Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Anticancer Res ; 43(11): 4897-4904, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37909980

ABSTRACT

BACKGROUND/AIM: The transcription factors NFATc2 and Sp1 play a key role in the progression of pancreatic cancer because they interact inside the cells and exert their carcinogenic effect through transcriptional modification. Drugs can also induce a variety of oncogenic signalling cascades. The risk of tumour progression and metastasis seems to be significantly increased in the perioperative period. Our research group has previously demonstrated the function of the interaction between NFATc2 and Sp1 in pancreatic cancer and has identified the proto-oncogene cFos as a target gene. We also found that the anaesthetic drug propofol has anti-tumour properties. The aim of the present study was to investigate the effect of propofol on the expression of the transcription factors NFATc2, Sp1 and cFos in the pancreatic cancer cell lines PaTu 8988t and PANC-1 and to analyse the relevance of this effect for the cells. MATERIALS AND METHODS: Stimulation with propofol and its effects on the expression of NFATc2, Sp1 and cFos were assessed by immunoblot. Cell cycle distribution was analysed by flow cytometry, and cell proliferation was measured with the ELISA BrdU assay. Propofol and siRNA against cFos were used for stimulation. RESULTS: Propofol regulated the expression of NFATc2, Sp1 and cFos. Stimulation with 250 µM or 500 µM propofol decreased NFATc2, Sp1 and cFos signalling in the Western blot analysis. At the same time, propofol significantly inhibited proliferation and activated cell cycle. The same proliferation behaviour was observed after transient cFos inhibition. These effects were potentiated by simultaneous stimulation with propofol and transient inhibition of cFos, further inhibiting cell proliferation. Interestingly, the cell cycle activation observed after stimulation with propofol alone was reversed in both cell lines. CONCLUSION: Anaesthetists only see oncological patients in a short time window. However, the perioperative period is increasingly recognised as a very vulnerable time with a major impact on tumour progression. Further studies are needed to identify the underlying mechanisms and to verify their clinical relevance, especially in anaesthesia.


Subject(s)
Pancreatic Neoplasms , Propofol , Humans , Pancreas , Pancreatic Neoplasms/genetics , Propofol/pharmacology , Sp1 Transcription Factor/genetics , Transcription Factors , Pancreatic Neoplasms
2.
Cancer Genomics Proteomics ; 20(6suppl): 706-711, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38035702

ABSTRACT

BACKGROUND/AIM: One in two people will develop a tumor during their lifetime. Adenocarcinoma of the pancreas is one of the most aggressive types of cancer in humans with very poor long-term survival. A central role in the carcinogenesis of pancreatic cancer has been attributed to NFAT transcription factors. Previous studies have identified the transcription factor Sp1 as a binding partner of NFATc2 in pancreatic cancer. Using expression profile analysis, our group was able to identify the tumor necrosis factor TNFalpha as a target gene of the interaction between NFATc2 and Sp1. The present study investigated the effect of TNFalpha over-expression via the transcription factors NFATc2 and Sp1 on the pancreatic cancer cell lines PaTu 8988t and PANC-1. MATERIALS AND METHODS: Transient transfection of NFATc2, Sp1, and TNFalpha siRNAs and their effects on the expression were investigated with immunoblot. Cell proliferation was measured with the ELISA BrdU assay. Cell migration was assayed with a Cell Migration Assay Kit using a Boyden chamber. RESULTS: Inhibition of the transfection factors NFATc2, Sp1, or TNFalpha by siRNA significantly inhibited proliferation, which was exacerbated when using the combination of NFATc2 and Sp1. TNFalpha was able to counterbalance this effect. In contrast to proliferation, migration of pancreatic cancer cells was increased by inhibiting these transfection factors. CONCLUSION: Tumor progression is strongly influenced by transcriptional changes in signaling cascades and oncogene mutations as well as by changes in tumor suppressor genes. Further studies are needed to understand the underlying mechanisms of these processes.


Subject(s)
Pancreatic Neoplasms , Sp1 Transcription Factor , Tumor Necrosis Factor-alpha , Humans , Cell Line, Tumor , Cell Proliferation , Gene Expression Regulation, Neoplastic , Pancreas/metabolism , Pancreas/pathology , Pancreatic Neoplasms/pathology , Sp1 Transcription Factor/genetics , Tumor Necrosis Factor-alpha/genetics , NFATC Transcription Factors/genetics , Pancreatic Neoplasms
3.
Front Neurol ; 14: 1325950, 2023.
Article in English | MEDLINE | ID: mdl-38425753

