Skip to main content
Erschienen in: BMC Cancer 1/2014

Open Access 01.12.2014 | Research article

Inhibitory effects of prostaglandin E2on collagen synthesis and cell proliferation in human stellate cells from pancreatic head adenocarcinoma

verfasst von: Ewa Pomianowska, Dagny Sandnes, Krzysztof Grzyb, Aasa R Schjølberg, Monica Aasrum, Ingun H Tveteraas, Vegard Tjomsland, Thoralf Christoffersen, Ivar P Gladhaug

Erschienen in: BMC Cancer | Ausgabe 1/2014

Abstract

Background

Several studies have described an increased cyclooxygenase-2 (COX-2) expression in pancreatic cancer, but the role of COX-2 in tumour development and progression is not clear. The aim of the present study was to examine expression of COX-2 in cancer cells and stromal cells in pancreatic cancer specimens, and to explore the role of PGE2 in pancreatic stellate cell proliferation and collagen synthesis.

Methods

Immunohistochemistry and immunofluorescence was performed on slides from whole sections of tissue blocks using antibodies against COX-2 and α-smooth muscle actin (αSMA). Pancreatic stellate cells (PSC) were isolated from surgically resected tumour tissue by the outgrowth method. Cells were used between passages 4 and 8. Collagen synthesis was determined by [3H]-proline incorporation, or by enzyme immunoassay measurement of collagen C-peptide. DNA synthesis was measured by incorporation of [3H]-thymidine in DNA. Cyclic AMP (cAMP) was determined by radioimmunoassay. Collagen 1A1 mRNA was determined by RT-qPCR.

Results

Immunohistochemistry staining showed COX-2 in pancreatic carcinoma cells, but not in stromal cells. All tumours showed positive staining for αSMA in the fibrotic stroma. Cultured PSC expressed COX-2, which could be further induced by interleukin-1β (IL-1β), epidermal growth factor (EGF), thrombin, and PGE2, but not by transforming growth factor-β1 (TGFβ). Indirect coculture with the adenocarcinoma cell line BxPC-3, but not HPAFII or Panc-1, induced COX-2 expression in PSC. Treatment of PSC with PGE2 strongly stimulated cAMP accumulation, mediated by EP2 receptors, and also stimulated phosphorylation of extracellular signal-regulated kinase (ERK). Treatment of PSC with PGE2 or forskolin suppressed both TGFβ-stimulated collagen synthesis and PDGF-stimulated DNA synthesis.

Conclusions

The present results show that COX-2 is mainly produced in carcinoma cells and suggest that the cancer cells are the main source of PGE2 in pancreatic tumours. PGE2 exerts a suppressive effect on proliferation and fibrogenesis in pancreatic stellate cells. These effects of PGE2 are mediated by the cAMP pathway and suggest a role of EP2 receptors.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​1471-2407-14-413) contains supplementary material, which is available to authorized users.
Ewa Pomianowska, Dagny Sandnes contributed equally to this work.

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

EP, DS, TC, IPG conceived and planned the study. EP and KG isolated the pancreatic stellate cells. EP and DS did the cell culturing work. EP and ARS did the immunohistochemistry and immunofluorescence work. DS, EP, IHT, MA and VT conducted the experimental work. DS, EP and IPG analysed and discussed the results. DS, EP and IPG drafted the manuscript. All authors read and approved the final manuscript.

Background

Pancreatic adenocarcinoma is one of the most lethal cancers of all solid malignancies with a 5 year survival of less than 5% [13]. A particular feature of primary pancreatic adenocarcinoma is the extensive fibrotic stromal reaction known as tumour desmoplasia surrounding these tumours [46]. There is increasing evidence that stromal cells are of major importance for tumour progression, by interacting in many ways with the malignant cells, such as reciprocal paracrine proliferative stimulation and angiogenesis, contributing to the early invasive growth and metastasis of this tumour [6]. These observations have raised the possibility that targeting the stromal cells to interrupt paracrine stromal signalling mechanisms may represent a new treatment strategy in pancreatic cancer. Animal studies have also indicated that targeting the tumour stroma of pancreatic cancer may improve drug delivery [79].
Multiple lines of evidence suggest that pancreatic stellate cells (PSC) have a major role in the development of pancreatic cancer desmoplasia [46, 10]. These cells, which are normally quiescent cells in the pancreas, are induced during pancreatic injury to undergo transformation into a myofibroblast-like phenotype expressing alpha smooth muscle actin (αSMA). Studies of human and rat PSC in culture have identified a number of growth factors, cytokines, and hormones as regulators of pancreatic stellate cell activation [6]. Activation promotes PSC proliferation, migration, and extracellular matrix (ECM) deposition.
Overexpression of COX-2 has been reported in a number of epithelial cancers, including pancreatic cancer [1116]. Transgenic mouse models have suggested that COX-2 overexpression in pancreatic ductal cells contributes to pancreatic tumour development [17, 18]. Upregulation of COX-2 leads to increased production of prostaglandins, in particular PGE2. PGE2 may affect both cancer cells and different stromal cells through its effects on EP and FP receptors [19, 20]. While EP2 and EP4 receptors are Gs-coupled receptors that stimulate adenylyl cyclase activity, EP3 receptors are Gi-coupled and inhibit adenylyl cyclase activity. EP1 receptors elevate the intracellular Ca2+-levels through mechanisms that may involve both phospholipase C-dependent and independent mechanisms [1921], and FP receptors are Gq-coupled and elevate intracellular Ca2+-levels [19, 20]. In addition, several of these receptors may signal via G protein-independent mechanisms [22].
Fibroblasts may be stimulated by PGE2. Elevation of the intracellular level of cAMP in response to PGE2 or other stimuli in fibroblasts from different tissues has been found to limit their proliferation, migration, and collagen secretion, as well as the differentiation of fibroblasts to myofibroblasts [2325]. These effects appear to be mediated via EP2 and EP4 receptors. It has also been reported that PGE2 may promote fibroblast proliferation through activation of EP1, EP3, or FP signalling [2629]. In hepatic stellate cells, PGE2 has been found to inhibit transforming growth factor β (TGFβ)-mediated induction of collagen mRNA [30], as well as proliferation induced by platelet-derived growth factor (PDGF) or thrombin [31, 32]. However, the role of PGE2 in pancreatic fibrosis is not well known. The aim of the present study was to examine further the effects of PGE2 on pancreatic stellate cell proliferation and collagen synthesis.

Methods

Patients

The study protocol and patient consent documents were approved by the Regional Committee for Medical and Health Research Ethics (REC South East, project number S-05081), and was in compliance with the Helsinki Declaration. Written informed consent was obtained from all study participants. The study included only adults.

Chemicals

Dulbecco’s Modified Eagle’s Medium, Ham’s F12 medium, RPMI 1640 medium, glutamine, and Pen-Strep (10.000 U/ml) were obtained from Lonza (Verviers, Belgium). HEPES, amphotericin, and heat-inactivated fetal bovine serum (FBS) was purchased from Gibco (Grand Island, NY, USA). Epidermal growth factor (EGF), adenosine 3’:5’-cyclic monophosphate (cAMP), 3-isobutyl 1-methylxanthine (IBMX), L-ascorbic acid, and 3-aminopropionitrile fumarate salt were obtained from Sigma-Aldrich (St.Louis, MO, USA). Human platelet derived growth factor (PDGF), recombinant human transforming growth factor-β (TGF-β), and recombinant human interleukin-1β (IL-1β) were obtained from R&D Systems Europe, Ltd (Abingdon, England). Recombinant interleukin-1 receptor antagonist (Anakinra®) was a gift from Swedish Orphan Biovitrum AS, [6-3H] thymidine (20–30 Ci/mmol), [2,8-3H] adenosine 3’,5’-cyclic phosphate ammonium salt (33.0 Ci/mmol), and L-[2,3-3H] proline (55.0 Ci/mmol) were purchased from PerkinElmer (Boston, MA, USA). L161982 (N-[[4’-[[3-butyl-1,5-dihydro-5-oxo-1-[2-(trifluoromethyl)phenyl]-4 H-1,2,4-triazol-4-yl]methyl][1,1'-biphenyl]-2-yl]sulfonyl]-3-methyl-2-thiophenecarboxamide, AH6809 (6-isopropoxy-9-oxoxanthene-2carboxylic acid), and prostaglandin E2 (PGE2) were obtained from Cayman Chemical (Ann Arbor, MI, USA). Procollagen Type I C-peptide enzyme immunoassay kit was purchased from Takara Bio Inc., Japan. All other chemicals were of analytical quality. Antibodies against phosphorylated AktSer473, total Akt, dually phosphorylated ERKThr202/Tyr204, and GAPDH were obtained from Cell Signaling Technology (Boston, MA, USA). Antibodies against COX-2 were obtained from Cayman Chemical (Ann Arbor, MI, USA) or from Thermo Fischer Scientific Inc (Fremont, CA, USA). Anti-ERK antibody was from Upstate/Millipore (Billerica, MA, USA). Antibodies against TGF-β receptor II and PDGF receptor β were purchased from Cell Signaling Technology (Boston, MA, USA). Antibody against EP2 receptor was obtained from Cayman Chemical (Ann Arbor, MI, USA). Secondary antibodies were purchased from Bio-Rad Laboratories (Hercules, CA, USA). Antibodies against vimentin and cytokeratins 7 and 19 were provided by DAKO (Glostrup, Denmark).

