Although existing PPIs have shown great efficacy, many scientists are still devoted to developing more effective PPIs with better safety profile. Herein, we aimed to compare the safety and efficacy of anaprazole in duodenal mucosal healing, a novel PPI, to that of rabeprazole. Methods: In this multicenter, randomized, positive-controlled, double-blinded, parallel-group phase II clinical trial, a total of 150 qualified patients with endoscopically confirmed active duodenal ulcers were randomized (1:1:1) to receive rabeprazole 10 mg, anaprazole 20 mg or anaprazole 40 mg for 4 weeks. The ulcer healing rates after 4 weeks of treatment were compared between groups by independent central review and investigator review. In addition, symptoms and safety were evaluated. Results: Based on the independent central review, the ulcer healing rates of the 10 mg rabeprazole, 20 mg anaprazole and 40 mg anaprazole groups were 88.0, 85.1, and 87.5%, respectively, in the FAS population and 88.9, 86.0, and 90.9%, respectively, in the PPS population. The ulcer healing rate difference between anaprazole 20 mg and Rabeprazole 10 mg is -2.9% (95% CI, -16.5-10.7%), and -0.5% (95% CI, -13.5-12.5%) between anaprazole 40 mg and Rabeprazole 10 mg, in the FAS population. Based on the investigator review, the ulcer healing rates of the 10 mg rabeprazole, 20 mg anaprazole, and 40 mg anaprazole groups were 72.0, 70.2, and 77.1%, respectively, in the FAS population and 75.6, 72.1, and 79.5%, respectively, in the PPS population. The ulcer healing rate difference between anaprazole 20 mg and Rabeprazole 10 mg is -1.8% (95% CI, -19.8-16.3%), and 5.1% (95% CI, -12.2-22.3%) between anaprazole 40 mg and Rabeprazole 10 mg, in the FAS population. Most patients (>90%) eventually achieved complete symptom relief. The incidence rates of adverse events were of no significant differences among the treatment groups. Potential possible better liver tolerance was observed in two anaprazole dose groups than rabeprazole 10 mg group. Conclusion: Both at a dosage of 20 and 40 mg daily, anaprazole, is effective with good safety profile in the treatment of active duodenal ulcers in this Phase 2 study, which allows anaprazole to be advanced to a phase III clinical trial. Clinical Trial Registration: https://www.clinicaltrials.gov/ct2/results?cond=&term=NCT04503629&cntry=&state=&city=&dist=, Identifier: CTR20181464, NCT04503629.

8.
J Diabetes Complications ; 35(2): 107766, 2021 02.
Article in English | MEDLINE | ID: mdl-33168395

ABSTRACT

INTRODUCTION: The epithelial tight junctions of intestine were impaired in murine model of type 2 diabetes mellitus (T2DM). The aim of this work was to investigate the alteration of intestinal barrier in T2DM patients. METHODS: 90 patients with T2DM and 28 healthy controls were recruited. Serum lipopolysaccharide (LPS), Zonulin, and intestinal fatty acid binding protein (IFABP) were measured by ELISA, based on which a derived permeability risk score (PRS) was calculated. Subgroup analyses were conducted based on the glycemic control (HbA1c < 7%, or HbA1c ≥ 7%), the amount of chronic diabetic complications, and the use of aspirin at the time. RESULTS: Serum LPS, Zonulin, and IFABP, and PRS of T2DM group were significantly higher than those of the control group (p < 0.05 for all). Serum LPS and PRS was higher in T2DM patients with poor glycemic control (both p < 0.05). Patients with more chronic complications of diabetes had higher serum LPS and IFABP, and PRS (all p < 0.05). No differences were found in these serum markers between T2DM patients being treated with aspirin or not. CONCLUSIONS: Intestinal barrier function was impaired in T2DM patients. Poor glycemic control and more chronic complications of diabetes were associated with worse intestinal barrier function. Treatment with aspirin did not aggravate the impairment of intestinal barrier in T2DM patients.


