Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters











Publication year range
1.
Endoscopy ; 50(1): 85, 2018 01.
Article in English | MEDLINE | ID: mdl-29268296
2.
Medicine (Baltimore) ; 96(33): e7780, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28816962

ABSTRACT

Relationships between the capsule endoscopy Lewis score (LS) and clinical disease activity indices and C-reactive protein (CRP) are controversial in adult patients with Crohn's disease (CD). Also, data on pediatric patients are relatively less. However, correlation between LS and small bowel transit time (SBTT) remains investigational. The aim of the present study was to explore the correlations between LS and clinical disease activity indices, CRP, SBTT in pediatric, and adult patients with small bowel CD.Retrospective, single-center study on consecutive inpatients with established small bowel CD was conducted. The clinical disease activity index was determined using the abbreviated Pediatric Crohn's Disease Activity Index (aPCDAI) in patients aged <18 years and the Harvey-Bradshaw Simple Index (HBI) in adults. Spearman's rank correlation coefficient was used to assess the correlations of LS with aPCDAI, HBI, CRP, and SBTT, respectively.150 patients were enrolled (30 children and adolescents). In pediatric patients, correlations between LS and aPCDAI, CRP were moderate (r1 = 0.413; r2 = 0.379; P1 = .023; P2 = .044). There was no correlation between LS and SBTT (r = -0.029; P = .88). In adults, weak correlations were found between LS and HBI, SBTT (r1 = 0.213; r2 = 0.237; P1 = .019; P2 = .009). Correlation between LS and CRP was moderate (r = 0.326; P < .001). Strong correlations were found between CRP and HBI, aPCDAI (r1 = 0.522; r2 = 0.650; P < .001). The follow-up patients were all in clinical remission after treatment within 4 months, whereas only a minority reached mucosal healing. HBI, aPCDAI, CRP, and LS in all patients were reduced after treatment, whereas difference in CRP in pediatric patients and difference in LS in adults between baseline and follow-up were not found to be statistically significant. Also, the average SBTT at baseline was not found to be different from that at follow-up in all patients.The role of capsule endoscopy should be emphasized both in pediatric and adult patients with small bowel CD. Furthermore, the small bowel transit time may not be affected by the grade of small intestinal inflammation.


Subject(s)
Capsule Endoscopy/methods , Crohn Disease/pathology , Gastrointestinal Transit , Severity of Illness Index , Adolescent , Adult , Biomarkers , C-Reactive Protein/analysis , Child , China , Female , Humans , Male , Middle Aged , Retrospective Studies , Young Adult
3.
Sci Rep ; 7(1): 3709, 2017 06 16.
Article in English | MEDLINE | ID: mdl-28623323

ABSTRACT

RUFY3 is highly expressed in brain tissue and has a role in neuronal development. Transcriptional factor FOXK1 is involved in cell growth and metabolism. We knew that RUFY3 or FOXK1 has been correlated with the malignant of tumor cells. However, the role of these molecules in colorectal cancer (CRC) progression remains unknown. We investigated the protein expression levels by Western blot, immunofluorescence and immunohistochemistry analyses. The migration and invasive abilities of CRC cells were assessed using shRNA-mediated inhibition in vitro and in vivo. We showed that RUFY3 expression was up-regulated in CRC compared with its expression in a normal human colon cell line (FHC). RUFY3 suppression inhibited anchorage independent cell tumorigenesis. RUFY3 induced elevated expression of eight major oncogenes. Moreover, RUFY3 physically interacts with FOXK1 in CRC. A positive correlation was observed between the expression patterns of RUFY3 and FOXK1. Furthermore, RUFY3 and FOXK1 expression were correlated with tumor progression and represented significant predictors of overall survival in CRC patients. SiRNA-mediated repression of FOXK1 in RUFY3-overexpressing cells reversed the epithelial-mesenchymal transition (EMT) and metastatic phenotypes. In vivo, FOXK1 promoted RUFY3-mediated metastasis via orthotopic implantation. These findings suggest that the RUFY3-FOXK1 axis might promote the development and progression of human CRC.


