Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
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
2.
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.

3.
Ann Rheum Dis ; 80(3): 356-366, 2021 03.
Article in English | MEDLINE | ID: mdl-33109602

ABSTRACT

OBJECTIVES: Despite preclinical studies involving miRNA therapeutics conducted in osteoarthritis (OA) over the years, none of these miRNAs have yet translated to clinical applications, owing largely to the lack of efficient intra-articular (IA) delivery systems. Here, we investigated therapeutic efficacy of the chondrocyte-specific aptamer-decorated PEGylated polyamidoamine nanoparticles (NPs)-based miRNAs delivery for OA. METHODS: The role of miR-141/200c cluster during skeletal and OA development was examined by miR-141/200cflox/flox mice and Col2a1-CreERT2; miR-141/200cflox/flox mice. Histological analysis was performed in mouse joints and human cartilage specimens. Chondrocyte-specific aptamer-decorated NPs was designed, and its penetration, stability and safety were evaluated. OA progression was assessed by micro-CT analysis, X-ray and Osteoarthritis Research Society International scores after destabilising the medial meniscus surgery with miR-141/200c manipulation by NPs IA injection. Mass spectrometry analysis, molecular docking and molecular dynamics simulations were performed to investigate the interaction between aptamer and receptor. RESULTS: Increased retention of NPs inside joint space is observed. The NPs are freely and deeply penetrant to mice and human cartilage, and unexpectedly persist in chondrocytes for at least 5 weeks. OA chondrocytes microenviroment improves endo/lysosomal escape of microRNAs (miRNAs). Therapeutically, IA injection of miR-141/200c inhibitors provides strong chondroprotection, whereas ectopic expression of miR-141/200c exacerbates OA. Mechanistically, miR-141/200c promotes OA by targeting SIRT1, which acetylates histone in the promoters of interleukin 6 (IL-6), thereby activating IL-6/STAT3 pathway. CONCLUSIONS: Our findings indicate that this nanocarrier can optimise the transport kinetics of miR-141/200c into chondrocytes, fostering miRNA-specific disease-modifying OA drugs development.


Subject(s)
Chondrocytes/metabolism , MicroRNAs , Osteoarthritis , Animals , Humans , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , Molecular Docking Simulation , Osteoarthritis/pathology
4.
Oncol Lett ; 20(3): 2191-2198, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32782536

ABSTRACT

N6-methyladenosine (m6A) RNA modification regulates multiple biological functions. Methyltransferase like 3 (METTL3), one of the major N6-methyltransferases, is highly expressed in gastric cancer, but its potential role in disease is unclear. The current study knocked out METTL3 (METTL3-KO) in human gastric cancer AGS cells using CRISPR/Cas9. METTL3-KO AGS cells exhibited decreased m6A methylation levels. A significant inhibition of cell proliferation was observed in METTL3-KO AGS cells. Silencing METTL3 in AGS cells altered the expression profile of many effector molecules that were previously demonstrated to serve key roles in AGS cell proliferation, including the suppressor of cytokine signaling (SOCS) family of proteins. The results further demonstrated that SOCS2 upregulation in METTL3-KO AGS cells was associated with a decreased RNA decay rate. Furthermore, SOCS2 KO or SOCS2 overexpression caused a significant increase and decrease in AGS cell proliferation, respectively. The current data suggested that METTL3-KO in gastric cancer cells resulted in the suppression of cell proliferation by inducing SOCS2, suggesting a potential role of elevated METTL3 expression in gastric cancer progression.

5.
Methods Mol Biol ; 2052: 205-218, 2020.
Article in English | MEDLINE | ID: mdl-31452164

ABSTRACT

MicroRNAs (miRNAs) represent a subclass of endogenous small noncoding RNAs that have been identified in both mammalian and nonmammalian cells. miRNAs are an essential part of the complex regulatory networks that control numerous biological processes and may play an important role in host defense and/or microbial offense during host-parasite interactions. Here, several methodologies to explore the role for miRNAs in host-parasite interactions are briefly summarized, including the detection, quantification, and intracellular localization of miRNAs, identification and validation of miRNA targets, and functional manipulation of specific miRNAs.


