Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
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.
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
4.
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
5.
Cell Death Discov ; 7(1): 129, 2021 Jun 01.
Article in English | MEDLINE | ID: mdl-34075026

ABSTRACT

Colorectal cancer (CRC) is the most common form of gastrointestinal malignancies. A growing number of reports focusing on oxaliplatin (OXA) resistance in CRC treatment have revealed that drug resistance is an urgent issue in clinical applications, especially for finding effective therapeutic targets. Recently, microRNAs (miRNAs) are reported to play a critical role in tumor progressions and multi-drug resistance. The main aim of this study is to establish whether miR-5000-3p is an oncogene that is resistant to OXA and further confirm its underlying regulatory role in CRC. The OXA-associated gene expression dataset in CRC cells was downloaded from Gene Expression Omnibus (GEO) database. Statistical software R was used for significance analysis of differentially expressed genes (DEGs) between OXA-resistant (OR)-CRC cells and CRC cells, and results indicated ubiquitin-specific peptidase 49 (USP49) was upregulated in OR-CRC cells. Luciferase reporter assay showed that USP49 was verified to act as a downstream target gene of miR-5000-3p. From the results of TCGA database, miR-5000-3p expression was upregulated and USP49 was downregulated in patients with CRC. The function of miR-5000-3p was detected using MTT assay, wound healing, Transwell, and flow cytometry assays. Moreover, through in vitro and in vivo experiments, miR-5000-3p expression was confirmed to be upregulated in CRC cells or OR-CRC cells comparing to normal cell lines. Molecular mechanism assays revealed that USP49 binds to the miR-5000-3p promoter to increase the expression of miR-5000-3p, resulting in cancer cells sensitized to OXA. To sum up, these results suggest that miR-5000-3p may be a novel biomarker involved in drug-resistance progression of CRC. Moreover, the drug-resistance mechanism of miR-5000-3p/USP49 axis provides new treatment strategies for CRC in clinical trials.

7.
Cell Death Dis ; 12(6): 529, 2021 05 22.
Article in English | MEDLINE | ID: mdl-34023852

ABSTRACT

At present, colorectal cancer (CRC) has become a serious threat to human health in the world. Dipeptidyl peptidase 3 (DPP3) is a zinc-dependent hydrolase that may be involved in several physiological processes. However, whether DPP3 affects the development and progression of CRC remains a mystery. This study is the first to demonstrate the role of DPP3 in CRC. Firstly, the results of immunohistochemistry analysis showed the upregulation of DPP3 in CRC tissues compared with normal tissues, which is statistically analyzed to be positively correlated with lymphatic metastasis, pathological stage, positive number of lymph nodes. Moreover, the high expression of DPP3 predicts poor prognosis in CRC patients. In addition, the results of cell dysfunction experiments clarified that the downregulation of DPP3 significantly inhibited cell proliferation, colony formation, cell migration, and promoted apoptosis in vitro. DPP3 depletion could induce cell apoptosis by upregulating the expression of BID, BIM, Caspase3, Caspase8, HSP60, p21, p27, p53, and SMAC. In addition, downregulation of DPP3 can reduce tumorigenicity of CRC cells in vivo. Furthermore, CDK1 is determined to be a downstream target of DPP3-mediated regulation of CRC by RNA-seq, qPCR, and WB. The interaction between DPP3 and CDK1 shows mutual regulation. Specifically, downregulation of DPP3 can accentuate the effects of CDK1 knockdown on the function of CRC cells. Overexpression of CDK1 alleviates the inhibitory effects of DPP3 knockdown in CRC cells. In summary, DPP3 has oncogene-like functions in the development and progression of CRC by targeting CDK1, which may be an effective molecular target for the prognosis and treatment of CRC.


