Skip to main content
Erschienen in: BMC Cancer 1/2019

Open Access 01.12.2019 | Research article

Secretion of fibronectin by human pancreatic stellate cells promotes chemoresistance to gemcitabine in pancreatic cancer cells

Erschienen in: BMC Cancer | Ausgabe 1/2019

Abstract

Background

Gemcitabine remains a cornerstone in chemotherapy of pancreatic ductal adenocarcinoma (PDAC) despite suboptimal clinical effects that are partly due to the development of chemoresistance. Pancreatic stellate cells (PSCs) of the tumor stroma are known to interact with pancreatic cancer cells (PCCs) and influence the progression of PDAC through a complex network of signaling molecules that involve extracellular matrix (ECM) proteins. To understand tumor-stroma interactions regulating chemosensitivity, the role of PSC-secreted fibronectin (FN) in the development of gemcitabine resistance in PDAC was examined.

Methods

PSC cultures obtained from ten different human PDAC tumors were co-cultured with PCC lines (AsPC-1, BxPC-3, Capan-2, HPAF-II, MIA PaCa-2, PANC-1 and SW-1990) either directly, or indirectly via incubation with PSC-conditioned medium (PSC-CM). Gemcitabine dose response cytotoxicity was determined using MTT based cell viability assays. Protein expression was assessed by western blotting and immunofluorescence. PSC-CM secretome analysis was performed by proteomics-based LC-MS/MS, and FN content in PSC-CM was determined with ELISA. Radiolabeled gemcitabine was used to determine the capacity of PCCs to uptake the drug.

Results

In both direct and indirect co-culture, PSCs induced varying degrees of resistance to the cytotoxic effects of gemcitabine among all cancer cell lines examined. A variable degree of increased phosphorylation of ERK1/2 was observed across all PCC lines upon incubation with PSC-CM, while activation of AKT was not detected. Secretome analysis of PSC-CM identified 796 different proteins, including several ECM-related proteins such as FN and collagens. Soluble FN content in PSC-CM was detected in the range 175–350 ng/ml. Neither FN nor PSC-CM showed any effect on PCC uptake capacity of gemcitabine. PCCs grown on FN-coated surface displayed higher resistance to gemcitabine compared to cells grown on non-coated surface. Furthermore, a FN inhibitor, synthetic Arg-Gly-Asp-Ser (RGDS) peptide significantly inhibited PSC-CM-induced chemoresistance in PCCs via downregulation of ERK1/2 phosphorylation.

Conclusions

The findings of this study suggest that FN secreted by PSCs in the ECM plays a key role in the development of resistance to gemcitabine via activation of ERK1/2. FN-blocking agents added to gemcitabine-based chemotherapy might counteract chemoresistance in PDAC and provide better clinical outcomes.
Begleitmaterial
Hinweise

Electronic supplementary material

The online version of this article (https://​doi.​org/​10.​1186/​s12885-019-5803-1) contains supplementary material, which is available to authorized users.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
CAFs
Cancer-associated fibroblasts
ECM
Extracellular matrix
FN
Fibronectin
PCC
Pancreatic cancer cell
PDAC
Pancreatic ductal adenocarcinoma
PSC
Pancreatic stellate cell
PSC-CM
PSC-conditioned medium
SFM
Serum-free DMEM

Background

Pancreatic ductal adenocarcinoma (PDAC), generally referred to as pancreatic cancer, is a highly malignant disease characterized by late diagnosis, early metastasis, limited response to chemotherapy, and an extremely poor prognosis [13]. Despite significant advances in the understanding of the pathobiology of the disease over the past decades, overall 5-year survival rate has only marginally improved to merely 7%, making it the fourth leading cause of cancer-related mortality in the Western world [4]. Moreover, it is projected to rank second by 2030 [5]. While surgery offers the only potential curative treatment, only 15–20% of patients have resectable disease at the time of diagnosis, and these patients also carry the risk of disease recurrence within a year post-surgery [6]. Chemotherapy is therefore a crucial component in the treatment of the disease, both for unresectable (locally advanced or metastatic) disease or in the neoadjuvant and adjuvant settings [7]. Gemcitabine (dFdC) is a nucleoside pyrimidine analogue that has long been the backbone of chemotherapy for PDAC in all stages of the disease [8, 9]. However, gemcitabine has suboptimal clinical outcomes caused by molecular mechanisms limiting its cellular uptake, activation and overall efficacy, as well as by the development of chemoresistance within weeks of treatment initiation [10, 11].
The presence of a dense desmoplastic stroma that constitutes the bulk of the tumor mass is a prominent feature of PDAC [12, 13]. The stroma is composed of several different cell types and various extracellular matrix (ECM) components such as collagens and fibronectin (FN), which provide structural integrity and initiate signaling cascades that promote tumor cell survival, proliferation, and migration [1416]. The activated pancreatic stellate cell (PSC) – a type of cancer-associated fibroblast (CAF) – is the primary source of ECM components and also the major driver and organizer of the desmoplastic reaction in PDAC [17]. In the healthy pancreas, PSCs exist in a quiescent state, but in response to various pathophysiological stimuli such as injury, inflammation and induction of neoplasia in ductal epithelium, the PSCs are activated [14, 18]. Through the secretion of excessive amounts of ECM components, activated PSCs induce the fibrosis that is characteristically found in PDAC [18, 19].
Several lines of evidence suggest that the tumor stroma contributes to gemcitabine chemoresistance through a range of mechanisms, including impairment of drug delivery due to decreased functional microvascularity, activation of anti-apoptotic pathways, promotion of epithelial-mesenchymal transition, and upregulation of tumor-supporting inflammatory cytokines [10, 2022]. While the cause of gemcitabine chemoresistance is clearly multifactorial and PSCs are implicated in several of these processes, the precise role of the PSCs in the development of chemoresistance is still not fully understood [2326]. Previous studies have also indicated that pancreatic cancer cells (PCCs) embedded in collagen gels [27] or cultured on FN-coated dishes [28], show increased resistance to gemcitabine, suggesting the possibility that ECM-induced signals might be involved.
In the present study we hypothesized that secreted soluble factors implicated in gemcitabine chemoresistance in pancreatic cancer may be identified through analysis of the PSC secretome. To this end, we examined conditioned medium from ten primary human PSC cultures for its secretome content and evaluated its effects on PCC’s gemcitabine chemosensitivity in order to explore the effects of ECM proteins on pancreatic cancer chemoresistance.