Isolation and culture of human pancreatic stellate cells

Human pancreatic stellate cells (PSC) were isolated by the outgrowth method developed by Bachem et al. [33]. Pancreatic tissue blocks (100–150 mg) were obtained during pancreatic surgery from patients with resectable pancreatic head adenocarcinoma. Altogether, stellate cell cultures were established from a total of 20 different patients. Briefly, the tissue blocks were cut using a razor blade (0.5–1 mm3) and seeded in 10 cm2 uncoated culture wells (6 per plate; 3–5 pieces per well) in a 1:1 (vol/vol) mixture of Dulbecco’s modified Eagle medium (DMEM) with Ham’s F12 medium, supplemented with l-glutamine (2 mmol/L), 100 U/ml Pen-Strep, 2.5 μg/ml amphotericin, and 10% FBS. Tissue blocks were cultured at 37°C in a 5% CO2/air humidified atmosphere. Twenty-four hours after seeding, the small tissue blocks were transferred to new culture plates. Culture medium was changed every third day. The PSCs grew out from the tissue blocks 7 to 10 days later. The small tissue blocks were removed after 2–3 weeks. After reaching confluence, monolayers were trypsinized and passaged 1:3. The purity of the cells was assessed by morphology (most cells were stellate-like, with long cytoplasmatic extensions; some were also spindle shaped) and cytofilament staining of αSMA and vimentin. None of the cells were positive for cytokeratins 7 or 19 (data not shown). All experiments were performed using cell populations between passage 4 and 8.

Pancreatic adenocarcinoma cell lines

BxPC-3, HPAFII, and Panc-1 pancreatic adenocarcinoma cell lines were purchased from ATCC (Manassas, VA, USA). BxPC-3 cells were cultured in RPMI medium containing 4.5 g/l glucose, HPAFII cells were cultured in Dulbecco’s modified Eagle’s medium containing 1 g/l glucose, and Panc-1 cells were cultured in Dulbecco’s modified Eagle’s medium containing 4.5 g/l glucose. The media were supplemented with glutamine (2 mM, or 4 mM in the case of Panc-1), 100 U/ml Pen-Strep, and 10% fetal bovine serum (FBS). Cells were plated in Transwell® inserts (Corning Incorporated, Corning, NY, USA) at a density of 100.000/cm2 in serum-containing medium and cultured overnight. The next day, medium was replaced with fresh, serum-free medium, and cells were cultured overnight. The following day, the Transwells were transferred to 12 well Costar plates containing stellate cells in the lower compartment, and cells were cocultured for 48 hours.

Coculture of pancreatic stellate cells with pancreatic adenocarcinoma cell lines

Pancreatic stellate cells were plated at a density of 10.000 cells/cm2 in 12 well Costar plates with serum-containing medium and cultured overnight. The following day, medium was replaced with fresh, serum-free medium, and cells were cultured overnight. The next day, the serum-free medium was changed, and Transwells containing pancreatic adenocarcinoma cell lines were placed on top. Cells were cocultured for 48 hours before harvesting for immunoblotting.

Measurement of DNA synthesis

Pancreatic stellate cells were seeded into 12 well Costar plates at a density of 10.000 cells/cm2 in serum-containing medium and cultured overnight. On the following day, medium was replaced with fresh, serum-free medium. The next day, the serum-free medium was changed 30 minutes before addition of agonists. The cells were harvested after pulsing for 6 hours with [3H]thymidine (18–24 hours after addition of agonists), and DNA synthesis was measured as the amount of radioactivity incorporated into DNA as previously described [34]. Briefly, medium was removed, and cells were washed twice with 0.9% NaCl. The cellular material was dissolved with 1 ml 0.5 N NaOH for 3 hours at 37°C, collected, mixed with 1 ml H2O, and precipitated with 0.5 ml 50% trichloroacetic acid (TCA). The acid-precipitable material was transferred to glass fiber filters (GF/C Whatman, GE Healthcare, UK) and washed twice with 5.0 ml 5% TCA, followed by liquid scintillation counting of the filters in a Packard Tri-Carb 1900 TR liquid scintillation counter.

Measurement of collagen synthesis

Collagen synthesis was assessed by quantification of [3H] proline incorporation into acetic acid-soluble proteins as described by Jaster et al. [35]. Pancreatic stellate cells were plated in 24 well Costar plates at a density of 10.000 cells/cm2 in serum-containing medium and cultured overnight. The following day, medium was replaced with fresh, serum-free medium. The next day, serum-free medium was changed, and agonists and/or antagonist were added. After 24 hours, the medium was replaced with fresh serum-free medium containing 100 μg/ml ascorbic acid, 100 μg/ml 3-aminopropionitrile, and 2 μCi/ml [3H] proline, and fresh agonists were added. The reaction was stopped 24 hours later, by addition of 50 μl/ml 10 N acetic acid. After an overnight incubation at 4°C, culture supernatants were transferred to microcentrifuge tubes, mixed with 100 μl/ml FBS, 5 μg/ml rat tail collagen and 250 μl/ml 25% NaCl dissolved in 0.5 N acetic acid, and incubated at 4°C for 30 minutes. Protein precipitates collected by centrifugation (30 min, 10,000 g) were washed twice with 5% NaCl, followed by dissolution of the pellet in 0.5 N acetic acid. [3H] proline incorporation was determined by liquid scintillation counting in a Packard Tri-Carb 1900 TR scintillation counter. In initial experiments, collagen synthesis was determined in parallell samples by measurement of procollagen type I C-peptide by an enzyme immunoassay. The two methods yielded similar results (data not shown).

RNA extraction and real-time quantitative RT-qPCR

Pancreatic stellate cells were plated at a density of 10.000/cm2 in 20 cm2 wells in serum-containing medium and cultured overnight. On the following day, medium was replaced with serum-free medium. The next day the medium was changed 30 minutes before agonists and/or antagonist were added, as indicated. The cells were stimulated for 24 hours. Total RNA was prepared from the samples using RNA Easy Mini kit (Qiagen Inc, Valencia, CA, USA) and cDNA was synthesized with SuperScript III Reverse Transcriptase First-Strand cDNA Synthesis kit according to the manufacturer’s protocol (InVitrogen, Carlsbad, CA, USA). Quantitative PCR was performed with Platinum SYBR Green Master Mix (Life Technologies, Oslo, Norway) on 7900 Real-Time PCR system with 7900 System SDS 2.3 Software (Applied Biosystems) according to the manufacturer’s protocol. Specific primers for collagen 1A1 were: forward, 5’-TGACGTGATCTGTGACGAGAC-3’ and reverse, 5’- GGTTTCTTGGTCGGTGGGT −3’ (Life Technologies Oslo, Norway). Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) was utilized as housekeeping gene, and specific primers were: forward, 5’-CCACCATGGAGAAGGCTGGGGCTC-3’ and reverse 5’-AGTGATGGCATGGACTGTGGTCAT3’ (Life Technologies, Oslo, Norway). The primers were designed using Primer-BLAST [36]. All reactions were performed in triplicates including non-template controls. The results were analyzed using the ΔΔCt method [37]. Results for collagen 1A1 were normalized to GAPDH, and controls were assigned a value of 100%.

Cyclic AMP measurement

Pancreatic stellate cells were plated in 12 well Costar wells at a density of 10.000 cells/cm2 in serum-containing medium. On the following day, medium was replaced with fresh, serum-free medium. The next day, medium was replaced with Krebs-Ringer-Hepes buffer, pH 7.4, containing 10 mM glucose. After preincubation for 30 minutes, cells were stimulated with PGE2 or forskolin as indicated in the figure legends. The reaction was stopped by removing the buffer and adding 5% TCA. cAMP in the neutralized TCA extract was determined by radioimmunoassay as previously described [38].

Immunoblotting

Aliquots with approximately 7000 cells (total cell lysate prepared in Laemmli buffer) were electrophoresed on 12% (w/v) polyacrylamide gels (acrylamide: N’N’-bis-methylene acrylamide 30:1). This was followed by protein electrotransfer to nitrocellulose membranes and immunoblotting with antibodies against phospho-Akt, total Akt, phospho ERK1/2, total ERK, COX-2, and GAPDH, respectively. Immunoreactive bands were visualized with enhanced chemiluminescence using LumiGLO (KPL Protein research Products, Gaithersburg, MD, USA).

Immunohistochemistry

Formalin-fixed, paraffin-embedded tissues from pancreatectomy specimens were sectioned (3 μm), and dried at 60°C. Further processing was carried out in the Ventana BenchMark Ultra machine (Ventana Medical Systems Inc. (Tucson Arizona USA) according to the manufacturer’s recommendations. Slides were incubated with monoclonal anti-COX-2 antibodies (Thermo Fischer Scientific rabbit), Universal Alkaline Phosphatase Red Detection Kit (Ultra View 760–501) and a-SMA (Dako M.0851, DAB (Ultra View 760–500). Finally, slides were counterstained with haematoxylin, fixed, mounted and analyzed using an inverted light microscope (Olympus, Center Valley, PA, USA).

Immunofluorescence staining

Immunofluorescence staining was performed to examine COX-2 expression in the tumour slides. Formalin-fixed, paraffin-embedded tissues from pancreatectomy specimens were sectioned (3 μm), dried at 60°C and hydrated. Slides were incubated with monoclonal anti-COX-2 antibody (Thermo sp21 rabbit) and anti-αSMA (DAKO 1A4 mouse) for 30 min at room temperature in Ventana diluents. After washing with PBS, slides were incubated with secondary antibody conjugates (Alexa 555 anti-rabbit and Alexa 488 anti - mouse) in the dark for 1 hour in Dako diluents. After three washes with PBS, slides were mounted in VECTASHIELD containing DAPI (Vector Laboratories Inc., Burlingame, CA, USA). Fixed cells were observed under a fluorescence microscope.
Immunofluorescence staining was also performed on the cultured pancreatic stellate cells. Cells were first seeded into a Lab-Tek®II Chamber Slide™ System (Nunc International, Naperville, IL, USA) and were cultured for 24 hours before they were fixed in 4% paraformaldehyde at room temperature for 15 minutes. Cells were then washed three times and incubated with 5% BSA for 30 minutes to block non-specific binding. Slides were further processed as describe for tumour tissue.

