Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
J Dig Dis ; 24(11): 630-637, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37823361

ABSTRACT

OBJECTIVES: Laser lithotripsy under fluoroscopic guidance is difficult to perform and risky due to its invisibility. In this study we aimed to investigate the efficacy and safety of a novel endoscopic auxiliary system (NEAS)-assisted lithotripsy under fluoroscopy for treating difficult common bile duct (CBD) stones. METHODS: Patients with difficult CBD stones who were treated with NEAS-assisted laser lithotripsy (NEAS group) or conventional mechanical lithotripsy (ML) under fluoroscopy (ML group) were retrospectively evaluated. The primary outcome was the complete stone clearance rate and the secondary outcomes included operation time, complications, and medical cost. RESULTS: Seventeen patients were treated with NEAS-assisted laser lithotripsy and 144 patients underwent ML. Using the propensity score matching analysis, 17 pairs of cases treated with NEAS-assisted lithotripsy and ML were included. Patients in the NEAS group showed a higher stone clearance rate than the ML group (94.1% vs 58.8%, P = 0.039), as well as shorter operation time (41.9 min vs 49.4 min, P < 0.001) and lower medical cost (USD 4607 vs USD 5014, P < 0.001). There was no significant difference in the complication rate between the two groups (5.9% vs 17.6%, P = 0.601). CONCLUSION: NEAS-assisted fluoroscopy-guided laser lithotripsy is feasible and safe, which may be a promising technique in fluoroscopy-guided laser lithotripsy for difficult CBD stones.


Subject(s)
Gallstones , Lithotripsy , Humans , Gallstones/diagnostic imaging , Gallstones/therapy , Gallstones/etiology , Pilot Projects , Retrospective Studies , Treatment Outcome , Lithotripsy/adverse effects , Cholangiopancreatography, Endoscopic Retrograde/methods , Fluoroscopy
2.
Neoplasma ; 69(4): 820-831, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35532294

ABSTRACT

Obesity is closely related to the initiation and development of hepatocellular carcinoma (HCC). The regulatory mechanism of obesity-associated HCC remains unclear. HepG2 cells treated with palmitic acid (PA) and diethylnitrosamine (DEN)-induced HCC mice fed a high-fat diet (HFD) were established. The expression of miR-27a and B-cell translocation gene 2 (BTG2) mRNA and protein were detected via qPCR and western blotting. Prediction software and luciferase assays were employed to verify the miR-27a/BTG2 axis. The biological effects of HepG2 cells were evaluated with ORO staining, MTT assays, Transwell assays, Mito-Timer, and Mito-SOX staining. Significantly upregulated miR-27a and downregulated BTG2 mRNA and protein were observed in HepG2 cells and liver tissues of HCC mice. Overexpressing miR-27a (mi-miR-27a) markedly promoted cellular lipid accumulation, proliferation, and invasion, accompanied by aggravated mitochondrial dysfunction (increased fading and ROS products of mitochondria) in HepG2 cells. Additionally, these effects were further reinforced in HepG2 cells treated with mi-miR-27a and PA. BTG2 was identified as a direct target and was negatively regulated by miR-27a. Similarly, BTG2 knockdown (sh-BTG2) had effects identical to those of mi-miR-27a on HepG2 cells. Additionally, PA evidently enhanced these effects of sh-BTG2 in HepG2 cells. Moreover, BTG2 overexpression effectively reversed the effects of miR-27a, including lipotropic and oncogenic effects, and simultaneously promoted mitochondrial imbalance in HepG2 cells. Thus, obesity-associated miR-27a acts as an oncogene to promote lipid accumulation, proliferation, and invasion by negatively regulating BTG2-mediated mitochondrial dysfunction in HCC.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , MicroRNAs , Animals , Carcinogenesis/genetics , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , Lipids , Liver Neoplasms/pathology , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , Mitochondria , Obesity/complications , Oncogenes , RNA, Messenger
3.
Mol Ther Nucleic Acids ; 26: 1241-1254, 2021 Dec 03.
Article in English | MEDLINE | ID: mdl-34853724

