Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Front Med (Lausanne) ; 11: 1387043, 2024.
Article in English | MEDLINE | ID: mdl-39021822

ABSTRACT

Background: An acute abdomen is a medical emergency that requires early diagnosis and treatment. In pregnancy, this process is significantly more challenging, and radiological findings are sometimes unclear due to the enlarged uterus displacing other structures. Moreover, endometriosis-related complications are rare, and the disease is often undiagnosed. Case presentation: We report a case of acute perforation of the cecum and appendix during pregnancy (35 weeks of gestation) caused by a previously unknown, deep infiltrating endometriosis with focal ulceration of the affected bowel wall, which sonographically seemed to be acute appendicitis. Conclusion: Despite the relatively low risk, clinicians should be aware of possible endometriosis-associated complications in pregnancy with potentially life-threatening events, even in previously unknown endometriosis. Further studies should evaluate intestinal complications during pregnancy in relation to previous treatment of intestinal endometriosis (conservative vs. surgical).

2.
J Clin Ultrasound ; 51(6): 1112-1114, 2023.
Article in English | MEDLINE | ID: mdl-37313859

ABSTRACT

The wash out behavior of focal liver lesions in contrast-enhanced ultrasound plays a central role in tumor classification. Besides hepatocellular carcinomas, other hypervascular tumor entities like renal cell carcinomas may also show a very late wash out, possibly caused by portal-venous tumor vessels. There is need to observe long enough in the late phase in order to classify them correctly.


Subject(s)
Carcinoma, Hepatocellular , Carcinoma, Renal Cell , Kidney Neoplasms , Liver Neoplasms , Humans , Carcinoma, Renal Cell/diagnostic imaging , Contrast Media , Liver Neoplasms/pathology , Carcinoma, Hepatocellular/pathology , Ultrasonography , Neovascularization, Pathologic/diagnostic imaging , Kidney Neoplasms/diagnostic imaging
5.
Endoscopy ; 53(4): 424-428, 2021 Apr.
Article in English | MEDLINE | ID: mdl-32894866

ABSTRACT

BACKGROUND: Conventional endoscopic resection of lesions affecting the appendiceal orifice is difficult. Endoscopic full-thickness resection (EFTR) is a novel technique in interventional endoscopy. As EFTR near the appendiceal orifice is associated with a subtotal appendectomy, it remains unclear whether the risk of developing appendicitis is increased. We conducted a retrospective analysis of lesions involving the appendiceal orifice treated by EFTR. METHODS: This was a multicenter retrospective analysis of patients (n = 50) treated with EFTR for lesions involving the appendiceal orifice between 2014 and 2019. The objective was to evaluate the occurrence of appendicitis. RESULTS: Acute appendicitis occurred in seven patients (14 %) during follow-up. Conservative treatment was sufficient in four cases, and three patients underwent appendectomy. CONCLUSIONS: EFTR of lesions involving the appendiceal orifice may be associated with an imminent risk of developing appendicitis and a consecutive need for appendectomy. Patients should be informed about this specific risk prior to resection. It is unclear why some patients develop appendicitis while the majority remains asymptomatic.


Subject(s)
Appendicitis , Appendix , Endoscopic Mucosal Resection , Appendicitis/etiology , Appendicitis/surgery , Appendix/diagnostic imaging , Appendix/surgery , Endoscopy , Humans , Retrospective Studies
6.
Surg Endosc ; 35(7): 3339-3353, 2021 07.
Article in English | MEDLINE | ID: mdl-32648038