Statistical analyses

Results are presented as mean ± standard error of the mean (S.E.M). DNA and collagen synthesis data were analyzed by one-way ANOVA, and post test using Bonferroni correction to compare groups, using GraphPad Prism (version 5.01, GraphPad Software, San Diego, CA, USA).

Results

COX-2 expression in pancreatic cancer cells

COX-2 expression in tumour tissue from pancreatic cancer was examined by double staining immunohistochemistry for COX-2 and αSMA. The cancer cells generally exhibited strong COX-2 staining (Figure 1A). We also found strong αSMA staining in the tumour stroma, indicating the presence of activated PSC. However, we could not detect double staining with COX-2 and αSMA in the stroma (Figure 1A). This was examined further by immunofluorescence, which failed to detect any COX-2 staining in the stroma (Figure 1B).

COX-2 expression in cultured human PSC

During culture of PSC, immunofluorescence staining in different passages revealed perinuclear staining with the COX-2 antibody in cells that were αSMA positive (Figure 1C). The expression of COX-2, αSMA, EP2 receptors, TGFβ receptors and PDGF receptors was found to be stable as a function of cell passage number as assessed by Western blotting (Figure 1D, 1E). Treatment of PSC with EGF and PGE2 increased the expression of COX-2, whereas treatment with TGFβ did not. This expression pattern was observed in cells of both low and high passage numbers (Figure 1F). PDGF had no significant effect. Thrombin also induced COX-2 expression (data not shown). Interleukin-1β (IL-1β) was found to be a potent inducer of COX-2 expression, with maximal induction obtained at 0.1 ng/ml (Figure 1G). Coculture of pancreatic adenocarcinoma cell lines with pancreatic stellate cells was previously found to upregulate COX-2 mRNA in both stellate cells and adenocarcinoma cell lines [39]. We examined the effect of coculture of stellate cells with the adenocarcinoma cell lines BxPC-3, Panc-1, and HPAFII. Of these, only BxPC-3 cells induced COX-2 protein in the stellate cells (Figure 2A). Furthermore, this effect was abolished when the stellate cells were pretreated for one hour with an IL-1 receptor antagonist (Figure 2B).

PGE2stimulates EP2-mediated cAMP accumulation in PSC

PGE2 may affect cells through both EP and FP receptors. Because fibroblasts from different tissues have been found to express mainly EP2 and EP4 receptors [24, 40, 41], we examined the effect of PGE2 on cAMP accumulation in the stellate cells. When stellate cells were stimulated for 5 min with 100 μM PGE2 or 50 μM forskolin, a direct activator of adenylyl cyclase [42], in the presence of the phosphodiesterase inhibitor isobutylmethylxanthine (IBMX), cAMP levels were elevated 16.8 ± 5.8-fold (mean ± S.E.M.) above basal levels with PGE2, and 33.0 ± 11.7-fold above basal with forskolin (n = 7). PGE2 induced a strong, dose-dependent accumulation of cAMP, both in the absence and presence of IBMX (Figure 3A). When cells were preincubated with the EP4 receptor antagonist L-161982 [43], no significant inhibition of PGE2-stimulated cAMP accumulation was observed. In contrast, AH6809, which is commonly used as en EP2 receptors antagonist [19], almost abolished the cAMP response, suggesting that cAMP accumulation in these cells is mediated mainly by EP2 receptors (Figure 3B).

PGE2inhibits DNA synthesis in PSC

We next examined how PGE2 affected stellate cells proliferation. In agreement with previous studies [6, 44, 45], PDGF strongly stimulated DNA synthesis (Figure 4A). Epidermal growth factor (EGF) also stimulated DNA synthesis, although to a lesser extent than PDGF, whereas TGFβ had non-significant effect. (Figure 4A). In agreement with these findings, PDGF and EGF, but not TGFβ, significantly stimulated phosphorylation of both ERK and Akt in the stellate cells (Figure 4C). Interestingly, PGE2, the FP selective receptor agonist fluprostenol, and thrombin also stimulated ERK phosphorylation in the stellate cells (Figure 4D), while they did not induce Akt phosphorylation (data not shown). The effect of PGE2 and fluprostenol on ERK phosphorylation did not seem to involve cAMP, since forskolin did not stimulate ERK phosphorylation.
In human hepatic stellate cells several growth-stimulatory agents, including PDGF and thrombin, stimulate an acute PGE2 production, as well as a delayed induction of COX-2, and pretreatment with a COX inhibitor enhances their growth stimulatory effect [31]. We examined the effect of pretreatment with indomethacin on PDGF-stimulated DNA synthesis in the pancreatic stellate cells. These experiments showed that pretreatment with indomethacin did not affect PDGF-stimulated DNA synthesis in the pancreatic stellate cells (Figure 4B).
Treating the stellate cells with PGE2 did not significantly affect the basal DNA synthesis, but attenuated PDGF-stimulated DNA synthesis. PGE2 exerted an inhibitory effect, which was significant at a concentration of 1 μM (Figure 5A). This effect was mimicked by forskolin (Figure 5B). cAMP levels were elevated above the basal level for at least 60 minutes following stimulation with PGE2 (Figure 5C) or forskolin (Figure 5D). Neither fluprostenol nor thrombin had any effect on DNA synthesis, alone or in combination with PDGF (data not shown).

PGE2inhibits collagen synthesis in PSC

In agreement with previous findings [6, 45, 46] treating the stellate cells with TGFβ enhanced collagen synthesis, whereas PDGF or EGF did not significantly affect collagen synthesis (Figure 6A). In agreement with the lack of induction of COX-2 by TGFβ in the stellate cells, pretreatment with indomethacin did not affect TGFβ-induced collagen synthesis (Figure 6B). Both PGE2 and forskolin inhibited TGFβ-stimulated collagen synthesis, suggesting that this was a cAMP-mediated effect (Figure 7A, B, C). While we were preparing this manuscript, Charo et al. reported that PGE2 stimulated the mRNA expression of collagen 1A1 in an immortalized human pancreatic stellate cell line [40]. To examine this further, RNA was extracted from cultured pancreatic stellate cells and assessed for elevated gene expression of collagen 1A1 by real time RT-qPCR. While TGFβ increased gene expression, PGE2 alone showed a slight inhibitory effect, and significantly attenuated TGFβ-stimulated increase in gene expression of collagen 1A1 at a concentration of 1 μM (Figures 7D, 7E). Since PGE2 might elevate cAMP levels through EP2 or EP4 receptors, we examined the effect of EP2 and EP4 receptor antagonists on collagen synthesis. We found that the EP4 receptor antagonist L161982 did not abrogate the effect of PGE2 on TGFβ-induced collagen synthesis (Figure 7F) whereas results with the EP2 receptor antagonist AH 6809 were not conclusive (data not shown).