ABSTRACT

The role of exosome-mediated mitophagy in the crosstalk between hepatocytes (HCs) and hepatic stellate cells (HSCs) in metabolic-associated fatty liver disease (MAFLD) remains unknown. Serum exosomal miR-27a levels were markedly increased and positively correlated with liver fibrosis in MAFLD patients and mice. Exosomal miR-27a was released from lipotoxic HCs and specifically transmitted to recipient-activated HSCs. PINK1, the key target of miR-27a, primarily mediates mitophagy. Overexpression of miR-27a or knockdown of PINK1 or lipotoxic HC-exosomal miR-27a impaired mitochondria (inhibiting mitophagy, respiration, membrane potential, and transcription while promoting reactive oxygen species production) in activated HSCs and stimulated HSC-derived fibroblasts (promoting activation and proliferation while inhibiting autophagy). High exosomal miR-27a serum levels and a lack of hepatic PINK1-mediated mitophagy were directly related to liver fibrosis in MAFLD mice. Lipotoxic HC exosome transplantation aggravated the degree of PINK1-mediated mitophagy suppression, steatohepatitis, lipidosis, and fibrosis in the livers of MAFLD mice with cirrhosis. Both in vitro and in vivo, exosomes derived from miR-27a-knockdown HCs could not facilitate the abovementioned deteriorating effects. In conclusion, lipotoxic HC-exosomal miR-27a plays a pivotal role in inhibiting mitophagy and in promoting MAFLD-related liver fibrosis by negatively regulating PINK1 expression.

5.
World J Gastroenterol ; 27(14): 1419-1434, 2021 Apr 14.
Article in English | MEDLINE | ID: mdl-33911465

ABSTRACT

BACKGROUND: Exosomes play an important role in metabolic-associated fatty liver disease (MAFLD), but the mechanism by which exosomes participate in MAFLD still remain unclear. AIM: To figure out the function of lipotoxic exosomal miR-1297 in MAFLD. METHODS: MicroRNA sequencing was used to detect differentially expressed miRNAs (DE-miR) in lipotoxic exosomes derived from primary hepatocytes. Bioinformatic tools were applied to analyze the target genes and pathways regulated by the DE-miRs. Quantitative real-time PCR (qPCR) was conducted for the verification of DE-miRs. qPCR, western blot, immunofluorescence staining and ethynyl-20-deoxyuridine assay were used to evaluate the function of lipotoxic exosomal miR-1297 on hepatic stellate cells (LX2 cells). A luciferase reporter experiment was performed to confirm the relationship of miR-1297 and its target gene PTEN. RESULTS: MicroRNA sequencing revealed that there were 61 exosomal DE-miRs (P < 0.05) with a fold-change > 2 from palmitic acid treated primary hepatocytes compared with the vehicle control group. miR-1297 was the most highly upregulated according to the microRNA sequencing. Bioinformatic tools showed a variety of target genes and pathways regulated by these DE-miRs were related to liver fibrosis. miR-1297 was overexpressed in exosomes derived from lipotoxic hepatocytes by qPCR. Fibrosis promoting genes (α-SMA, PCNA) were altered in LX2 cells after miR-1297 overexpression or miR-1297-rich lipotoxic exosome incubation via qPCR and western blot analysis. Immunofluorescence staining and ethynyl-20-deoxyuridine staining demonstrated that the activation and proliferation of LX2 cells were also promoted after the above treatment. PTEN was found to be the target gene of miR-1297 and knocking down PTEN contributed to the activation and proliferation of LX2 cells via modulating the PI3K/AKT signaling pathway. CONCLUSION: miR-1297 was overexpressed in exosomes derived from lipotoxic hepatocytes. The lipotoxic hepatocyte-derived exosomal miR-1297 could promote the activation and proliferation of hepatic stellate cells through the PTEN/PI3K/AKT signaling pathway, accelerating the progression of MAFLD.