ABSTRACT

Background: The vaso- and psychoactive endogenous Neuropeptide Y (NPY) has repeatedly been shown to be excessively released after subarachnoid hemorrhage and in numerous psychiatric disorders. NPY is stored in sympathetic perivascular nerve fibers around the major cerebral arteries. This prospective study was designed to analyze the impact of microsurgical and endovascular manipulation of the cerebral vasculature versus cranio- and durotomy alone on the serum levels of NPY. Methods: 58 patients (drop-out n = 3; m:f = 26:29; mean age 52.0 ± 14.1 years) were prospectively enrolled. The vascular group underwent repair for unruptured intracranial aneurysms (UIA) of the anterior circulation [endovascular aneurysm occlusion (EV) n = 13; microsurgical clipping (MS) n = 17]; in the non-vascular group, 14 patients received microsurgical resection of a small-sized convexity meningioma (CM), and 11 patients with surgically treated degenerative lumbar spine disease (LD) served as control. Plasma was drawn (1) before treatment (t0), (2) periprocedurally (t1), (3) 6 h postprocedurally (t2), (4) 72 h postprocedurally (t3), and (5) at the 6-week follow-up (FU; t4) to determine the NPY levels via competitive enzyme immunoassay in duplicate serum samples. We statistically evaluated differences between groups by calculating one-way ANOVA and for changes along the time points using repeated measure ANOVA. Results: Except for time point t0, the serum concentrations of NPY ranged significantly higher in the vascular than in the non-vascular group (p < 0.001), with a slight decrease in both vascular subgroups 6 h postprocedurally, followed by a gradual increase above baseline levels until FU. At t3, the EV subgroup showed significantly higher NPY levels (mean ± standard deviation) than the MS subgroup (0.569 ± 0.198 ng/mL vs. 0.415 ± 0.192 ng/mL, p = 0.0217). The highest NPY concentrations were measured in the EV subgroup at t1, t3, and t4, reaching a climax at FU (0.551 ± 0.304 ng/mL). Conclusion: Our study reveals a first insight into the short-term dynamics of the serum levels of endogenous NPY in neurosurgical and endovascular procedures, respectively: Direct manipulation within but also next to the major cerebral arteries induces an excessive release of NPY into the serum. Our findings raise the interesting question of the potential capacity of NPY in modulating the psycho-behavioral outcome of neurovascular patients.

4.
In Vivo ; 36(6): 2722-2729, 2022.
Article in English | MEDLINE | ID: mdl-36309382

ABSTRACT

BACKGROUND/AIM: The influence of surgical interventions and anaesthesiological procedures on tumour progression was investigated as early as the 1920s. In current cancer management, the perioperative phase is increasingly being considered a vulnerable period with an increased risk of tumour cell dissemination due to medication, surgical manipulation, and immunosuppression. The extent to which narcotics administered in the perioperative setting influence the oncological outcomes of patients with pancreatic cancer is still unclear. MATERIALS AND METHODS: To investigate the effect of propofol and etomidate on the proliferation, cell-cycle distribution, apoptosis, and necrosis of pancreatic tumour cells in vitro, PaTu 8988t and Panc-1 pancreatic cancer cells were treated with 0-1,000 µM propofol or etomidate for 24 h each. Cell proliferation was measured with enzyme-linked immunosorbent-bromodeoxyuridine assay. The apoptosis rate was analysed with annexin V staining and the cell-cycle distribution with flow cytometry. RESULTS: Propofol at 1,000 µM induced apoptosis and inhibited cell proliferation. The cell cycle showed an increased S-phase and reduced cells in the G1-phase. At 100 µM, propofol significantly inhibited proliferation of the pancreatic cancer cell line PaTu 8988t and reduced cells in the G2-phase in the cell cycle. Etomidate had no effects on cell-cycle distribution, proliferation, apoptosis, and necrosis at the concentrations used. CONCLUSION: In this study, propofol was shown to have anticancer effects by induction of apoptosis and inhibition of cell proliferation, while etomidate did not affect pancreatic cancer cells. However, it is too early to make any recommendation for changes in clinical practice and further clinical studies are warranted to investigate the effect of anaesthetics on cancer progression.


Subject(s)
Etomidate , Pancreatic Neoplasms , Propofol , Humans , Etomidate/pharmacology , Etomidate/therapeutic use , Propofol/pharmacology , Propofol/therapeutic use , Apoptosis , Necrosis , Pancreatic Neoplasms/pathology , Cell Cycle , Cell Proliferation , Pancreatic Neoplasms
6.
In Vivo ; 36(2): 576-581, 2022.
Article in English | MEDLINE | ID: mdl-35241509