Subject(s)
Diabetes Mellitus, Type 2 , Fatty Acid-Binding Proteins/blood , Intestinal Mucosa/physiopathology , Lipopolysaccharides , Protein Precursors/blood , Aspirin/therapeutic use , Diabetes Mellitus, Type 2/complications , Glycated Hemoglobin , Glycemic Control , Haptoglobins , Humans , Intestinal Mucosa/metabolism , Lipopolysaccharides/blood
9.
Oncol Rep ; 44(3): 973-986, 2020 09.
Article in English | MEDLINE | ID: mdl-32705223

ABSTRACT

N6­methyladenosine (m6A) RNA modification maintained by N6­methyltransferases and demethylases is involved in multiple biological functions. Methyltransferase like 3 (METTL3) is a major N6­methyltransferase. However, the role of METTL3 and its installed m6A modification in colorectal tumorigenesis remains to be fully elucidated. METTL3 is highly expressed as indicated in colorectal cancer samples in the TCGA and Oncomine databases, implying its potential role in colon tumorigenesis. SW480 cell line with stable METTL3 knockout (METTL3­KO) was generated using CRISPR/Cas9 and were confirmed by the loss of METTL3 expression and suppression of m6A modification. The proliferation of METTL3­KO cells was significantly inhibited compared with that of control cells. METTL3­KO decreased the decay rate of suppressor of cytokine signaling 2 (SOCS2) RNA, resulting in elevated SOCS2 protein expression. m6A­RNA immunoprecipitation­qPCR (MeRIP­qPCR) revealed that SOCS2 mRNA was targeted by METTL3 for m6A modification. Similar to METTL3­KO SW480 cells, SW480 cells treated with 3­deazaadenosine, an RNA methylation inhibitor, exhibited elevated SOCS2 protein expression. Increased levels of SOCS2 in METTL3­KO SW480 cells were associated with decreased expression of leucine­rich repeat­containing G protein­coupled receptor 5 (LGR5), contributing to the inhibition of cell proliferation. The underlying associations among METTL3, SOCS2, and LGR5 were further confirmed in SW480 cells transfected with si­METTL3 and in tumor samples from patients with colorectal cancer. Taken together, our data demonstrate that an increased level of METTL3 may maintain the tumorigenicity of colon cancer cells by suppressing SOCS2.


Subject(s)
Carcinogenesis/genetics , Colonic Neoplasms/genetics , Gene Expression Regulation, Neoplastic/genetics , Methyltransferases/metabolism , Suppressor of Cytokine Signaling Proteins/genetics , Adenosine/analogs & derivatives , Adenosine/metabolism , Aged , Carcinogenesis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Colectomy , Colon/pathology , Colon/surgery , Colonic Neoplasms/diagnosis , Colonic Neoplasms/pathology , Colonic Neoplasms/surgery , Datasets as Topic , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Gene Knockout Techniques , Humans , Male , Methylation/drug effects , Methyltransferases/genetics , Middle Aged , Neoplasm Staging , RNA Stability/drug effects , RNA Stability/genetics , RNA, Messenger/metabolism , Receptors, G-Protein-Coupled/genetics , Spheroids, Cellular , Tubercidin/pharmacology
10.
J Diabetes ; 12(3): 224-236, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31503404

ABSTRACT

BACKGROUND: Impaired intestinal barrier structure and function have been validated as an important pathogenic process in type 2 diabetes mellitus (T2DM). Gut dysbiosis is thought to be the critical factor in diabetic intestinal pathogenesis. As the most abundant commensal bacteria, Faecalibacterium prausnitzii (F. prausnitzii) play important roles in gut homeostasis. The microbial anti-inflammatory molecule (MAM), an F. prausnitzii metabolite, has anti-inflammatory potential in inflammatory bowel disease (IBD). Thus, we aimed to explore the function and mechanism of MAM on the diabetic intestinal epithelium. METHODS: 16S high-throughput sequencing was used to analyze the gut microbiota of db/db mice (T2DM mouse model). We transfected a FLAG-tagged MAM plasmid into human colonic cells to explore the protein-protein interactions and observe cell monolayer permeability. For in vivo experiments, db/db mice were supplemented with recombinant His-tagged MAM protein from E. coli BL21 (DE3). RESULTS: The abundance of F. prausnitzii was downregulated in the gut microbiota of db/db mice. Immunoprecipitation (IP) and mass spectroscopy (MS) analyses revealed that MAM potentially interacts with proteins in the tight junction pathway, including zona occludens 1 (ZO-1). FLAG-tagged MAM plasmid transfection stabilized the cell permeability and increased ZO-1 expression in NCM460, Caco2, and HT-29 cells. The db/db mice supplemented with recombinant His-tagged MAM protein showed restored intestinal barrier function and elevated ZO-1 expression. CONCLUSIONS: Our study shows that MAM from F. prausnitzii can restore the intestinal barrier structure and function in DM conditions via the regulation of the tight junction pathway and ZO-1 expression.