Methods

Reagents

Reagents were purchased from the following sources: Dulbecco’s modified Eagle’s medium containing 4.5 g/l glucose (DMEM), penicillin-streptomycin (Pen-Strep), amphotericin B, Trypsin/EDTA (1X), fetal bovine serum (FBS) and Pierce™ BCA protein assay kit from Thermo Fisher Scientific (Waltham, MA, USA); bovine serum albumin (BSA), Collagen type I solution from rat tail, FN, gemcitabine hydrochloride, 3-(4,5-Dimethylthiazol-2-yl)-2,5-Diphenyltetrazolium Bromide (MTT), phosphate buffered saline (PBS) and human fibronectin ELISA kit from Sigma-Aldrich (St Louis, MO, USA); FN inhibitor (RGDS) from Santa Cruz Biotechnology (Santa Cruz, CA, USA); [3H]-gemcitabine from Moravek Biochemicals Inc. (Brea, CA, USA); Ultima Gold from Perkin Elmer (Waltham, MA, USA); PD98059 from Calbiochem (La Jolla, CA); human anti-alpha smooth muscle actin (αSMA; BS66) from Nordic Biosite AB (Taby, Sweden); anti-AKT, anti-phospho-AKT, anti-glial fibrillary acidic protein (GFAP; GA5), anti-epithelial cell adhesion molecule (EpCAM; VU1D9), anti-p44/42 MAPK (ERK1/2), anti-phospho-ERK1/2, anti-vimentin (D21H3) and anti-GAPDH from Cell Signaling Technology (Beverly, MA, USA); anti-phosphoinositol-3-kinases (PI3K) from Merck Life Science AS (Oslo, Norway); secondary HRP-conjugated antibodies goat anti-mouse and goat anti-rabbit IgG from Bio-Rad Laboratories (Hercules, CA, USA); secondary Alexa Fluor-conjugated antibodies (anti-mouse and anti-rabbit) and DAPI from Jackson ImmunoResearch (West Grove, PA, USA).

Cell isolation and culture

Primary human PSCs obtained from tumor tissue sampled from surgical resection specimens with PDAC, were isolated and cultured by the outgrowth method as previously described [29, 30]. Cultures were established and propagated from ten different patients (designated as PSC-1 -2, − 3, − 4, − 5, − 6, − 7, − 8, − 9, and − 10). None of the ten patients received neoadjuvant therapy. Clinicopathological features of tumors are provided in Additional file 1: Table S1. Assessment of morphology and expression analysis of αSMA and vimentin was performed to determine the purity of PSCs. PSC cultures between passage 3 and 8 were used for all experiments. The PSC cultures as well as the commercial PCC lines AsPC-1 (ATCC® CRL-1682™), BxPC-3 (ATCC® CRL-1687™), Capan-2 (ATCC® HTB-80™), HPAF-II (ATCC® CRL-1997™), MIA PaCa-2 (ATCC® CRL-1420™), PANC-1 (ATCC® CRL-1469™) and SW-1990 (ATCC® CRL-2172™) obtained from American Type Culture Collection (ATCC, Manassas, VA, USA) were cultured and maintained in DMEM supplemented with 10% FBS, 1% Pen-Strep and 1% amphotericin B and tested for mycoplasma contamination. None of the PCC lines are listed in the database of the International Cell Line Authentication Committee (ICLAC). For direct co-culture equal numbers of PSCs were seeded together with PCCs, while in indirect co-culture PCCs were incubated with PSC-conditioned medium (PSC-CM), as described in Additional file 2: Figure S1.

Immunocytochemistry

Immunocytochemistry of PSCs was performed as described previously [31]. Briefly, PSCs cultured in 96-well plates were fixed in 4% formaldehyde, blocked in BSA and incubated overnight with primary antibodies against αSMA (1:50) and vimentin (1:200). The next day, cells were stained with Alexa Fluor-conjugated secondary antibodies (1:200) and DAPI was used for nuclear staining. Images were captured using EVOS FLoid Cell Imaging Station (Thermo Fisher Scientific).

Preparation of conditioned medium

The PSC-CM samples were prepared as described previously [31]. Briefly, sub-confluent PSC cultures were washed thoroughly with PBS and incubated for 48 h with fresh serum-free DMEM (SFM), conditioned medium was collected, centrifuged and stored at - 20 °C until further use.

Chemosensitivity

Both PCCs and PSCs were cultured in 96-well plates at a density of 3000 cells/well and treated for 48 h with gemcitabine at a final concentration range of 0.01–100 μM. The cell viability and IC50 values were determined using the MTT assay, in which the degree of formazan crystals formation is relative to the number of viable cells. IC50 values were determined by calculating the amount of gemcitabine required for inhibition of cell growth by 50% compared to untreated controls. Furthermore, PCCs were either cultured together with PSCs for 96 h (direct co-culture) or incubated with PSC-CM (indirect co-culture) for 24 h, prior to treatment with gemcitabine for 48 h at a final concentration of 10 μM. The response to gemcitabine was evaluated with the MTT assay. The PSC-CM induced resistance to gemcitabine in various PCC lines was calculated using the following formula:
$$ \mathrm{Drug}\ \mathrm{resistance}\ \left(\%\right)=\frac{\%\mathrm{viability}\ \mathrm{in}\ \mathrm{PSC}-\mathrm{CM}-\%\mathrm{viability}\ \mathrm{in}\ \mathrm{SFM}}{\%\mathrm{viability}\ \mathrm{in}\ \mathrm{SFM}}\mathrm{X}\ 100 $$

Secretome analysis

Proteomics-based analysis of PSC-CM samples was performed as described previously with some modifications [31]. Briefly, the PSC-CM samples were concentrated down to 5% of the original volume using 10 kDa cut off Amicon Ultra centrifugal filter devices. Subsequently, the proteins were reduced, alkylated and digested overnight using trypsin (Promega). The next day, the peptides were desalted and concentrated before they were submitted to mass spectrometry (MS). Each peptide mixture was analyzed in three technical replicates by nEASY-LC coupled to QExactive Plus (ThermoElectron, Bremen, Germany) with EASY Spray PepMap®RSLC column (C18, 2 μm, 100 Å, 75 μm × 50 cm). Protein identification was performed using Proteome Discoverer 2.1 (Thermo Fisher Scientific) and Mascot 2.6 (MatrixScience, London, UK) search engine. The search criteria for Mascot searches were: trypsin digestion with two missed cleavage allowed, Carbamidomethyl (C) as fixed modification and Acetyl (N-term), Gln- > pyro-Glu (N-term Q), Oxidation (M) as dynamic modifications. The parent mass tolerance was 10 ppm and MS/MS tolerance 0.1 Da. The SwissProt database for human entries supplemented with known contaminants provided by MaxQuant was used for the database searches. All of the reported protein identifications were statistically significant (p < 0.05) in Mascot and filtered in Proteome Discoverer for at least medium confidence identifications. The list of identified proteins were subjected to Kyoto Encyclopedia of Genes and Genomes (KEGG) database for pathway analysis [32] and Gene Ontology (GO) analysis was conducted using the DAVID Bioinformatics Database [33, 34]. The workflow of the procedure is illustrated in Additional file 3: Figure S2A.