ABSTRACT

BACKGROUND/AIM: Adenocarcinoma of the pancreas is one of the most aggressive malignant diseases in humans. Characteristics of this tumour type are poor response to radiotherapy and chemotherapeutic agents as well as metastasis in the absence of an organ capsule. The best therapeutic option is surgical removal of the tumour followed by chemotherapy or radiotherapy. Yet, even after surgical R0-resection, the 5-year survival probability is only about 20% because of the high recurrence rate of this tumour and complications due to metastases. Furthermore, recent studies have shown that the perioperative period is a particularly vulnerable phase, during which tumour progression and metastasis may be facilitated. The effects of analgesics administered during the perioperative period are still unknown. The present work investigated the effects of analgesics on pancreatic cancer cell migration in vitro. MATERIALS AND METHODS: The migratory potential of pancreatic cancer cells was analysed using a Cell Migration Assay Kit with a Boyden chamber, in which cells migrate through a semi-permeable membrane under different stimuli. Cell concentration was measured by reading fluorescence (Ex/Em=530/590 nm) in a plate reader. RESULTS: Migration in PANC-1 pancreatic cancer cells was significantly decreased after 24 h stimulation with 100 µM of ropivacaine, 100 nM of sufentanil, 1,000 µM of ropivacaine and 1,000 nM of sufentanil. In the PaTu 8988t cell line, incubation with 10 µM of ropivacaine caused a slight but statistically significant increase in migration, whereas lidocaine, metamizole and paracetamol did not significantly affect migration. CONCLUSION: The risk of tumour progression and metastasis seems to be increased during major oncological surgical interventions. The recent advances in the molecular and biological understanding of pathogenesis of pancreatic cancer have not yet significantly improved patient outcome. Therefore, further studies are needed to identify the underlying mechanisms of this aggressive tumour and establish new therapeutic options for the future.


Subject(s)
Pancreatic Neoplasms , Analgesics , Cell Line, Tumor , Humans , Pancreas/pathology , Pancreatic Neoplasms/pathology , Ropivacaine
7.
J Cancer Sci Clin Ther ; 6(4): 446-451, 2022.
Article in English | MEDLINE | ID: mdl-36777697

ABSTRACT

Introduction: Pancreatic ductal adenocarcinoma is one of the most aggressive malignancies in humans. The main reason for its unfavourable prognosis is the combination of rapid tumour growth, early-onset metastasis and currently still inadequate diagnostic and therapeutic options. Thus, only very few patients are eligible for radical resection of the primary tumour as the only curative treatment option available so far. In the perioperative period, tumour progression and metastasis are facilitated by the activation of key signalling pathways and the altered regulation of transcription factors. Various tumour entities have shown increased expression of the integrin-3 receptor subunit, which correlates with more rapid tumour progression and metastasis through advanced migration, invasion and proliferation. The influence of perioperative medication and postoperative pain management remains unclear. To investigate the effects of ketamine, s-ketamine and MK 801 on integrin beta-3-mediated cell migration in pancreatic cancer cells in vitro. Methods: The effects of ketamine, s-ketamine and MK 801 on integrin beta-3 expression were investigated with immunoblot. Cell migratory potentials were analysed using a Cell Migration Assay Kit with a Boyden chamber, in which cells migrate through a semipermeable membrane under different stimuli. Results: Stimulation with ketamine and MK 801 significantly promoted migration in pancreatic cancer cells, increasing the expression of integrin beta-3. Conclusion: Novel therapeutic approaches target the effective modulation of specific signalling and transcription pathways. The prerequisite for such 'target therapies' is comprehensive knowledge about the respective carcinogenesis. Further studies are required to identify the underlying disease mechanisms of pancreatic carcinoma.

8.
Anticancer Res ; 39(11): 6049-6055, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31704831

ABSTRACT

BACKGROUND/AIM: For patients undergoing cancer surgery, the risk for cancer progression is enhanced during the perioperative period. To what extent the type of anesthetic can affect the metastatic process and finally the outcome of patients with cancer is under debate. For this reason, the aim of this study was to investigate the effects of the volatile anesthetics sevoflurane and desflurane on colon cancer cells in vitro. MATERIALS AND METHODS: SW480 colon carcinoma cells were exposed for 3 or 6 h to sevoflurane (1 or 2.5 vol%) or desflurane (6 or 12 vol%). Cell cycle distribution was analyzed by flow cytometry after a 24-72 h recovery and apoptosis was detected by annexin V staining after a 0-48 h recovery. Viability was tested by measuring ATP content after 0 and 24 h recovery. RESULTS: Treatment with sevoflurane or desflurane caused no or only slight changes in cell-cycle distribution and apoptosis rate. Desflurane at 12vol% significantly reduced cell viability by 17±25% and 11±22% after 3 and 6 h incubation and 24 h recovery, respectively, while 2.5 vol% sevoflurane slightly increased viability. CONCLUSION: At clinically relevant concentrations, sevoflurane and desflurane had only slight effects on SW480 colon cancer cells in vitro.