Discussion

In the present study we have demonstrated that PGE2 inhibits both collagen and DNA synthesis in human pancreatic stellate cells from pancreatic adenocarcinoma. These effects are mediated by increased cAMP production. It is well known that in fibroblasts from lung and other tissues, PGE2 inhibits proliferation by activating Gs-coupled EP2 and/or EP4 receptors [2325, 41, 47, 48]. Since EP4 inhibition affected neither the cAMP response nor the effect on collagen synthesis by PGE2 in our study, it is most likely that EP2 receptors mediate these inhibitory effects of PGE2 on cAMP and collagen synthesis. However, due to inconclusive results with the EP2 receptor antagonist, these mechanisms require further experimental confirmation.
In human hepatic stellate cells, thrombin and PDGF stimulate the release of PGE2, which exerts an inhibitory effect on DNA synthesis induced by PDGF and thrombin [31]. However, PGE2 appeared to mediate the mitogenic effect of EGF in BALB/c 3 T3 cells, and of PDGF in Swiss 3 T3 cells [49, 50]. In our study, EGF, PGE2 and thrombin, but not PDGF, consistently induced COX-2 protein expression in the stellate cells.
Pretreatment of the cells with indomethacin did not affect PDGF-stimulated DNA synthesis, suggesting that COX-2 induction and PGE2 production neither mediated nor modulated PDGF-stimulated DNA synthesis. While we did not measure production of PGE2, studies in various cells, including pancreatic stellate cells [40], indicate that levels are in the nanomolar range. We did not detect an effect of PGE2 on DNA synthesis in the stellate cells when it was added alone, however, PGE2, as well as forskolin, inhibited PDGF-stimulated DNA synthesis, suggesting that this effect was mediated by cAMP. This is in contrast to findings in rat pancreatic stellate cells, where treatment of the cells with conditioned medium from the Panc-1 adenocarcinoma cell line induced COX-2 expression and stimulated DNA synthesis [51]. Furthermore, inhibition of COX-2 activity with the COX-2 specific inhibitor NS-398 attenuated DNA synthesis in the rat stellate cells, albeit at high concentrations of the inhibitor, which may lead to nonspecific effects. Thus, at high concentrations of NS-398, inhibition of DNA synthesis has been reported in COX-2 expressing cell lines as well as in cell lines without COX-2 expression [5254].
Pancreatic stellate cells are believed to be essential in the development of fibrosis associated with chronic pancreatitis and pancreatic cancer [46, 10]. However, the role of PGE2 in pancreatic fibrosis is unknown. TGFβ has been found to induce COX-2, which attenuates the profibrotic effect of TGFβ, in lung fibroblasts and hepatic stellate cells [30, 48], and exogenous addition of PGE2 inhibited TGFβ-induced collagen expression in hepatic stellate cells [30]. However, we found no induction of COX-2 by TGFβ in the pancreatic stellate cells, and preincubation of the cells with indomethacin did not affect TGFβ-stimulated collagen synthesis. In the lung, PGE2 has been found to inhibit collagen synthesis by activating EP2 receptors and stimulating cAMP accumulation. In patients with idiopathic pulmonary fibrosis, lung fibroblasts display a diminished capacity to express COX-2 and to synthesize PGE2. This results in decreased levels of PGE2 and excessive fibroblast activation with massive fibrosis [41, 47, 48]. Our findings in the pancreatic stellate cells are consistent with these studies. Treatment with PGE2, as well as forskolin, suppressed the increase in collagen synthesis stimulated by TGFβ, suggesting that this effect was mediated by cAMP. Our observations are thus in disagreement with findings in an immortalized human pancreatic stellate cell line, where 100 nM PGE2 was found to induce mRNA of collagen 1A1 as well as other structural genes involved in extracellular matrix formation [40]. We therefore examined the effect of PGE2 in our stellate cells, and found no evidence of collagen 1A1 mRNA induction. Rather, PGE2 (1 μM) attenuated the TGFβ-induced expression of collagen 1A1, which is in agreement with our findings of an inhibitory effect of PGE2 on collagen synthesis. The possibility that immortalized pancreatic stellate cells behave differently from primary cell lines needs consideration. Interestingly, the effects of PGE2 on immortalized stellate cells were mediated by activation of EP4 receptors [40]. We have found no evidence of EP4 receptor involvement in the cAMP response in our primary stellate cells, however, we can presently not exclude the possibility that EP4 receptors signal via G protein-independent pathways [22].
We observed that PGE2 stimulated ERK phosphorylation in the stellate cells. This effect was mimicked by thrombin and the FP selective agonist fluprostenol, but not by forskolin, suggesting that it was a cAMP-independent effect. Thus, the stellate cells may express other EP receptors or FP receptors that mediate this effect. PGE2 has been reported to stimulate fibroblast proliferation through activation of EP1, EP3, or FP signalling in lung and cardiac fibroblasts, as well as in NIH 3 T3 cells [2629]. If other prostaglandin receptors could stimulate proliferation of pancreatic stellate cells, the inhibitory effect of cAMP induced by EP2 receptors, appear to suppress these effects. It is notable that the inhibitory effect of PGE2 on collagen and DNA synthesis was only significant at a concentration of 1 μM, whereas in lung fibroblasts effects have been observed at concentrations as low as 10 nM [41]. In a comparative study of fibroblasts from lung and gingiva, it was observed that stimulation with PGE2 resulted in less cAMP accumulation in gingival fibroblasts than in lung fibroblasts [55]. Furthermore, EP3 receptor activation induced phosphorylation of c-Jun NH2-terminal kinase (JNK), which also mediated TGFβ-stimulated fibrosis. Thus, simultaneous EP3 receptor activation might reduce EP2-stimulated cAMP accumulation and blunt the inhibitory effect on DNA and collagen synthesis. Further studies, using subtype-specific agonists, or knockdown of prostaglandin receptors, are required to explore the role of other prostaglandin receptors on proliferation and fibrosis in the stellate cells.
Several previous studies have demonstrated that COX-2 is overexpressed in most human pancreatic cancers [1216, 5660]. However, only a few publications have addressed COX-2 expression in pancreatic stellate cells and they reported no detectable COX-2 expression in the stroma [16, 60]. In our study, immunohistochemical analysis carried out with a specific monoclonal antibody revealed no detectable COX-2 expression in the stroma – neither in the normal pancreas nor in the pancreatic cancer. In contrast Charo et al. [40] reported COX-2 expression in the stroma. One reason for the discrepancy in the results could be the use of different antibodies. For immunohistochemical staining in the study presented by Charo [40] the polyclonal rabbit antihuman COX-2 antibody was used. It is known that polyclonal antibodies are more sensitive, but do not show as high specificity, as monoclonal antibodies [61]. To confirm the expression of COX-2 in pancreatic stroma, Charo at al [40] performed RT-PCR on isolated stellate cells. However, it is likely that the isolation process itself could cause activation of the stellate cells and increase the COX-2 expression [62].
Expression of COX-2 in cultured pancreatic stellate cells is well documented [40, 51, 63] and our results support these findings. In the immunofluorescence double staining of the cultured pancreatic stellate cells, only cells with positive expression for αSMA were additionally positive for COX-2. The COX-2 staining was perinuclear and was constant in different passages (data not shown). COX-2 expression could be further induced by stimulating the stellate cells with IL-1β, EGF, thrombin, and PGE2. Also, indirect coculture with the BxPC-3 cell line, but not HPAFII or Panc-1 cells, induced COX-2 expression. Pretreatment of the stellate cells with IL-1 receptor antagonist blocked the induction of COX-2 induced by BxPC-3 cells, which is consistent with the fact that the BxPC-3 cell line is known to produce IL-1α [64]. Interestingly, conditioned medium from Panc-1 cells induced COX-2 in rat pancreatic stellate cells, however, how this was mediated was not examined [51].

Conclusions

The present results show that COX-2 is mainly expressed in carcinoma cells, and suggest that the cancer cells are the main source of PGE2 in pancreatic tumours. In the pancreatic stellate cells, PGE2 exerts both antiproliferative and antifibrotic effects. These effects of PGE2 are mediated by the cAMP pathway and suggests a role of EP2 receptors. Inhibition of COX-2 may inadvertently accelerate fibrosis progression in pancreatic cancer.

Acknowledgements

This study was supported by the Norwegian Cancer Society. We thank Ole Petter F. Clausen for help with the immunohistochemistry work, Eva Østby Magnussen for help with the collagen synthesis experiments, and John Ødegård and Magne Bryne for help with the initial immunocytochemistry work.
This article is published under license to BioMed Central Ltd. This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://​creativecommons.​org/​licenses/​by/​2.​0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly credited.