ELISA

The amount of soluble FN present in the conditioned medium obtained from ten different PSC cultures was measured using human fibronectin ELISA kit, according to the manufacturer’s protocol.

Western blot analysis

Total cell lysates were prepared using Laemmli buffer, and aliquots of protein were separated by electrophoresis (SDS-PAGE), as described previously [31]. The proteins were transferred to nitro-cellulose membranes, blocked in 5% non-fat dry milk solution and incubated overnight with the primary antibodies as indicated. The next day, blots were washed and incubated with HRP-conjugated secondary antibodies at room temperature for 1 h. The blots were visualized with LumiGLO® (KPL, Gaithersburg, MD, USA) and their densitometric analysis was performed using Labworks Software (UVP, Cambridge, UK).

Gemcitabine uptake

PCCs cultured to confluence in 96-well plates were washed with PBS and incubated with 100 μL transport buffer (20 mM Tris, 3 mM K2HPO4, 5 mM glucose, 145 mM NaCl, 1 mM MgCl2, and 1.2 mM CaCl2) containing 50 nM [3H]-gemcitabine at 37 °C for 4 h. Subsequently, cells were lysed by incubation with 0.2 M NaOH for 15 min and lysates were then added to the scintillation tubes containing Ultima Gold. Cell-associated radioactivity in counts per minute (CPM) was determined using a liquid scintillation counter. Protein concentration of cell lysates was determined using BCA protein assay kit. [3H]-gemcitabine transport was calculated by normalizing CPM to protein concentration for each well.

Statistical analysis

All values are expressed as a mean ± standard error of mean (SEM). Statistical analysis of the results was performed using GraphPad Prism 6 (GraphPad Software), by an unpaired two-tailed Student’s t test or by two-way ANOVA, followed by Tukey’s post hoc test, with a value of p < 0.05 considered statistically significant.

Results

Phenotypic characterization of the human primary PSC cultures

Expression of proteins considered characteristic of PSCs was analyzed by immunofluorescence and western blot analysis. All primary PSC cultures examined exhibited a fibroblast-like morphology that is characteristic of PSCs, and showed strong expression of the activation marker αSMA and the mesenchymal marker vimentin, consistent with activated PSCs (Additional file 4: Figure S3A, B). GFAP was not detected in any of the PSC cultures (data not shown), consistent with the reported loss of expression during culturing [35]. None of the PSC cultures showed expression of the epithelial marker EpCAM (data not shown), excluding the possibility of contamination with epithelial cells during isolation.

Gemcitabine dose response chemosensitivity in PSCs and PCCs

MTT-based evaluation of dose response to gemcitabine in terms of cell viability in four PSC cultures revealed that these cells were resistant to the cytotoxic actions of gemcitabine. At a single concentration of gemcitabine (10 μM) an average reduction of cell viability by merely 12% was observed (Fig. 1a). In contrast, gemcitabine differentially reduced the cell viability in the seven PCC lines studied (Fig. 1b). MTT assay revealed that gemcitabine dose dependently inhibited PCC viability and, at a single concentration of gemcitabine (10 μM), the decrease in cell viability was 50, 70, 52, 66, 49, 41 and 51% in AsPC-1, BxPC-3, Capan-2, HPAF-II, MIA PaCa-2, PANC-1 and SW-1990, respectively (Fig. 1b). Notably, a significant cell population in each PCC line was observed to be inherently resistant to gemcitabine, whereby the size of this subpopulation ranged from 14% of the total cell population in SW-1990 to 33% in AsPC-1 (Fig. 1b). The IC50 values suggested that PANC-1 (IC50 = ~ 20 μM) and BxPC-3 (~ 2 μM) were least and most sensitive to gemcitabine, respectively (Table 1), which is in accordance with previous data [36].
Table 1
IC50 values for gemcitabine in different PCC lines were calculated using GraphPad Prism software 6.0
Cell line
IC50 (μM)
PANC-1
20.5
AsPC-1
13.9
Capan-2
9.7
MIA PaCa-2
9.5
SW-1990
6.2
HPAF-II
4.3
BxPC-3
2.0

Effect of PSCs and their secretome on gemcitabine chemosensitivity of PCCs

Each of the seven different PCCs were each co-cultured with four different PSC cultures for 96 h and evaluated for the response to gemcitabine at single concentration of 10 μM, using the MTT assay. Compared to the cell survival observed in PCCs alone (Fig. 1b), the cancer cells in co-cultures developed a considerable degree of resistance to gemcitabine, as indicated by a high cell survival than in monoculture (Fig. 2). For instance, co-culture with PSC-1 reduced cell viability by a mere 9, 35, 9, 19, 15, 7 and 16% for AsPC-1, BxPC-3, Capan-2, HPAF-II, MIA PaCa-2, PANC-1 and SW-1990, respectively. Similar responses were observed for co-cultures with three further PSC culture (Fig. 2).
To evaluate the effect of PSCs-secreted soluble factors on cancer cell chemosensitivity, PCCs were incubated with PSC-CM for 24 h and analyzed for the response to the cytotoxic effects of gemcitabine using the MTT assay (Fig. 3a). All PCC lines studied developed a certain degree of resistance to gemcitabine, with the highest resistance being observed in HPAF-II (~ 75%). Although PANC-1 and BxPC-3 cells displayed a modest resistance (20–25%), this change was not significant (Fig. 3a and b). Notably, across the cell lines that were studied, co-culture with PSC-CM had no significant effect on the uptake of radiolabeled gemcitabine in PCCs (Fig. 3c).
Of the many important signaling pathways that play a role in the pathobiology of PDAC, the ERK and PI3K/AKT signaling pathways are known to be also implicated in gemcitabine resistance [37, 38]. Substantiating the putative signaling effects of soluble factors in PSC-CM on the PCCs, a variable degree of increased phosphorylation of ERK1/2 was observed in all PCC lines upon incubation with PSC-CM (Fig. 3d), whereas activity of AKT or PI3K remained largely unaltered in the PCCs with the exception of HPAF-II, which displayed an increase in AKT phosphorylation (Fig. 3d).