Subject(s)
Anti-Inflammatory Agents/metabolism , Diabetes Mellitus, Type 2/metabolism , Faecalibacterium prausnitzii/metabolism , Intestinal Mucosa/metabolism , Tight Junction Proteins/metabolism , Tight Junctions/metabolism , Animals , Anti-Inflammatory Agents/pharmacology , Caco-2 Cells , Diabetes Mellitus, Type 2/genetics , Dysbiosis/genetics , Dysbiosis/physiopathology , Gastrointestinal Microbiome/drug effects , Gastrointestinal Microbiome/genetics , Gastrointestinal Microbiome/physiology , Gene Expression/drug effects , HEK293 Cells , HT29 Cells , Humans , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/metabolism , Intestinal Mucosa/drug effects , Mice , Permeability/drug effects , Tight Junction Proteins/genetics , Tight Junctions/drug effects
11.
Biomed Pharmacother ; 118: 109131, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31545226

ABSTRACT

Berberine and metformin, both established pharmaceutical agents with herbal origins, have incidental beneficial effects on multiple diseases, including diabetes. These effects have been speculated to occur via the gut microbiome. In this study, we administered either berberine or metformin to db/db mice and investigated changes in body weight, food intake, and blood glucose levels. Fresh stool samples were analyzed using 16 s rDNA high-throughput sequencing to evaluate the gut microbiome. Short-chain fatty acids (SCFA) in the stool were quantified using gas chromatography. The expression of NF-κB signaling pathway and tight junction (ZO1 and occludin) proteins in the intestinal epithelium was determined using qPCR and western blotting. The intestinal barrier structure was examined using transmission electron microscopy and serum lipopolysaccharide (LPS) was measured using a commercial kit. Both berberine and metformin reduced food intake, body weight, and blood glucose and HbA1c levels. Both treatments effectively restored the intestinal SCFA content, reduced the level of serum LPS, relieved intestinal inflammation, and repaired intestinal barrier structure. Intervention with metformin or berberine modified the gut microbiome in db/db mice, increasing the number of SCFA-producing bacteria (e.g., Butyricimonas, Coprococcus, Ruminococcus) and reducing opportunistic pathogens (e.g., Prevotella, Proteus). An increased abundance of other probiotics including Lactobacillus and Akkermansia was also observed. Berberine and metformin can modulate the composition of the gut microbiome and reduce body weight, blood glucose levels, and intestinal inflammation in db/db mice, which demonstrates their effectiveness in the reduction of diabetic complications in this model.


Subject(s)
Berberine/pharmacology , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Type 2/drug therapy , Gastroenteritis/prevention & control , Gastrointestinal Microbiome/drug effects , Hypoglycemic Agents/pharmacology , Metformin/pharmacology , Animals , Blood Glucose/analysis , Body Weight/drug effects , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Experimental/microbiology , Diabetes Mellitus, Type 2/immunology , Diabetes Mellitus, Type 2/microbiology , Eating/drug effects , Gastroenteritis/microbiology , Glycated Hemoglobin/analysis , Intestinal Mucosa/drug effects , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Obesity/immunology , Obesity/microbiology , Obesity/prevention & control , Permeability
12.
Int J Mol Med ; 43(6): 2471-2480, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30942388

ABSTRACT

The majority of Musashi 1 (Msi1)­positive cells derived from mouse embryonic stem cells (mESCs) are prone to differentiate into neural epithelial­like cells, and only a small proportion of Msi1­positive cells differentiate into intestinal epithelial­like cells. Whether inhibiting the phosphatidylinositol 3­kinase (PI3K) signaling of mESCs can promote the differentiation of Msi1­positive cells into intestinal epithelial­like cells remains to be fully elucidated. In the present study, to inhibit PI3K signaling, mESCs were treated with LY294002. A pMsi1­green fluorescence protein reporter plasmid was used to sort the Msi1­positive cells from mESCs treated and untreated with LY294002 (5 µmol/l). The Msi1­positive cells were hypodermically engrafted into the backs of non­obese diabetic/severe combined immunodeficient mice. The presence of neural and intestinal epithelial­like cells in the grafts was detected by reverse transcription­quantitative polymerase chain reaction analysis and immunohistochemistry. Compared with the Msi1­positive cells derived from mESCs without LY294002 treatment, Msi1­positive cells derived from mESCs treated with LY294002 expressed higher levels of leucine­rich repeat­containing G­protein coupled receptor, a marker of intestinal epithelial stem cells, and lower levels of Nestin, a marker of neural epithelial stem cells. The grafts from Msi1­positive cells treated with LY294002 contained more intestinal epithelial­like tissues and fewer neural epithelial­like tissues, compared with those from untreated Msi1­positive cells. LY294002 had the ability to promote the differentiation of mESCs into intestinal epithelial­like tissues. The Msi1­positive cells selected from the cell population derived from mESCs treated with LY294002 exhibited more characteristics of intestinal epithelial stem cells than those from the untreated group.