Subject(s)
Exosomes , MicroRNAs , Exosomes/genetics , Exosomes/metabolism , Hepatic Stellate Cells/metabolism , Hepatocytes/metabolism , MicroRNAs/genetics , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction
6.
J Exp Clin Cancer Res ; 40(1): 26, 2021 Jan 09.
Article in English | MEDLINE | ID: mdl-33422101

ABSTRACT

BACKGROUND: Increasing evidence has demonstrated that long noncoding RNAs (lncRNAs) have regulatory functions in hepatocellular carcinoma (HCC). The link between lincSCRG1 and HCC remains unclear. METHODS: To explore the lincSCRG1 regulation axis, bioinformatics, RIP and luciferase reporter assay were performed. The expressions of lincSCRG1-miR26a-SKP2 were detected in HCC tissues and cell lines through qPCR and western blot. The functions of HCC cells were investigated through in vitro assays (MTT, colony formation, transwell and flow cytometry) and the inner effect of lincSCRG1-miR26a in vivo was evaluated by xenografts and liver metatstatic nude mice models. RESULTS: LincSCRG1 was found to be strongly elevated in human HCC tissues and cell lines. MiR26a and S phase kinase-related protein 2 (SKP2) were predicted as the target miRNA for lincSCRG1 and the target gene for miR26a with direct binding sites, respectively. LincSCRG1 was verified as a competing endogenous RNA (ceRNA) via negative regulation of miR26a and derepression of SKP2 in HCC cells. Both overexpression of lincSCRG1 (ov-lincSCRG1) and inhibition of miR26a (in-miR26a) obviously stimulated cellular viability, colony formation, migration and proliferation of S phase cells and also significantly increased the protein levels of cyclinD1, CDK4, MMP2/3/9, Vimentin, and N-cadherin or inhibited the protein level of E-cadherin of HCC cells, while knockdown of lincSCRG1 (sh-lincSCRG1) and upregulation of miR26a (mi-miR26a) had the opposite effects on HCC cells. Cotransfection of in-miR26a or overexpression of SKP2 (ov-SKP2) with sh-lincSCRG1 could rescue the anticancer functions of sh-lincSCRG1, including suppressing proliferation and migration of HCC cells. Additionally, sh-lincSCRG1 could effectively inhibit the growth of subcutaneous xenograft tumours and lung metastasis, while the anticancer effect of sh-lincSCRG1 could be reversed by cotransfection of in-miR26a. CONCLUSIONS: LincSCRG1 acts as a ceRNA of miR26a to restrict its ability to derepress SKP2, thereby inducing the proliferation and migration of HCC cells in vitro and in vivo. Depletion of lincSCRG1 could be used as a potential therapeutic approach in HCC.


Subject(s)
Carcinoma, Hepatocellular/genetics , Liver Neoplasms/genetics , MicroRNAs/metabolism , RNA, Long Noncoding/metabolism , S-Phase Kinase-Associated Proteins/metabolism , Carcinoma, Hepatocellular/mortality , Disease Progression , Humans , Liver Neoplasms/mortality
7.
World J Gastroenterol ; 25(36): 5451-5468, 2019 Sep 28.
Article in English | MEDLINE | ID: mdl-31576092

ABSTRACT

BACKGROUND: Zinc-α2-glycoprotein 1 (AZGP1) plays important roles in metabolism-related diseases. The underlying molecular mechanisms and therapeutic effects of AZGP1 remain unknown in non-alcoholic fatty liver disease (NAFLD). AIM: To explore the effects and potential mechanism of AZGP1 on NAFLD in vivo and in vitro. METHODS: The expression of AZGP1 and its effects on hepatocytes were examined in NAFLD patients, CCl4-treated mice fed a high fat diet (HFD), and human LO2 cells. RESULTS: AZGP1 levels were significantly decreased in liver tissues of NAFLD patients and mice. AZGP1 knockdown was found to activate inflammation; enhance steatogenesis, including promoting lipogenesis [sterol regulatory element-binding protein (SREBP)-1c, liver X receptor (LXR), fatty acid synthase (FAS), acetyl-CoA carboxylase (ACC), and stearoyl CoA desaturase 1 (SCD)-1], increasing lipid transport and accumulation [fatty acid transport protein (FATP), carnitine palmitoyl transferase (CPT)-1A, and adiponectin], and reducing fatty acid ß-oxidation [farnesoid X receptor (FXR) and peroxisome proliferator-activated receptor (PPAR)-α]; accelerate proliferation; and reverse apoptosis in LO2 cells. AZGP1 overexpression (OV-AZGP1) had the opposite effects. Furthermore, AZGP1 alleviated NAFLD by blocking TNF-α-mediated inflammation and intracellular lipid deposition, promoting proliferation, and inhibiting apoptosis in LO2 cells. Finally, treatment with OV-AZGP1 plasmid dramatically improved liver injury and eliminated liver fat in NAFLD mice. CONCLUSION: AZGP1 attenuates NAFLD with regard to ameliorating inflammation, accelerating lipolysis, promoting proliferation, and reducing apoptosis by negatively regulating TNF-α. AZGP1 is suggested to be a novel promising therapeutic target for NAFLD.