Secretome analysis of PSC-CM

The composition of conditioned medium from various PSC cultures was investigated by proteomics-based analysis of the secretome preparations. A total of 796 different proteins (Homo sapiens) were identified, after the protein grouping performed by MaxQuant software. A complete list of all proteins together with their identification parameters are provided in Additional file 5: Table S2. The proteins with a number of peptides with high-medium confidence identified was between 200 and 600, across the ten PSC cultures studied (Additional file 3: Figure S2B). The identified proteins mainly belonged to the structural cellular components such as exosomes, extracellular vesicles and membrane-bound vesicles. Furthermore, to facilitate the interpretation of the results, a functional interaction network of the identified proteins was searched using the STRING software. The top five enriched KEGG pathway categories of the identified proteins are listed in Fig. 4a. To determine the functional role of the PSC-secreted proteins, a GO analysis was undertaken using the DAVID bioinformatic resource tool, which revealed a significant representation of biological process categories that were mainly related to ECM structure and organization, cytoskeleton organization, cell motility and adhesion, metabolism, regulation of cell death, and response to stimuli (Fig. 4b). A GO analysis also revealed a significant representation of molecular processes categories that were mainly related to carbohydrate and polysaccharide binding, enzyme activity, and calcium biding (Additional file 3: Figure S2C). GO functional annotation chart for KEGG analysis is presented in Additional file 6: Table S3. Furthermore, a total of 47 of all the identified proteins that are involved in ECM remodeling (Table 2), were grouped based on molecular functions as indicated (Fig. 4c).
Table 2
List of ECM remodeling proteins identified in PSC secretome
Accession
Protein description
Gene symbol
Molecular weight [kDa]
Coverage
P02751
Fibronectin
FN1
262.5
66.7
P08123
Collagen alpha-2(I) chain
COL1A2
129.2
82.3
P02452
Collagen alpha-1(I) chain
COL1A1
138.9
81.1
P02461
Collagen alpha-1(III) chain
COL3A1
138.5
55.3
P12109
Collagen alpha-1(VI) chain
COL6A1
108.5
61.1
P35555
Fibrillin-1
FBN1
312.0
44.7
P12111
Collagen alpha-3(VI) chain
COL6A3
343.5
48.1
P08253
72 kDa type IV collagenase
MMP2
73.8
73.5
Q99715
Collagen alpha-1(XII) chain
COL12A1
332.9
35.0
P11047
Laminin subunit gamma-1
LAMC1
177.5
38.1
P98160
Heparan sulfate proteoglycan core protein
HSPG2
468.5
26.3
P08572
Collagen alpha-2(IV) chain
COL4A2
167.4
41.1
P03956
Interstitial collagenase
MMP1
54.0
65.9
P05997
Collagen alpha-2(V) chain
COL5A2
144.8
50.9
P01033
Metalloproteinase inhibitor 1
TIMP1
23.2
58.9
P20908
Collagen alpha-1(V) chain
COL5A1
183.4
21.0
P07585
Decorin
DCN
39.7
54.3
P12110
Collagen alpha-2(VI) chain
COL6A2
108.5
44.2
P16035
Metalloproteinase inhibitor 2
TIMP2
24.4
67.7
P07942
Laminin subunit beta-1
LAMB1
197.9
32.7
P02462
Collagen alpha-1(IV) chain
COL4A1
160.5
15.5
P14543
Nidogen-1
NID1
136.3
23.4
P12107
Collagen alpha-1(XI) chain
COL11A1
181.0
14.2
P01023
Alpha-2-macroglobulin
A2M
163.2
33.2
P07858
Cathepsin B
CTSB
37.8
46.3
P13497
Bone morphogenetic protein 1
BMP1
111.2
26.7
P16112
Aggrecan core protein
ACAN
250.0
10.2
P16070
CD44 antigen
CD44
81.5
6.3
Q13443
Disintegrin and metalloproteinase domain-containing protein 9
ADAM9
90.5
5.5
P13796
Plastin-2
LCP1
70.2
12.8
Q07092
Collagen alpha-1(XVI) chain
COL16A1
157.7
5.9
P27658
Collagen alpha-1(VIII) chain
COL8A1
73.3
18.1
P07339
Cathepsin D
CTSD
44.5
38.3
P08254
Stromelysin-1
MMP3
53.9
24.1
P35556
Fibrillin-2
FBN2
314.6
4.1
Q05707
Collagen alpha-1(XIV) chain
COL14A1
193.4
9.4
Q13751
Laminin subunit beta-3
LAMB3
129.5
8.7
Q13753
Laminin subunit gamma-2
LAMC2
130.9
8.5
O14672
Disintegrin and metalloproteinase domain-containing protein 10
ADAM10
84.1
15.1
P07711
Cathepsin L1
CTSL1
37.5
20.4
P10451
Osteopontin
SPP1
35.4
19.1
P17813
Endoglin
ENG
70.5
8.2
Q16787
Laminin subunit alpha-3
LAMA3
366.4
2.8
P39059
Collagen alpha-1(XV) chain
COL15A1
141.6
6.2
P39060
Collagen alpha-1(XVIII) chain
COL18A1
178.1
5.2
P25940
Collagen alpha-3(V) chain
COL5A3
172.0
2.7
P43235
Cathepsin K
CTSK
36.9
5.5

Effect of PSC-secreted FN on PCC chemoresistance

In the PSC secretome, FN was detected with one of the highest number of peptides. The levels of soluble FN in conditioned medium from the ten PSCs cultures were in the range 175–350 ng/ml (Fig. 5a). Western blot analysis revealed that only the PSCs expressed FN, whereas all the PCC lines studied were negative (Fig. 5b), which is in accordance with previous findings [39]. Gemcitabine uptake capacity was not affected by FN-coating in any of the PCC lines studied (Fig. 5c). To evaluate the effect of FN on chemoresistance development, PCCs were cultured on FN-coated plates, and all cell lines with the exception of BxPC-3, displayed partial loss of the chemosensitivity to gemcitabine (Fig. 6a and b). However, the observed effect was considerably weaker than that of co-culture with PSC-CM (Fig. 3). To evaluate the effect of collagen - another major PSC-CM component – on chemoresistance development, PCCs were cultured on collagen-coated plates, which showed that none of the cell lines displayed reduction or loss of chemosensitivity to gemcitabine (Additional file 7: Figure S4). Furthermore, PCCs pre-incubated with PSC-CM and further incubated with RGDS, a FN inhibitor (H-Arg-Gly-Asp-Ser-OH), regained sensitivity to gemcitabine to some extent (Fig. 6a and b). Interestingly, in all PCC lines, treatment with RGDS also reversed the increase in phosphorylation of ERK1/2 to basal levels that had occurred during co-culture with PSC-CM (Fig. 6c). These findings were further confirmed by treatment with PD98059, a MEK/ERK inhibitor, which resulted in regained sensitivity to gemcitabine to an extent similar to that of RGDS (Additional file 8: Figure S5).