Subject(s)
Colorectal Neoplasms/metabolism , Forkhead Transcription Factors/metabolism , rab5 GTP-Binding Proteins/metabolism , Biomarkers, Tumor , Cell Line, Tumor , Cell Movement/genetics , Colorectal Neoplasms/mortality , Colorectal Neoplasms/pathology , Cytoskeletal Proteins , Epithelial-Mesenchymal Transition , Genes, Reporter , Humans , Neoplasm Metastasis , Prognosis , Protein Binding , Protein Interaction Mapping
4.
Cancer Lett ; 390: 30-38, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28089833

ABSTRACT

Rufy3 is a RUN domain-containing protein that has been associated with gastric cancers; however, the role of Rufy3 in the progression of colorectal cancer (CRC) remains unknown. We demonstrated that Rufy3 expression was higher in 11/12 fresh CRC tissues than in adjacent normal tissues. Rufy3 induced elevated expression and transactivity of four major oncogenes in CRC. Moreover, siRNA-mediated repression of Rufy3 induced G0/G1 cell cycle arrest, and Rufy3 overexpression enhanced CRC cell proliferation in vitro and in vivo. Furthermore, Rufy3 up-regulation promoted epithelial-mesenchymal transition (EMT) and metastatic phenotypes. Using an established in vitro cell model of 5-fluorouracil-resistant (5-FU) CRC cells, we assessed cellular morphology, molecular changes, and invasion and found that these characteristics were consistent with EMT. Silencing of Rufy3 by siRNA reversed EMT and greatly diminished the invasion of 5-FU-treated cells. In addition, TGF-ß1 induced Rufy3 expression in a dose-dependent manner, and Rufy3 knockdown inhibited TGF-ß1-induced EMT. In vivo, higher expression of Rufy3 promoted CRC cell invasion and metastasis and induced EMT. Taken together, this work identified that Rufy3 promoted cancer metastasis in CRC cells through EMT induction.


Subject(s)
Colorectal Neoplasms/physiopathology , Epithelial-Mesenchymal Transition/genetics , rab5 GTP-Binding Proteins/metabolism , Colorectal Neoplasms/genetics , Cytoskeletal Proteins , Humans , Models, Biological , Neoplasm Metastasis/genetics , Prognosis
5.
Cell Death Dis ; 7(11): e2480, 2016 11 24.
Article in English | MEDLINE | ID: mdl-27882939

ABSTRACT

Forkhead box (FOX) K1 is a member of the FOX transcription factor superfamily. High FOXK1 expression is associated with several cancers. However, whether FOXK1 expression contributes to gastric cancer (GC) development and progression remains unknown. We analyzed the FOXK1 promoter using the Promo software and found several binding sequence transcription factors, including c-jun. However, the molecular mechanism by which FOXK1 affects the c-jun-mediated malignant phenotype is poorly understood. Here, we found that FOXK1 protein expression was higher in 8/10 (80.0%) fresh cancer tissues compared with that in adjacent normal tissues. FOXK1 overexpression enhanced the proliferation, migration and invasion of GC cells. Moreover, FOXK1 expression was stimulated by transforming growth factor-ß1 (TGF-ß1). FOXK1 acted as a potential epithelial-to-mesenchymal transition (EMT) inducer by stimulating vimentin expression and inducing the loss of E-cadherin in stable FOXK1-transfected cells. The results of promoter reporter and chromatin immunoprecipitation assays demonstrated that c-jun directly binds to and activates the human FOXK1 gene promoter. A positive correlation was observed between the expression patterns of FOXK1 and c-jun in GC cells and tissue. FOXK1 and c-jun expression were correlated with tumor progression and represented significant predictors of overall survival in GC patients. However, the siRNA-mediated repression of c-jun in FOXK1-overexpressing cells reversed EMT, as well as the proliferative and metastatic phenotypes. In vivo, c-jun promoted FOXK1-mediated proliferation and metastasis via orthotopic implantation. The evidence presented here suggests that FOXK1-directed regulation by c-jun promote the development and progression of human GC.


Subject(s)
Forkhead Transcription Factors/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Base Sequence , Carcinogenesis/drug effects , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Disease Progression , Epithelial-Mesenchymal Transition/drug effects , Humans , Neoplasm Invasiveness , Neoplasm Metastasis , Phenotype , Prognosis , Promoter Regions, Genetic/genetics , Proto-Oncogene Proteins c-jun/genetics , Stomach Neoplasms/genetics , Transcriptional Activation/drug effects , Transcriptional Activation/genetics , Transforming Growth Factor beta1/pharmacology
6.
Oncotarget ; 7(32): 51150-51162, 2016 08 09.
Article in English | MEDLINE | ID: mdl-27223064