ABSTRACT

INTRODUCTION: Endoscopic full-thickness resection (eFTR) using the full-thickness resection device (FTRD®) is a novel minimally invasive procedure that allows the resection of various lesions in the gastrointestinal tract including the colorectum. Real-world data outside of published studies are limited. The aim of this study was a detailed analysis of the outcomes of colonoscopic eFTR in different hospitals from different care levels in correlation with the number of endoscopists performing eFTR. MATERIAL AND METHODS: In this case series, the data of all patients who underwent eFTR between November 2014 and June 2019 (performed by a total of 22 endoscopists) in 7 hospitals were analyzed retrospectively regarding rates of technical success, R0 resection, and procedure-related complications. RESULTS: Colonoscopic eFTR was performed in 229 patients (64.6% men; average age 69.3 ± 10.3 years) mainly on the basis of the following indication: 69.9% difficult adenomas, 21.0% gastrointestinal adenocarcinomas, and 7.9% subepithelial tumors. The average size of the lesions was 16.3 mm. Technical success rate of eFTR was achieved in 83.8% (binominal confidence interval 78.4-88.4%). Overall, histologically complete resection (R0) was achieved in 77.2% (CI 69.8-83.6%) while histologically proven full-wall excidate was confirmed in 90.0% (CI 85.1-93.7%). Of the resectates obtained (n = 210), 190 were resected en bloc (90.5%). We did not observe a clear improvement of technical success and R0 resection rate over time by the performing endoscopists. Altogether, procedure-related complications were observed in 17.5% (mostly moderate) including 2 cases of acute gangrenous appendicitis requiring operation. DISCUSSION: In this pooled analysis, eFTR represents a feasible, effective, and safe minimally invasive endoscopic technique.


Subject(s)
Adenoma , Colonoscopy , Aged , Female , Hospitals , Humans , Male , Retrospective Studies , Treatment Outcome
8.
Gastrointest Endosc ; 89(6): 1180-1189.e1, 2019 06.
Article in English | MEDLINE | ID: mdl-30653939

ABSTRACT

BACKGROUND AND AIMS: Current international guidelines recommend endoscopic resection for T1 colorectal cancer (CRC) with low-risk histology features and oncologic resection for those at high risk of lymphatic metastasis. Exact risk stratification is therefore crucial to avoid under-treatment as well as over-treatment. Endoscopic full-thickness resection (EFTR) has shown to be effective for treatment of non-lifting benign lesions. In this multicenter, retrospective study we aimed to evaluate efficacy, safety, and clinical value of EFTR for early CRC. METHODS: Records of 1234 patients undergoing EFTR for various indications at 96 centers were screened for eligibility. A total of 156 patients with histologic evidence of adenocarcinoma were identified. This cohort included 64 cases undergoing EFTR after incomplete resection of a malignant polyp (group 1) and 92 non-lifting lesions (group 2). Endpoints of the study were: technical success, R0-resection, adverse events, and successful discrimination of high-risk versus low-risk tumors. RESULTS: Technical success was achieved in 144 out of 156 (92.3%). Mean procedural time was 42 minutes. R0 resection was achieved in 112 of 156 (71.8%). Subgroup analysis showed a R0 resection rate of 87.5% in Group 1 and 60.9% in Group 2 (P < .001). Severe procedure-related adverse events were recorded in 3.9% of patients. Discrimination between high-risk versus low-risk tumor was successful in 155 of 156 cases (99.3%). In Group 1, 84.1% were identified as low-risk lesions, whereas 16.3% in group 2 had low-risk features. In total, 53 patients (34%) underwent oncologic resection due to high-risk features whereas 98 patients (62%) were followed endoscopically. CONCLUSIONS: In early colorectal cancer, EFTR is technically feasible and safe. It allows exact histological risk stratification and can avoid surgery for low-risk lesions. Prospective studies are required to further define indications for EFTR in malignant colorectal lesions and to evaluate long-term outcome.


Subject(s)
Adenocarcinoma/surgery , Colonoscopy/methods , Colorectal Neoplasms/surgery , Adenocarcinoma/pathology , Aged , Aged, 80 and over , Colorectal Neoplasms/pathology , Female , Humans , Male , Middle Aged , Neoplasm Staging , Retrospective Studies , Risk Assessment , Treatment Outcome
9.
J Affect Disord ; 208: 384-391, 2017 Jan 15.
Article in English | MEDLINE | ID: mdl-27810722