Discussion

Crosstalk between mesenchymal stromal cells and epithelial cancer cells has recently been implicated in the development of chemoresistance in pancreatic cancer. In particular, increasing evidence suggest that PSCs in the tumor stroma are both direct and indirect drivers of pancreatic cancer chemoresistance [23]. Since the 1997 report by Burris et al. [8], gemcitabine has remained a cornerstone of pancreatic cancer chemotherapy despite only marginal effects on patient survival, to a large extent caused by the high intrinsic (de novo) and acquired (therapy-induced) chemoresistance [40]. Thus, knowledge about the underlying mechanisms of drug resistance in PDAC is critical in attempts to develop new and effective treatment regimens for this lethal condition.
In the present study we performed a proteomics-based analysis of the PSC secretome to identify PSC-secreted factors that possibly affect PCCs in signaling pathways involved in the development of resistance to gemcitabine. The analysis resulted in identification of 796 proteins, the majority of which were ECM related proteins including FN and collagens. When cultured on FN-coated surface, all PCC lines tested displayed to a varying degree an increased resistance to gemcitabine, suggesting that ECM proteins are implicated in resistance-induction, in accordance with a previous report by Miyamoto et al. [28]. We observed that both in indirect and direct co-culture, PSCs induced a varying degree of gemcitabine resistance in all PCC lines tested. Notably, a synthetic FN inhibitor (RGDS peptide, which blocks integrin receptor function) counteracted to a limited extent resistance-induction by the PSCs, suggesting that PSC-secreted FN plays a key role in the development of gemcitabine resistance. However, the effect of PSC-CM on the development of chemoresistance was significantly higher compared to the effect of FN alone, which suggests that secretome components other than FN may also contribute to chemoresistance to gemcitabine.
Several tumor-modifying interactions between PSCs and PCCs have been reported in the literature, including effects of PSCs on cancer cell proliferation, migration and invasion as well as resistance to chemotherapeutic agents [10, 35, 4144]. The panel of PCCs examined in the present study differ in their sensitivity to gemcitabine, as the IC50 for gemcitabine was approximately ten times higher in the relatively resistant PANC-1 cells compared to the most sensitive BxPC-3 cells. Upon direct co-culture with the tumor-derived PSCs, all the PCC lines examined became relatively resistant, albeit to a variable degree, indicating possible cell-line specific effects. In this context it should be recognized that the cancer cells differ in their mutation status; notably the BxPC-3 cell line is the only one with wild type KRAS [45]. Although several mechanisms could conceivably account for the observed loss of sensitivity to gemcitabine during direct co-culture, effects of PSC-secreted components on cancer cells seems an obvious possibility. This was substantiated with the findings from indirect co-culture experiments in which incubation of PCCs with PSC-CM resulted in the development of varying degrees of resistance to gemcitabine across all cell lines examined. Notably, direct co-culture experiments also suggest that, despite the differences in origin (tumor biopsies from different patients), PSCs modulated the chemosensitivity to gemcitabine of neighboring cancer cells to a similar extent.
Shortcomings of PDAC chemotherapy have been ascribed to poor drug availability as well as stromal factors with the ability to induce chemoresistance. Intracellular uptake of gemcitabine requires nucleoside transporters hENTs and hCNTs, and their expression correlate with gemcitabine sensitivity and overall survival among gemcitabine-treated PDAC patients. Loss of nucleoside transporters leads to development of resistance to gemcitabine [10, 39, 46, 47]. In the present study, neither PSC-CM nor FN had any significant effect on the overall gemcitabine uptake, suggesting that the observed loss of chemosensitivity may not be an effect of PSCs on the gemcitabine uptake capacity of PCCs. Furthermore, KEGG pathways analysis of the PSC secretome indicated that PSC secretion of FN could be a major determinant for induction of gemcitabine chemoresistance. Indeed, FN is a major ECM component that is primarily secreted by fibroblasts and cancer-associated fibroblasts [48, 49], and most of the biological effects of FN in as diverse processes as embryogenesis, wound healing and tissue homeostasis [48, 5053] are mediated through integrins. The latter are a family of heterodimeric and transmembrane receptors that lead to activation of several signal transduction pathways, including ERK, p38 and PI3K/AKT [54, 55].
Both the extracellular signal-regulated kinase (ERK) pathway and the PI3K/AKT pathway are known to be important in the pathobiology of PDAC [56, 57] and also implicated in gemcitabine chemoresistance [37, 38]. High ERK1/2 activity has also been reported to contribute to the gemcitabine resistance in pancreatic cancer, by protecting tumor cells from chemotherapy-induced apoptosis [58, 59]. In this study, incubation of PCCs with PSC-CM-induced an increase in phosphorylation of ERK1/2, and this effect was counteracted by the synthetic FN inhibitor, suggesting that of many possible signaling pathways, PSCs mediate the resistance to gemcitabine via ERK1/2 activity. This was further confirmed by treatment with PD98059, a MEK/ERK inhibitor, which resulted in regained chemosensitivity for gemcitabine in PCCs. Notably, FN displayed profound inhibitory effects on ERK signaling whereas the gemcitabine sensitivity was not inhibited to the same extent, suggesting that other mechanisms or signaling pathways also might be involved. The PI3K/AKT pathway has been reported to be linked to anti-apoptotic signal transduction and chemoresistance in the PCC lines PK1 and PK8 as well as in an orthotopic xenograft model from the same cells [60, 61]. However, it was later reported by Arlt et al. [62], that no such role of PI3K/AKT was found in a study employing BxPc-3, Capan-1 and PancTu-1 cells. In the present study, no change in activity of AKT or PI3K was observed in response to PSC-CM across the seven different PCCs examined. A previous study by Sawai et al. [63] showed that the basement membrane proteins FN and collagen IV induced invasion of pancreatic cancer cells via increased ERK1/2 phosphorylation and that this effect was inhibited by RGDS peptide and β1-integrin antibody as well as by the MEK/ERK inhibitor PD98059. Thus, the results of the present study are consistent with the notion that fibronectin-integrin mediated ERK signaling is involved in pancreatic cancer cell proliferation and survival.
In addition to FN, other matrix components, particularly collagens, may contribute to stroma-induced chemoresistance [64]. High levels of expression of collagen genes were associated with drug resistance in ovarian and breast cancer cell lines [65, 66]. Knowledge regarding the role of different collagens in chemoresistance in pancreatic cancer is very limited. PANC-1 cells cultured in 3D collagen I have been reported to show increased gemcitabine resistance due to increased histone acetylation, also possibly affecting gene expression through activation of PI3K/AKT and ERK signaling pathways, leading to increased proliferation despite drug treatment [27, 67].
Despite recent advances in combination therapies, gemcitabine remains a cornerstone of neo-adjuvant, adjuvant, and palliative therapy for advanced PDAC [68]. A better understanding of the causes of gemcitabine chemoresistance, including the role of the tumor stroma, is critical to the development of novel treatment strategies. It seems obvious that developing new therapies for PDAC is a much more complex challenge than merely targeting the cancer cells alone. Recent studies in genetically modified mouse models and clinical trials targeting PSCs have shown conflicting results, with both positive and negative effects on cancer progression and responses to gemcitabine [69]. Understanding the aspects of PSC biology that promote therapy resistance and identifying the relevant signaling mechanisms are important steps in the development of effective stroma-targeting treatment strategies.