ABSTRACT

Transcriptional factor FOXK1 is a member of the FOX family, involved in the cell growth and metabolism. The higher expression of FOXK1 leads to a variety of diseases and may play an important role in the development of various tumors. However, the role of FOXK1 in the progression of colorectal cancer (CRC) remains unknown. We demonstrated that FOXK1 was overexpressed in 16 types of solid tumor tissues via tissue multi-array (TMA). We found that FOXK1 induced elevated expressions and transactivities of five major oncogenes in CRC. Moreover, the elevated expression of FOXK1 was showed to be correlated with tumor progression and was a significant predictor of overall survival in CRC patients. Furthermore, it was showed that the depletion of FOXK1 expression could inhibit the migratory and invasive abilities of CRC cells. In contrast, ectopic expression of FOXK1 elicited the opposite effects on these phenotypes in vitro. FOXK1 promoted tumor metastasis through EMT program induction. In addition, TGF-ß1 induced FOXK1 expression in a time-dependent pattern and the knockdown of FOXK1 inhibited TGF-ß1-induced EMT. In vivo, higher expression of FOXK1 promotes CRC cell invasion and metastasis, and induces EMT in CRC as well. Alltogether, it was concluded that the higher expression of FOXK1 could indicate a poor prognosis in CRC patients since that FOXK1 induces EMT and promotes CRC cell invasion in vitro and in vivo.


Subject(s)
Cell Movement/genetics , Cell Proliferation/genetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Epithelial-Mesenchymal Transition/genetics , Forkhead Transcription Factors/physiology , Adult , Aged , Aged, 80 and over , Animals , Cell Line, Tumor , Disease Progression , Female , Forkhead Transcription Factors/genetics , Gene Expression Regulation, Neoplastic , HEK293 Cells , HT29 Cells , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Neoplasm Invasiveness , Neoplasm Metastasis
7.
Oncotarget ; 6(42): 44538-50, 2015 Dec 29.
Article in English | MEDLINE | ID: mdl-26517090

ABSTRACT

Chemoresistance is often associated with other clinical characteristics such as enhanced migratory/invasive potential. However, the correlation and underlying molecular mechanisms remain unclear. The aim of this study was to elucidate the function of the miR-222-ABCG2 pathway in the correlation between cisplatin (DDP) resistance and enhanced cell migration/invasion in tongue squamous cell carcinoma (TSCC). Using TSCC cell lines and primary cultures from TSCC cases, we first confirmed the correlation among DDP resistance (measured by IC50 values and ABCG2/ERCC1 expression), migratory/invasive potential (assessed by migration/invasion assays) and miR-222 expression. In TSCC cells, siRNA-mediated ABCG2 knockdown led to enhanced DDP responsiveness and reduced migratory/invasive potential, whereas ABCG2 overexpression induced DDP resistance and enhanced cell migration/invasion. Luciferase assays revealed that ABCG2 is a direct target of miR-222. In addition to reducing cell migration/invasion, functional analyses in TSCC cells indicated that miR-222 can reduce expression of the ABCG2 gene and enhance DDP responsiveness. However, co-transfection with ABCG2 cDNA restored both DDP resistance and migration/invasion. Moreover, miR-222 mimics and ABCG2 siRNA inhibited tumor growth and lung metastasis in vivo. Thus, our results verified that DDP resistance is correlated with enhanced migratory/invasive potential in TSCC. ABCG2 is a direct target of miR-222,and deregulation of the miR-222-ABCG2 regulatory module in TSCC contributes to both DDP resistance and enhanced migratory/invasive potential.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Antineoplastic Agents/pharmacology , Carcinoma, Squamous Cell/drug therapy , Cell Movement/drug effects , Cisplatin/pharmacology , Drug Resistance, Neoplasm , Head and Neck Neoplasms/drug therapy , Lung Neoplasms/prevention & control , MicroRNAs/metabolism , Neoplasm Proteins/metabolism , Tongue Neoplasms/drug therapy , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/genetics , Animals , Antineoplastic Agents/metabolism , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/secondary , Cell Line, Tumor , Cisplatin/metabolism , Dose-Response Relationship, Drug , Female , Gene Expression Regulation, Neoplastic , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/secondary , Humans , Inhibitory Concentration 50 , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Mice, Inbred BALB C , Mice, Nude , MicroRNAs/genetics , Neoplasm Invasiveness , Neoplasm Proteins/genetics , RNA Interference , Signal Transduction/drug effects , Squamous Cell Carcinoma of Head and Neck , Time Factors , Tongue Neoplasms/genetics , Tongue Neoplasms/metabolism , Tongue Neoplasms/pathology , Transfection , Tumor Burden/drug effects , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
8.
Tissue Eng Part A ; 15(11): 3261-70, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19382853

