Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Dig Endosc ; 2024 Feb 21.
Article in English | MEDLINE | ID: mdl-38380564

ABSTRACT

OBJECTIVES: The aim of this study was to compare endoscopic ultrasound-guided choledochoduodenostomy (EUS-CDS) vs. EUS-gallbladder drainage (EUS-GBD) in cases of failed endoscopic retrograde cholangiopancreatography (ERCP) for jaundice resulting from malignant distal biliary obstruction (MDBO). METHODS: This multicenter retrospective study included patients with obstructive jaundice secondary to MDBO who underwent EUS-GBD or EUS-CDS with lumen-apposing metal stents after failed ERCP. The primary end-point was clinical success rate. Secondary end-points were technical success, periprocedural adverse events rate (<24 h), late adverse events rate (>24 h), overall survival, and time to recurrent biliary obstruction. RESULTS: A total of 78 patients were included: 41 underwent EUS-GBD and 37 underwent EUS-CDS. MDBO was mainly the result of pancreatic cancer (n = 63/78, 80.7%). Clinical success rate was similar for both procedures: 87.8% for EUS-GBD and 89.2% for EUS-CDS (P = 0.8). Technical success rate was 100% for EUS-GBD and 94.6% for EUS-CDS (P = 0.132). Periprocedural morbidity (<24 h) rates were similar between both groups: 4/41 (9.8%) for EUS-GBD and 5/37 (13.5%) for EUS-CDS (P = 0.368). There was a significantly higher rate of late morbidity (>24 h) among patients in the EUS-CDS group (8/37 [21.6%]) than in the EUS-GBD group (3/41 [7.3%]) (P = 0.042). The median follow-up duration was 4.7 months. Overall survival and time to recurrent biliary obstruction did not significantly differ between the groups. DISCUSSION: After failed ERCP for MDBO, EUS-GBD and EUS-CDS show comparable clinical success rates and technical success. EUS-GBD appears to be a promising alternative for MDBO, even as a second-line treatment after failed ERCP. Further studies are needed to validate these findings and compare the long-term outcomes of EUS-GBD and EUS-CDS.

2.
Endoscopy ; 55(9): 796-803, 2023 09.
Article in English | MEDLINE | ID: mdl-36849106

ABSTRACT

BACKGROUND : The diagnosis of cholangiocarcinoma in patients with a biliary stricture without mass syndrome can be obtained by biliary brushing with a sensitivity of ~50 %. We performed a multicenter randomized crossover trial comparing the aggressive Infinity brush with the standard RX Cytology Brush. The aims were to compare sensitivity for cholangiocarcinoma diagnosis and cellularity obtained. METHODS : Biliary brushing was performed consecutively with each brush, in a randomized order. Cytological material was studied with blinding to the brush type used and order. The primary end point was sensitivity for cholangiocarcinoma diagnosis; the secondary end point was the abundance of cellularity obtained with each brush, with cellularity quantified in order to determine if one brush strongly outperformed the other. RESULTS : 51 patients were included. Final diagnoses were cholangiocarcinoma (n = 43; 84 %), benign (n = 7; 14 %), and indeterminate (n = 1; 2 %). Sensitivity for cholangiocarcinoma was 79 % (34 /43) for the Infinity brush versus 67 % (29/43) for the RX Cytology Brush (P = 0.10). Cellularity was rich in 31/51 cases (61 %) with the Infinity brush and in 10/51 cases (20 %) with the RX Cytology Brush (P < 0.001). In terms of quantification of cellularity, the Infinity brush strongly outperformed the RX Cytology Brush in 28/51 cases (55 %), while the RX Cytology Brush strongly outperformed the Infinity brush in 4/51 cases (8 %; P < 0.001). CONCLUSIONS : This randomized crossover trial showed that the Infinity brush is not significantly more effective than the RX Cytology Brush for biliary stenosis without mass syndrome in terms of sensitivity for cholangiocarcinoma diagnosis, but does offer a significantly higher abundance of cellularity.


Subject(s)
Bile Duct Neoplasms , Cholangiocarcinoma , Cholestasis , Humans , Constriction, Pathologic/diagnosis , Cholangiopancreatography, Endoscopic Retrograde/methods , Sensitivity and Specificity , Cholestasis/diagnosis , Cholestasis/etiology , Cholangiocarcinoma/diagnosis , Cholangiocarcinoma/pathology , Bile Duct Neoplasms/diagnosis , Bile Duct Neoplasms/pathology , Bile Ducts, Intrahepatic/pathology
3.
J Clin Monit Comput ; 37(5): 1219-1228, 2023 10.
Article in English | MEDLINE | ID: mdl-36840793