Subject(s)
Anesthetics, Inhalation/pharmacology , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Proliferation/drug effects , Colonic Neoplasms/pathology , Desflurane/pharmacology , Sevoflurane/pharmacology , Colonic Neoplasms/drug therapy , Humans , In Vitro Techniques , Tumor Cells, Cultured
9.
Anticancer Res ; 39(9): 4721-4728, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31519571

ABSTRACT

BACKGROUND/AIM: Recent research has identified the transcription factors NFATc2 and Sp1 as key regulators in the carcinogenesis of pancreatic carcinoma. This study aimed to examine the effect of clinically achievable dosages of analgesics including ketamine, s-ketamine, metamizole, and paracetamol as well as that of sufentanil, ropicavaine, and lidocaine on pancreatic carcinoma cells and the expression of NFATc2 and Sp1. MATERIALS AND METHODS: The effects of analgesics on the expression of NFATc2 and Sp1 were investigated with immunoblotting. Cell proliferation was measured with the ELISA BrdU assay. RESULTS: In PaTu8988t pancreatic carcinoma cells, 48 h stimulation with ketamine and s-ketamine significantly inhibited proliferation and decreased expression of NFATc2 in the nucleus. The addition of metamizole and lidocaine reduced proliferation of PaTu8988t cells after 48 h. CONCLUSION: New treatment concepts target specific signaling and transcription pathways. The extent to which drugs influence these mechanisms in pancreatic carcinoma cells needs to be investigated in future studies.


Subject(s)
Analgesics/pharmacology , Anesthetics, Local/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , NFATC Transcription Factors/metabolism , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Sp1 Transcription Factor/metabolism , Biomarkers , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Pancreatic Neoplasms/pathology , Transcription, Genetic , Pancreatic Neoplasms
10.
Int J Neurosci ; 129(10): 986-994, 2019 Oct.
Article in English | MEDLINE | ID: mdl-30957600

ABSTRACT

Background: Numerous experimental studies show that anesthetics are potentially toxic to the immature brain. Even though benzodiazepines are widely used in pediatric anesthesia and intensive care medicine, only a few studies examine the effects of these drugs on immature neurons. Methods: Hippocampal neuronal cell cultures of embryonic Wistar rats (15 days in culture) were incubated with midazolam 100 or 300 nM for either 30 min or 4 h. The time course of the mRNA expression of the glutamate receptors subunits NR1, NR2A and NR2B of the NMDA receptor, the GluA-1 and A-2 subunits of the AMPA receptor as well as the alpha 1 subunit of the GABAA receptor were examined by PCR. Apoptosis was detected using Western blot analysis for BAX, Bcl-2 and Caspase-3. Results: Midazolam at 100 and 300 nM applied for 30 min and 100 nM for 4 h affected glutamate receptor and GABAA receptor subunit expression. However, these effects were reversible within 72 h following washout. When 300 nM midazolam was applied for 4 h a significant increase in the NR 1 and NR 2A mRNA subunit expression could be detected. The increase in NR 2B receptor subunit expression as well as the GluA1 subunit expression was not reversible within 72 h following washout. This increase in mRNA glutamate receptor subunit expression was associated with a significant increase in neuronal apoptosis. Conclusion: In immature neurons midazolam altered GABA and glutamate mRNA receptor subunit expression. Prolonged increase in midazolam-induced glutamate receptor expression was associated with apoptosis.


Subject(s)
GABA Modulators/pharmacology , Hippocampus/metabolism , Midazolam/pharmacology , Neurons/metabolism , RNA, Messenger/biosynthesis , Receptors, GABA-A/biosynthesis , Allosteric Regulation/drug effects , Allosteric Regulation/physiology , Animals , Apoptosis/drug effects , Apoptosis/physiology , Cells, Cultured , Dose-Response Relationship, Drug , Gene Expression , Hippocampus/drug effects , Hippocampus/embryology , Neurons/drug effects , RNA, Messenger/genetics , Rats , Rats, Wistar , Receptors, GABA-A/genetics
11.
Mediators Inflamm ; 2019: 8274903, 2019.
Article in English | MEDLINE | ID: mdl-30804708