Subject(s)
Cell Differentiation/drug effects , Chromones/pharmacology , Intestinal Mucosa/cytology , Morpholines/pharmacology , Mouse Embryonic Stem Cells/drug effects , Nerve Tissue Proteins/analysis , Protein Kinase Inhibitors/pharmacology , RNA-Binding Proteins/analysis , Animals , Cell Culture Techniques , Cell Line , Cell Survival/drug effects , Gene Expression Regulation/drug effects , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Mice , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/metabolism , Nerve Tissue Proteins/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , RNA-Binding Proteins/genetics , Signal Transduction/drug effects
13.
J Pharmacol Sci ; 139(4): 266-274, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30871870

ABSTRACT

Butyrate is widely accepted as a proliferation inhibitor in colon cancer but less thoroughly characterized in the colonic epithelium of objects with type 2 diabetes mellitus. The present study investigated the regulatory effect of butyrate on proliferation, the related molecule high-mobility group box 1 (HMGB1) and the receptor for advanced glycation end products (RAGE) in the colon of db/db type 2 diabetic model mice and non-cancerous NCM460 colon cells. Proliferation and the expression of HMGB1 and RAGE were increased and could be partially reversed by butyrate treatment in the colon of db/db mice, which were consistent in NCM460 cells under a high glucose state. In NCM460 cells, under the normal glucose state, proliferation increased by overexpression of HMGB1. Under a high glucose state, increased expression of HMGB1 was accompanied with a release from cell nuclei into the cytoplasm and extracellular matrix. Down-regulation of HMGB1 could lower the expression of RAGE and attenuate the abnormally increased proliferation. And overexpression of HMGB1 reversed the suppressing effect of butyrate on abnormally increased proliferation. Conclusively, butyrate suppressed the abnormally increased proliferation in colonic epithelial cells under diabetic state by targeting HMGB1.


Subject(s)
Butyrates/pharmacology , Cell Proliferation/drug effects , Colon/cytology , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/pathology , Epithelial Cells/physiology , Gene Expression/drug effects , HMGB1 Protein/metabolism , Animals , Cells, Cultured , Disease Models, Animal , Epithelial Cells/metabolism , HMGB1 Protein/genetics , Male , Mice, Inbred Strains , Receptor for Advanced Glycation End Products/genetics , Receptor for Advanced Glycation End Products/metabolism
14.
Exp Cell Res ; 368(1): 101-110, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29689277

ABSTRACT

BACKGROUND: Intestinal barrier dysfunctions are related to dysbacteriosis and chronic gut inflammation in type 2 diabetes. Although there is emerging evidence that the chronic gut inflammatory response is stimulated by nucleotide-binding oligomerization domain-like receptors (NLRs), the relationship and precise mechanism between NLRC3 and the colonic epithelial barrier remains largely elusive. METHODS: We investigated the function and mechanism of NLRC3 in the colonic tissues of diabetic mice and colonic epithelial cell lines. The regulatory mechanism between NLRC3, butyrate and tight junctions was elucidated via a transepithelial electrical resistance measurement, transmission electron microscopy, RNA interference and western blotting. RESULTS: In this study, we found that NLRC3 expression was decreased in the colonic tissues of diabetic mice. NLRC3 over-expression ameliorated colonic epithelial barrier integrity and up-regulated tight junction proteins in colonic epithelial cells. Knockdown of TRAF6 diminished NLRC3-induced ZO-1/occludin expression. In addition, we demonstrated that butyrate could stimulate NLRC3 expression in both diabetic mice and colonic epithelial cells. GPR43 on colonic epithelial cells is involved in the activation of NLRC3 induced by butyrate. CONCLUSION: Our findings demonstrated that NLCR3 could ameliorate colonic epithelial barrier integrity in diabetes mellitus in a TRAF6-dependent manner, and NLCR3 was stimulated by butyrate via binding GPR43 on colonic epithelial cells.