ABSTRACT

BACKGROUND & AIMS: Ascites is a frequent complication of cirrhosis. In intensive care units, initial hemodynamic assessment is frequently performed by echocardiography. This study evaluated the feasibility and usefulness of early hemodynamic assessment in the gastroenterology ward. METHODS: This observational cohort study prospectively included all patients admitted to a teaching hospital's gastroenterology unit for decompensated cirrhosis. A gastroenterologist with minimal training and an intensivist both performed an echocardiography exam. The primary outcome was inter-rater agreement and reliability for three echocardiography parameters: visual LVEF (Left Ventricular Ejection Fraction), subaortic VTI (velocity time integral) and E wave velocity. Secondary outcomes were agreement for presence of pleural effusion, description of 3 hemodynamics profiles (hypovolemic, hyperkinetic and intermediate), and 28-day mortality. RESULTS: From March 2018 to March 2020, 53 patients were included. The median age was 62 years and 81% were men. Patients presented mostly advanced liver disease, with 43% Child-Pugh C and median MELD score of 15.2. The limits of agreement between intensivists and gastroenterologists for subaortic VTI were - 6.6 to 7.2 cm, and ranged from - 0.6 to 0.37 m.s-1 for E wave velocity. Clinically significant differences between intensivists and gastroenterologists were found in 22% for subaortic VTI and 24.5% for E wave velocity. Reliability was good for subaortic VTI (ICC: 0.79, 95% CI [0.58; 0.9;]) and moderate for E wave velocity (0.53, 95% CI [0.19; 0.74]). The three hemodynamics profiles had different prognosis, with a 28-day mortality for Hypovolemic, Intermediate and Hyperkinetic group of 31, 18, and 4%, respectively. CONCLUSION: Reliability of hemodynamic assessment by gastroenterologists was good, while agreement was unsatisfactory, advocating for further training. Transthoracic echocardiography can differentiate hypovolemia from hyperkinetic states. The role of transthoracic echocardiography in managing decompensated cirrhosis requires further study. CLINICAL TRIAL NUMBER: NCT03650660.


Subject(s)
Gastroenterologists , Male , Humans , Middle Aged , Female , Stroke Volume , Ventricular Function, Left , Hypovolemia , Reproducibility of Results , Echocardiography , Hemodynamics
5.
Gut ; 66(10): 1802-1810, 2017 10.
Article in English | MEDLINE | ID: mdl-27456153

ABSTRACT

OBJECTIVE: Although counting of circulating tumour cells (CTC) has attracted a broad interest as potential markers of tumour progression and treatment response, the lack of functional characterisation of these cells had become a bottleneck in taking these observations to the clinic. Our objective was to culture these cells in order to understand them and exploit their therapeutic potential to the full. DESIGN: Here, hypothesising that some CTC potentially have cancer stem cell (CSC) phenotype, we generated several CTC lines from the blood of patients with advanced metastatic colorectal cancer (CRC) based on their self-renewal abilities. Multiple standard tests were then employed to characterise these cells. RESULTS: Our CTC lines self-renew, express CSC markers and have multilineage differentiation ability, both in vitro and in vivo. Patient-derived CTC lines are tumorigenic in subcutaneous xenografts and are also able to colonise the liver after intrasplenic injection. RNA sequencing analyses strikingly demonstrate that drug metabolising pathways represent the most upregulated feature among CTC lines in comparison with primary CRC cells grown under similar conditions. This result is corroborated by the high resistance of the CTC lines to conventional cytotoxic compounds. CONCLUSIONS: Taken together, our results directly demonstrate the existence of patient-derived colorectal CTCs that bear all the functional attributes of CSCs. The CTC culture model described here is simple and takes <1 month from blood collection to drug testing, therefore, routine clinical application could facilitate access to personalised medicine. CLINICAL TRIAL REGISTRATION: ClinicalTrial.gov NCT01577511.


Subject(s)
Colorectal Neoplasms/blood , Colorectal Neoplasms/pathology , Liver Neoplasms/pathology , Neoplastic Cells, Circulating/metabolism , Neoplastic Stem Cells/enzymology , RNA, Neoplasm/analysis , Aldehyde Dehydrogenase/genetics , Aldehyde Dehydrogenase/metabolism , Aldehyde Dehydrogenase 1 Family , Animals , Antineoplastic Agents/metabolism , Cell Differentiation , Cell Self Renewal , Colorectal Neoplasms/genetics , Dipeptidyl Peptidase 4/genetics , Dipeptidyl Peptidase 4/metabolism , Drug Resistance, Neoplasm/genetics , Humans , Hyaluronan Receptors/genetics , Hyaluronan Receptors/metabolism , Inactivation, Metabolic/genetics , Liver Neoplasms/secondary , Mice , Neoplasm Transplantation , Neoplastic Stem Cells/physiology , Phenotype , Primary Cell Culture , Retinal Dehydrogenase , Sequence Analysis, RNA , Tumor Cells, Cultured , Up-Regulation
6.
Cancer Cell Int ; 16: 6, 2016.
Article in English | MEDLINE | ID: mdl-26877710

ABSTRACT

BACKGROUND: This last decade, a lot of emphasis has been placed on developing new cancer cell culture models, closer to in vivo condition, in order to test new drugs and therapies. In the case of colorectal cancer, the use of patient biopsies to seed 3D primary cultures and mimic tumor initiation necessitates the use of antibiotics to prevent microbial intestinal contamination. However, not only long term use of antibiotics may mask the presence of low levels of microbial contamination, it may also impact cancer cell phenotype. METHODS: In this study we tested the impact of penicillin-streptomycin cocktail addition in both monolayer and suspension culture. To ensure the reliability of our observations we used six different cell lines and each experiment was performed in triplicate. Results were analyzed with Student's t test. RESULTS: We show that penicillin-streptomycin cocktail inhibits the sphere-forming ability of six cancer cell lines in suspension culture though it has no impact in monolayer culture. We correlate this effect with a significant decrease of cancer stem cells pool which holds self-renewal potential. CONCLUSIONS: Overall, this study warns against systematic addition of antibiotics in growth medium and raises the interesting possibility of using antibiotics to target cancer stem cells.