ABSTRACT

Synovial membrane has been shown to contain mesenchymal stem cells. We hypothesized that an enriched population of synovial fibroblasts would undergo chondrogenic differentiation and secrete cartilage extracellular matrix to a greater extent than would a mixed synovial cell population (MSCP). The optimum doses of transforming growth factor beta 1 (TGF-beta1) and insulin-like growth factor 1 (IGF-1) for chondrogenesis were investigated. CD14-negative isolation was used to obtain a porcine cell population enriched in type-B synovial fibroblasts (SFB) from an MSCP. The positive cell surface markers in SFB were CD90, CD44, and cadherin-11. SFB and MSCP were cultured in the presence of 20 ng/mL TGF-beta1 for 7 days, and SFB were demonstrated to have higher chondrogenic potential. Further dose-response studies were carried out using the SFB cells and several doses of TGF-beta1 (2, 10, 20, and 40 ng/mL) and/or IGF-1 (1, 10, 100, and 500 ng/mL) for 14 days. TGF-beta1 supplementation was essential for chondrogenesis and prevention of cell death, whereas IGF-1 did not have a significant effect on the SFB cell number or glycosaminoglycan production. This study demonstrates that the CD14-negative isolation yields an enhanced cell population SFB that is more potent than MSCP as a cell source for cartilage tissue engineering.


Subject(s)
Chondrogenesis/physiology , Fibroblasts/cytology , Fibroblasts/physiology , Hyaluronan Receptors/metabolism , Synovial Membrane/cytology , Synovial Membrane/physiopathology , Transforming Growth Factor beta1/pharmacology , Animals , Cell Culture Techniques/methods , Cells, Cultured , Chondrogenesis/drug effects , Fibroblasts/drug effects , Swine , Synovial Membrane/drug effects , Tissue Engineering/methods , Transforming Growth Factor beta1/metabolism
9.
Cancer Res ; 68(16): 6587-97, 2008 Aug 15.
Article in English | MEDLINE | ID: mdl-18701482

ABSTRACT

Alveolar rhabdomyosarcomas (ARMS) are highly malignant soft-tissue sarcomas that arise in children, adolescents, and young adults. Although formation and expression of the PAX-FKHR fusion genes is thought to be the initiating event in this cancer, the role of PAX-FKHR in the neoplastic process remains largely unknown in a progenitor cell that is undefined. We hypothesize that PAX-FKHR determine the ARMS progenitor to the skeletal muscle lineage, which when coupled to the inactivation and/or activation of critical cell signaling pathways leads to the formation of ARMS. Because a number of studies have proposed that mesenchymal stem cells (MSC) are the progenitor for several of the sarcomas, we tested this hypothesis in MSCs. We show that PAX-FKHR induce skeletal myogenesis in MSCs by transactivating MyoD and myogenin. Despite exhibiting enhanced growth in vitro, the PAX-FKHR-expressing populations do not form colonies in soft agar or tumors in mice. Expression of dominant-negative p53, or the SV40 early region, elicits tumor formation in some of the PAX-FKHR-expressing populations. Additional activation of the Ras signaling pathway leads to highly malignant tumor formation for all of the populations. The PAX-FKHR-expressing tumors were shown to have histologic, immunohistochemical, and gene expression profiles similar to human ARMS. Our results show the critical role played by PAX-FKHR in determining the molecular, myogenic, and histologic phenotype of ARMS. More importantly, we identify MSCs as a progenitor that can give rise to ARMS.