Subject(s)
Carrier Proteins/metabolism , Glycoproteins/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Tumor Necrosis Factor-alpha/metabolism , Adipokines , Animals , Carbon Tetrachloride/toxicity , Carrier Proteins/genetics , Diet, High-Fat/adverse effects , Disease Models, Animal , Down-Regulation , Gene Knockdown Techniques , Glycoproteins/genetics , Humans , Lipogenesis , Liver/drug effects , Liver/pathology , Male , Mice , Non-alcoholic Fatty Liver Disease/etiology , Oxidation-Reduction , Signal Transduction
8.
World J Gastroenterol ; 25(31): 4468-4480, 2019 Aug 21.
Article in English | MEDLINE | ID: mdl-31496625

ABSTRACT

BACKGROUND: Activation of hepatic stellate cells (HSCs) is a pivotal event in the onset and progression of liver fibrosis. Loss of microRNA-194 (miR-194) has been reported in activated HSCs, but the actual role of miR-194 in liver fibrosis remains uncertain. AIM: To explore the role and potential mechanism of miR-194-mediated regulation of liver fibrosis in vitro and in vivo. METHODS: The expression of miR-194 was examined in human fibrotic liver tissues, activated HSCs, and a carbon tetrachloride (CCl4) mouse model by qPCR. The effects of AKT2 regulation by miR-194 on the activation and proliferation of HSCs were assessed in vitro. For in vivo experiments, we reintroduced miR-194 in mice using a miR-194 agomir to investigate the functions of miR-194 in liver fibrosis. RESULTS: MiR-194 expression was notably lacking in activated HSCs from both humans and mice. Overexpression of miR-194 (OV-miR-194) inhibited α-smooth muscle actin (α-SMA) and type I collagen (Col I) expression and suppressed cell proliferation in HSCs by causing cell cycle arrest in G0/G1 phase. AKT2 was predicted to be a target of miR-194. Notably, the effects of miR-194 knockdown in HSCs were almost blocked by AKT2 deletion, indicating that miR-194 plays a role in HSCs via regulation of AKT2. Finally, miR-194 agomir treatment dramatically ameliorated liver fibrosis in CCl4-treated mice. CONCLUSION: We revealed that miR-194 plays a protective role by inhibiting the activation and proliferation of HSCs via AKT2 suppression. Our results further propose miR-194 as a potential therapeutic target for liver fibrosis.


Subject(s)
Hepatic Stellate Cells/pathology , Liver Cirrhosis, Experimental/genetics , MicroRNAs/metabolism , Proto-Oncogene Proteins c-akt/genetics , Animals , Carbon Tetrachloride/toxicity , Cell Line , Cell Proliferation/genetics , Cohort Studies , Down-Regulation , G1 Phase Cell Cycle Checkpoints/genetics , Humans , Liver/cytology , Liver/drug effects , Liver/pathology , Liver Cirrhosis, Experimental/chemically induced , Liver Cirrhosis, Experimental/pathology , Male , Mice , MicroRNAs/agonists , MicroRNAs/genetics , RNA, Small Interfering/metabolism , Signal Transduction/genetics
9.
FASEB J ; 33(2): 2105-2115, 2019 02.
Article in English | MEDLINE | ID: mdl-30226813