7.
Endoscopy ; 48(12): 1084-1095, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27760437

ABSTRACT

Background and study aims: The hemostatic powder TC-325 (Hemospray; Cook Medical, Winston-Salem, North Carolina, USA) has shown promising results in the treatment of upper gastrointestinal bleeding (UGIB) in expert centers in pilot studies. The aim of this study was to evaluate the feasibility and efficacy of TC-325 in a large prospective registry of use in routine practice. Patients and methods: The data of all patients treated with TC-325 were prospectively collected through a national registry. Outcomes were the immediate feasibility and efficacy of TC-325 application, as well as the rates of rebleeding at Day 8 and Day 30. Multivariate analysis was performed to determine predictive factors of rebleeding. Results: A total of 202 patients were enrolled and 64 endoscopists participated from 20 centers. TC-325 was used as salvage therapy in 108 patients (53.5 %). The etiology of bleeding was an ulcer in 75 patients (37.1 %), tumor in 61 (30.2 %), postendoscopic therapy in 35 (17.3 %), or other in 31 (15.3 %). Application of the hemostatic powder was found to be very easy or easy in 31.7 % and 55.4 %, respectively. The immediate efficacy rate was 96.5 %. Recurrence of UGIB was noted at Day 8 and Day 30 in 26.7 % and 33.5 %, respectively. Predictive factors of recurrence at Day 8 were melena at initial presentation and use of TC-325 as salvage therapy. Conclusion: These multicenter data confirmed the high rate of immediate hemostasis, excellent feasibility, and good safety profile of TC-325, which could become the treatment of choice in bleeding tumors or postendoscopic bleeding but not in bleeding ulcers where randomized studies are needed. TRIAL REGISTRATION: ClinicalTrials.gov (NCT02595853).


Subject(s)
Gastrointestinal Hemorrhage/therapy , Gastrointestinal Neoplasms/complications , Hemostasis, Endoscopic , Hemostatics/therapeutic use , Minerals/therapeutic use , Aged , Aged, 80 and over , Endoscopy, Gastrointestinal/adverse effects , Feasibility Studies , Female , Gastrointestinal Hemorrhage/etiology , Humans , Male , Middle Aged , Peptic Ulcer/complications , Powders/therapeutic use , Prospective Studies , Recurrence , Registries , Risk Factors
8.
Endoscopy ; 46(12): 1063-70, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25098612

ABSTRACT

BACKGROUND AND STUDY AIMS: A new core biopsy needle for endoscopic ultrasound (EUS)-guided sampling has recently been developed. The aim of this prospective multicenter study was to compare this needle with a standard needle in patients with solid pancreatic masses. PATIENTS AND METHODS: Consecutive patients with solid pancreatic masses referred to 17 centers for EUS-guided sampling were included. Each patient had two passes with a standard 22G needle and a single pass with a 22G core needle performed in a randomized order. Samples from both needles were separately processed for liquid-based cytology and cell-block preparation and were assessed independently by two blinded expert pathologists. The primary endpoint was the accuracy of the detection of malignancy. The reference standard was based on further cytohistological analysis obtained under ultrasound or computed tomography scanning, endoscopic or surgical guidance, and/or by clinical follow-up with repeated imaging examinations for at least 12 months. The secondary endpoints were the rate of technical failure and the quality of the cytohistological samples obtained. RESULTS: Of the 80 patients included (49 men; mean age 67.1 ±â€Š11.1), 87.5 % had final malignant diagnoses (adenocarcinoma n = 62, 77.5 %). There was no difference between the needles in diagnostic accuracy (standard needle 92.5 % vs. core needle 90 %; P = 0.68) or technical failure. Both pathologists found the overall sample quality significantly better for the standard needle (expert 1, P = 0.009; expert 2, P = 0.002). CONCLUSIONS: The diagnostic accuracy of EUS sampling for solid pancreatic masses using standard and core needles seems comparable but with a better overall histological sample quality for the former. ClinicalTrial.gov identifier: NCT01479803.