Subject(s)
CDC2 Protein Kinase/physiology , Colorectal Neoplasms/pathology , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/physiology , Adult , Aged , Aged, 80 and over , Animals , Apoptosis/drug effects , Apoptosis/genetics , CDC2 Protein Kinase/genetics , Case-Control Studies , Cell Movement/drug effects , Cell Movement/genetics , Cell Proliferation/drug effects , Cell Proliferation/genetics , Colorectal Neoplasms/diagnosis , Colorectal Neoplasms/genetics , Colorectal Neoplasms/mortality , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/antagonists & inhibitors , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/genetics , Disease Progression , Female , Gene Expression Regulation, Neoplastic/drug effects , HCT116 Cells , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Prognosis , RNA, Small Interfering/pharmacology , Tumor Cells, Cultured
8.
J Anim Physiol Anim Nutr (Berl) ; 103(5): 1374-1379, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31309620

ABSTRACT

The ruminal microbiota of 15 dairy buffalo was characterized using high-throughput 16S rRNA gene amplicon sequencing. Results showed that Bacteroidetes was the dominant bacterial phylum in all rumen samples, followed by Firmicutes, Proteobacteria, Tenericutes and Verrucomicrobia. Butyrivibrio was positively correlated with average milk fat yield (R = 0.55; p = 0.03), average milk total solid yield (R = 0.56; p = 0.03) and standard milk yield (R = 0.52; p = 0.05). Acinetobacter were positively correlated with average milk protein yield (R = 0.56; p = 0.03), average milk total solid yield (R = 0.60; p = 0.02) and standard milk yield (R = 0.57; p = 0.03). Acetobacter was positively correlated with acetate (R = 0.63; p = 0.01), propionate content (R = 0.55; p = 0.03), butyrate content (R = 0.61; p = 0.02) and total VFA (R = 0.62; p = 0.01). The phyla Proteobacteria (R = 0.53; p = 0.04) and genus Prevotella (R = 0.52; p = 0.05) were positively correlated with butyrate content. Correlation analysis suggested that increased Butyrivibrio and Acinetobacter residing in the buffalo rumen could improve milk performance.


Subject(s)
Bacteria/classification , Buffaloes/physiology , Lactation/physiology , Rumen/microbiology , Animals , Buffaloes/microbiology , China , Female , Fermentation
9.
Biomed Pharmacother ; 116: 109000, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31152930

ABSTRACT

Aberrant expression of SNHG8 has been observed in some types of cancers. However, whether SNHG8 was aberrantly expressed in colorectal cancer and whether it could exert any function on the development of colorectal cancer remains largely elusive. In this study, we first investigated the expression pattern and biological function of SNHG8 in colorectal cancer. The expression level of SNHG8 was investigated in colorectal cancer tissues as well as in colorectal cancer cell lines by real-time PCR. Next, CCK8 assays were performed to evaluate the effects of SNHG8 on the proliferation of colorectal cancer cells and transwell assays were employed to evaluate migration and invasion. Bioinformatics were used for predicting the sponging miRNAs that interact with SNHG8. A dual luciferase reporter assay was adopted for the verification of interaction between SNHG8 and miRNA. Our data showed that SNHG8 was significantly up-regulated in colorectal cancer tissues and cell lines. In addition, knockdown of SNHG8 significantly inhibited the growth, migration, and invasion of colorectal cancer cells. It was predicted that miR-663 might interact with SNHG8 and the direct sponging was verified by dual luciferase reporter assay. Moreover, rescue experiments revealed that SNHG8 played a tumor promoting role by regulating miR-663. In the present study, we revealed that SNHG8 was up-regulated in colorectal cancer and promoted the proliferation, migration, and invasion of colorectal cancer by sponging miR-663, which helps to further reveal the underlying developmental mechanism of action and provides a potential therapeutic molecule for colorectal cancer therapy in the future.


Subject(s)
Cell Movement/genetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Gene Knockdown Techniques , MicroRNAs/metabolism , RNA, Long Noncoding/metabolism , Base Sequence , Cell Line, Tumor , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , Humans , MicroRNAs/genetics , Prognosis , RNA, Long Noncoding/genetics , Up-Regulation
10.
Bioresour Technol ; 285: 121315, 2019 Aug.
Article in English | MEDLINE | ID: mdl-30965280