ABSTRACT

The biologic roles of long noncoding RNAs (lncRNAs) in liver fibrosis remained unknown. Through microarray analysis, linc-SCRG1 (a lncRNA with transcript length 3118 bp) was found up-regulated 13.62-fold in human cirrhotic tissues. Quantitative PCR verified that linc-SCRG1 increased along with liver fibrosis progression in human tissues and in activated LX2 cells induced by TGF-ß1. Knockdown of linc-SCRG1 significantly reversed the effects of TGF-ß1 on LX2, including inhibiting activation, promoting apoptosis, reducing proliferation, lessening invasion, and down-regulating genes [fibrosis-related mRNA: α-smooth muscle actin ( α-SMA), type I collagen, and B-cell lymphoma-2; invasion-related mRNA: matrix metallopeptidase-2 ( MMP-2), MMP-9, and MMP-13; inflammation-related mRNA: TNF-α, IL-6, and IL-10]. linc-SCRG1 had binding sites with tristetraprolin (TTP), a kind of RNA-binding protein, and specifically combined to TTP proteins. Overexpression of linc-SCRG1 would cause TTP mRNA unstably and proteins decreasing. TTP mRNA was proved having negative relevance with linc-SCRG1 and was gradually reduced during human liver fibrosis progression. Overexpressing TTP resulted in knockdown of lincSCRG1 and degraded downstream target genes ( MMP-2 and TNF-α) in activated LX2. Overexpressing TTP had the same effects as small interfering RNA-lincSCRG1 (si- lincSCRG1), whereas knockdown of TTP had reversal effects on si- lincSCRG1 in activated LX2. In summary, linc-SCRG1 reduced TTP and restricted its degradation of target genes TNF-α and MMP-2. Therefore, linc-SCRG1 had a repressing TTP-elicited inactivation effect on hepatic stellate cell (HSC) phenotypes. Inhibition of linc-SCRG1 may be a novel therapeutic approach to inactivate HSCs and extenuate human liver fibrosis.-Wu, J.-C., Luo, S.-Z., Liu, T., Lu, L.-G., Xu, M.-Y. linc-SCRG1 accelerates liver fibrosis by decreasing RNA-binding protein tristetraprolin.


Subject(s)
Gene Expression Regulation , Hepatic Stellate Cells/cytology , Liver Cirrhosis/pathology , Nerve Tissue Proteins/genetics , RNA, Long Noncoding/genetics , Tristetraprolin/metabolism , Cell Proliferation , Cells, Cultured , Down-Regulation , Hepatic Stellate Cells/metabolism , Humans , Liver Cirrhosis/genetics , Liver Cirrhosis/metabolism , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism , Tristetraprolin/genetics
10.
Pancreatology ; 18(4): 438-445, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29653723

ABSTRACT

An increasing number of reports have demonstrated that there is an association between the presence of pathogenic microorganisms and pancreatic cancer. However, the role of the duodenal microbiota in pancreatic carcinogenesis remains unknown. In this study, duodenal mucosal microbiota was analyzed in 14 patients with pancreatic head cancer and 14 healthy controls using 16S rRNA gene pyrosequencing methods. Plasma endotoxin activity and the concentrations of the proinflammatory cytokine IL-6 and C-reactive protein (CRP) were measured in blood samples. The urea breath test was used to detect Helicobacter pylori infections. Endoscopic duodenal mucosal biopsies were evaluated by histological examinations. Statistical comparisons of inflammatory factors revealed significantly higher levels of CRP and IL-6 in the pancreatic cancer group as compared to healthy controls. Patients with pancreatic cancer also had a higher incidence of H. pylori infections and showed mucosal changes, including villous abnormalities and diffuse inflammatory cell infiltration in the lamina propria. The sequences analysis showed that based on linear discriminant analysis effect size (LEfSe) analysis at the genus level, Acinetobacter, Aquabacterium, Oceanobacillus, Rahnella, Massilia, Delftia, Deinococcus, and Sphingobium were more abundant in the duodenal mucosa of pancreatic cancer patients, whereas the duodenal microbiotas of healthy controls were enriched with Porphyromonas, Paenibacillus, Enhydrobacter, Escherichia, Shigella, and Pseudomonas. These results reveal a picture of duodenal microbiota in pancreatic head cancer patients that could be useful in future trials investigating the role of gut microbiota in pancreatic cancer.