ABSTRACT

BACKGROUND: Like other mental illnesses, depression is a culturally sensitive topic. Hence, findings cannot be transferred between countries. We investigated the frequency of depressed mood and its association with diabetes-related factors in a large type 2 diabetes (T2D) cohort from real-life care in Germany. METHODS: 17,563 adults (median [IQR]: 64.5[55.9-71.1] years) from the multicenter diabetes follow-up registry, DPV (diabetes prospective follow-up), were investigated. All had completed the WHO-5 questionnaire, a screening tool for depression. Logistic regression was applied to study the association of demographic and diabetes-related factors with depressed mood (SAS 9.4). P<0.05 was considered significant. RESULTS: Using a WHO-5 cut-off of <13, 27.4% of patients were at risk for depressed mood. A clinical depression diagnosis was recognized in 8.4%. Female sex (OR: 1.5[95%-CI: 1.4-1.6]), young age (1.2[1.1-1.4]), longer diabetes duration (1.2[1.1-1.3]), and living in Northern Germany (1.3[1.2-1.4]) were each associated with increased odds for depressed mood. After adjusting for these confounders, worse glycemic control (1.4[1.3-1.5]), insulin use (1.3[1.2-1.4]), myocardial infarction (1.3[1.2-1.5]), stroke (1.8[1.5-2.0]), retinopathy (1.4[1.3-1.6]), renal failure (1.4[1.2-1.8]), diabetic foot syndrome (1.3[1.2-1.4]), severe hypoglycemia (1.5[1.2-1.9]), two or more inpatient admissions (2.1[1.8-2.4]), and longer duration of hospital stay (1-<14 days: 1.3[1.2-2.3]; >14 days: 2.1[1.9-2.3]) were related to depressed mood. LIMITATION: Due to the cross-sectional design, no causality can be drawn. CONCLUSIONS: In T2D, depressed mood is not uncommon. However, in routine care a clinical depression might be missed and regular screening is advisable. Besides the well-known associations with depressed mood, northern German residence and mainly life-compromising diabetes comorbidities were identified as related factors.


Subject(s)
Depression/etiology , Diabetes Mellitus, Type 2/psychology , Adult , Age Factors , Aged , Cross-Sectional Studies , Depression/diagnosis , Diabetes Complications , Diabetes Mellitus, Type 2/complications , Female , Germany , Hospitalization , Humans , Hypoglycemia/psychology , Length of Stay , Logistic Models , Male , Middle Aged , Prospective Studies , Registries , Sex Factors , Surveys and Questionnaires , Young Adult
11.
Int J Cancer ; 137(3): 678-85, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-25530271

ABSTRACT

Perioperative treatment is a standard of care in locally advanced gastroesophageal cancer (GEC) (gastric adenocarcinoma and gastroesophageal junction (GEJ) adenocarcinoma). While preoperative treatment can be applied to the majority of patients, postoperative chemotherapy can be given only to a fraction. The NeoFLOT-study therefore investigates the application of prolonged neoadjuvant chemotherapy (NACT). Patients with T3, T4, and/or node-positive adenocarcinoma (GEC) were eligible for this multicenter phase II trial. NACT consisted of 6 cycles of oxaliplatin 85 mg/m(2) , leucovorin 200 mg/m(2) , 5-fluorouracil 2600 mg/m(2) and docetaxel 50 mg/m(2) (FLOT) applied q 2 wks. Application of adjuvant chemotherapy was explicitly not part of the protocol. R0-resection rate was evaluated as a primary endpoint. Of 59 enrolled patients, 50 patients underwent surgery and were assessable for the primary endpoint. R0-resection rate was 86.0% (43/50). Pathologic complete response (pCR) was 20.0% (10/50) and a further 20% (10/50) of patients achieved near complete histological remission (<10% residual tumor). Among these very good responders, 85% (17/20) had intestinal type tumors, 10% (2/20) had diffuse and 5% (1/20) had mixed type tumors. After 3 cycles of NACT, 6.9% (4/58) of patients developed progressive disease. Median disease-free survival was 32.9 months. The 1-year survival-rate was 79.3%. Grade 3-4 toxicities included neutropenia 29.3%, febrile neutropenia 1.7%, diarrhea 12.1% and mucositis 6.9%. This study indicates that intensified NACT with 6 cycles of FLOT is highly effective and tolerable in resectable GEC. Very good response (pCR and <10% residual tumor) was predominantly observed in patients with intestinal type tumors.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Esophageal Neoplasms/drug therapy , Esophagogastric Junction/pathology , Stomach Neoplasms/drug therapy , Adenocarcinoma/mortality , Adenocarcinoma/pathology , Adenocarcinoma/surgery , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Chemotherapy, Adjuvant , Esophageal Neoplasms/mortality , Esophageal Neoplasms/pathology , Female , Follow-Up Studies , Germany , Humans , Male , Middle Aged , Neoadjuvant Therapy , Neoplasm Grading , Neoplasm Staging , Perioperative Period , Stomach Neoplasms/mortality , Stomach Neoplasms/pathology , Stomach Neoplasms/surgery , Treatment Outcome
12.
PLoS Pathog ; 9(4): e1003251, 2013.
Article in English | MEDLINE | ID: mdl-23592983