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

EP, DS, TC, IPG conceived and planned the study. EP and KG isolated the pancreatic stellate cells. EP and DS did the cell culturing work. EP and ARS did the immunohistochemistry and immunofluorescence work. DS, EP, IHT, MA and VT conducted the experimental work. DS, EP and IPG analysed and discussed the results. DS, EP and IPG drafted the manuscript. All authors read and approved the final manuscript.
Literatur
1.
2.
Zurück zum Zitat Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D: Global cancer statistics. CA Cancer J Clin. 2011, 61 (2): 69-90.CrossRefPubMed Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D: Global cancer statistics. CA Cancer J Clin. 2011, 61 (2): 69-90.CrossRefPubMed
4.
Zurück zum Zitat Chu GC, Kimmelman AC, Hezel AF, DePinho RA: Stromal biology of pancreatic cancer. J Cell Biochem. 2007, 101 (4): 887-907.CrossRefPubMed Chu GC, Kimmelman AC, Hezel AF, DePinho RA: Stromal biology of pancreatic cancer. J Cell Biochem. 2007, 101 (4): 887-907.CrossRefPubMed
5.
Zurück zum Zitat Apte MV, Park S, Phillips PA, Santucci N, Goldstein D, Kumar RK, Ramm GA, Buchler M, Friess H, McCarroll JA, Keogh G, Merrett N, Pirola R, Wilson JS: Desmoplastic reaction in pancreatic cancer: role of pancreatic stellate cells. Pancreas. 2004, 29 (3): 179-187.CrossRefPubMed Apte MV, Park S, Phillips PA, Santucci N, Goldstein D, Kumar RK, Ramm GA, Buchler M, Friess H, McCarroll JA, Keogh G, Merrett N, Pirola R, Wilson JS: Desmoplastic reaction in pancreatic cancer: role of pancreatic stellate cells. Pancreas. 2004, 29 (3): 179-187.CrossRefPubMed
7.
Zurück zum Zitat Olive KP, Jacobetz MA, Davidson CJ, Gopinathan A, McIntyre D, Honess D, Madhu B, Goldgraben MA, Caldwell ME, Allard D, Frese KK, Denicola G, Feig C, Combs C, Winter SP, Ireland-Zecchini H, Reichelt S, Howat WJ, Chang A, Dhara M, Wang L, Ruckert F, Grutzmann R, Pilarsky C, Izeradjene K, Hingorani SR, Huang P, Davies SE, Plunkett W, Egorin M, et al: Inhibition of hedgehog signaling enhances delivery of chemotherapy in a mouse model of pancreatic cancer. Science. 2009, 324 (5933): 1457-1461.CrossRefPubMedPubMedCentral Olive KP, Jacobetz MA, Davidson CJ, Gopinathan A, McIntyre D, Honess D, Madhu B, Goldgraben MA, Caldwell ME, Allard D, Frese KK, Denicola G, Feig C, Combs C, Winter SP, Ireland-Zecchini H, Reichelt S, Howat WJ, Chang A, Dhara M, Wang L, Ruckert F, Grutzmann R, Pilarsky C, Izeradjene K, Hingorani SR, Huang P, Davies SE, Plunkett W, Egorin M, et al: Inhibition of hedgehog signaling enhances delivery of chemotherapy in a mouse model of pancreatic cancer. Science. 2009, 324 (5933): 1457-1461.CrossRefPubMedPubMedCentral
8.
Zurück zum Zitat Beatty GL, Chiorean EG, Fishman MP, Saboury B, Teitelbaum UR, Sun W, Huhn RD, Song W, Li D, Sharp LL, Torigian DA, O’Dwyer PJ, Vonderheide RH: CD40 agonists alter tumor stroma and show efficacy against pancreatic carcinoma in mice and humans. Science. 2011, 331 (6024): 1612-1616.CrossRefPubMedPubMedCentral Beatty GL, Chiorean EG, Fishman MP, Saboury B, Teitelbaum UR, Sun W, Huhn RD, Song W, Li D, Sharp LL, Torigian DA, O’Dwyer PJ, Vonderheide RH: CD40 agonists alter tumor stroma and show efficacy against pancreatic carcinoma in mice and humans. Science. 2011, 331 (6024): 1612-1616.CrossRefPubMedPubMedCentral
9.
Zurück zum Zitat Jacobetz MA, Chan DS, Neesse A, Bapiro TE, Cook N, Frese KK, Feig C, Nakagawa T, Caldwell ME, Zecchini HI, Lolkema MP, Jiang P, Kultti A, Thompson CB, Maneval DC, Jodrell DI, Frost GI, Shepard HM, Skepper JN, Tuveson DA: Hyaluronan impairs vascular function and drug delivery in a mouse model of pancreatic cancer. Gut. 2013, 62 (1): 112-120.CrossRefPubMed Jacobetz MA, Chan DS, Neesse A, Bapiro TE, Cook N, Frese KK, Feig C, Nakagawa T, Caldwell ME, Zecchini HI, Lolkema MP, Jiang P, Kultti A, Thompson CB, Maneval DC, Jodrell DI, Frost GI, Shepard HM, Skepper JN, Tuveson DA: Hyaluronan impairs vascular function and drug delivery in a mouse model of pancreatic cancer. Gut. 2013, 62 (1): 112-120.CrossRefPubMed
10.
Zurück zum Zitat Vonlaufen A, Joshi S, Qu C, Phillips PA, Xu Z, Parker NR, Toi CS, Pirola RC, Wilson JS, Goldstein D, Apte MV: Pancreatic stellate cells: partners in crime with pancreatic cancer cells. Cancer Res. 2008, 68 (7): 2085-2093.CrossRefPubMed Vonlaufen A, Joshi S, Qu C, Phillips PA, Xu Z, Parker NR, Toi CS, Pirola RC, Wilson JS, Goldstein D, Apte MV: Pancreatic stellate cells: partners in crime with pancreatic cancer cells. Cancer Res. 2008, 68 (7): 2085-2093.CrossRefPubMed
12.
Zurück zum Zitat Okami J, Yamamoto H, Fujiwara Y, Tsujie M, Kondo M, Noura S, Oshima S, Nagano H, Dono K, Umeshita K, Ishikawa O, Sakon M, Matsuura N, Nakamori S, Monden M: Overexpression of cyclooxygenase-2 in carcinoma of the pancreas. Clin Cancer Res. 1999, 5 (8): 2018-2024.PubMed Okami J, Yamamoto H, Fujiwara Y, Tsujie M, Kondo M, Noura S, Oshima S, Nagano H, Dono K, Umeshita K, Ishikawa O, Sakon M, Matsuura N, Nakamori S, Monden M: Overexpression of cyclooxygenase-2 in carcinoma of the pancreas. Clin Cancer Res. 1999, 5 (8): 2018-2024.PubMed
13.
Zurück zum Zitat Tucker ON, Dannenberg AJ, Yang EK, Zhang F, Teng L, Daly JM, Soslow RA, Masferrer JL, Woerner BM, Koki AT, Fahey TJ: Cyclooxygenase-2 expression is Up-regulated in human pancreatic cancer. Cancer Res. 1999, 59 (5): 987-990.PubMed Tucker ON, Dannenberg AJ, Yang EK, Zhang F, Teng L, Daly JM, Soslow RA, Masferrer JL, Woerner BM, Koki AT, Fahey TJ: Cyclooxygenase-2 expression is Up-regulated in human pancreatic cancer. Cancer Res. 1999, 59 (5): 987-990.PubMed
14.
Zurück zum Zitat Yip-Schneider MT, Barnard DS, Billings SD, Cheng L, Heilman DK, Lin A, Marshall SJ, Crowell PL, Marshall MS, Sweeney CJ: Cyclooxygenase-2 expression in human pancreatic adenocarcinomas. Carcinogenesis. 2000, 21 (2): 139-146.CrossRefPubMed Yip-Schneider MT, Barnard DS, Billings SD, Cheng L, Heilman DK, Lin A, Marshall SJ, Crowell PL, Marshall MS, Sweeney CJ: Cyclooxygenase-2 expression in human pancreatic adenocarcinomas. Carcinogenesis. 2000, 21 (2): 139-146.CrossRefPubMed
15.
Zurück zum Zitat Juuti A, Louhimo J, Nordling S, Ristimaki A, Haglund C: Cyclooxygenase-2 expression correlates with poor prognosis in pancreatic cancer. J Clin Pathol. 2006, 59 (4): 382-386.CrossRefPubMedPubMedCentral Juuti A, Louhimo J, Nordling S, Ristimaki A, Haglund C: Cyclooxygenase-2 expression correlates with poor prognosis in pancreatic cancer. J Clin Pathol. 2006, 59 (4): 382-386.CrossRefPubMedPubMedCentral
16.
Zurück zum Zitat Matsubayashi H, Infante JR, Winter J, Klein AP, Schulick R, Hruban R, Visvanathan K, Goggins M: Tumor COX-2 expression and prognosis of patients with resectable pancreatic cancer. Cancer Biol Ther. 2007, 6 (10): 1569-1575.CrossRefPubMed Matsubayashi H, Infante JR, Winter J, Klein AP, Schulick R, Hruban R, Visvanathan K, Goggins M: Tumor COX-2 expression and prognosis of patients with resectable pancreatic cancer. Cancer Biol Ther. 2007, 6 (10): 1569-1575.CrossRefPubMed
17.
Zurück zum Zitat Muller-Decker K, Furstenberger G, Annan N, Kucher D, Pohl-Arnold A, Steinbauer B, Esposito I, Chiblak S, Friess H, Schirmacher P, Berger I: Preinvasive duct-derived neoplasms in pancreas of keratin 5-promoter cyclooxygenase-2 transgenic mice. Gastroenterology. 2006, 130 (7): 2165-2178.CrossRefPubMed Muller-Decker K, Furstenberger G, Annan N, Kucher D, Pohl-Arnold A, Steinbauer B, Esposito I, Chiblak S, Friess H, Schirmacher P, Berger I: Preinvasive duct-derived neoplasms in pancreas of keratin 5-promoter cyclooxygenase-2 transgenic mice. Gastroenterology. 2006, 130 (7): 2165-2178.CrossRefPubMed
18.
Zurück zum Zitat Colby JK, Klein RD, McArthur MJ, Conti CJ, Kiguchi K, Kawamoto T, Riggs PK, Pavone AI, Sawicki J, Fischer SM: Progressive metaplastic and dysplastic changes in mouse pancreas induced by cyclooxygenase-2 overexpression. Neoplasia. 2008, 10 (8): 782-796.CrossRefPubMedPubMedCentral Colby JK, Klein RD, McArthur MJ, Conti CJ, Kiguchi K, Kawamoto T, Riggs PK, Pavone AI, Sawicki J, Fischer SM: Progressive metaplastic and dysplastic changes in mouse pancreas induced by cyclooxygenase-2 overexpression. Neoplasia. 2008, 10 (8): 782-796.CrossRefPubMedPubMedCentral
19.
Zurück zum Zitat Breyer RM, Bagdassarian CK, Myers SA, Breyer MD: Prostanoid receptors: subtypes and signaling. Annu Rev Pharmacol Toxicol. 2001, 41: 661-690.CrossRefPubMed Breyer RM, Bagdassarian CK, Myers SA, Breyer MD: Prostanoid receptors: subtypes and signaling. Annu Rev Pharmacol Toxicol. 2001, 41: 661-690.CrossRefPubMed
20.
21.
Zurück zum Zitat Ji R, Chou CL, Xu W, Chen XB, Woodward DF, Regan JW: EP1 prostanoid receptor coupling to G I/o Up-regulates the expression of hypoxia-inducible factor-1 alpha through activation of a phosphoinositide-3 kinase signaling pathway. Mol Pharmacol. 2010, 77 (6): 1025-1036.CrossRefPubMedPubMedCentral Ji R, Chou CL, Xu W, Chen XB, Woodward DF, Regan JW: EP1 prostanoid receptor coupling to G I/o Up-regulates the expression of hypoxia-inducible factor-1 alpha through activation of a phosphoinositide-3 kinase signaling pathway. Mol Pharmacol. 2010, 77 (6): 1025-1036.CrossRefPubMedPubMedCentral
23.
Zurück zum Zitat Yokoyama U, Patel HH, Lai NC, Aroonsakool N, Roth DM, Insel PA: The cyclic AMP effector epac integrates Pro- and anti-fibrotic signals. Proc Natl Acad Sci U S A. 2008, 105 (17): 6386-6391.CrossRefPubMedPubMedCentral Yokoyama U, Patel HH, Lai NC, Aroonsakool N, Roth DM, Insel PA: The cyclic AMP effector epac integrates Pro- and anti-fibrotic signals. Proc Natl Acad Sci U S A. 2008, 105 (17): 6386-6391.CrossRefPubMedPubMedCentral
25.
Zurück zum Zitat Insel PA, Murray F, Yokoyama U, Romano S, Yun H, Brown L, Snead A, Lu D, Aroonsakool N: CAMP and Epac in the regulation of tissue fibrosis. Br J Pharmacol. 2012, 166 (2): 447-456.CrossRefPubMedPubMedCentral Insel PA, Murray F, Yokoyama U, Romano S, Yun H, Brown L, Snead A, Lu D, Aroonsakool N: CAMP and Epac in the regulation of tissue fibrosis. Br J Pharmacol. 2012, 166 (2): 447-456.CrossRefPubMedPubMedCentral
26.
Zurück zum Zitat White KE, Ding Q, Moore BB, Peters-Golden M, Ware LB, Matthay MA, Olman MA: Prostaglandin E2 mediates IL-1beta-related fibroblast mitogenic effects in acute lung injury through differential utilization of prostanoid receptors. J Immunol. 2008, 180 (1): 637-646.CrossRefPubMedPubMedCentral White KE, Ding Q, Moore BB, Peters-Golden M, Ware LB, Matthay MA, Olman MA: Prostaglandin E2 mediates IL-1beta-related fibroblast mitogenic effects in acute lung injury through differential utilization of prostanoid receptors. J Immunol. 2008, 180 (1): 637-646.CrossRefPubMedPubMedCentral