ABSTRACT

Investigating the dynamic changes in bacterial community composition during sugarcane top silage production starting in late March and finishing in late June (storage temperature: 20 to 35 °C) will advance our understanding of ensilage in hot ambient temperatures. The results showed that, the fermentation process was dominated by Leuconostocaceae (until d 5), followed by Lactobacillaceae (from d 5 to d 30), and finally Lactobacillaceae and Clostridium (from d 60 to d 90). As the fermentation process progressed, there was a significant increase in Lactobacillaceae abundance, and on d 60 there was a sharp increase in Clostridiaceae abundance. Spearman's correlation showed that, Lactococcus and Leuconostoc abundance were negatively correlated with acetate, propionate, butyrate, and ammonia-N levels. Clostridiaceae and Lactobacillaceae abundance were positively correlated with acetate, propionate, butyrate, and ammonia-N levels. The high moisture content (DM 24.31%) of sugarcane tops stored in hot ambient temperatures may result in butyric acid fermentation.


Subject(s)
Saccharum , Silage , Acetic Acid , Bacteria , Fermentation
11.
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
12.
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.

13.
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
14.
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
15.
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
16.
Int J Pharm ; 505(1-2): 24-34, 2016 May 30.
Article in English | MEDLINE | ID: mdl-27039150

ABSTRACT

RNA interfere (RNAi)-based technology holds great promise in cancer treatment. The use of small interfering RNA (siRNA), however, is hampered by its low delivery efficiency in vivo when they are diluted in blood biofluids and in the presence of serum and salt. In this study, we developed the polyglutamate derivative polymer brush, poly(ethyleneglycol) monomethyl ether-b-polyglutamate-g-spermine (mPEG-b-PG-g-spermine, PPGS), which could efficiently deliver survivin-siRNA under ultra-high dilution and in the presence of salt (NaCl 150mM) and serum (10% FBS), most likely due to its PEG-shelled polymer brush structure. On the contrary, aggregation occurred when PEI/siRNA polyplex dispersed in saline and serum-containing media and PEI polyplex dissociated after making a 256-fold dilution. PPGS/si-survivin polyplex exhibited high cellular uptake efficiency and efficiently down-regulated the expression of survivin mRNA in the cisplatin-resistance of non-small cell human lung adenocarcinoma (A549/DDP) cells in the presence of serum. However, either PEI polyplex or Lipofectmine 2000 complex was unstable in serum and salt-containing media and at high dilution rates, which resulted in their dramatical decrease of cellular uptake and gene-silencing efficiency in these conditions. The PPGS/si-survivin polyplex also exhibited synergistic effects of killing the cancer cells by combination treatment with cisplatin. Therefore, the PPGS gene carrier showed great potential in systemic siRNA delivery, and its combination with chemotherapeutic drug is promising in treating drug resistant cancers.


Subject(s)
Cisplatin/pharmacology , Inhibitor of Apoptosis Proteins/genetics , Lung Neoplasms/drug therapy , RNA, Small Interfering/administration & dosage , Adenocarcinoma/drug therapy , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cisplatin/administration & dosage , Down-Regulation/genetics , Drug Resistance, Neoplasm , Gene Transfer Techniques , Humans , Lipids/chemistry , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Polyethylene Glycols/chemistry , Polyglutamic Acid/chemistry , RNA Interference , Spermine/chemistry , Survivin
17.
Mol Cell Biochem ; 413(1-2): 165-78, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26724951

ABSTRACT

In previous studies, we have reported the abnormal proliferation and differentiation of intestinal epithelial cells (IECs) in diabetes mellitus (DM) mice. The insulin receptor (IR) and its downstream mitogen-activated protein kinase kinase (MAPKK also known as MEK)/extracellular-regulated protein kinase (ERK) pathway is a classic pathway associated with cell proliferation and differentiation. The purpose of the present study is to investigate the role of the MEK/ERK pathway in abnormal proliferation and differentiation of IECs in DM mice. DM mouse models were induced by intraperitoneal injection of streptozotocin. The expression levels of the IR and its isoforms in IECs of DM mice and in IEC-6 cells were investigated. To ensure that the downstream pathways were monitored, QPCR and Western blotting were performed to detect the expression levels of MEK1/2, ERK1/2, PI3K, and Akt. Moreover, siRNA for IR-A and U0126, a specific inhibitor of MEK, were used to further investigate the relationship between the IR/MEK/ERK pathway and abnormal proliferation and differentiation of IECs in DM mice. In DM mice, excessive proliferation, disturbed differentiation, and a high ratio of IR-A/IR-B were detected in IECs. The expression levels of MEK1, MEK2, and ERK1/2 and their phosphorylated proteins in DM mice were significantly higher than those in the control group (P < 0.05), which could be offset by using siRNA for IR-A. The abnormal proliferation and differentiation of IECs in DM mice were normalized after the in vivo administration of U0126. The abnormal proliferation and differentiation of IECs in DM mice are associated with high IR-A/IR-B ratio and increased IR/MEK/ERK pathway activity.