Subject(s)
Butyrates/pharmacology , Epithelial Cells/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Receptors, Cell Surface/drug effects , Receptors, G-Protein-Coupled/drug effects , Animals , Diabetes Mellitus, Experimental/metabolism , Humans , Intestinal Mucosa/metabolism , Mice, Transgenic , Protective Agents/pharmacology , Receptors, Cell Surface/metabolism , Receptors, G-Protein-Coupled/metabolism , Tight Junctions/metabolism
15.
Mol Cell Biochem ; 438(1-2): 85-96, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28744818

ABSTRACT

The intestinal epithelium cells (IECs) in diabetes mellitus (DM) patients have been proven to be abnormally differentiated. During the differentiation of IECs, epigenetic modification acts as an important regulator. In this study, we aimed to examine the epigenetic alteration of Transducin-like Enhancer of Split 1 (TLE1), a multitask transcriptional co-repressor, contributing to the differentiation homeostasis in IECs of DM mice. The IECs of type 2 diabetic mice model were isolated and collected. Methylation states of whole genomic DNA promoter regions were investigated by microarray. Methylated-specific PCR was used to detect the methylation state of TLE1 promoter in DM mice IECs. The expression of TLE1, Hes1, and differentiated cell markers were measured through real-time PCR, Western blots, and immunohistochemistry; by transfection assay, TLE1 or Hes1 was independently down-regulated in intestinal epithelium cell line, IEC-6. Subsequent modulation on TLE1, Hes1, and differentiated intestinal cell markers were detected. Global gene promoter regions in DM intestinal epithelium were less methylated comparing to normal control. The expression of TLE1 was significantly increased via hypomethylated activation in DM mice IECs. Hes1 was significantly suppressed and the terminal cell markers abnormally expressed in DM mice IECs (P < 0.05). Inhibition or induction on the abundance of TLE1 in IEC-6 cell line resulted in the corresponding dysregulation of Hes1 and intestinal epithelium differentiation (P < 0.05). Demethylation of TLE1 promoter region activates the self-expression in diabetic mice IECs. Subsequently, TLE1, through the transcriptional suppression on expression of Hes1, contributes to the aberrant differentiation of IECs in DM mice.


Subject(s)
Co-Repressor Proteins/biosynthesis , DNA Methylation , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 2/metabolism , Epigenesis, Genetic , Intestinal Mucosa/metabolism , Promoter Regions, Genetic , Animals , Co-Repressor Proteins/genetics , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/pathology , Intestinal Mucosa/pathology , Mice
16.
Gastroenterol Res Pract ; 2017: 5715403, 2017.
Article in English | MEDLINE | ID: mdl-28883831

ABSTRACT

BACKGROUND: Extragastric manifestations of Helicobacter pylori (H. pylori) infection have been reported in many diseases. However, there are still controversies about whether H. pylori infection is associated with diabetes mellitus (DM). This study was aimed at answering the question. METHODS: A systematic search of the literature from January 1996 to January 2016 was conducted in PubMed, Embase databases, Cochrane Library, Google Scholar, Wanfang Data, China national knowledge database, and SinoMed. Published studies reporting H. pylori infection in both DM and non-DM individuals were recruited. RESULTS: 79 studies with 57,397 individuals were included in this meta-analysis. The prevalence of H. pylori infection in DM group (54.9%) was significantly higher than that (47.5%) in non-DM group (OR = 1.69, P < 0.001). The difference was significant in comparison between type 2 DM group and non-DM group (OR = 2.05), but not in that between type 1 DM group and non-DM group (OR = 1.23, 95% CI: 0.77-1.96, P = 0.38). CONCLUSION: Our meta-analysis suggested that there is significantly higher prevalence of H. pylori infection in DM patients as compared to non-DM individuals. And the difference is associated with type 2 DM but not type 1 DM.