Subject(s)
Antigens, Polyomavirus Transforming/genetics , Forkhead Transcription Factors/genetics , Genes, ras/physiology , Mesenchymal Stem Cells/pathology , Mutation/genetics , Oncogene Proteins, Fusion/metabolism , PAX7 Transcription Factor/genetics , Paired Box Transcription Factors/genetics , Rhabdomyosarcoma, Alveolar/genetics , Rhabdomyosarcoma, Alveolar/pathology , Animals , Antigens, Viral, Tumor/genetics , Blotting, Western , Bone Marrow/metabolism , Bone Marrow/pathology , Cell Differentiation , Child , Fibroblasts/cytology , Fibroblasts/metabolism , Forkhead Box Protein O1 , Forkhead Transcription Factors/metabolism , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Macrophages/cytology , Macrophages/metabolism , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Mice, Nude , Muscle Development/physiology , Muscle, Skeletal , Myogenin/metabolism , PAX3 Transcription Factor , PAX7 Transcription Factor/metabolism , Paired Box Transcription Factors/metabolism , Rhabdomyosarcoma, Alveolar/metabolism , Tumor Suppressor Protein p53/physiology
10.
Mech Dev ; 125(5-6): 396-410, 2008.
Article in English | MEDLINE | ID: mdl-18359204

ABSTRACT

Vertebrate embryos generate striking Ca(2+) patterns, which are unique regulators of dynamic developmental events. In the present study, we used zebrafish embryos as a model system to examine the developmental roles of Ca(2+) during gastrulation. We found that gastrula stage embryos maintain a distinct pattern of cytosolic Ca(2+) along the dorsal-ventral axis, with higher Ca(2+) concentrations in the ventral margin and lower Ca(2+) concentrations in the dorsal margin and dorsal forerunner cells. Suppression of the endoplasmic reticulum Ca(2+) pump with 0.5 microM thapsigargin elevates cytosolic Ca(2+) in all embryonic regions and induces a randomization of laterality in the heart and brain. Affected hearts, visualized in living embryos by a subtractive imaging technique, displayed either a reversal or loss of left-right asymmetry. Brain defects include a left-right reversal of pitx2 expression in the dorsal diencephalon and a left-right reversal of the prominent habenular nucleus in the brain. Embryos are sensitive to inhibition of the endoplasmic reticulum Ca(2+) pump during early and mid gastrulation and lose their sensitivity during late gastrulation and early segmentation. Suppression of the endoplasmic reticulum Ca(2+) pump during gastrulation inhibits expression of no tail (ntl) and left-right dynein related (lrdr) in the dorsal forerunner cells and affects development of Kupffer's vesicle, a ciliated organ that generates a counter-clockwise flow of fluid. Previous studies have shown that Ca(2+) plays a role in Kupffer's vesicle function, influencing ciliary motility and translating the vesicle's counter-clockwise flow into asymmetric patterns of gene expression. The present results suggest that Ca(2+) plays an additional role in the formation of Kupffer's vesicle.


Subject(s)
Brain/embryology , Calcium/physiology , Dyneins/biosynthesis , Endoplasmic Reticulum/metabolism , Gastrula/physiology , Heart/embryology , Sodium-Calcium Exchanger/biosynthesis , T-Box Domain Proteins/biosynthesis , Zebrafish Proteins/biosynthesis , Animals , Body Patterning , Calcium/metabolism , Dyneins/physiology , Fetal Proteins , Gene Expression Regulation, Developmental , In Situ Hybridization , Models, Biological , Sodium-Calcium Exchanger/physiology , T-Box Domain Proteins/physiology , Thapsigargin/pharmacology , Zebrafish , Zebrafish Proteins/physiology
11.
Di Yi Jun Yi Da Xue Xue Bao ; 24(9): 1045-7, 2004 Sep.
Article in Chinese | MEDLINE | ID: mdl-15447859

ABSTRACT

OBJECTIVE: To investigate the effects of recombinant Helicobacter pylori catalase (rHpCAT)on oxidative stress in rat colonic mucosal epithelial cells. METHODS: Oxidative stress model was established by hydroxyl generated from Fenton reaction in cultured colonic mucosal epithelial cells isolated from normal rats, in the model of which the effects of rHpCAT were observed. The cells were divided into normal control, model, 5-aminosalicylic acid (5-ASA, 0.1 mmol/L), and rHpCAT (1 x 10(5), 1 x 10(6), and 1 x 10(7) U/kg, respectively) groups. At the end of the experiment, the content of lactic dehydrogenase (LDH), malondialdehyde (MDA), myeloperoxidase (MPO), glutathione peroxidase (GSH-Px), catalase (CAT) and, superoxide dismutase (SOD) were detected in the culture supernatant. RESULTS: The contents of LDH, MDA and MPO were elevated while those of GSH-Px, CAT and SOD reduced in the model group. rHpCAT at different doses reduced the release of LDH, depressed the contents of MDA and MPO, and increased the contents of GSH-Px, SOD and CAT. CONCLUSION: rHpCAT has protective effects against rat colonic mucosal oxidative damage.