ABSTRACT

Caveolin-1 (Cav1) is a scaffold protein and pathogen receptor in the mucosa of the gastrointestinal tract. Chronic infection of gastric epithelial cells by Helicobacter pylori (H. pylori) is a major risk factor for human gastric cancer (GC) where Cav1 is frequently down-regulated. However, the function of Cav1 in H. pylori infection and pathogenesis of GC remained unknown. We show here that Cav1-deficient mice, infected for 11 months with the CagA-delivery deficient H. pylori strain SS1, developed more severe gastritis and tissue damage, including loss of parietal cells and foveolar hyperplasia, and displayed lower colonisation of the gastric mucosa than wild-type B6129 littermates. Cav1-null mice showed enhanced infiltration of macrophages and B-cells and secretion of chemokines (RANTES) but had reduced levels of CD25+ regulatory T-cells. Cav1-deficient human GC cells (AGS), infected with the CagA-delivery proficient H. pylori strain G27, were more sensitive to CagA-related cytoskeletal stress morphologies ("humming bird") compared to AGS cells stably transfected with Cav1 (AGS/Cav1). Infection of AGS/Cav1 cells triggered the recruitment of p120 RhoGTPase-activating protein/deleted in liver cancer-1 (p120RhoGAP/DLC1) to Cav1 and counteracted CagA-induced cytoskeletal rearrangements. In human GC cell lines (MKN45, N87) and mouse stomach tissue, H. pylori down-regulated endogenous expression of Cav1 independently of CagA. Mechanistically, H. pylori activated sterol-responsive element-binding protein-1 (SREBP1) to repress transcription of the human Cav1 gene from sterol-responsive elements (SREs) in the proximal Cav1 promoter. These data suggested a protective role of Cav1 against H. pylori-induced inflammation and tissue damage. We propose that H. pylori exploits down-regulation of Cav1 to subvert the host's immune response and to promote signalling of its virulence factors in host cells.


Subject(s)
Caveolin 1/metabolism , Gastritis/immunology , Gastritis/microbiology , Helicobacter Infections/immunology , Helicobacter Infections/microbiology , Helicobacter pylori/pathogenicity , Animals , Antigens, Bacterial/genetics , B-Lymphocytes/immunology , Bacterial Proteins/genetics , Caveolin 1/deficiency , Caveolin 1/genetics , Cell Line , Dogs , Enzyme Activation , GTPase-Activating Proteins/metabolism , Gastric Mucosa/immunology , Gastric Mucosa/metabolism , Gastric Mucosa/microbiology , Gastritis/prevention & control , HEK293 Cells , Helicobacter Infections/prevention & control , Helicobacter pylori/genetics , Helicobacter pylori/immunology , Helicobacter pylori/metabolism , Humans , Macrophages/immunology , Madin Darby Canine Kidney Cells , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Parietal Cells, Gastric , Sterol Regulatory Element Binding Protein 1/metabolism , T-Lymphocytes, Regulatory/immunology , Tumor Suppressor Proteins/metabolism , p120 GTPase Activating Protein/metabolism
13.
Am J Physiol Gastrointest Liver Physiol ; 304(2): G181-92, 2013 Jan 15.
Article in English | MEDLINE | ID: mdl-23139224