Subject(s)
Diabetes Mellitus, Experimental/pathology , Epithelial Cells/cytology , Intestinal Mucosa/cytology , MAP Kinase Signaling System , Receptor, Insulin/metabolism , Animals , Butadienes/administration & dosage , Butadienes/pharmacology , Cell Differentiation/drug effects , Cell Line , Cell Proliferation/drug effects , Diabetes Mellitus, Experimental/metabolism , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , MAP Kinase Signaling System/drug effects , Mice , Nitriles/administration & dosage , Nitriles/pharmacology , Protein Isoforms/genetics , Protein Isoforms/metabolism , Receptor, Insulin/genetics , Streptozocin
18.
Cell Prolif ; 49(1): 102-14, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26786283

ABSTRACT

OBJECTIVES: Depression of the Notch/Hes1 pathway has been reported to play a role in abnormal differentiation of intestinal epithelial cells (IECs) in diabetes mellitus (DM). However, the mechanism by which this pathway influences IEC differentiation has remained unclear. In this study, we have investigated the role of microRNAs (miRNAs) in regulating the Notch/Hes1 pathway in IECs of DM mice. MATERIALS AND METHODS: Integrated comparative miRNA microarray technology was used to determine the expression profile of miRNAs in IECs of DM mice. After bioinformatic analysis, an miRNA with altered expression levels, miRNA-30e, was identified as a candidate for regulating the Notch pathway in DM. A luciferase reporter assay confirmed that miRNA-30e targeted 3'-UTR of the Notch gene. The role of miRNA-30e in regulating Notch signalling was then explored by up- and downregulating its expression in vitro and in vivo. RESULTS: Abnormal differentiation of IECs in DM mice was associated with reduced activity of the Dll4/NICD/Hes1 signal pathway. Based on bioinformatic analyses, increased expression of miRNA-30e was identified as a potential candidate for regulating Notch signalling. miRNA-30e targeted the 3'-UTR of Dll4 and downregulated Dll4 expression in primary IECs and IEC-6 cells. Exogenous miRNA-30e reduced activity of the Dll4/NICD/Hes1 pathway, and induced abnormal differentiation of IECs in normal mice. Conversely, treatment with miRNA-30e antagonist upregu-lated activity of the Dll4/NICD/Hes1 pathway in vivo, and normalized IEC differentiation in DM mice. CONCLUSIONS: Increased levels of miRNA-30e downregulated activity of the Dll4/NICD/Hes1 signalling pathway by targeting the 3'-UTR of Dll4, which contributed to abnormal differentiation in small intestinal epithelia of DM mice.


Subject(s)
Cell Differentiation/genetics , Diabetes Mellitus, Experimental/genetics , Down-Regulation/genetics , Enterocytes/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , MicroRNAs/metabolism , 3' Untranslated Regions/genetics , Adaptor Proteins, Signal Transducing , Animals , Base Sequence , Calcium-Binding Proteins , Cell Line , Diabetes Mellitus, Experimental/pathology , Enterocytes/pathology , Gene Expression Profiling , Intestinal Mucosa/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Mice, Inbred C57BL , Molecular Sequence Data , Oligonucleotide Array Sequence Analysis , Signal Transduction/genetics
19.
Clin Res Hepatol Gastroenterol ; 40(1): 110-23, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26162991