27.
Zurück zum Zitat Watanabe T, Satoh H, Togoh M, Taniguchi S, Hashimoto Y, Kurokawa K: Positive and negative regulation of cell proliferation through prostaglandin receptors in NIH-3 T3 cells. J Cell Physiol. 1996, 169 (2): 401-409.CrossRefPubMed Watanabe T, Satoh H, Togoh M, Taniguchi S, Hashimoto Y, Kurokawa K: Positive and negative regulation of cell proliferation through prostaglandin receptors in NIH-3 T3 cells. J Cell Physiol. 1996, 169 (2): 401-409.CrossRefPubMed
28.
Zurück zum Zitat Oga T, Matsuoka T, Yao C, Nonomura K, Kitaoka S, Sakata D, Kita Y, Tanizawa K, Taguchi Y, Chin K, Mishima M, Shimizu T, Narumiya S: Prostaglandin F (2alpha) receptor signaling facilitates bleomycin-induced pulmonary fibrosis independently of transforming growth factor-beta. Nat Med. 2009, 15 (12): 1426-1430.CrossRefPubMed Oga T, Matsuoka T, Yao C, Nonomura K, Kitaoka S, Sakata D, Kita Y, Tanizawa K, Taguchi Y, Chin K, Mishima M, Shimizu T, Narumiya S: Prostaglandin F (2alpha) receptor signaling facilitates bleomycin-induced pulmonary fibrosis independently of transforming growth factor-beta. Nat Med. 2009, 15 (12): 1426-1430.CrossRefPubMed
29.
Zurück zum Zitat Harding P, LaPointe MC: Prostaglandin E2 increases cardiac fibroblast proliferation and increases cyclin D expression Via EP1 receptor. Prostaglandins Leukot Essent Fatty Acids. 2011, 84 (5–6): 147-152.CrossRefPubMedPubMedCentral Harding P, LaPointe MC: Prostaglandin E2 increases cardiac fibroblast proliferation and increases cyclin D expression Via EP1 receptor. Prostaglandins Leukot Essent Fatty Acids. 2011, 84 (5–6): 147-152.CrossRefPubMedPubMedCentral
30.
Zurück zum Zitat Hui AY, Dannenberg AJ, Sung JJ, Subbaramaiah K, Du B, Olinga P, Friedman SL: Prostaglandin E2 inhibits transforming growth factor beta 1-mediated induction of collagen alpha 1(I) in hepatic stellate cells. J Hepatol. 2004, 41 (2): 251-258.CrossRefPubMed Hui AY, Dannenberg AJ, Sung JJ, Subbaramaiah K, Du B, Olinga P, Friedman SL: Prostaglandin E2 inhibits transforming growth factor beta 1-mediated induction of collagen alpha 1(I) in hepatic stellate cells. J Hepatol. 2004, 41 (2): 251-258.CrossRefPubMed
31.
Zurück zum Zitat Mallat A, Gallois C, Tao J, Habib A, Maclouf J, Mavier P, Preaux AM, Lotersztajn S: Platelet-derived growth factor-BB and thrombin generate positive and negative signals for human hepatic stellate cell proliferation. Role of a prostaglandin/cyclic AMP pathway and cross-talk with endothelin receptors. J Biol Chem. 1998, 273 (42): 27300-27305.CrossRefPubMed Mallat A, Gallois C, Tao J, Habib A, Maclouf J, Mavier P, Preaux AM, Lotersztajn S: Platelet-derived growth factor-BB and thrombin generate positive and negative signals for human hepatic stellate cell proliferation. Role of a prostaglandin/cyclic AMP pathway and cross-talk with endothelin receptors. J Biol Chem. 1998, 273 (42): 27300-27305.CrossRefPubMed
32.
Zurück zum Zitat Koide S, Kobayashi Y, Oki Y, Nakamura H: Prostaglandin E2 inhibits platelet-derived growth factor-stimulated cell proliferation through a prostaglandin E receptor EP2 subtype in Rat hepatic stellate cells. Dig Dis Sci. 2004, 49 (9): 1394-1400.CrossRefPubMed Koide S, Kobayashi Y, Oki Y, Nakamura H: Prostaglandin E2 inhibits platelet-derived growth factor-stimulated cell proliferation through a prostaglandin E receptor EP2 subtype in Rat hepatic stellate cells. Dig Dis Sci. 2004, 49 (9): 1394-1400.CrossRefPubMed
33.
Zurück zum Zitat Bachem MG, Schneider E, Gross H, Weidenbach H, Schmid RM, Menke A, Siech M, Beger H, Grunert A, Adler G: Identification, culture, and characterization of pancreatic stellate cells in rats and humans. Gastroenterology. 1998, 115 (2): 421-432.CrossRefPubMed Bachem MG, Schneider E, Gross H, Weidenbach H, Schmid RM, Menke A, Siech M, Beger H, Grunert A, Adler G: Identification, culture, and characterization of pancreatic stellate cells in rats and humans. Gastroenterology. 1998, 115 (2): 421-432.CrossRefPubMed
34.
Zurück zum Zitat Refsnes M, Thoresen GH, Dajani OF, Christoffersen T: Stimulation of hepatocyte DNA synthesis by prostaglandin E2 and prostaglandin F2 alpha: additivity with the effect of norepinephrine, and synergism with epidermal growth factor. J Cell Physiol. 1994, 159 (1): 35-40.CrossRefPubMed Refsnes M, Thoresen GH, Dajani OF, Christoffersen T: Stimulation of hepatocyte DNA synthesis by prostaglandin E2 and prostaglandin F2 alpha: additivity with the effect of norepinephrine, and synergism with epidermal growth factor. J Cell Physiol. 1994, 159 (1): 35-40.CrossRefPubMed
35.
Zurück zum Zitat Jaster R, Hilgendorf I, Fitzner B, Brock P, Sparmann G, Emmrich J, Liebe S: Regulation of pancreatic stellate cell function in vitro: biological and molecular effects of all-trans retinoic acid. Biochem Pharmacol. 2003, 66 (4): 633-641.CrossRefPubMed Jaster R, Hilgendorf I, Fitzner B, Brock P, Sparmann G, Emmrich J, Liebe S: Regulation of pancreatic stellate cell function in vitro: biological and molecular effects of all-trans retinoic acid. Biochem Pharmacol. 2003, 66 (4): 633-641.CrossRefPubMed
36.
Zurück zum Zitat Ye J, Coulouris G, Zaretskaya I, Cutcutache I, Rozen S, Madden TL: Primer-BLAST: a tool to design target-specific primers for polymerase chain reaction. BMC Bioinforma. 2012, 13: 134-CrossRef Ye J, Coulouris G, Zaretskaya I, Cutcutache I, Rozen S, Madden TL: Primer-BLAST: a tool to design target-specific primers for polymerase chain reaction. BMC Bioinforma. 2012, 13: 134-CrossRef
37.
Zurück zum Zitat Livak KJ, Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2 (−delta delta C (T)) method. Methods. 2001, 25 (4): 402-408.CrossRefPubMed Livak KJ, Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2 (−delta delta C (T)) method. Methods. 2001, 25 (4): 402-408.CrossRefPubMed
38.
Zurück zum Zitat Skomedal T, Grynne B, Osnes JB, Sjetnan AE, Oye I: A radioimmunoassay for cyclic AMP (CAMP) obtained by acetylation of both unlabeled and labeled (3H-CAMP) ligand, or of unlabeled ligand only. Acta Pharmacol Toxicol (Copenh). 1980, 46 (3): 200-204.CrossRef Skomedal T, Grynne B, Osnes JB, Sjetnan AE, Oye I: A radioimmunoassay for cyclic AMP (CAMP) obtained by acetylation of both unlabeled and labeled (3H-CAMP) ligand, or of unlabeled ligand only. Acta Pharmacol Toxicol (Copenh). 1980, 46 (3): 200-204.CrossRef
39.
Zurück zum Zitat Sato N, Maehara N, Goggins M: Gene expression profiling of tumor-stromal interactions between pancreatic cancer cells and stromal fibroblasts. Cancer Res. 2004, 64 (19): 6950-6956.CrossRefPubMed Sato N, Maehara N, Goggins M: Gene expression profiling of tumor-stromal interactions between pancreatic cancer cells and stromal fibroblasts. Cancer Res. 2004, 64 (19): 6950-6956.CrossRefPubMed
40.
Zurück zum Zitat Charo C, Holla V, Arumugam T, Hwang R, Yang P, Dubois RN, Menter DG, Logsdon CD, Ramachandran V: Prostaglandin E2 regulates pancreatic stellate cell activity Via the EP4 receptor. Pancreas. 2013, 42 (3): 467-474.CrossRefPubMedPubMedCentral Charo C, Holla V, Arumugam T, Hwang R, Yang P, Dubois RN, Menter DG, Logsdon CD, Ramachandran V: Prostaglandin E2 regulates pancreatic stellate cell activity Via the EP4 receptor. Pancreas. 2013, 42 (3): 467-474.CrossRefPubMedPubMedCentral
41.
Zurück zum Zitat Huang S, Wettlaufer SH, Hogaboam C, Aronoff DM, Peters-Golden M: Prostaglandin E (2) inhibits collagen expression and proliferation in patient-derived normal lung fibroblasts Via E prostanoid 2 receptor and CAMP signaling. Am J Physiol Lung Cell Mol Physiol. 2007, 292 (2): L405-L413.CrossRefPubMed Huang S, Wettlaufer SH, Hogaboam C, Aronoff DM, Peters-Golden M: Prostaglandin E (2) inhibits collagen expression and proliferation in patient-derived normal lung fibroblasts Via E prostanoid 2 receptor and CAMP signaling. Am J Physiol Lung Cell Mol Physiol. 2007, 292 (2): L405-L413.CrossRefPubMed
42.
Zurück zum Zitat Seamon KB, Padgett W, Daly JW: Forskolin: unique diterpene activator of adenylate cyclase in membranes and in intact cells. Proc Natl Acad Sci U S A. 1981, 78 (6): 3363-3367.CrossRefPubMedPubMedCentral Seamon KB, Padgett W, Daly JW: Forskolin: unique diterpene activator of adenylate cyclase in membranes and in intact cells. Proc Natl Acad Sci U S A. 1981, 78 (6): 3363-3367.CrossRefPubMedPubMedCentral
43.
Zurück zum Zitat Machwate M, Harada S, Leu CT, Seedor G, Labelle M, Gallant M, Hutchins S, Lachance N, Sawyer N, Slipetz D, Metters KM, Rodan SB, Young R, Rodan GA: Prostaglandin receptor EP (4) mediates the bone anabolic effects of PGE(2). Mol Pharmacol. 2001, 60 (1): 36-41.PubMed Machwate M, Harada S, Leu CT, Seedor G, Labelle M, Gallant M, Hutchins S, Lachance N, Sawyer N, Slipetz D, Metters KM, Rodan SB, Young R, Rodan GA: Prostaglandin receptor EP (4) mediates the bone anabolic effects of PGE(2). Mol Pharmacol. 2001, 60 (1): 36-41.PubMed
44.
Zurück zum Zitat Bachem MG, Schunemann M, Ramadani M, Siech M, Beger H, Buck A, Zhou S, Schmid-Kotsas A, Adler G: Pancreatic carcinoma cells induce fibrosis by stimulating proliferation and matrix synthesis of stellate cells. Gastroenterology. 2005, 12847 (4): 907-921.CrossRef Bachem MG, Schunemann M, Ramadani M, Siech M, Beger H, Buck A, Zhou S, Schmid-Kotsas A, Adler G: Pancreatic carcinoma cells induce fibrosis by stimulating proliferation and matrix synthesis of stellate cells. Gastroenterology. 2005, 12847 (4): 907-921.CrossRef
45.
Zurück zum Zitat Luttenberger T, Schmid-Kotsas A, Menke A, Siech M, Beger H, Adler G, Grunert A, Bachem MG: Platelet-derived growth factors stimulate proliferation and extracellular matrix synthesis of pancreatic stellate cells: implications in pathogenesis of pancreas fibrosis. Lab Invest. 2000, 80 (1): 47-55.CrossRefPubMed Luttenberger T, Schmid-Kotsas A, Menke A, Siech M, Beger H, Adler G, Grunert A, Bachem MG: Platelet-derived growth factors stimulate proliferation and extracellular matrix synthesis of pancreatic stellate cells: implications in pathogenesis of pancreas fibrosis. Lab Invest. 2000, 80 (1): 47-55.CrossRefPubMed
46.
Zurück zum Zitat Menke A, Adler G: TGFbeta-induced fibrogenesis of the pancreas. Int J Gastrointest Cancer. 2002, 31 (1–3): 41-46.CrossRefPubMed Menke A, Adler G: TGFbeta-induced fibrogenesis of the pancreas. Int J Gastrointest Cancer. 2002, 31 (1–3): 41-46.CrossRefPubMed
47.