ABSTRACT

The endocannabinoid system has been shown to mediate beneficial effects on gastrointestinal inflammation via cannabinoid receptors 1 (CB(1)) and 2 (CB(2)). These receptors have also been reported to activate the MAP kinases p38 and c-Jun NH(2)-terminal kinase (JNK), which are involved in early acinar events leading to acute pancreatitis and induction of proinflammatory cytokines. Our aim was to examine the role of cannabinoid receptor activation in an experimental model of acute pancreatitis and the potential involvement of MAP kinases. Cerulein pancreatitis was induced in wild-type, CB(1)-/-, and MK2-/- mice pretreated with selective cannabinoid receptor agonists or antagonists. Severity of pancreatitis was determined by serum amylase and IL-6 levels, intracellular activation of pancreatic trypsinogen, lung myeloperoxidase activity, pancreatic edema, and histological examinations. Pancreatic lysates were investigated by Western blotting using phospho-specific antibodies against p38 and JNK. Quantitative PCR data, Western blotting experiments, and immunohistochemistry clearly show that CB(1) and CB(2) are expressed in mouse pancreatic acini. During acute pancreatitis, an upregulation especially of CB(2) on apoptotic cells occurred. The unselective CB(1)/CB(2) agonist HU210 ameliorated pancreatitis in wild-type and CB(1)-/- mice, indicating that this effect is mediated by CB(2). Furthermore, blockade of CB(2), not CB(1), with selective antagonists engraved pathology. Stimulation with a selective CB(2) agonist attenuated acute pancreatitis and an increased activation of p38 was observed in the acini. With use of MK2-/- mice, it could be demonstrated that this attenuation is dependent on MK2. Hence, using the MK2-/- mouse model we reveal a novel CB(2)-activated and MAP kinase-dependent pathway that modulates cytokine expression and reduces pancreatic injury and affiliated complications.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Cannabinoids/pharmacology , Dronabinol/analogs & derivatives , Intracellular Signaling Peptides and Proteins/metabolism , Pancreas, Exocrine/drug effects , Pancreatitis/prevention & control , Protein Serine-Threonine Kinases/metabolism , Receptor, Cannabinoid, CB2/agonists , p38 Mitogen-Activated Protein Kinases/metabolism , Amylases/blood , Animals , Apoptosis , Blotting, Western , Ceruletide , Disease Models, Animal , Dronabinol/pharmacology , Edema/chemically induced , Edema/enzymology , Edema/prevention & control , Enzyme Activation , Immunohistochemistry , Interleukin-6/blood , Intracellular Signaling Peptides and Proteins/deficiency , Intracellular Signaling Peptides and Proteins/genetics , Lung/drug effects , Lung/enzymology , Mice , Mice, Inbred C57BL , Mice, Knockout , Pancreas, Exocrine/enzymology , Pancreatitis/blood , Pancreatitis/chemically induced , Pancreatitis/enzymology , Pancreatitis/genetics , Peroxidase/metabolism , Phosphorylation , Polymerase Chain Reaction , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , Receptor, Cannabinoid, CB1/agonists , Receptor, Cannabinoid, CB1/genetics , Receptor, Cannabinoid, CB1/metabolism , Receptor, Cannabinoid, CB2/genetics , Receptor, Cannabinoid, CB2/metabolism , Trypsinogen/metabolism
14.
Z Med Phys ; 23(1): 21-32, 2013 Feb.
Article in German | MEDLINE | ID: mdl-22917569

ABSTRACT

For LDR-brachytherapy, a limited number of implant geometries and materials are available. To avoid wound healing related hyper-proliferation (stenosis, keloids) a novel radioactive foil system was developed based on beta emitting (32)P, which can be easily integrated in existing implants such as urethral catheters or bile duct stents. As substrate material for these foils PEEK (polyetherethercetone) was chosen because of its radiation hardness during neutron activation of (32)P. The activity was determined by liquid scintillation counting and gamma spectroscopy, dose distributions were measured with scintillation detectors and radiochromic films. The correlation between activity and dose was checked by Monte-Carlo-simulations (Geant4). Prototypes of the (32)P-implants have shown in wash-out tests the required tightness for sealed radioactive sources. In animal tests on urethra and bile duct, the uncomplicated and save application of (32)P-foils mounted on standard implants has been demonstrated, which is almost unchanged due to the simple radiation protection with plexiglass. This concept of radioactive implants with integrated (32)P-foils could extend essentially the application possibilities of LDR-brachytherapy.