ABSTRACT

OBJECTIVE: Recent studies proved that patients with diabetes were at significantly higher risk of developing colorectal cancer. However, the association between diabetes mellitus and the risk of colorectal adenoma remains undefined. Thus we conducted an updated meta-analysis to identify the association between diabetes mellitus and the risk of colorectal neoplasia including adenoma and cancer. METHODS: We conducted a search in databases including Pubmed, Web of Science, EMBASE Databases, Cochrane CENTRAL, Wanfang Data, and CNKI database. Case-control and cohort studies were included. All articles were published before January 2015 and the quality of each study was evaluated by the Newcastle-Ottawa Scale. Odds ratios (ORs) or relative risks (RRs) and its corresponding 95% confidence intervals (CIs) for each study were calculated and summary relative risk estimates with corresponding 95% CIs were generated using the random-effects model. Heterogeneity and publication bias were assessed. RESULTS: Twenty-nine articles including ten case-control studies and nineteen cohort studies were included in this meta-analysis. In a pooled analysis of all studies, diabetes mellitus was associated with increased risk of colorectal neoplasia (RR=1.35, 95% CI=1.28-1.42). The risk increased significantly for both colorectal cancer (RR=1.37, 95% CI=1.30-1.45) and adenoma (RR=1.26, 95% CI=1.11-1.44). Subgroup analyses on study design, gender, geographical region, and type of diabetes mellitus further evidenced these findings. CONCLUSIONS: Diabetes mellitus was associated with an increased risk of colorectal neoplasia. Not only the increased risk of colorectal cancer but also the higher risk of adenoma was identified in patients with diabetes mellitus.


Subject(s)
Colorectal Neoplasms/epidemiology , Colorectal Neoplasms/etiology , Diabetes Complications/epidemiology , Humans , Risk Factors
20.
Dev Growth Differ ; 57(6): 453-465, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26122164

ABSTRACT

Intestinal epithelial stem cells (IESCs) can differentiate into all types of intestinal epithelial cells (IECs) and Leucine-rich repeat-containing G protein-coupled receptor 5 (Lgr5) is a marker for IESC. Previous studies reported enhanced proliferation of IECs in diabetic mice. In this study, the in vitro differentiation of Lgr5 positive IESCs sorted from diabetic mice was further investigated. The diabetic mouse model was induced by streptozotocin (STZ), and crypt IECs were isolated from small intestines. Subsequently, Lgr5 positive IESCs were detected by flow cytometry (FCM) and sorted by magnetic activated cell sorting (MACS). Differentiation of the sorted IESCs was investigated by detecting the IEC markers in the diabetic mice using immunostaining, quantitative real-time reverse-transcription polymerase chain reaction (qRT-PCR), and Western blot analysis, which was compared with normal mice. We found that the proportion of Lgr5 positive cells in the crypt IECs of diabetic mice was higher than that of control mice (P < 0.05). Lgr5 positive IESCs could be significantly enriched in Lgr5 positive cell fraction sorted by MACS. Furthermore, the absorptive cell marker sucrase-isomaltase (SI) and the Paneth cell marker lysozyme 1 (Lyz1) were more highly expressed in the differentiated cells derived from Lgr5 positive IESCs of diabetic mice in vitro (P < 0.05). We demonstrate that the number of Lgr5 positive IESCs is significantly increased in the small intestines of STZ-induced diabetic mice. Lgr5 positive IESCs sorted from the diabetic mice can differentiate into a higher proportion of absorptive cells and Paneth cells in vitro. We characterized the expression of Lgr5 in the small intestine of diabetic mice, and sorted Lgr5 positive intestinal epithelial stem cells (IESCs) for investigating their differentiation in vitro. We proved that the quantity of Lgr5 positive IESCs was significantly increased in the small intestines of diabetic mice. IESCs sorted from the diabetic mice can differentiate into a higher proportion of absorptive cells and Paneth cells in vitro.


Subject(s)
Cell Differentiation , Diabetes Mellitus, Experimental/pathology , Intestine, Small/pathology , Paneth Cells/physiology , Receptors, G-Protein-Coupled/metabolism , Stem Cells , Animals , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/metabolism , Female , Immunomagnetic Separation , Intestinal Absorption , Intestinal Mucosa/pathology , Intestinal Mucosa/physiology , Intestine, Small/metabolism , Mice , Mice, Inbred C57BL , Paneth Cells/pathology , Stem Cells/metabolism , Stem Cells/pathology , Stem Cells/physiology , Streptozocin
SELECTION OF CITATIONS
SEARCH DETAIL
...