Subject(s)
Duodenum/microbiology , Gastrointestinal Microbiome , Pancreatic Neoplasms/microbiology , Aged , C-Reactive Protein/analysis , Endotoxins/blood , Enteritis/epidemiology , Enteritis/etiology , Enteritis/microbiology , Female , Healthy Volunteers , Helicobacter Infections/epidemiology , Helicobacter Infections/microbiology , Helicobacter pylori , Humans , Incidence , Interleukin-6/analysis , Male , Middle Aged , Pancreas/pathology , Pancreatic Neoplasms/epidemiology , Pancreatic Neoplasms/pathology , RNA, Ribosomal, 16S/analysis
11.
Biomed Pharmacother ; 98: 214-221, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29268242

ABSTRACT

Signal transducer and activator of transcription 3 (STAT3) has been shown to affect epithelial-to-mesenchymal transition (EMT) in cancers. We investigated the underlying molecular mechanisms of STAT3 crosstalk with Snail-Smad3/transforming growth factor (TGF)-ß1 signaling pathways during the EMT in hepatocellular carcinoma (HCC). STAT3 and TGF-ß1 expressions are examined in liver tissues of HCC patients and rats. The effect of IL-6/ STAT3 crosstalk with Snail-Smad3/TGF-ß1 on EMT, carcinogenesis, migration and invasion are tested in vitro and in vivo. Phosphorylation of STAT3 and TGF-ß1 proteins are universally high and positively co-expressed in HCC tissues from human and rats. Hepatic lower p-STAT3 proteins are related to earlier tumor stages in HCC patients. AG490 (a JAK2-specific inhibitor) treatment could reduce tumor numbers and sizes depending on suppression of STAT3 signaling in HCC rats. TGF-ß1 could induce EMT along with an E-cadherin decrease, while vimentin, Snail, p-Smad2/3, and p-STAT3/STAT3 increase in HepG2. SIS3 (a specific inhibitor of Smad3) could markedly inhibit Snail, Vim and p-STAT3 along with blocking phosphorylation of Smad3, but E-cadherin could be activated in HepG2. IL-6 activates STAT3 signaling and then has cascading consequences for activating Snail-Smad3/TGF-ß1 and vimentin as well as migration and invasion in liver cancer cells. In contrast, AG490 has an effect that inhibits phosphorylation of STAT3, lowers Snail-p-Smad3 protein levels, decreases TGF-ß1-related PAI-1 promoter activation and then reduces migration or invasion of liver cancer cells. STAT3 functions as a positive regulator to activate TGF-ß1-induced EMT and metastasis of HCC. STAT3 and the Snail-Smad3/TGF-ß1 signaling pathways synergistically augment EMT and migration in HCC.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Cell Movement/physiology , Epithelial-Mesenchymal Transition/physiology , Liver Neoplasms/metabolism , STAT3 Transcription Factor/metabolism , Transforming Growth Factor beta1/pharmacology , Animals , Cell Movement/drug effects , Epithelial-Mesenchymal Transition/drug effects , Female , Hep G2 Cells , Humans , Male , Rats , Rats, Wistar , STAT3 Transcription Factor/pharmacology , Transforming Growth Factor beta1/antagonists & inhibitors
12.
J Dig Dis ; 15(2): 78-84, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24131862