Subject(s)
Brachytherapy/instrumentation , Brachytherapy/methods , Gastroenterology/instrumentation , Phosphorus Radioisotopes/therapeutic use , Urology/instrumentation , Constriction, Pathologic/radiotherapy , Equipment Design , Equipment Failure Analysis , Humans , Prostheses and Implants , Radiopharmaceuticals/therapeutic use
15.
J Cell Mol Med ; 16(8): 1776-91, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22004109

ABSTRACT

A role of heat shock protein 27 (HSP27) as a potential biomarker has been reported in various tumour entities, but comprehensive studies in pancreatic cancer are lacking. Applying tissue microarray (TMA) analysis, we correlated HSP27 protein expression status with clinicopathologic parameters in pancreatic ductal adenocarcinoma specimens from 86 patients. Complementary, we established HSP27 overexpression and RNA-interference models to assess the impact of HSP27 on chemo- and radiosensitivity directly in pancreatic cancer cells. In the TMA study, HSP27 expression was found in 49% of tumour samples. Applying univariate analyses, a significant correlation was found between HSP27 expression and survival. In the multivariate Cox-regression model, HSP27 expression emerged as an independent prognostic factor. HSP27 expression also correlated inversely with nuclear p53 accumulation, indicating either protein interactions between HSP27 and p53 or TP53 mutation-dependent HSP27-regulation in pancreatic cancer. In the sensitivity studies, HSP27 overexpression rendered HSP27 low-expressing PL5 pancreatic cancer cells more susceptible towards treatment with gemcitabine. Vice versa, HSP27 protein depletion in HSP27 high-expressing AsPC-1 cells caused increased gemcitabine resistance. Importantly, HSP27 expression was inducible in pancreatic cancer cell lines as well as primary cells. Taken together, our study suggests a role for HSP27 as a prognostic and predictive marker in pancreatic cancer. Assessment of HSP27 expression could thus facilitate the identification of specific patient subpopulations that might benefit from individualized treatment options. Additional studies need to clarify whether modulation of HSP27 expression could represent an attractive concept to support the incorporation of hyperthermia in clinical treatment protocols for pancreatic cancer.


Subject(s)
Adenocarcinoma/metabolism , Carcinoma, Pancreatic Ductal/metabolism , HSP27 Heat-Shock Proteins/metabolism , Adenocarcinoma/drug therapy , Adenocarcinoma/pathology , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/metabolism , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Deoxycytidine/analogs & derivatives , Deoxycytidine/therapeutic use , Drug Screening Assays, Antitumor , Female , Heat-Shock Proteins , Heat-Shock Response/drug effects , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Male , Middle Aged , Molecular Chaperones , Multivariate Analysis , Pancreas/metabolism , Pancreas/pathology , Prognosis , Tissue Array Analysis , Gemcitabine
16.
Am J Physiol Gastrointest Liver Physiol ; 301(5): G773-82, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21778463

ABSTRACT

In acute pancreatitis, endoplasmic reticulum (ER) stress prompts an accumulation of malfolded proteins inside the ER, initiating the unfolded protein response (UPR). Because the ER chaperone tauroursodeoxycholic acid (TUDCA) is known to inhibit the UPR in vitro, this study examined the in vivo effects of TUDCA in an acute experimental pancreatitis model. Acute pancreatitis was induced in Wistar rats using caerulein, with or without prior TUDCA treatment. UPR components were analyzed, including chaperone binding protein (BiP), phosphorylated protein kinase-like ER kinase (pPERK), X-box binding protein (XBP)-1, phosphorylated c-Jun NH(2)-terminal kinase (pJNK), CCAAT/enhancer binding protein homologues protein, and caspase 12 and 3 activation. In addition, pancreatitis biomarkers were measured, such as serum amylase, trypsin activation, edema formation, histology, and the inflammatory reaction in pancreatic and lung tissue. TUDCA treatment reduced intracellular trypsin activation, edema formation, and cell damage, while leaving amylase levels unaltered. The activation of myeloperoxidase was clearly reduced in pancreas and lung. Furthermore, TUDCA prevented caerulein-induced BiP upregulation, reduced XBP-1 splicing, and caspase 12 and 3 activation. It accelerated the downregulation of pJNK. In controls without pancreatitis, TUDCA showed cytoprotective effects including pPERK signaling and activation of downstream targets. We concluded that ER stress responses activated in acute pancreatitis are grossly attenuated by TUDCA. The chaperone reduced the UPR and inhibited ER stress-associated proapoptotic pathways. TUDCA has a cytoprotective potential in the exocrine pancreas. These data hint at new perspectives for an employment of chemical chaperones, such as TUDCA, in prevention of acute pancreatitis.