Conclusions

In conclusion, the findings of the present study suggest that PSC-secreted FN promotes high ERK1/2 activity in cancer cells and thereby protects the PCCs from gemcitabine-induced cytotoxicity (Fig. 7). Combined use of an FN inhibitor and gemcitabine might result in reduced chemoresistance to gemcitabine. Further studies are needed to elucidate the mechanisms by which FN blocking agents added to gemcitabine-based chemotherapy might counteract chemoresistance in PDAC thereby providing better clinical outcomes.

Acknowledgements

Mass spectrometry (secretome) analysis was performed at the Proteomics Core Facility, Department of Immunology, University of Oslo and Oslo University hospital, Norway.
The study protocol and patient consent documents were approved by the Regional Committee for Medical and Health Research Ethics (REC South East, project number 2015/738) and followed the Helsinki Declaration. Written informed consent was obtained from all study participants, whose tumor tissue was used for the study. Commercial cancer cell lines were purchased from ATCC, and did not require ethics approval for their use in this study.
Not applicable.

Competing interests

The authors declare that they have no competing interests.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Anhänge

Additional files

Literatur
1.
2.
Zurück zum Zitat Zijlstra M, Bernards N, de Hingh IH, van de Wouw AJ, Goey SH, Jacobs EM, et al. Does long-term survival exist in pancreatic adenocarcinoma? Acta Oncol. 2016;55(3):259–64.PubMedCrossRef Zijlstra M, Bernards N, de Hingh IH, van de Wouw AJ, Goey SH, Jacobs EM, et al. Does long-term survival exist in pancreatic adenocarcinoma? Acta Oncol. 2016;55(3):259–64.PubMedCrossRef
3.
Zurück zum Zitat Ying H, Dey P, Yao W, Kimmelman AC, Draetta GF, Maitra A, et al. Genetics and biology of pancreatic ductal adenocarcinoma. Genes Dev. 2016;30(4):355–85.PubMedPubMedCentralCrossRef Ying H, Dey P, Yao W, Kimmelman AC, Draetta GF, Maitra A, et al. Genetics and biology of pancreatic ductal adenocarcinoma. Genes Dev. 2016;30(4):355–85.PubMedPubMedCentralCrossRef
4.
Zurück zum Zitat Kleeff J, Korc M, Apte M, La Vecchia C, Johnson CD, Biankin AV, et al. Pancreatic cancer. Nat Rev Dis Primers. 2016;2:16022.PubMedCrossRef Kleeff J, Korc M, Apte M, La Vecchia C, Johnson CD, Biankin AV, et al. Pancreatic cancer. Nat Rev Dis Primers. 2016;2:16022.PubMedCrossRef
5.
Zurück zum Zitat Rahib L, Smith BD, Aizenberg R, Rosenzweig AB, Fleshman JM, Matrisian LM. Projecting cancer incidence and deaths to 2030: the unexpected burden of thyroid, liver, and pancreas cancers in the United States. Cancer Res. 2014;74(11):2913–21.CrossRef Rahib L, Smith BD, Aizenberg R, Rosenzweig AB, Fleshman JM, Matrisian LM. Projecting cancer incidence and deaths to 2030: the unexpected burden of thyroid, liver, and pancreas cancers in the United States. Cancer Res. 2014;74(11):2913–21.CrossRef
6.
Zurück zum Zitat Labori KJ, Katz MH, Tzeng CW, Bjornbeth BA, Cvancarova M, Edwin B, et al. Impact of early disease progression and surgical complications on adjuvant chemotherapy completion rates and survival in patients undergoing the surgery first approach for resectable pancreatic ductal adenocarcinoma - a population-based cohort study. Acta Oncol. 2016;55(3):265–77.PubMedCrossRef Labori KJ, Katz MH, Tzeng CW, Bjornbeth BA, Cvancarova M, Edwin B, et al. Impact of early disease progression and surgical complications on adjuvant chemotherapy completion rates and survival in patients undergoing the surgery first approach for resectable pancreatic ductal adenocarcinoma - a population-based cohort study. Acta Oncol. 2016;55(3):265–77.PubMedCrossRef
7.
Zurück zum Zitat Neoptolemos JP, Kleeff J, Michl P, Costello E, Greenhalf W, Palmer DH. Therapeutic developments in pancreatic cancer: current and future perspectives. Nat Rev Gastroenterol Hepatol. 2018;15(6):333–48.PubMedCrossRef Neoptolemos JP, Kleeff J, Michl P, Costello E, Greenhalf W, Palmer DH. Therapeutic developments in pancreatic cancer: current and future perspectives. Nat Rev Gastroenterol Hepatol. 2018;15(6):333–48.PubMedCrossRef
8.
Zurück zum Zitat Burris HA, Moore MJ, Andersen J, Green MR, Rothenberg ML, Modiano MR, et al. Improvements in survival and clinical benefit with gemcitabine as first-line therapy for patients with advanced pancreas cancer: a randomized trial. J Clin Oncol. 1997;15(6):2403–13.PubMedCrossRef Burris HA, Moore MJ, Andersen J, Green MR, Rothenberg ML, Modiano MR, et al. Improvements in survival and clinical benefit with gemcitabine as first-line therapy for patients with advanced pancreas cancer: a randomized trial. J Clin Oncol. 1997;15(6):2403–13.PubMedCrossRef
9.
Zurück zum Zitat Wong A, Soo RA, Yong WP, Innocenti F. Clinical pharmacology and pharmacogenetics of gemcitabine. Drug Metab Rev. 2009;41(2):77–88.PubMedCrossRef Wong A, Soo RA, Yong WP, Innocenti F. Clinical pharmacology and pharmacogenetics of gemcitabine. Drug Metab Rev. 2009;41(2):77–88.PubMedCrossRef
11.
Zurück zum Zitat Kim MP, Gallick GE. Gemcitabine resistance in pancreatic cancer: picking the key players. Clin Cancer Res. 2008;14(5):1284–5.PubMedCrossRef Kim MP, Gallick GE. Gemcitabine resistance in pancreatic cancer: picking the key players. Clin Cancer Res. 2008;14(5):1284–5.PubMedCrossRef
12.
Zurück zum Zitat Neesse A, Michl P, Frese KK, Feig C, Cook N, Jacobetz MA, et al. Stromal biology and therapy in pancreatic cancer. Gut. 2011;60(6):861–8.PubMedCrossRef Neesse A, Michl P, Frese KK, Feig C, Cook N, Jacobetz MA, et al. Stromal biology and therapy in pancreatic cancer. Gut. 2011;60(6):861–8.PubMedCrossRef