Subject(s)
Catalase/pharmacology , Colon/metabolism , Helicobacter pylori/enzymology , Oxidative Stress/drug effects , Animals , Catalase/biosynthesis , Catalase/genetics , Cells, Cultured , Colon/cytology , Female , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , Male , Rats , Rats, Sprague-Dawley , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology
12.
World J Gastroenterol ; 10(11): 1630-3, 2004 Jun 01.
Article in English | MEDLINE | ID: mdl-15162538

ABSTRACT

AIM: To observe competitive inhibition of adherence of enterotoxigenic Escherichia coli (ETEC), enteropathogenic Escherichia coli (EPEC) and Clostridium difficile (C. difficile) to intestinal epithelial cell line Lovo by purified adhesin of Bifidobacterium adolescentis 1027 (B. ado 1027). METHODS: The binding of bacteria to intestinal epithelial cell line Lovo was counted by adhesion assay. The inhibition of adherence of ETEC, EPEC and C. difficile to intestinal epithelial cell line Lovo by purified adhesin of B. ado 1027 was evaluated quantitatively by flow cytometry. RESULTS: The purified adhesin at the concentration of 10 microg/mL, 20 microg/mL and 30 microg/mL except at 1 microg/mL and 5 microg/mL could inhibit significantly the adhesion of ETEC, EPEC and C. difficile to intestinal epithelial cell line Lovo. Moreover, we observed that a reduction in bacterial adhesion was occurred with increase in the concentration of adhesin, and MFI (Mean fluorescent intensity) was decreased with increase in the concentration of adhesin. CONCLUSION: The purified adhesin of B. ado 1027 can inhibit the adhesion of ETEC, EPEC and C. difficile to intestinal epithelial cell line Lovo in a dose-dependent manner.


Subject(s)
Adhesins, Bacterial/pharmacology , Bacterial Adhesion/drug effects , Clostridioides difficile/physiology , Enterocolitis, Pseudomembranous/prevention & control , Escherichia coli Infections/prevention & control , Escherichia coli/physiology , Bifidobacterium , Cell Line , Clostridioides difficile/pathogenicity , Diarrhea/prevention & control , Escherichia coli/pathogenicity , Flow Cytometry , Humans , Intestinal Mucosa/cytology , Intestinal Mucosa/microbiology , Virulence
13.
World J Gastroenterol ; 10(6): 881-4, 2004 Mar 15.
Article in English | MEDLINE | ID: mdl-15040037

ABSTRACT

AIM: To investigate frequency and clinical significance of K-ras mutations in pancreatic diseases and to identify its diagnostic values in pancreatic carcinoma. METHODS: 117 ductal lesions were identified in the available sections from pancreatic resection specimens of pancreatic ductal adenocarcinoma, comprising 24 pancreatic ductal adenocarcinoma, 19 peritumoral ductal atypical hyperplasia, 58 peritumoral ductal hyperplasia and 19 normal duct at the tumor free resection margin. 24 ductal lesions were got from 24 chronic pancreatitis. DNA was extracted. Codon 12 K-ras mutations were examined using the two-step polymerase chain reaction (PCR) combined with restriction enzyme digestion, followed by nonradioisotopic single-strand conformation polymorphism (SSCP) analysis and by means of automated DNA sequencing. RESULTS: K-ras mutation rate of the pancreatic carcinoma was 79%(19/24) which was significantly higher than that in the chronic pancreatitis 33%(8/24) (P<0.01). It was also found that K-ras mutation rate was progressively increased from normal duct at the tumor free resection margin, peritumoral ductal hyperplasia, peritumoral ductal atypical hyperplasia to pancreatic ductal adenocarcinoma. The mutation pattern of K-ras 12 codon of chronic pancreatitis was GGT-GAT, GGT and CGT, which is identical to that in pancreatic carcinoma. CONCLUSION: K-ras mutation may play a role in the malignant transformation of pancreatic ductal cell. K-ras mutation was not specific enough to diagnose pancreatic carcinoma.


Subject(s)
Codon/genetics , Genes, ras/genetics , Pancreatic Diseases/genetics , Point Mutation , Humans , Pancreatic Neoplasms/diagnosis , Pancreatic Neoplasms/genetics , Polymerase Chain Reaction , Polymorphism, Single-Stranded Conformational
SELECTION OF CITATIONS
SEARCH DETAIL