Subject(s)
Biopsy, Large-Core Needle/instrumentation , Endoscopic Ultrasound-Guided Fine Needle Aspiration/instrumentation , Needles , Pancreas/pathology , Pancreatic Neoplasms/diagnosis , Adult , Aged , Aged, 80 and over , Cross-Over Studies , Diagnosis, Differential , Equipment Design , Female , Humans , Male , Middle Aged , Prospective Studies
9.
Proc Natl Acad Sci U S A ; 107(6): 2628-33, 2010 Feb 09.
Article in English | MEDLINE | ID: mdl-20133805

ABSTRACT

Symplekin is a ubiquitously expressed protein involved in cytoplasmic RNA polyadenylation and transcriptional regulation and is localized at tight junctions (TJs) in epithelial cells. Nuclear symplekin cooperates with the Y-box transcription factor zonula occludens 1-associated nucleic acid-binding protein (ZONAB) to increase the transcription of cell cycle-related genes and also inhibits differentiation of intestinal cells. We detected high levels of nuclear symplekin in 8 of 12 human colorectal cancer (CRC) samples. shRNA-mediated reduction of symplekin expression was sufficient to decrease significantly the anchorage-independent growth and proliferation of HT-29 CRC cells as well as their tumorigenicity when injected into immunodeficient animals. Symplekin down-regulation also was found to alter ion transport through TJs, to promote the localization of ZONAB in the membrane rather than the nucleus, and strongly to enhance cell polarization in a 3D matrix, leading to the formation of spheroids organized around a central lumen. Claudin-2 expression was reduced following symplekin down-regulation, an effect mimicked when ZONAB expression was down-regulated using selective siRNA. Virus-mediated restoration of claudin-2 expression was found to restore nuclear expression of ZONAB in HT29DeltaSym cells and to rescue the phenotypic alterations induced by symplekin down-regulation of cell polarity, paracellular transport, ZONAB localization, cyclin D1 expression, proliferation, and anchorage-independent growth. Finally, siRNA-mediated claudin-2 down-regulation increased the transepithelial resistance and decreased cyclin D1 expression and ZONAB nuclear localization, similar to observations in symplekin-depleted cells. Our results suggest that nuclear overexpression of symplekin promotes tumorigenesis in the human colon and that the regulation of claudin-2 expression is instrumental in this effect.


Subject(s)
Colorectal Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Membrane Proteins/genetics , Nuclear Proteins/genetics , Animals , Blotting, Western , CCAAT-Enhancer-Binding Proteins/genetics , CCAAT-Enhancer-Binding Proteins/metabolism , Cell Proliferation , Claudins , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Cyclin D1/genetics , Cyclin D1/metabolism , Fluorescent Antibody Technique , HT29 Cells , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Humans , Membrane Proteins/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Nuclear Proteins/metabolism , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Transplantation, Heterologous , Tumor Burden , Up-Regulation
10.
Clin Res Hepatol Gastroenterol ; 47(5): 102107, 2023 05.
Article in English | MEDLINE | ID: mdl-36906225

ABSTRACT

OBJECTIVES: The emergence of biologics has improved the course of inflammatory bowel diseases (IBD) in the elderly population despite a potential higher risk of infections. We conducted a one-year, prospective, multicenter, observational study to determine the frequency of occurrence of at least one infectious event in elderly IBD patients under anti-TNF therapy compared with that in elderly patients under vedolizumab or ustekinumab therapies. METHODS: All IBD patients over 65 years exposed to anti-TNF, vedolizumab or ustekinumab therapies were included. The primary endpoint was the prevalence of at least one infection during the whole one year follow-up. RESULTS: Among the 207 consecutive elderly IBD patients prospectively enrolled, 113 were treated with anti-TNF and 94 with vedolizumab (n=63) or ustekinumab (n=31) (median age 71 years, 112 Crohn's disease). The Charlson index was similar between patients under anti-TNF and those under vedolizumab or ustekinumab as well as the proportion of patients under combination therapy and under concomitant steroid therapy did not differ between both both groups. The prevalence of infections was similar in patients under anti-TNF and in those under vedolizumab or ustekinumab (29% versus 28%, respectively; p=0.81). There was no difference in terms of type and severity of infection and of infection-related hospitalization rate. In multivariate regression analysis, only the Charlson comorbidity index (≥ 1) was identified as a significant and independent risk factor of infection (p=0.03). CONCLUSION: Around 30 % of elderly patients with IBD under biologics experienced at least one infection during the one-year study follow-up period. The risk of occurrence of infection does not differ between anti-TNF and vedolizumab or ustekinumab therapies, and only the associated comorbidity was linked with the risk of infection.


Subject(s)
Biological Products , Inflammatory Bowel Diseases , Humans , Aged , Ustekinumab/adverse effects , Follow-Up Studies , Biological Products/adverse effects , Prospective Studies , Tumor Necrosis Factor Inhibitors , Inflammatory Bowel Diseases/complications , Inflammatory Bowel Diseases/drug therapy , Treatment Outcome , Retrospective Studies
11.
Stem Cell Reports ; 17(4): 835-848, 2022 04 12.
Article in English | MEDLINE | ID: mdl-35276090

ABSTRACT

Tumor recurrence is often attributed to cancer stem cells (CSCs). We previously demonstrated that down-regulation of Pregnane X Receptor (PXR) decreases the chemoresistance of CSCs and prevents colorectal cancer recurrence. Currently, no PXR inhibitor is usable in clinic. Here, we identify miR-148a as a targetable element upstream of PXR signaling in CSCs, which when over-expressed decreases PXR expression and impairs tumor relapse after chemotherapy in mouse tumor xenografts. We then develop a fluorescent reporter screen for miR-148a activators and identify the anti-helminthic drug niclosamide as an inducer of miR-148a expression. Consequently, niclosamide decreased PXR expression and CSC numbers in colorectal cancer patient-derived cell lines and synergized with chemotherapeutic agents to prevent CSC chemoresistance and tumor recurrence in vivo. Our study suggests that endogenous miRNA inducers is a viable strategy to down-regulate PXR and illuminates niclosamide as a neoadjuvant repurposing strategy to prevent tumor relapse in colon cancer.