17.
Oncol Lett ; 14(2): 1875-1883, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28781635

ABSTRACT

Colon cancer is one of the most common cancers in the world. Multidrug resistance is one of the main reasons for failure of therapy in patients with advanced colon cancer. In previous studies, multiple methods were investigated to reverse the multidrug resistance of colon cancer cells. However, to date, no clinical method has been identified to be satisfactory. Therefore, successful reversal of drug resistance in colon cancer cells still requires new therapeutic strategies or pharmaceuticals. Wild-type p53-induced phosphatase (Wip1), a member of the 2C type serine/threonine protein phosphatase family, is closely associated with the p53 gene, which is the most important tumor-suppressor gene. Wip1 was reported to be associated with the chemosensitivity of breast cancer cells. However, the correlation between the expression of Wip1 gene and the chemosensitivity of colon cancer cells has not been reported yet. In the present study, Wip1-811 small interfering RNA (siRNA) targeting Wip1 was investigated to reverse the multidrug resistance of colon cancer cells. The siRNA duplexes were transfected into RKO colon cancer cells. The messenger RNA (mRNA) expression of Wip1 was measured by reverse transcription-quantitative polymerase chain reaction. The protein level of Wip1 was detected by western blotting. The cell viability was measured by MTS assay. The cell apoptosis and cell cycle were analyzed by flow cytometry. Intracellular adriamycin cumulative concentration was determined using flow cytometry. Wip1-811 siRNA efficiently inhibited the expression of Wip1 at the mRNA and protein levels, and enhanced the sensitivity of RKO colon cancer cells towards chemotherapy, which was accompanied by increased cell apoptosis, following the inhibition of Wip1 gene expression. These results indicate that Wip1 gene silencing could enhance the chemosensitivity of colon cancer cells, which may provide a new potential approach for the reversal of multidrug resistance in colon cancer cells.

18.
Med Sci Monit ; 23: 3942-3951, 2017 Aug 15.
Article in English | MEDLINE | ID: mdl-28809820

ABSTRACT

BACKGROUND Functional dyspepsia (FD) refers to a group of upper gastrointestinal syndromes, subdivided into two types: postprandial distress syndrome (PDS) and epigastric pain syndrome (EPS). The etiology of FD remains unclear; however, unhealthy dietary habit is one potential underlying cause. We aim to explore the association of poor dietary habits with FD and its subtypes. MATERIAL AND METHODS A validated epidemiological questionnaire was designed to investigate dietary habits and gastrointestinal syndromes. Citizens in the Baotun community of Dongguan were invited to complete the study questionnaire. All participants were asked to undergo a physical examination and a blinded physician interview. The study was conducted from June 2015 to June 2016. FD was diagnosed using ROME III criteria. The association between investigated dietary habits and dyspeptic symptoms were explored. RESULTS There were 1,304 adult residents recruited for the study survey; 165 residents had existing organic dyspepsia (OD), 203 residents were diagnosed with FD, and the other 936 participants, who were without dyspeptic symptoms or functional gastrointestinal diseases, were regarded as the control group. Subtype diagnosis indicated 61 participants had EPS, 66 participants had PDS, and 76 participants had coexisting EPS and PDS. Unhealthy dietary habits were more prevalent in the FD group than in the control groups (75.86% versus 37.50%; p<0.001). FD was found to be associated with irregular mealtime, dining out, fatty food, sweet food, and coffee (p<0.05). The impact of each dietary factor varied with FD subtypes. CONCLUSIONS Certain types of dietary habits were positively correlated with the prevalence of FD. FD subtypes showed relatively different associations with dietary factors.


Subject(s)
Diet/adverse effects , Dyspepsia/etiology , Feeding Behavior , Gastrointestinal Diseases/etiology , Abdominal Pain/diet therapy , Abdominal Pain/epidemiology , Abdominal Pain/etiology , Adult , China/epidemiology , Diagnosis, Differential , Diet/statistics & numerical data , Dyspepsia/diet therapy , Dyspepsia/epidemiology , Female , Gastrointestinal Diseases/epidemiology , Gastrointestinal Diseases/metabolism , Helicobacter pylori/isolation & purification , Humans , Male , Middle Aged , Postprandial Period/physiology , Prevalence , Rural Population , Surveys and Questionnaires
19.
Stem Cell Res Ther ; 8(1): 60, 2017 03 09.
Article in English | MEDLINE | ID: mdl-28279198