Subject(s)
Acinar Cells/drug effects , Endoplasmic Reticulum Stress/drug effects , Inflammation/drug therapy , Pancreatitis/drug therapy , Taurochenodeoxycholic Acid/therapeutic use , Unfolded Protein Response/drug effects , Acinar Cells/metabolism , Acinar Cells/pathology , Animals , Ceruletide , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/physiology , Inflammation/metabolism , Inflammation/pathology , Male , Pancreas/drug effects , Pancreas/metabolism , Pancreas/pathology , Pancreatitis/chemically induced , Pancreatitis/metabolism , Pancreatitis/pathology , Rats , Rats, Wistar , Taurochenodeoxycholic Acid/pharmacology
17.
Biochem J ; 438(2): 315-23, 2011 Sep 01.
Article in English | MEDLINE | ID: mdl-21619550

ABSTRACT

Bile acids from duodenogastric reflux promote inflammation and increase the risk for gastro-oesophageal cancers. FXR (farnesoid X receptor/NR1H4) is a transcription factor regulated by bile acids such as CDCA (chenodeoxycholic acid). FXR protects the liver and the intestinal tract against bile acid overload; however, a functional role for FXR in the stomach has not been described. We detected FXR expression in the normal human stomach and in GC (gastric cancer). FXR mRNA and protein were also present in the human GC cell lines MKN45 and SNU5, but not in the AGS cell line. Transfection of FXR into AGS cells protected against TNFα (tumour necrosis factor α)-induced cell damage. We identified K13 (keratin 13), an anti-apoptotic protein of desmosomes, as a novel CDCA-regulated FXR-target gene. FXR bound to a conserved regulatory element in the proximal human K13 promoter. Gastric expression of K13 mRNA was increased in an FXR-dependent manner by a chow diet enriched with 1% (w/w) CDCA and by indomethacin (35 mg/kg of body weight intraperitoneal) in C57BL/6 mice. FXR-deficient mice were more susceptible to indomethacin-induced gastric ulceration than their WT (wild-type) littermates. These results suggest that FXR increases the resistance of human and murine gastric epithelial cells to inflammation-mediated damage and may thus participate in the development of GC.


Subject(s)
Cytoprotection , Epithelial Cells/pathology , Inflammation/pathology , Receptors, Cytoplasmic and Nuclear/metabolism , Stomach/pathology , Animals , Apoptosis/genetics , Cell Line , Disease Susceptibility , Epithelial Cells/metabolism , Gene Expression Regulation, Neoplastic , Humans , Inflammation/metabolism , Keratin-13/genetics , Keratin-13/metabolism , Mice , Mice, Inbred C57BL , Promoter Regions, Genetic/genetics , Protein Binding , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Transcriptional Activation/genetics
18.
Mol Cancer ; 9: 127, 2010 May 28.
Article in English | MEDLINE | ID: mdl-20509860