Subject(s)
Colonic Neoplasms , MicroRNAs , Animals , Cell Line, Tumor , Colonic Neoplasms/drug therapy , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Humans , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/metabolism , Neoplasm Recurrence, Local/pathology , Neoplastic Stem Cells/metabolism , Niclosamide/metabolism , Niclosamide/pharmacology , Niclosamide/therapeutic use , Pregnane X Receptor/genetics , Pregnane X Receptor/metabolism
12.
Gastroenterology ; 137(1): 156-64, 164.e1-3, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19328795

ABSTRACT

BACKGROUND & AIMS: Symplekin is a ubiquitously expressed protein involved in RNA polyadenylation and transcriptional regulation that localizes at tight junctions in epithelial cells. The association between symplekin and the Y-box transcription factor ZONAB activates proliferation in intestinal and kidney cells. We analyzed symplekin expression in human colonic crypts and investigated its function in differentiation. METHODS: Expression of differentiation markers and transcription factors was assessed in HT29-Cl.16E cells that expressed inducible symplekin short hairpin RNA or were transfected with ZONAB small interfering RNAs. Intestines of AML1(Delta/Delta) mice were stained with alcian blue and analyzed for expression of AML1/Runx1, GAPDH, KLF-4, and Muc-2. Mobility shift and chromatin immunoprecipitation were used to detect AML1 and ZONAB/DbpA binding to promoter regions of the Krüppel-like factor 4 (KLF4) and acute myeloid leukemia-1 (AML1) genes, respectively. RESULTS: The gradient of nuclear symplekin expression decreased from the proliferative toward the differentiated compartment of colonic crypts; symplekin down-regulation promoted the differentiation of HT29-Cl.16E colorectal carcinoma cells into goblet cells. Down-regulation of symplekin or ZONAB/Dbpa induced de novo expression of the transcription factor AML1/Runx1, thereby increasing the expression of KLF4 and promoting goblet cell differentiation. Furthermore, increased AML1 expression was required for the induction of goblet cell differentiation after symplekin down-regulation. KLF4 expression and goblet cell numbers were reduced in the intestines of AML1(Delta/Delta) mice, confirming the role of AML1 as a promoter of intestinal differentiation in vivo. CONCLUSIONS: Symplekin cooperates with ZONAB to negatively regulate intestinal goblet cell differentiation, acting by repression of AML1 and KLF4.


Subject(s)
CCAAT-Enhancer-Binding Proteins/metabolism , Cell Differentiation , Core Binding Factor Alpha 2 Subunit/metabolism , Goblet Cells/metabolism , Heat-Shock Proteins/metabolism , Intestinal Mucosa/metabolism , Nuclear Proteins/metabolism , Animals , CCAAT-Enhancer-Binding Proteins/genetics , Cell Proliferation , Core Binding Factor Alpha 2 Subunit/genetics , Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , Goblet Cells/enzymology , HT29 Cells , Heat-Shock Proteins/genetics , Humans , Intestines/enzymology , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/metabolism , Mice , Mice, Transgenic , Mucin-2/metabolism , Nuclear Proteins/genetics , RNA Interference , Transfection
13.
Mol Cancer Res ; 5(11): 1147-57, 2007 Nov.
Article in English | MEDLINE | ID: mdl-18025260

ABSTRACT

Chronic alcohol consumption is associated with increased risk of gastrointestinal cancer. High concentrations of ethanol trigger mucosal hyperregeneration, disrupt cell adhesion, and increase the sensitivity to carcinogens. Most of these effects are thought to be mediated by acetaldehyde, a genotoxic metabolite produced from ethanol by alcohol dehydrogenases. Here, we studied the role of low ethanol concentrations, more likely to mimic those found in the intestine in vivo, and used intestinal cells lacking alcohol dehydrogenase to identify the acetaldehyde-independent biological effects of ethanol. Under these conditions, ethanol did not stimulate the proliferation of nonconfluent cells, but significantly increased maximal cell density. Incorporation of phosphatidylethanol, produced from ethanol by phospholipase D, was instrumental to this effect. Phosphatidylethanol accumulation induced claudin-1 endocytosis and disrupted the claudin-1/ZO-1 association. The resulting nuclear translocation of ZONAB was shown to mediate the cell density increase in ethanol-treated cells. In vivo, incorporation of phosphatidylethanol and nuclear translocation of ZONAB correlated with increased proliferation in the colonic epithelium of ethanol-fed mice and in adenomas of chronic alcoholics. Our results show that phosphatidylethanol accumulation after chronic ethanol exposure disrupts signals that normally restrict proliferation in highly confluent intestinal cells, thus facilitating abnormal intestinal cell proliferation.