13.
Zurück zum Zitat Kadaba R, Birke H, Wang J, Hooper S, Andl CD, Di Maggio F, et al. Imbalance of desmoplastic stromal cell numbers drives aggressive cancer processes. J Pathol. 2013;230(1):107–17.PubMedPubMedCentralCrossRef Kadaba R, Birke H, Wang J, Hooper S, Andl CD, Di Maggio F, et al. Imbalance of desmoplastic stromal cell numbers drives aggressive cancer processes. J Pathol. 2013;230(1):107–17.PubMedPubMedCentralCrossRef
14.
Zurück zum Zitat Lunardi S, Muschel RJ, Brunner TB. The stromal compartments in pancreatic cancer: are there any therapeutic targets? Cancer Lett. 2014;343(2):147–55.PubMedCrossRef Lunardi S, Muschel RJ, Brunner TB. The stromal compartments in pancreatic cancer: are there any therapeutic targets? Cancer Lett. 2014;343(2):147–55.PubMedCrossRef
15.
Zurück zum Zitat Topalovski M, Brekken RA. Matrix control of pancreatic cancer: new insights into fibronectin signaling. Cancer Lett. 2016;381(1):252–8.PubMedCrossRef Topalovski M, Brekken RA. Matrix control of pancreatic cancer: new insights into fibronectin signaling. Cancer Lett. 2016;381(1):252–8.PubMedCrossRef
16.
Zurück zum Zitat Grzesiak JJ, Ho JC, Moossa AR, Bouvet M. The integrin-extracellular matrix axis in pancreatic cancer. Pancreas. 2007;35(4):293–301.PubMedCrossRef Grzesiak JJ, Ho JC, Moossa AR, Bouvet M. The integrin-extracellular matrix axis in pancreatic cancer. Pancreas. 2007;35(4):293–301.PubMedCrossRef
17.
Zurück zum Zitat Pothula SP, Xu Z, Goldstein D, Pirola RC, Wilson JS, Apte MV. Key role of pancreatic stellate cells in pancreatic cancer. Cancer Lett. 2016;381(1):194–200.PubMedCrossRef Pothula SP, Xu Z, Goldstein D, Pirola RC, Wilson JS, Apte MV. Key role of pancreatic stellate cells in pancreatic cancer. Cancer Lett. 2016;381(1):194–200.PubMedCrossRef
19.
Zurück zum Zitat Apte MV, Wilson JS. Dangerous liaisons: pancreatic stellate cells and pancreatic cancer cells. J Gastroenterol Hepatol. 2012;27(Suppl 2):69–74.PubMedCrossRef Apte MV, Wilson JS. Dangerous liaisons: pancreatic stellate cells and pancreatic cancer cells. J Gastroenterol Hepatol. 2012;27(Suppl 2):69–74.PubMedCrossRef
20.
Zurück zum Zitat Liang C, Shi S, Meng Q, Liang D, Ji S, Zhang B, et al. Complex roles of the stroma in the intrinsic resistance to gemcitabine in pancreatic cancer: where we are and where we are going. Exp Mol Med. 2017;49(12):e406.PubMedPubMedCentralCrossRef Liang C, Shi S, Meng Q, Liang D, Ji S, Zhang B, et al. Complex roles of the stroma in the intrinsic resistance to gemcitabine in pancreatic cancer: where we are and where we are going. Exp Mol Med. 2017;49(12):e406.PubMedPubMedCentralCrossRef
21.
Zurück zum Zitat Arumugam T, Ramachandran V, Fournier KF, Wang H, Marquis L, Abbruzzese JL, et al. Epithelial to mesenchymal transition contributes to drug resistance in pancreatic cancer. Cancer Res. 2009;69(14):5820–8.PubMedPubMedCentralCrossRef Arumugam T, Ramachandran V, Fournier KF, Wang H, Marquis L, Abbruzzese JL, et al. Epithelial to mesenchymal transition contributes to drug resistance in pancreatic cancer. Cancer Res. 2009;69(14):5820–8.PubMedPubMedCentralCrossRef
22.
Zurück zum Zitat Toste PA, Nguyen AH, Kadera BE, Duong M, Wu N, Gawlas I, et al. Chemotherapy-induced inflammatory gene signature and Protumorigenic phenotype in pancreatic CAFs via stress-associated MAPK. Mol Cancer Res. 2016;14(5):437–47.PubMedPubMedCentralCrossRef Toste PA, Nguyen AH, Kadera BE, Duong M, Wu N, Gawlas I, et al. Chemotherapy-induced inflammatory gene signature and Protumorigenic phenotype in pancreatic CAFs via stress-associated MAPK. Mol Cancer Res. 2016;14(5):437–47.PubMedPubMedCentralCrossRef
23.
Zurück zum Zitat McCarroll JA, Naim S, Sharbeen G, Russia N, Lee J, Kavallaris M, et al. Role of pancreatic stellate cells in chemoresistance in pancreatic cancer. Front Physiol. 2014;5:141.PubMedPubMedCentral McCarroll JA, Naim S, Sharbeen G, Russia N, Lee J, Kavallaris M, et al. Role of pancreatic stellate cells in chemoresistance in pancreatic cancer. Front Physiol. 2014;5:141.PubMedPubMedCentral
24.
Zurück zum Zitat Zhang H, Wu H, Guan J, Wang L, Ren X, Shi X, et al. Paracrine SDF-1α signaling mediates the effects of PSCs on GEM chemoresistance through an IL-6 autocrine loop in pancreatic cancer cells. Oncotarget. 2015;6(5):3085–97.PubMed Zhang H, Wu H, Guan J, Wang L, Ren X, Shi X, et al. Paracrine SDF-1α signaling mediates the effects of PSCs on GEM chemoresistance through an IL-6 autocrine loop in pancreatic cancer cells. Oncotarget. 2015;6(5):3085–97.PubMed
25.
Zurück zum Zitat Liu Y, Li F, Gao F, Xing L, Qin P, Liang X, et al. Periostin promotes the chemotherapy resistance to gemcitabine in pancreatic cancer. Tumour Biol. 2016;37(11):15283–91.PubMedCrossRef Liu Y, Li F, Gao F, Xing L, Qin P, Liang X, et al. Periostin promotes the chemotherapy resistance to gemcitabine in pancreatic cancer. Tumour Biol. 2016;37(11):15283–91.PubMedCrossRef
26.
Zurück zum Zitat Zhao X, Fan W, Xu Z, Chen H, He Y, Yang G, et al. Inhibiting tumor necrosis factor-alpha diminishes desmoplasia and inflammation to overcome chemoresistance in pancreatic ductal adenocarcinoma. Oncotarget. 2016;7(49):81110–22.PubMedPubMedCentralCrossRef Zhao X, Fan W, Xu Z, Chen H, He Y, Yang G, et al. Inhibiting tumor necrosis factor-alpha diminishes desmoplasia and inflammation to overcome chemoresistance in pancreatic ductal adenocarcinoma. Oncotarget. 2016;7(49):81110–22.PubMedPubMedCentralCrossRef
27.