ABSTRACT

BACKGROUND: Inactivation of the Fanconi anemia (FA) pathway through defects in one of 13 FA genes occurs at low frequency in various solid cancer entities among the general population. As FA pathway inactivation confers a distinct hypersensitivity towards DNA interstrand-crosslinking (ICL)-agents, FA defects represent rational targets for individualized therapeutic strategies. Except for pancreatic cancer, however, the prevalence of FA defects in gastrointestinal (GI) tumors has not yet been systematically explored. RESULTS: A panel of GI cancer cell lines was screened for FA pathway inactivation applying FANCD2 monoubiquitination and FANCD2/RAD51 nuclear focus formation and a newly identified FA pathway-deficient cell line was functionally characterized. The hepatocellular carcinoma (HCC) line HuH-7 was defective in FANCD2 monoubiquitination and FANCD2 nuclear focus formation but proficient in RAD51 focus formation. Gene complementation studies revealed that this proximal FA pathway inactivation was attributable to defective FANCC function in HuH-7 cells. Accordingly, a homozygous inactivating FANCC nonsense mutation (c.553C > T, p.R185X) was identified in HuH-7, resulting in partial transcriptional skipping of exon 6 and leading to the classic cellular FA hypersensitivity phenotype; HuH-7 cells exhibited a strongly reduced proliferation rate and a pronounced G2 cell cycle arrest at distinctly lower concentrations of ICL-agents than a panel of non-isogenic, FA pathway-proficient HCC cell lines. Upon retroviral transduction of HuH-7 cells with FANCC cDNA, FA pathway functions were restored and ICL-hypersensitivity abrogated. Analyses of 18 surgical HCC specimens yielded no further examples for genetic or epigenetic inactivation of FANCC, FANCF, or FANCG in HCC, suggesting a low prevalence of proximal FA pathway inactivation in this tumor type. CONCLUSIONS: As the majority of HCC are chemoresistant, assessment of FA pathway function in HCC could identify small subpopulations of patients expected to predictably benefit from individualized treatment protocols using ICL-agents.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/genetics , Drug Resistance, Neoplasm/genetics , Fanconi Anemia Complementation Group C Protein/genetics , Liver Neoplasms/genetics , Cell Line, Tumor , Gene Expression , Gene Expression Profiling , Gene Silencing , Humans , Immunoblotting , RNA, Messenger/analysis
19.
Oncology ; 77(1): 40-8, 2009.
Article in English | MEDLINE | ID: mdl-19556808

ABSTRACT

BACKGROUND: No data have previously been available regarding the current treatment of patients with pancreatic cancer (PC) in German hospitals and medical practices. METHODS: Between February 2007 and March 2008 we conducted a national survey [on behalf of the Arbeitsgemeinschaft Internistische Onkologie (AIO) and the Chirurgische Arbeitsgemeinschaft Onkologie (CAO)] regarding the current surgical and oncological treatment of PC in Germany. Standardized questionnaires were sent via mailing lists to members of the AIO and CAO (n = 1,130). The data were analyzed using SPSS software (version 16.0). Pre-defined subgroup analysis was performed by grouping the results of each question with regard to the professional site of the responding physician and to the number of patients treated in their institution by year. RESULTS: 181 (16%) of the oncological questionnaires were sent back. For 61% of the participating centers, a histological confirmation of PC diagnosis is obligatory. 21% of physicians offer neoadjuvant therapy to patients with potentially resectable PC. In the adjuvant treatment after curative-intent surgery, gemcitabine (Gem) is regarded as standard of care by 71% after R0 resection and 62% after R1 resection. For patients with locally advanced PC, 52% of the participating centers recommend systemic chemotherapy, 17% prefer combined primary chemoradiotherapy. Most centers (59%) base their decision of combination regimens for metastatic disease on the performance status of their patients. In patients with a good status, 28% apply single-agent Gem, 3% use Gem + capecitabine, 12% Gem + erlotinib, 16% Gem + oxaliplatin, and 8% Gem + cisplatin. Only 28% of the survey doctors offer second-line treatment to the majority of their patients with advanced PC. CONCLUSION: Not every PC patient in Germany is treated according to the present S3 guidelines. Diagnosis and treatment of PC in Germany still need to be improved.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Health Care Surveys , Pancreatic Neoplasms/therapy , Adult , Capecitabine , Chemotherapy, Adjuvant , Combined Modality Therapy , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Erlotinib Hydrochloride , Fluorouracil/administration & dosage , Fluorouracil/analogs & derivatives , Follow-Up Studies , Germany , Humans , Lymphatic Metastasis , Male , Neoplasm Staging , Organoplatinum Compounds/administration & dosage , Oxaliplatin , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/radiotherapy , Practice Guidelines as Topic , Prognosis , Quinazolines/administration & dosage , Radiotherapy Dosage , Surveys and Questionnaires , Treatment Outcome , Young Adult , Gemcitabine
SELECTION OF CITATIONS
SEARCH DETAIL
...