ABSTRACT

INTRODUCTION: Recent studies have shown that acetylcholinesterase (AChE) and butyrylcholinesterase (BChE) may serve as important diagnostic and therapeutic targets in sepsis. Since polymorphonuclear neutrophils (PMNs) play a pivotal role in the early phase of sepsis, we evaluated the potential therapeutic effects of cholinesterase inhibitors on PMN functions during cecal ligation and puncture- (CLP-) induced sepsis and investigated the roles of AChE and BChE as inflammatory markers under standardized experimental conditions. METHODS: Sham surgery or CLP was performed in male Wistar rats (n = 60). Animals were randomized into four groups: physostigmine, 100 µg/kg; neostigmine, 75 µg/kg; 0.9% saline (control group); and sham group, each applied four times over 24 h. The levels of reactive oxygen species (ROS) production and CD11b/CD62l expression were quantified by flow cytometry at t = 0, 6, 15, 20, and 24 h. Blood gas analysis as well as AChE and BChE activity levels was measured by validated point-of-care measurements. Clinical scores and survival times were determined. RESULTS: CLP induced a significant increase in ROS production and CD11b upregulation by rat PMNs. Treatment with physostigmine or neostigmine significantly reduced ROS production and CD11b upregulation by PMNs 20 h after CLP induction. In physostigmine-treated animals, survival times were significantly improved compared to the control animals, but not in neostigmine-treated animals. While AChE activity significantly decreased in the control animals at t > 6 h, AChE activity did not change in the sham group. BChE activity decreased at t > 20 h in the control animals. CONCLUSION: While AChE activity may serve as an acute inflammatory marker, BChE activity shows a delayed decrease. Administration of centrally acting physostigmine in CLP-induced sepsis in rats has protective effects on PMN functions and improves survival times, which may be of interest in clinical practice.


Subject(s)
Acetylcholinesterase/metabolism , Biomarkers/metabolism , Butyrylcholinesterase/metabolism , Neostigmine/therapeutic use , Neutrophils/drug effects , Physostigmine/therapeutic use , Sepsis/drug therapy , Sepsis/metabolism , Animals , Blood Gas Analysis , Male , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism
12.
BMC Biochem ; 20(1): 2, 2019 01 29.
Article in English | MEDLINE | ID: mdl-30696421

ABSTRACT

BACKGROUND: Adenocarcinoma of the pancreas is one of the most aggressive tumor diseases affecting the human body. The oncogenic potential of pancreatic cancer is mainly characterized by extremely rapid growth triggered by the activation of oncogenic signaling cascades, which suggests a change in the regulation of important transcription factors. Amongst others, NFAT transcription factors are assumed to play a central role in the carcinogenesis of pancreatic cancer. Recent research has shown the importance of the transcription factor Sp1 in the transcriptional activity of NFATc2 in pancreatic cancer. However, the role of the interaction between these two binding partners remains unclear. The current study investigated the role of Sp1 proteins in the expression of NFATc2 target genes and identified new target genes and their function in cells. A further objective was the domain of the Sp1 protein that mediates interaction with NFATc2. The involvement of Sp1 proteins in NFATc2 target genes was shown by means of a gene expression profile analysis, and the results were confirmed by quantitative RT-PCR. The functional impact of this interaction was shown in a thymidine incorporation assay. A second objective was the physical interaction between NFATc2 and different Sp1 deletion mutants that was investigated by means of immunoprecipitation. RESULTS: In pancreatic cancer, the proto-oncogene c-Fos, the tumor necrosis factor TNF-alpha, and the adhesion molecule integrin beta-3 are target genes of the interaction between Sp1 and NFATc2. Loss of just one transcription factor inhibits oncogenic complex formation and expression of cell cycle-regulating genes, thus verifiably decreasing the carcinogenic effect. The current study also showed the interaction between the transcription factor NFATc2 and the N-terminal domain of Sp1 in pancreatic cancer cells. Sp1 increases the activity of NFATc2 in the NFAT-responsive promoter. CONCLUSIONS: The regulation of gene promotors during transcription is a rather complex process because of the involvement of many proteins that - as transcription factors or co-factors - regulate promotor activity as required and control cell function. NFATc2 and Sp1 seem to play a key role in the progression of pancreatic cancer.


Subject(s)
Gene Expression Regulation, Neoplastic , NFATC Transcription Factors/metabolism , Pancreatic Neoplasms/genetics , Sp1 Transcription Factor/genetics , Humans , Immunoprecipitation , Pancreatic Neoplasms/etiology , Promoter Regions, Genetic , Protein Binding , Proto-Oncogene Mas , Sp1 Transcription Factor/metabolism
13.
Eur J Med Res ; 24(1): 5, 2019 Jan 28.
Article in English | MEDLINE | ID: mdl-30686270