Subject(s)
Cryptosporidium/genetics , Host-Parasite Interactions/genetics , MicroRNAs/genetics , Blotting, Northern/methods , Blotting, Western/methods , Cell Line , Cryptosporidium/pathogenicity , Databases, Genetic , Epithelial Cells/metabolism , Epithelial Cells/parasitology , Genes, Reporter , Humans , In Situ Hybridization/methods , Luciferases/genetics , Luciferases/metabolism , MicroRNAs/isolation & purification , MicroRNAs/metabolism , RNA Interference , Real-Time Polymerase Chain Reaction/methods , Workflow
6.
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
7.
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
8.
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
9.
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
10.
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.

11.
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
12.
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
13.
Cell Tissue Res ; 366(2): 341-352, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27299781

ABSTRACT

Diabetes mellitus (DM) is a group of metabolic diseases characterised by insulin deficiency/resistance and hyperglycaemia. We previously reported the presence of an impaired tight junction and decreased expression of occludin (Ocln) and zonula occludens-1 (ZO-1) in the intestinal epithelial cells (IECs) of type 1 DM mice, but the exact mechanism remains unclear. In this study, we investigated the role of microRNAs (miRNAs) in impairing the tight junction in IECs of DM mice. Using an integrated comparative miRNA microarray, miR-429 was found to be up-regulated in IECs of type 1 DM mice. Then, miR-429 was confirmed to directly target the 3'-UTR of Ocln, although it did not target ZO-1. Moreover, miR-429 down-regulated the Ocln expression in IEC-6 cells in vitro. Finally, exogenous agomiRNA-429 was shown to down-regulate Ocln and induce intestinal barrier dysfunction in normal mice, while exogenous antagomiRNA-429 up-regulated Ocln in vivo and improved intestinal barrier function in DM mice. In conclusion, increased miR-429 could down-regulate the expression of Ocln by targeting the Ocln 3'-UTR, which impaired intestinal barrier function in DM mice.


Subject(s)
Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/pathology , Down-Regulation , Intestines/pathology , MicroRNAs/metabolism , Occludin/genetics , 3' Untranslated Regions/genetics , Animals , Antagomirs/metabolism , Base Sequence , Binding Sites , Cell Membrane Permeability , Down-Regulation/genetics , Epithelial Cells/metabolism , Gene Expression Profiling , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Occludin/metabolism , Oligonucleotide Array Sequence Analysis , Rats
14.
Oncotarget ; 7(1): 961-75, 2016 Jan 05.
Article in English | MEDLINE | ID: mdl-26510906

ABSTRACT

Long intergenic noncoding RNAs (lincRNAs) play important roles in regulating the biological functions and underlying molecular mechanisms of colorectal cancer (CRC). Here, we investigated the association of linc-POU3F3 and prognosis in CRC. We demonstrated that linc-POU3F3 was overexpressed in CRC tissues and positively correlated with tumor grade and N stage. Inhibition of linc-POU3F3 resulted in inhibition of cell proliferation and G1 cell cycle arrest, which was mediated by cyclin D1, CDK4, p18, Rb, and phosphorylated Rb. Inhibition of linc-POU3F3 induced apoptosis, and suppressed migration and invasion in LOVO and SW480 cell lines. This inhibition also increased the expressions of epithelial markers and decreased the expressions of mesenchymal markers, thus inhibiting the cancer epithelial-mesenchymal transition. The decreased migration and invasion following linc-POU3F3 knockdown were mediated by an increased BMP signal. Furthermore, autophagy was enhanced by linc-POU3F3 knockdown, suggesting the involvement of autophagy in the induced apoptosis. Collectively, linc-POU3F3 might be crucial in pro-proliferation, anti-apoptosis, and metastasis in LOVO and SW480 cells by regulating the cell cycle, intrinsic apoptosis, BMP signaling and autophagy. Thus, linc-POU3F3 is a potential therapeutic target and novel molecular biomarker for CRC.


Subject(s)
Apoptosis/genetics , Cell Movement/genetics , Cell Proliferation/genetics , Colorectal Neoplasms/genetics , POU Domain Factors/genetics , RNA, Long Noncoding/genetics , Autophagy/genetics , Blotting, Western , Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/metabolism , Cell Line, Tumor , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Epithelial-Mesenchymal Transition/genetics , Fluorescent Antibody Technique , G1 Phase Cell Cycle Checkpoints/genetics , Gene Expression Regulation, Neoplastic , HCT116 Cells , Humans , Microscopy, Electron, Transmission , POU Domain Factors/metabolism , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...