Subject(s)
Adenocarcinoma/chemically induced , CCAAT-Enhancer-Binding Proteins/metabolism , Colonic Neoplasms/chemically induced , Ethanol/toxicity , Glycerophospholipids/metabolism , Heat-Shock Proteins/metabolism , Intestines/drug effects , Intestines/pathology , Adenocarcinoma/metabolism , Alcohol Dehydrogenase/deficiency , Alcohol Dehydrogenase/genetics , Animals , CCAAT-Enhancer-Binding Proteins/genetics , Caco-2 Cells , Cell Count , Claudin-1 , Colonic Neoplasms/metabolism , Endocytosis , Heat-Shock Proteins/genetics , Humans , Hyperplasia/chemically induced , Membrane Proteins/metabolism , Mice , Phospholipase D/metabolism , Phosphoproteins/metabolism , Zonula Occludens-1 Protein
14.
Gastroenterology ; 133(5): 1554-68, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17920061

ABSTRACT

BACKGROUND & AIMS: Aberrant activation of the beta-catenin/Tcf-4 transcriptional complex represents an initiating event for colorectal carcinogenesis, shifting the balance from differentiation toward proliferation in colonic crypts. Here, we assessed whether endogenous progastrin, encoded by a target gene of this complex, was in turn able to regulate beta-catenin/Tcf-4 activity in adenomatous polyposis coli (APC)-mutated cells, and we analyzed the impact of topical progastrin depletion on intestinal tumor growth in vivo. METHODS: Stable or transient RNA silencing of the GAST gene was induced in human tumor cells and in mice carrying a heterozygous Apc mutation (APCDelta14), which overexpress progastrin but not amidated or glycine-extended gastrin. RESULTS: Depletion of endogenous progastrin production strongly decreased intestinal tumor growth in vivo through a marked inhibition of constitutive beta-catenin/Tcf-4 activity in tumor cells. This effect was mediated by the de novo expression of the inhibitor of beta-catenin and Tcf-4 (ICAT), resulting from a down-regulation of integrin-linked kinase in progastrin-depleted cells. Accordingly, ICAT down-regulation was correlated with progastrin overexpression and Tcf-4 target gene activation in human colorectal tumors, and ICAT repression was detected in the colon epithelium of tumor-prone, progastrin-overexpressing mice. In APCDelta14 mice, small interfering RNA-mediated progastrin depletion not only reduced intestinal tumor size and numbers, but also increased goblet cell lineage differentiation and cell apoptosis in the remaining adenomas. CONCLUSIONS: Thus, depletion of endogenous progastrin inhibits the tumorigenicity of APC-mutated colorectal cancer cells in vivo by promoting ICAT expression, thereby counteracting Tcf-4 activity. Progastrin targeting strategies should provide an exciting prospect for the differentiation therapy of colorectal cancer.


Subject(s)
Adenoma/pathology , Cell Cycle Proteins/metabolism , Cell Proliferation , Colorectal Neoplasms/pathology , Gastrins/metabolism , Protein Precursors/metabolism , TCF Transcription Factors/metabolism , Transcription Factors/metabolism , beta Catenin/metabolism , Adaptor Proteins, Signal Transducing , Adenoma/genetics , Adenoma/metabolism , Adenomatous Polyposis Coli/genetics , Animals , Apoptosis/physiology , Cell Cycle Proteins/genetics , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Genes, APC , Humans , Mice , Mice, Nude , Phosphatidylinositol 3-Kinases/physiology , RNA, Small Interfering/pharmacology , Random Allocation , Repressor Proteins , Signal Transduction/physiology , Transcription Factor 7-Like 2 Protein , Transcription Factors/genetics , Transcriptional Activation , Transplantation, Heterologous
15.
Cancer Res ; 78(11): 2925-2938, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29510994

ABSTRACT

Posttreatment recurrence of colorectal cancer, the third most lethal cancer worldwide, is often driven by a subpopulation of cancer stem cells (CSC). The tight junction (TJ) protein claudin-2 is overexpressed in human colorectal cancer, where it enhances cell proliferation, colony formation, and chemoresistance in vitro While several of these biological processes are features of the CSC phenotype, a role for claudin-2 in the regulation of these has not been identified. Here, we report that elevated claudin-2 expression in stage II/III colorectal tumors is associated with poor recurrence-free survival following 5-fluorouracil-based chemotherapy, an outcome in which CSCs play an instrumental role. In patient-derived organoids, primary cells, and cell lines, claudin-2 promoted colorectal cancer self-renewal in vitro and in multiple mouse xenograft models. Claudin-2 enhanced self-renewal of ALDHHigh CSCs and increased their proportion in colorectal cancer cell populations, limiting their differentiation and promoting the phenotypic transition of non-CSCs toward the ALDHHigh phenotype. Next-generation sequencing in ALDHHigh cells revealed that claudin-2 regulated expression of nine miRNAs known to control stem cell signaling. Among these, miR-222-3p was instrumental for the regulation of self-renewal by claudin-2, and enhancement of this self-renewal required activation of YAP, most likely upstream from miR-222-3p. Taken together, our results indicate that overexpression of claudin-2 promotes self-renewal within colorectal cancer stem-like cells, suggesting a potential role for this protein as a therapeutic target in colorectal cancer.Significance: Claudin-2-mediated regulation of YAP activity and miR-222-3p expression drives CSC renewal in colorectal cancer, making it a potential target for therapy. Cancer Res; 78(11); 2925-38. ©2018 AACR.