ABSTRACT

BACKGROUND: Cancer, one of the leading causes of death worldwide, develops when the normal balance between mitosis and apoptosis is disrupted. The subsequently increased proliferation rate or decreased apoptosis rate of cells leads to uncontrolled cellular growth. Thus, the current aim of cancer research is to increase the apoptosis rate in tumor cells-while limiting the concurrent death of healthy cells-and to induce controlled apoptosis in abnormal cells. Staurosporine is a very potent inducer of apoptosis because it inhibits many different kinases. So far, many different kinase pathways of staurosporine-induced apoptosis have been discussed for various tumor entities. AIMS: To identify the effect of staurosporine in pancreatic and colorectal carcinoma cells and its apoptosis-inducing signaling pathway. METHODS: The apoptosis rate in pancreatic and colorectal carcinoma cells was analyzed by annexin V staining after staurosporine administration. Staurosporine stimulation and its effects on the expression of Bcl2, BAX, Bad, caspase-8, and caspase-9 were investigated with immunoblot. RESULTS: Staurosporine significantly increased apoptosis in pancreatic carcinoma cells. Western blot analysis showed activation of caspase-9 in PaTu 8988t and Panc-1 cells with 1 µM staurosporine. In addition, expression of Bcl2 and Bad was decreased in PaTu 8988t cells. In colorectal carcinoma cells SW 480, staurosporine stimulation did not induce apoptosis. CONCLUSION: Modern therapeutic strategies for tumor diseases target the efficient modulation of specific signaling and transcription pathways. In this respect, the therapeutic potential of protein kinase inhibitors has been repeatedly discussed. Our study showed that staurosporine induces apoptosis in pancreatic carcinoma cells via the intrinsic signaling pathway. Thus, staurosporine is a suitable positive control for in vitro apoptosis tests for the pancreatic cancer cell lines PaTu 8988t and Panc-1. Further clinical studies should analyze the impact of this finding on cancer treatment.


Subject(s)
Adenocarcinoma/pathology , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Pancreatic Neoplasms/pathology , Staurosporine/pharmacology , Cell Line, Tumor , Enzyme Inhibitors/pharmacology , Humans , Signal Transduction/drug effects , Pancreatic Neoplasms
14.
Int J Emerg Med ; 11(1): 9, 2018 Feb 17.
Article in English | MEDLINE | ID: mdl-29455310

ABSTRACT

BACKGROUND: Sudden cardiac death is one of the most frequent causes of death in Germany and the third leading cause of death in the industrialized world. Yet, the percentage of people providing first aid in the case of sudden cardiac arrest in Germany is alarmingly low by international comparison. Training Germans or reminding them of the simple but effective steps of resuscitation, so that everybody can save a live in an emergency. METHODS: For the campaign 'Resuscitation Week', physicians and paramedics trained passers-by in cardiovascular resuscitation free of charge. Skills were evaluated before and after the instruction by means of a questionnaire. RESULTS: Three hundred three people aged between 9 and 89 years were trained and evaluated. Forty-nine passers-by had never participated in a resuscitation course, and 46.8% had participated in a course more than 20 years ago. Before the instruction, 41.6% of the passers-by were confident to be capable of resuscitating a person; after the instruction, however, this percentage had risen to 100%! CONCLUSIONS: Saving a life is simple, but one has to know what to do in the case of sudden cardiac arrest. The German population is being gradually trained in resuscitation using campaigns such as 'Resuscitation Week' and 'Kids Save Lives' to break down barriers in the long term. However, lives are not only saved by training but also by refreshing knowledge and skills; thus, a further effective approach may be training all holders of a driving license in cardiopulmonary resuscitation in intervals of 5 years.

15.
Anticancer Res ; 38(2): 745-751, 2018 02.
Article in English | MEDLINE | ID: mdl-29374698

ABSTRACT

BACKGROUND/AIM: The perioperative phase is supposed to be a period with high vulnerability for cancer dissemination. Acetaminophen and metamizole are common analgesics administered during this phase. We investigated the effect of acetaminophen, metamizole and 4-methylaminoantipyrine (MAA) on proliferation and apoptosis of colon carcinoma cell lines (SW 480 and HT 29). MATERIALS AND METHODS: Proliferation was detected by cell proliferation ELISA BrdU, and apoptosis by Annexin V staining. Cytochrome c and caspase 3, 8 and 9 expression levels were detected by western blot. RESULTS: Acetaminophen, metamizole or MAA caused slight changes in proliferation. Acetaminophen, metamizole or the combination increased apoptosis in both cell lines. All agents decreased caspase 3 and 8 expression in SW480. Acetaminophen decreased caspase 9 expression in both cell lines. CONCLUSION: In clinically relevant doses, acetaminophen and/or metamizole induce apoptosis in both colon cancer cell lines. Both mitochondrial and death receptor pathways might be involved in acetaminophen-induced apoptosis.