ABSTRACT

OBJECTIVE: We aimed to determine the efficacy and safety of endoscopic nasobiliary drainage (ENBD) with or without endoscopic sphincterotomy (EST) for temporary biliary decompression in patients with acute obstructive cholangitis. METHODS: In total, 72 patients with acute obstructive cholangitis were prospectively randomized to undergo emergency ENBD with EST (EST group, n = 36) or without EST (non-EST group, n = 36). The clinical outcomes and complications between the two groups were compared. RESULTS: Endoscopic nasobiliary decompression was successful in all 72 patients. Four patients underwent a second endoscopic retrograde cholangiopancreatography (ERCP) to replace the nasobiliary catheter due to blockage (one in the EST group and two in the non-EST group) or migration (one in the EST group). The mean serum γ-glutamyltransferase and total bilirubin levels after treatment were significantly higher in the non-EST group than in the EST group (P < 0.05). However, no significant differences were observed for other parameters evaluated. The total complication rate was similar between the two groups (EST 25.0% vs non-EST 19.4%). Although hemorrhage occurred more frequently in the EST group and acute pancreatitis in the non-EST group, these differences were not significant. CONCLUSIONS: EST is helpful and safe for biliary drainage while ENBD without EST is the first choice for acute cholangitis. EST may increase the efficacy of ENBD in patients with papillary inflammatory stricture and thick bile.


Subject(s)
Cholangitis/surgery , Cholestasis/complications , Natural Orifice Endoscopic Surgery/methods , Sphincterotomy, Endoscopic/methods , Acute Disease , Adult , Aged , Cholangiopancreatography, Endoscopic Retrograde/adverse effects , Cholangiopancreatography, Endoscopic Retrograde/methods , Cholangitis/etiology , Decompression, Surgical/adverse effects , Decompression, Surgical/methods , Drainage/methods , Emergencies , Endoscopy, Gastrointestinal/adverse effects , Endoscopy, Gastrointestinal/methods , Female , Humans , Male , Middle Aged , Prospective Studies , Sphincterotomy, Endoscopic/adverse effects , Treatment Outcome
13.
Hepatobiliary Pancreat Dis Int ; 11(6): 643-9, 2012 Dec 15.
Article in English | MEDLINE | ID: mdl-23232637

ABSTRACT

BACKGROUND: Endoscopic biliary stent placement during the procedure of endoscopic retrograde cholangiopancreatography (ERCP) is preferred to provide biliary drainage for unresectable malignant biliary obstruction. There is considerable controversy over the use of endoscopic sphincterotomy (ES) prior to stent placement. This study aimed to determine whether ES before intraductal self-expanding metal stent (SEMS) placement affects the clinical outcome and complications in patients with proximal malignant obstructive biliary diseases. METHODS: In a prospective randomized controlled trial, 82 patients with inoperable malignant biliary strictures were randomly assigned to biliary stenting groups with or without ES. Resolution of jaundice and the incidence of complications including acute cholangitis, pancreatitis and stent occlusion within 6 months were evaluated. RESULTS: SEMSs were successfully deployed in all patients, resulting in clinical and biochemical improvement of obstructive symptoms in both groups. The incidence of cholangitis was higher in the ES group than in the non-ES group (58.5% vs 31.7%, P=0.015). The interval between stent placement and the first acute cholangitis was much shorter in the ES group than in the non-ES group (P=0.024). The use of ES increased the incidence of cholangitis (P=0.004, risk ratio, 8.196). The rate of post-ERCP pancreatitis after stent placement was greater in the non-ES group than in the ES group (31.7% vs 9.8%, P=0.014). No significant differences were found in the rate of restenosis and the mortality rate between the two groups. CONCLUSIONS: ES prior to intraductal SEMS placement was associated with an increased incidence of acute cholangitis. ES should be carefully evaluated prior to its use in patients with proximal malignant obstructive biliary diseases.


Subject(s)
Carcinoma/complications , Cholangitis/etiology , Cholestasis/surgery , Digestive System Neoplasms/complications , Jaundice, Obstructive/surgery , Sphincterotomy, Endoscopic/adverse effects , Aged , Cholangiopancreatography, Endoscopic Retrograde , Cholestasis/etiology , Drainage , Female , Humans , Jaundice, Obstructive/etiology , Kaplan-Meier Estimate , Male , Middle Aged , Multivariate Analysis , Pancreatitis/etiology , Stents , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...