Subject(s)
Cell Self Renewal/physiology , Claudin-2/metabolism , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/physiopathology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/physiology , Zonula Occludens-2 Protein/metabolism , Animals , Cell Line, Tumor , Cell Proliferation/physiology , Gene Expression Regulation, Neoplastic/physiology , Humans , Mice , Mice, Inbred NOD , Mice, SCID , MicroRNAs , Signal Transduction/physiology
16.
Gastroenterol Clin Biol ; 31(4): 431-5, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17483784

ABSTRACT

AIM: Acute pancreatitis is an important cause of morbidity and mortality, mainly due to sepsis. The aim of this study was to determine the incidence of infectious complications and their impact on mortality in patients hospitalized for acute pancreatitis. PATIENTS AND METHODS: Patients admitted for acute pancreatitis were retrospectively included within a period between 1995 and 2000. Incidence of abdominal and extra-abdominal sepsis and specific care were specifically analyzed. Risk factors for death were evaluated by uni- and multivariated analysis. RESULTS: Two hundreds and twelve consecutive patients (128 males, median age 54 years) were included. Mortality was 10.8%. At least one infectious episode was collected in 25% of the patients with an abdominal sepsis (26.8%), bacteriemia (24.4%), respiratory (24.4%) and urinary tracts (19.5%) infections. Infection was polymicrobial in 37.5%. An antibiotic prophylaxis was administered in 10.8%, more often in patients with severe pancreatitis. It did not alter mortality or incidence of infections but significantly delayed occurrence of sepsis. Mortality of patients treated with more than one line of antibiotics was higher. However in this study infectious complications were not an independent factor for mortality. CONCLUSION: Infections are frequent and polymicrobial but are not an independent prognostic factor during acute pancreatitis.


Subject(s)
Bacterial Infections/epidemiology , Pancreatitis/complications , APACHE , Acute Disease , Adult , Age Factors , Aged , Aged, 80 and over , Amoxicillin/administration & dosage , Amoxicillin/therapeutic use , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/therapeutic use , Antibiotic Prophylaxis , Azithromycin/administration & dosage , Azithromycin/therapeutic use , Bacterial Infections/drug therapy , Ceftriaxone/administration & dosage , Ceftriaxone/therapeutic use , Data Interpretation, Statistical , Drug Therapy, Combination , Female , Humans , Incidence , Male , Middle Aged , Pancreatitis/diagnosis , Pancreatitis/drug therapy , Pancreatitis/mortality , Pancreatitis/surgery , Prognosis , Retrospective Studies , Risk Factors , Sepsis/drug therapy , Sepsis/epidemiology , Time Factors
17.
Geriatr Psychol Neuropsychiatr Vieil ; 15(4): 377-382, 2017 Dec 01.
Article in English | MEDLINE | ID: mdl-29187327

ABSTRACT

Whilst endoscopic retrograde cholangiopancreatography (ERCP) is being practiced on increasingly older patients, its benefits have not been well studied. This study assessed the clinical benefit of ERCP for subjects aged 75 and over, including follow-up at 3 months of geriatric functional parameters and lifestyle. This is a prospective mono-centric, cohort study. All patients aged 75 and over receiving ERCP were enrolled and monitored for a period of 3 months. We recorded 53 inclusions between November 1st 2014 to 31 May 2015. Our "ill-type" was a fragile polypathology woman of 85 years, living at home, with loss of autonomy for instrumental activities of daily living (IADL). Lithiasis was diagnosed in 56.6% of cases, and malignant stenosis in 35.8% of cases. The success rate was 88.7% with a 5.7% complication rate. At 3 months, we observed no clinically significant change in ADL parameters, IADL and lifestyle. Our follow-up data support the practice of ERCP in geriatric populations.


Subject(s)
Cholangiopancreatography, Endoscopic Retrograde/statistics & numerical data , Aged , Aged, 80 and over , Cohort Studies , Female , Follow-Up Studies , France , General Practitioners/statistics & numerical data , Health Care Surveys , Humans , Lithiasis/therapy , Male , Prospective Studies , Treatment Outcome
18.
Clin Cancer Res ; 23(17): 5267-5280, 2017 Sep 01.
Article in English | MEDLINE | ID: mdl-28600477

ABSTRACT

Purpose: Patients with metastatic colorectal cancer suffer from disease relapse mainly due to cancer stem cells (CSC). Interestingly, they have an increased level of blood progastrin, a tumor-promoting peptide essential for the self-renewal of colon CSCs, which is also a direct ß-catenin/TCF4 target gene. In this study, we aimed to develop a novel targeted therapy to neutralize secreted progastrin to inhibit Wnt signaling, CSCs, and reduce relapses.Experimental Design: Antibodies (monoclonal and humanized) directed against progastrin were produced and selected for target specificity and affinity. After validation of their effectiveness on survival of colorectal cancer cell lines harboring B-RAF or K-RAS mutations, their efficacy was assessed in vitro and in vivo, alone or concomitantly with chemotherapy, on CSC self-renewal capacity, tumor recurrence, and Wnt signaling.Results: We show that anti-progastrin antibodies decrease self-renewal of CSCs both in vitro and in vivo, either alone or in combination with chemotherapy. Furthermore, migration and invasion of colorectal cancer cells are diminished; chemosensitivity is prolonged in SW620 and HT29 cells and posttreatment relapse is significantly delayed in T84 cells, xenografted nude mice. Finally, we show that the Wnt signaling activity in vitro is decreased, and, in transgenic mice developing Wnt-driven intestinal neoplasia, the tumor burden is alleviated, with an amplification of cell differentiation in the remaining tumors.Conclusions: Altogether, these data show that humanized anti-progastrin antibodies might represent a potential new treatment for K-RAS-mutated colorectal patients, for which there is a crucial unmet medical need. Clin Cancer Res; 23(17); 5267-80. ©2017 AACR.