Subject(s)
Acetaminophen/pharmacology , Analgesics, Non-Narcotic/pharmacology , Apoptosis/drug effects , Colonic Neoplasms/drug therapy , Dipyrone/pharmacology , Aminopyrine/analogs & derivatives , Aminopyrine/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Blotting, Western , Cell Line, Tumor , Cell Proliferation/drug effects , Colonic Neoplasms/pathology , HT29 Cells , Humans
16.
BMC Pharmacol Toxicol ; 18(1): 77, 2017 12 06.
Article in English | MEDLINE | ID: mdl-29208039

ABSTRACT

BACKGROUND: Adenocarcinoma of the pancreas is one of the most aggressive cancer diseases affecting the human body. Recent research has shown the importance of the perioperative phase in disease progression. Particularly during this vulnerable phase, substances such as metamizole and paracetamol are given as general anesthetics and postoperative analgesics. Therefore, the effects of metamizole and paracetamol on tumor progression should be investigated in more detail because the extent to which these substances influence the carcinogenesis of pancreatic carcinoma is still unclear. This study analyzed the influence of metamizole and its active metabolites MAA (4-N-methyl-aminoantipyrine) and paracetamol on the proliferation, apoptosis, and necrosis of the pancreatic cancer cell lines PaTu 8988t and Panc-1 in vitro. METHODS: Cell proliferation was measured by means of the ELISA BrdU assay and the rate of apoptosis by flow cytometry using the Annexin V assay. RESULTS: Metamizole and paracetamol significantly inhibited cell proliferation in pancreatic cancer cells. After the addition of metamizole to PaTu 8988t cells, the rate of apoptosis was reduced after 3 h of incubation but significantly increased after 9 h of incubation. CONCLUSION: The oncogenic potential of pancreatic adenocarcinoma is mainly characterized by its extreme growth rate. Non-opioid analgesics such as metamizole and paracetamol are given as general anesthetics and postoperative analgesics. The combination of metamizole or paracetamol with cytotoxic therapeutic approaches may achieve synergistic effects. Further studies are necessary to identify the underlying mechanisms so that new therapeutic options may be developed for the treatment of this aggressive tumor.


Subject(s)
Acetaminophen/pharmacology , Analgesics, Non-Narcotic/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Antineoplastic Agents/pharmacology , Dipyrone/analogs & derivatives , Dipyrone/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Synergism , Humans , Necrosis/chemically induced , Pancreatic Neoplasms/drug therapy
17.
BMC Mol Biol ; 18(1): 20, 2017 08 03.
Article in English | MEDLINE | ID: mdl-28774282

ABSTRACT

BACKGROUND: Nuclear factors of activated T-cells (NFATs) have been mainly characterized in the context of immune response regulation because, as transcription factors, they have the ability to induce gene transcription. NFAT proteins are found in several types of tumors, for instance, pancreatic carcinoma. The role of NFATs in carcinogenesis is regulating central genes in cell differentiation and cell growth. NFAT proteins are primarily located in cytoplasm and only transported to the cell nucleus after activation. Here, they interact with other transcription factors cooperating with NFAT proteins, thus influencing the selection and regulation of NFAT-controlled genes. To identify and characterize possible interaction partners of the transcription factor NFATc2 in pancreatic carcinoma cells PaTu 8988t. METHODS: NFATc2 expression and the mode of action of Ionomycin in the pancreatic tumor cell lines PaTu 8988t were shown with Western blotting and immunofluorescence tests. Potential partner proteins were verified by means of immunoprecipitation and binding partners, their physical interactions with DNA pull-down assays, siRNA technologies, and GST pull-down assays. Functional evidence was complemented by reporter-promoter analyses. RESULTS: NFATc2 and Sp1 are co-localized in cell nuclei and physically interact at the NFAT target sequence termed NFAT-responsive promotor construct. Sp1 increases the functional activity of its binding partner NFATc2. This interaction is facilitated by Ionomycin in the early stimulation phase (up to 60 min). CONCLUSIONS: Oncological therapy concepts are becoming more and more specific, aiming at the efficient modulation of specific signal and transcription pathways. The oncogenic transcription partner Sp1 is important for the transcriptional and functional activity of NFATc2 in pancreatic carcinoma. The binding partners interact in cells. Further studies are necessary to identify the underlying mechanisms and establish future therapeutic options for treating this aggressive type of tumor.


Subject(s)
NFATC Transcription Factors/metabolism , Pancreatic Neoplasms/metabolism , Sp1 Transcription Factor/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Line, Tumor , Cell Nucleus/metabolism , Gene Expression , Humans , Nigericin/analogs & derivatives , Nigericin/pharmacology , Pancreatic Neoplasms/genetics , Promoter Regions, Genetic , Protein Binding , Protein Transport , Pancreatic Neoplasms
18.
Anticancer Res ; 37(4): 1941-1945, 2017 04.
Article in English | MEDLINE | ID: mdl-28373464