Zurück zum Zitat Wilborn J, Crofford LJ, Burdick MD, Kunkel SL, Strieter RM, Peters-Golden M: Cultured lung fibroblasts isolated from patients with idiopathic pulmonary fibrosis have a diminished capacity to synthesize prostaglandin E2 and to express cyclooxygenase-2. J Clin Invest. 1995, 95 (4): 1861-1868.CrossRefPubMedPubMedCentral Wilborn J, Crofford LJ, Burdick MD, Kunkel SL, Strieter RM, Peters-Golden M: Cultured lung fibroblasts isolated from patients with idiopathic pulmonary fibrosis have a diminished capacity to synthesize prostaglandin E2 and to express cyclooxygenase-2. J Clin Invest. 1995, 95 (4): 1861-1868.CrossRefPubMedPubMedCentral
48.
Zurück zum Zitat Keerthisingam CB, Jenkins RG, Harrison NK, Hernandez-Rodriguez NA, Booth H, Laurent GJ, Hart SL, Foster ML, McAnulty RJ: Cyclooxygenase-2 deficiency results in a loss of the anti-proliferative response to transforming growth factor-beta in human fibrotic lung fibroblasts and promotes bleomycin-induced pulmonary fibrosis in mice. Am J Pathol. 2001, 158 (4): 1411-1422.CrossRefPubMedPubMedCentral Keerthisingam CB, Jenkins RG, Harrison NK, Hernandez-Rodriguez NA, Booth H, Laurent GJ, Hart SL, Foster ML, McAnulty RJ: Cyclooxygenase-2 deficiency results in a loss of the anti-proliferative response to transforming growth factor-beta in human fibrotic lung fibroblasts and promotes bleomycin-induced pulmonary fibrosis in mice. Am J Pathol. 2001, 158 (4): 1411-1422.CrossRefPubMedPubMedCentral
49.
Zurück zum Zitat Handler JA, Danilowicz RM, Eling TE: Mitogenic signaling by epidermal growth factor (EGF), but Not platelet-derived growth factor, requires arachidonic acid metabolism in BALB/c 3 T3 cells. Modulation of EGF-dependent C-Myc expression by prostaglandins. J Biol Chem. 1990, 265 (7): 3669-3673.PubMed Handler JA, Danilowicz RM, Eling TE: Mitogenic signaling by epidermal growth factor (EGF), but Not platelet-derived growth factor, requires arachidonic acid metabolism in BALB/c 3 T3 cells. Modulation of EGF-dependent C-Myc expression by prostaglandins. J Biol Chem. 1990, 265 (7): 3669-3673.PubMed
50.
Zurück zum Zitat Castano E, Bartrons R, Gil J: Inhibition of cyclooxygenase-2 decreases DNA synthesis induced by platelet-derived growth factor in Swiss 3 T3 fibroblasts. J Pharmacol Exp Ther. 2000, 293 (2): 509-513.PubMed Castano E, Bartrons R, Gil J: Inhibition of cyclooxygenase-2 decreases DNA synthesis induced by platelet-derived growth factor in Swiss 3 T3 fibroblasts. J Pharmacol Exp Ther. 2000, 293 (2): 509-513.PubMed
51.
Zurück zum Zitat Yoshida S, Ujiki M, Ding XZ, Pelham C, Talamonti MS, Bell RH, Denham W, Adrian TE: Pancreatic stellate cells (PSCs) express cyclooxygenase-2 (COX-2) and pancreatic cancer stimulates COX-2 in PSCs. Mol Cancer. 2005, 4 (1): 27-CrossRefPubMedPubMedCentral Yoshida S, Ujiki M, Ding XZ, Pelham C, Talamonti MS, Bell RH, Denham W, Adrian TE: Pancreatic stellate cells (PSCs) express cyclooxygenase-2 (COX-2) and pancreatic cancer stimulates COX-2 in PSCs. Mol Cancer. 2005, 4 (1): 27-CrossRefPubMedPubMedCentral
52.
Zurück zum Zitat Smith ML, Hawcroft G, Hull MA: The effect of Non-steroidal anti-inflammatory drugs on human colorectal cancer cells: evidence of different mechanisms of action. Eur J Cancer. 2000, 36 (5): 664-674.CrossRefPubMed Smith ML, Hawcroft G, Hull MA: The effect of Non-steroidal anti-inflammatory drugs on human colorectal cancer cells: evidence of different mechanisms of action. Eur J Cancer. 2000, 36 (5): 664-674.CrossRefPubMed
53.
Zurück zum Zitat Denkert C, Furstenberg A, Daniel PT, Koch I, Kobel M, Weichert W, Siegert A, Hauptmann S: Induction of G0/G1 cell cycle arrest in ovarian carcinoma cells by the anti-inflammatory drug NS-398, but Not by COX-2-specific RNA interference. Oncogene. 2003, 22 (54): 8653-8661.CrossRefPubMed Denkert C, Furstenberg A, Daniel PT, Koch I, Kobel M, Weichert W, Siegert A, Hauptmann S: Induction of G0/G1 cell cycle arrest in ovarian carcinoma cells by the anti-inflammatory drug NS-398, but Not by COX-2-specific RNA interference. Oncogene. 2003, 22 (54): 8653-8661.CrossRefPubMed
54.
Zurück zum Zitat Elder DJ, Halton DE, Hague A, Paraskeva C: Induction of apoptotic cell death in human colorectal carcinoma cell lines by a cyclooxygenase-2 (COX-2)-selective nonsteroidal anti-inflammatory drug: independence from COX-2 protein expression. Clin Cancer Res. 1997, 3 (10): 1679-1683.PubMed Elder DJ, Halton DE, Hague A, Paraskeva C: Induction of apoptotic cell death in human colorectal carcinoma cell lines by a cyclooxygenase-2 (COX-2)-selective nonsteroidal anti-inflammatory drug: independence from COX-2 protein expression. Clin Cancer Res. 1997, 3 (10): 1679-1683.PubMed
55.
Zurück zum Zitat Black SA, Palamakumbura AH, Stan M, Trackman PC: Tissue-specific mechanisms for CCN2/CTGF persistence in fibrotic gingiva: interactions between CAMP and MAPK signaling pathways, and prostaglandin E2-EP3 receptor mediated activation of the C-JUN N-terminal kinase. J Biol Chem. 2007, 282 (21): 15416-15429.CrossRefPubMedPubMedCentral Black SA, Palamakumbura AH, Stan M, Trackman PC: Tissue-specific mechanisms for CCN2/CTGF persistence in fibrotic gingiva: interactions between CAMP and MAPK signaling pathways, and prostaglandin E2-EP3 receptor mediated activation of the C-JUN N-terminal kinase. J Biol Chem. 2007, 282 (21): 15416-15429.CrossRefPubMedPubMedCentral
56.
Zurück zum Zitat Kokawa A, Kondo H, Gotoda T, Ono H, Saito D, Nakadaira S, Kosuge T, Yoshida S: Increased expression of cyclooxygenase-2 in human pancreatic neoplasms and potential for chemoprevention by cyclooxygenase inhibitors. Cancer. 2001, 91 (2): 333-338.CrossRefPubMed Kokawa A, Kondo H, Gotoda T, Ono H, Saito D, Nakadaira S, Kosuge T, Yoshida S: Increased expression of cyclooxygenase-2 in human pancreatic neoplasms and potential for chemoprevention by cyclooxygenase inhibitors. Cancer. 2001, 91 (2): 333-338.CrossRefPubMed
57.
Zurück zum Zitat Kong G, Kim EK, Kim WS, Lee KT, Lee YW, Lee JK, Paik SW, Rhee JC: Role of cyclooxygenase-2 and inducible nitric oxide synthase in pancreatic cancer. J Gastroenterol Hepatol. 2002, 17 (8): 914-921.CrossRefPubMed Kong G, Kim EK, Kim WS, Lee KT, Lee YW, Lee JK, Paik SW, Rhee JC: Role of cyclooxygenase-2 and inducible nitric oxide synthase in pancreatic cancer. J Gastroenterol Hepatol. 2002, 17 (8): 914-921.CrossRefPubMed
58.
Zurück zum Zitat Koshiba T, Hosotani R, Miyamoto Y, Wada M, Lee JU, Fujimoto K, Tsuji S, Nakajima S, Doi R, Imamura M: Immunohistochemical analysis of cyclooxygenase-2 expression in pancreatic tumors. Int J Pancreatol. 1999, 26 (2): 69-76.CrossRefPubMed Koshiba T, Hosotani R, Miyamoto Y, Wada M, Lee JU, Fujimoto K, Tsuji S, Nakajima S, Doi R, Imamura M: Immunohistochemical analysis of cyclooxygenase-2 expression in pancreatic tumors. Int J Pancreatol. 1999, 26 (2): 69-76.CrossRefPubMed
59.
Zurück zum Zitat Molina MA, Sitja-Arnau M, Lemoine MG, Frazier ML, Sinicrope FA: Increased cyclooxygenase-2 expression in human pancreatic carcinomas and cell lines: growth inhibition by nonsteroidal anti-inflammatory drugs. Cancer Res. 1999, 59 (17): 4356-4362.PubMed Molina MA, Sitja-Arnau M, Lemoine MG, Frazier ML, Sinicrope FA: Increased cyclooxygenase-2 expression in human pancreatic carcinomas and cell lines: growth inhibition by nonsteroidal anti-inflammatory drugs. Cancer Res. 1999, 59 (17): 4356-4362.PubMed
60.
Zurück zum Zitat Schlosser W, Schlosser S, Ramadani M, Gansauge F, Gansauge S, Beger HG: Cyclooxygenase-2 is overexpressed in chronic pancreatitis. Pancreas. 2002, 25 (1): 26-30.CrossRefPubMed Schlosser W, Schlosser S, Ramadani M, Gansauge F, Gansauge S, Beger HG: Cyclooxygenase-2 is overexpressed in chronic pancreatitis. Pancreas. 2002, 25 (1): 26-30.CrossRefPubMed
61.
Zurück zum Zitat Saukkonen K, Nieminen O, Van RB, Vilkki S, Harkonen M, Juhola M, Mecklin JP, Sipponen P, Ristimaki A: Expression of cyclooxygenase-2 in dysplasia of the stomach and in intestinal-type gastric adenocarcinoma. Clin Cancer Res. 2001, 7 (7): 1923-1931.PubMed Saukkonen K, Nieminen O, Van RB, Vilkki S, Harkonen M, Juhola M, Mecklin JP, Sipponen P, Ristimaki A: Expression of cyclooxygenase-2 in dysplasia of the stomach and in intestinal-type gastric adenocarcinoma. Clin Cancer Res. 2001, 7 (7): 1923-1931.PubMed
62.
Zurück zum Zitat Vonlaufen A, Phillips PA, Yang L, Xu Z, Fiala-Beer E, Zhang X, Pirola RC, Wilson JS, Apte MV: Isolation of quiescent human pancreatic stellate cells: a promising in vitro tool for studies of human pancreatic stellate cell biology. Pancreatology. 2010, 10 (4): 434-443.CrossRefPubMed Vonlaufen A, Phillips PA, Yang L, Xu Z, Fiala-Beer E, Zhang X, Pirola RC, Wilson JS, Apte MV: Isolation of quiescent human pancreatic stellate cells: a promising in vitro tool for studies of human pancreatic stellate cell biology. Pancreatology. 2010, 10 (4): 434-443.CrossRefPubMed
63.
Zurück zum Zitat Aoki H, Ohnishi H, Hama K, Shinozaki S, Kita H, Osawa H, Yamamoto H, Sato K, Tamada K, Sugano K: Cyclooxygenase-2 is required for activated pancreatic stellate cells to respond to proinflammatory cytokines. Am J Physiol Cell Physiol. 2007, 292 (1): C259-C268.CrossRefPubMed Aoki H, Ohnishi H, Hama K, Shinozaki S, Kita H, Osawa H, Yamamoto H, Sato K, Tamada K, Sugano K: Cyclooxygenase-2 is required for activated pancreatic stellate cells to respond to proinflammatory cytokines. Am J Physiol Cell Physiol. 2007, 292 (1): C259-C268.CrossRefPubMed
64.
Zurück zum Zitat Xu D, Matsuo Y, Ma J, Koide S, Ochi N, Yasuda A, Funahashi H, Okada Y, Takeyama H: Cancer cell-derived IL-1alpha promotes HGF secretion by stromal cells and enhances metastatic potential in pancreatic cancer cells. J Surg Oncol. 2010, 102 (5): 469-477.CrossRefPubMed Xu D, Matsuo Y, Ma J, Koide S, Ochi N, Yasuda A, Funahashi H, Okada Y, Takeyama H: Cancer cell-derived IL-1alpha promotes HGF secretion by stromal cells and enhances metastatic potential in pancreatic cancer cells. J Surg Oncol. 2010, 102 (5): 469-477.CrossRefPubMed
Metadaten
Titel
Inhibitory effects of prostaglandin E2on collagen synthesis and cell proliferation in human stellate cells from pancreatic head adenocarcinoma
verfasst von
Ewa Pomianowska
Dagny Sandnes
Krzysztof Grzyb
Aasa R Schjølberg
Monica Aasrum
Ingun H Tveteraas
Vegard Tjomsland
Thoralf Christoffersen
Ivar P Gladhaug
Publikationsdatum
01.12.2014
Verlag
BioMed Central
Erschienen in
BMC Cancer / Ausgabe 1/2014
Elektronische ISSN: 1471-2407
DOI
https://doi.org/10.1186/1471-2407-14-413