Subject(s)
Antibodies, Anti-Idiotypic/administration & dosage , Colorectal Neoplasms/drug therapy , Gastrins/antagonists & inhibitors , Protein Precursors/antagonists & inhibitors , Proto-Oncogene Proteins p21(ras)/genetics , Animals , Antibodies, Anti-Idiotypic/immunology , Antibodies, Monoclonal, Humanized/administration & dosage , Cell Proliferation/drug effects , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Gastrins/blood , Gastrins/immunology , Gene Expression Regulation, Neoplastic/drug effects , HT29 Cells , Humans , Mice , Mutation , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/pathology , Neoplastic Stem Cells/drug effects , Protein Precursors/blood , Protein Precursors/immunology , Wnt Signaling Pathway/drug effects
19.
Oncotarget ; 7(35): 56558-56573, 2016 Aug 30.
Article in English | MEDLINE | ID: mdl-27448961

ABSTRACT

Colorectal cancer lethality usually results from post-treatment relapse in the majority of stage II-IV patients, due to the enhanced resistance of Cancer Stem Cells (CSCs). Here, we show that the nuclear receptor Pregnane X Receptor (PXR, NR1I2), behaves as a key driver of CSC-mediated tumor recurrence. First, PXR is specifically expressed in CSCs, where it drives the expression of genes involved in self-renewal and chemoresistance. Clinically, high levels of PXR correlate with poor recurrence-free survival in a cohort of >200 stage II/III colorectal cancer patients treated with chemotherapy, for whom finding biomarkers of treatment outcome is an urgent clinical need. shRNA silencing of PXR increased the chemo-sensitivity of human colon CSCs, reduced their self-renewal and tumor-initiating potential, and drastically delayed tumor recurrence in mice following chemotherapy. This study uncovers PXR as a key factor for CSC self-renewal and chemoresistance and targeting PXR thus represents a promising strategy to minimize colorectal cancer relapse by selectively sensitizing CSCs to chemotherapy.


Subject(s)
Colonic Neoplasms/metabolism , Neoplasm Recurrence, Local/metabolism , Neoplastic Stem Cells/cytology , Receptors, Steroid/metabolism , Aged , Aldehyde Dehydrogenase/metabolism , Aldehyde Dehydrogenase 1 Family , Animals , Antineoplastic Agents/pharmacology , Biomarkers, Tumor , Cohort Studies , Disease-Free Survival , Drug Resistance, Neoplasm , Female , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Transplantation , Pregnane X Receptor , Prognosis , Retinal Dehydrogenase , Spheroids, Cellular/metabolism
20.
Cancer Res ; 76(12): 3618-28, 2016 06 15.
Article in English | MEDLINE | ID: mdl-27197176

ABSTRACT

Subpopulations of cancer stem-like cells (CSC) are thought to drive tumor progression and posttreatment recurrence in multiple solid tumors. However, the mechanisms that maintain stable proportions of self-renewing CSC within heterogeneous tumors under homeostatic conditions remain poorly understood. Progastrin is a secreted peptide that exhibits tumor-forming potential in colorectal cancer, where it regulates pathways known to modulate colon CSC behaviors. In this study, we investigated the role of progastrin in regulating CSC phenotype in advanced colorectal cancer. Progastrin expression and secretion were highly enriched in colon CSC isolated from human colorectal cancer cell lines and colon tumor biopsies. Progastrin expression promoted CSC self-renewal and survival, whereas its depletion by RNA interference-mediated or antibody-mediated strategies altered the homeostatic proportions of CSC cells within heterogeneous colorectal cancer tumors. Progastrin downregulation also decreased the frequency of ALDH(high) cells, impairing their tumor-initiating potential, and inhibited the high glycolytic activity of ALDH(high) CSC to limit their self-renewal capability. Taken together, our results show how colorectal CSC maintain their tumor-initiating and self-renewal capabilities by secreting progastrin, thereby contributing to the tumor microenvironment to support malignancy. Cancer Res; 76(12); 3618-28. ©2016 AACR.


Subject(s)
Colonic Neoplasms/pathology , Gastrins/physiology , Neoplastic Stem Cells/physiology , Protein Precursors/physiology , Aldehyde Dehydrogenase/metabolism , Animals , Cell Line, Tumor , Cell Survival , Humans , Mice , Tumor Microenvironment
SELECTION OF CITATIONS
SEARCH DETAIL