ABSTRACT

BACKGROUND: Distinctive structures called crypts harbor intestinal epithelial stem cells (IESCs) which generate progenitor and terminally differentiated cells in the intestinal epithelium. Mammalian IESCs and their daughter cells require the participation of DNA methylation and the transcription factor Sox9 for proliferation and differentiation. However, the association between Sox9 and DNA methylation in this process remains elusive. METHODS: The DNA methylation of small intestinal epithelial crypts in db/db mice was detected via combining methylated DNA immunoprecipitation with microarray hybridization. DNA methylation of Sox9 promoter in crypts and IESCs was validated using bisulfite sequence analysis. The target sequence of the transcription factor Sox9 in IESCs was investigated via chromatin immunoprecipitation (ChIP) combined with deep sequencing (ChIP-seq). RESULTS: Increased Sox9 expression is accompanied by the loss of methylation in its promoter in IESCs. Sox9 targets the enhancers of the Wnt signaling pathway-related genes. Sox9 predominantly acts as a transcriptional activator at proximal enhancers of Wnt4, Tab2, Sox4, and Fzd8, but also functions as a potential transcriptional inhibitor at a distant enhancer of Cdk1. Lack of Sox9 transcriptional activation in specific repressors of the Wnt signaling pathway leads to the loss of intrinsic inhibitory action and ultimately produces overactivation of this pathway in db/db mice. CONCLUSIONS: Our study sheds light on the connections among DNA methylation, transcription factor modulation, and Wnt signaling in IESCs in the diabetic state. Hypomethylation in the Sox9 promoter is correlated to increased Sox9 expression in db/db IESCs. Although there is increased expression of Sox9 in db/db IESCs, the loss of Sox9 transcriptional activation in specific repressors of the Wnt signaling pathway might result in abnormalities in this pathway.


Subject(s)
DNA Methylation/genetics , Diabetes Mellitus/therapy , SOX9 Transcription Factor/genetics , Stem Cell Transplantation , Animals , Cell Differentiation/genetics , Cell Proliferation/genetics , Diabetes Mellitus/genetics , Diabetes Mellitus/pathology , Gene Expression Regulation, Developmental , High-Throughput Nucleotide Sequencing , Humans , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Mice , Mice, Inbred NOD , Promoter Regions, Genetic , Stem Cells/metabolism , Transcriptional Activation/genetics , Wnt Signaling Pathway/genetics
20.
Med Sci Monit ; 22: 3062-72, 2016 Aug 30.
Article in English | MEDLINE | ID: mdl-27572949

ABSTRACT

BACKGROUND Type 1 diabetes mellitus (T1DM) is associated with increased risks of enteric infection. Paneth cells constitute the first line of the gut defense. Little is known about the impact of T1DM on the bactericidal function of intestinal Paneth cells. MATERIAL AND METHODS A T1DM mouse model was induced by intraperitoneal injection of streptozocin. The analysis of intestinal microbiota and the mucosal bactericidal assay were conducted to evaluate intestinal innate defense. Numbers of Paneth cells and their expression of related antimicrobial peptides were analyzed. Expression of total insulin receptor (IR) mRNA and relative levels of IR-A/IR-B were analyzed. The primary mouse small intestinal crypt culture was used to analyze the effect of insulin and glucose on the expression of related antimicrobial peptides of Paneth cells. RESULTS In T1DM mice, bacterial loads were increased and there was an alteration in the composition of the intestinal microflora. Exogenous bacteria had better survival in the small bowel of the T1DM mice. The expression of Paneth cell-derived antimicrobial peptides was significantly decreased in the T1DM mice, although the number of Paneth cells was increased. Relative levels of IR-A/IR-B in Paneth cells of diabetic mice were elevated, but the total IR mRNA did not change. Insulin treatment restored the expression of antimicrobial peptides and normalized the microbiota in the gut of T1DM mice. Subsequently, in vitro culture assay demonstrated that insulin rather than glucose was essential for the optimal expression of Paneth cell-derived antimicrobial peptides. CONCLUSIONS The bactericidal function of intestinal Paneth cells was impaired in STZ-induced diabetic mice, resulting in the altered intestinal flora, and insulin was essential for the optimal expression of Paneth cell-derived antimicrobial peptides.


Subject(s)
Diabetes Mellitus, Experimental/immunology , Insulin/deficiency , Paneth Cells/immunology , Animals , Antimicrobial Cationic Peptides/biosynthesis , Antimicrobial Cationic Peptides/immunology , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/microbiology , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/microbiology , Immunity, Innate , Insulin/administration & dosage , Insulin/blood , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Intestine, Small/immunology , Intestine, Small/microbiology , Mice , Mice, Inbred C57BL , Microbiota , Paneth Cells/microbiology , Random Allocation , Receptor, Insulin/biosynthesis , Receptor, Insulin/deficiency , Receptor, Insulin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...