ABSTRACT

BACKGROUND: Evidence is growing that the risk of cancer dissemination may be enhanced during the perioperative period. Whether particular anesthetic techniques influence oncological outcome is still under discussion. For pain management, lidocaine can be administered perioperatively by intravenous, intraperitoneal or epidural infusion. Here we investigated the effect of lidocaine on colon carcinoma cell lines (HT-29 and SW480) in vitro. MATERIALS AND METHODS: ELISA BrdU (Roche) for cell proliferation and FITC Annexin V detection kit (BD Pharming) for apoptosis analysis were applied. Cell-cycle profiles were investigated by flow cytometry. RESULTS: Cell-cycle arrest was induced in both cell lines by 1000 µM lidocaine, while no inhibition of cell proliferation was detected. Apoptosis decreased in SW480 but not in HT-29 cells. CONCLUSION: Lidocaine induces cell-cycle arrest in both colon carcinoma cell lines in vitro. The effective drug concentration can be obtained by local infiltration.


Subject(s)
Anti-Arrhythmia Agents/pharmacology , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Proliferation/drug effects , Colonic Neoplasms/pathology , Lidocaine/pharmacology , Colonic Neoplasms/drug therapy , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , In Vitro Techniques , Tumor Cells, Cultured
19.
BMC Anesthesiol ; 15: 111, 2015 Jul 29.
Article in English | MEDLINE | ID: mdl-26219286

ABSTRACT

BACKGROUND: Adenocarcinoma of the pancreas is one of the most aggressive cancer diseases affecting the human body. The oncogenic potential of this type of cancer is mainly characterized by its extreme growth rate triggered by the activation of signaling cascades. Modern oncological treatment strategies aim at efficiently modulating specific signaling and transcriptional pathways. Recently, anti-tumoral potential has been proven for several substances that are not primarily used in cancer treatment. In some tumor entities, for example, administration of glutamate antagonists inhibits cell proliferation, cell cycle arrest, and finally cell death. To attain endogenic proof of NMDA receptor type expression in the pancreatic cancer cell lines PaTu8988t and Panc-1 and to investigate the impact of ketamine, s-ketamine, and the NMDA receptor antagonist MK 801 on proliferation, apoptosis, and necrosis in pancreatic carcinoma. METHODS: Cell proliferation was measured by means of the ELISA BrdU assay, and the apoptosis rate was analyzed by annexin V staining. Immunoblotting were also used. RESULTS: The NMDA receptor type R2a was expressed in both pancreatic carcinoma cell lines. Furthermore, ketamine, s-ketamine, and MK 801 significantly inhibited proliferation and apoptosis. CONCLUSIONS: In this study, we showed the expression of the NMDA receptor type R2a in pancreatic cancer cells. The NMDA antagonists ketamine, s-ketamine, and MK 801 inhibited cell proliferation and cell death. Further clinical studies are warranted to identify the impact of these agents on the treatment of cancer patients.


Subject(s)
Adenocarcinoma/drug therapy , Dizocilpine Maleate/pharmacology , Ketamine/pharmacology , Pancreatic Neoplasms/drug therapy , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Enzyme-Linked Immunosorbent Assay , Humans , Ketamine/chemistry , Necrosis/pathology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Receptors, N-Methyl-D-Aspartate/genetics , Stereoisomerism
20.
BMC Res Notes ; 8: 117, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25890020

ABSTRACT

BACKGROUND: Natural latex rubber products have been known to cause severe anaphylactic reactions during surgery. Even 25 years after the first description of anaphylactic reactions in the literature, natural latex rubber products are still used in pediatric surgery. CASE PRESENTATION: The following article describes the case of a healthy 4.5-year old Caucasian boy who simultaneously developed severe hypotension, tachycardia and bronchospasm during surgery for congenital strabismus sursoadductorius under uneventful anesthesia. An allergy test conducted afterwards showed natural latex rubber as the trigger for this severe intraoperative anaphylactic reaction. This case was special because of the absence of any previous clinical or anamnestical evidence of natural latex rubber allergy. The fact that the child had been previously exposed to natural latex rubber - because the boy's mother used disposable gloves for her work as a cosmetician at home - was only discovered later. Such contact may have had a slight sensitizing effect that manifested after the initial contact with the conjunctiva through the surgeon's natural latex rubber gloves. CONCLUSION: Natural latex rubber products have caused severe anaphylactic reactions time and again. Diagnosis is impeded by the highly variable clinical symptoms of anaphylaxis, the non-responsivity of patients, anesthesia-induced changes in blood pressure, surgical drapes, and blood loss. Therefore, use of alternative products and implementation of the right course of action in clinical routine seems to be even more important than raising awareness for allergies to natural latex rubber.


Subject(s)
Anaphylaxis/etiology , Latex Hypersensitivity/complications , Surgical Procedures, Operative/adverse effects , Child, Preschool , Humans , Male
SELECTION OF CITATIONS
SEARCH DETAIL
...