Weitere Artikel der Ausgabe 1/2014

BMC Cancer 1/2014 Zur Ausgabe

Adjuvante Immuntherapie verlängert Leben bei RCC

25.04.2024 Nierenkarzinom Nachrichten

Nun gibt es auch Resultate zum Gesamtüberleben: Eine adjuvante Pembrolizumab-Therapie konnte in einer Phase-3-Studie das Leben von Menschen mit Nierenzellkarzinom deutlich verlängern. Die Sterberate war im Vergleich zu Placebo um 38% geringer.

Alectinib verbessert krankheitsfreies Überleben bei ALK-positivem NSCLC

25.04.2024 NSCLC Nachrichten

Das Risiko für Rezidiv oder Tod von Patienten und Patientinnen mit reseziertem ALK-positivem NSCLC ist unter einer adjuvanten Therapie mit dem Tyrosinkinase-Inhibitor Alectinib signifikant geringer als unter platinbasierter Chemotherapie.

Bei Senioren mit Prostatakarzinom auf Anämie achten!

24.04.2024 DGIM 2024 Nachrichten

Patienten, die zur Behandlung ihres Prostatakarzinoms eine Androgendeprivationstherapie erhalten, entwickeln nicht selten eine Anämie. Wer ältere Patienten internistisch mitbetreut, sollte auf diese Nebenwirkung achten.

ICI-Therapie in der Schwangerschaft wird gut toleriert

Müssen sich Schwangere einer Krebstherapie unterziehen, rufen Immuncheckpointinhibitoren offenbar nicht mehr unerwünschte Wirkungen hervor als andere Mittel gegen Krebs.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.