Zurück zum Zitat Dangi-Garimella S, Sahai V, Ebine K, Kumar K, Munshi HG. Three-dimensional collagen I promotes gemcitabine resistance in vitro in pancreatic cancer cells through HMGA2-dependent histone acetyltransferase expression. PLoS One. 2013;8(5):e64566.PubMedPubMedCentralCrossRef Dangi-Garimella S, Sahai V, Ebine K, Kumar K, Munshi HG. Three-dimensional collagen I promotes gemcitabine resistance in vitro in pancreatic cancer cells through HMGA2-dependent histone acetyltransferase expression. PLoS One. 2013;8(5):e64566.PubMedPubMedCentralCrossRef
28.
Zurück zum Zitat Miyamoto H, Murakami T, Tsuchida K, Sugino H, Miyake H, Tashiro S. Tumor-stroma interaction of human pancreatic cancer: acquired resistance to anticancer drugs and proliferation regulation is dependent on extracellular matrix proteins. Pancreas. 2004;28(1):38–44.PubMedCrossRef Miyamoto H, Murakami T, Tsuchida K, Sugino H, Miyake H, Tashiro S. Tumor-stroma interaction of human pancreatic cancer: acquired resistance to anticancer drugs and proliferation regulation is dependent on extracellular matrix proteins. Pancreas. 2004;28(1):38–44.PubMedCrossRef
29.
Zurück zum Zitat Pomianowska E, Sandnes D, Grzyb K, Schjolberg AR, Aasrum M, Tveteraas IH, et al. Inhibitory effects of prostaglandin E2 on collagen synthesis and cell proliferation in human stellate cells from pancreatic head adenocarcinoma. BMC Cancer. 2014;14:413.PubMedPubMedCentralCrossRef Pomianowska E, Sandnes D, Grzyb K, Schjolberg AR, Aasrum M, Tveteraas IH, et al. Inhibitory effects of prostaglandin E2 on collagen synthesis and cell proliferation in human stellate cells from pancreatic head adenocarcinoma. BMC Cancer. 2014;14:413.PubMedPubMedCentralCrossRef
30.
Zurück zum Zitat Bachem MG, Schneider E, Gross H, Weidenbach H, Schmid RM, Menke A, et al. Identification, culture, and characterization of pancreatic stellate cells in rats and humans. Gastroenterology. 1998;115(2):421–32.PubMedCrossRef Bachem MG, Schneider E, Gross H, Weidenbach H, Schmid RM, Menke A, et al. Identification, culture, and characterization of pancreatic stellate cells in rats and humans. Gastroenterology. 1998;115(2):421–32.PubMedCrossRef
33.
Zurück zum Zitat Dennis G Jr, Sherman BT, Hosack DA, Yang J, Gao W, Lane HC, et al. DAVID: database for annotation, visualization, and integrated discovery. Genome Biol. 2003;4(5):P3.PubMedCrossRef Dennis G Jr, Sherman BT, Hosack DA, Yang J, Gao W, Lane HC, et al. DAVID: database for annotation, visualization, and integrated discovery. Genome Biol. 2003;4(5):P3.PubMedCrossRef
34.
Zurück zum Zitat Huang d W, Sherman BT, Lempicki RA. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat Protoc. 2009;4(1):44–57.CrossRef Huang d W, Sherman BT, Lempicki RA. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat Protoc. 2009;4(1):44–57.CrossRef
35.
Zurück zum Zitat Tjomsland V, Aasrum M, Christoffersen T, Gladhaug IP. Functional heterogeneity in tumor-derived human pancreatic stellate cells: differential expression of HGF and implications for mitogenic signaling and migration in pancreatic cancer cells. Oncotarget. 2017;8(42):71672–84.PubMedPubMedCentralCrossRef Tjomsland V, Aasrum M, Christoffersen T, Gladhaug IP. Functional heterogeneity in tumor-derived human pancreatic stellate cells: differential expression of HGF and implications for mitogenic signaling and migration in pancreatic cancer cells. Oncotarget. 2017;8(42):71672–84.PubMedPubMedCentralCrossRef
36.
Zurück zum Zitat Pan X, Arumugam T, Yamamoto T, Levin PA, Ramachandran V, Ji B, et al. Nuclear factor-kappaB p65/relA silencing induces apoptosis and increases gemcitabine effectiveness in a subset of pancreatic cancer cells. Clin Cancer Res. 2008;14(24):8143–51.PubMedPubMedCentralCrossRef Pan X, Arumugam T, Yamamoto T, Levin PA, Ramachandran V, Ji B, et al. Nuclear factor-kappaB p65/relA silencing induces apoptosis and increases gemcitabine effectiveness in a subset of pancreatic cancer cells. Clin Cancer Res. 2008;14(24):8143–51.PubMedPubMedCentralCrossRef
37.
Zurück zum Zitat Fryer RA, Barlett B, Galustian C, Dalgleish AG. Mechanisms underlying gemcitabine resistance in pancreatic cancer and sensitisation by the iMiD lenalidomide. Anticancer Res. 2011;31(11):3747–56.PubMed Fryer RA, Barlett B, Galustian C, Dalgleish AG. Mechanisms underlying gemcitabine resistance in pancreatic cancer and sensitisation by the iMiD lenalidomide. Anticancer Res. 2011;31(11):3747–56.PubMed
38.
Zurück zum Zitat Kim R, Yamauchi T, Husain K, Sebti S, Malafa M. Triciribine phosphate monohydrate, an AKT inhibitor, enhances gemcitabine activity in pancreatic Cancer cells. Anticancer Res. 2015;35(9):4599–604.PubMed Kim R, Yamauchi T, Husain K, Sebti S, Malafa M. Triciribine phosphate monohydrate, an AKT inhibitor, enhances gemcitabine activity in pancreatic Cancer cells. Anticancer Res. 2015;35(9):4599–604.PubMed
39.
Zurück zum Zitat Hesler RA, Huang JJ, Starr MD, Treboschi VM, Bernanke AG, Nixon AB, et al. TGF-β-induced stromal CYR61 promotes resistance to gemcitabine in pancreatic ductal adenocarcinoma through downregulation of the nucleoside transporters hENT1 and hCNT3. Carcinogenesis. 2016;37(11):1041–51.PubMedPubMedCentralCrossRef Hesler RA, Huang JJ, Starr MD, Treboschi VM, Bernanke AG, Nixon AB, et al. TGF-β-induced stromal CYR61 promotes resistance to gemcitabine in pancreatic ductal adenocarcinoma through downregulation of the nucleoside transporters hENT1 and hCNT3. Carcinogenesis. 2016;37(11):1041–51.PubMedPubMedCentralCrossRef
40.
Zurück zum Zitat Duxbury MS, Ito H, Zinner MJ, Ashley SW, Whang EE. Inhibition of SRC tyrosine kinase impairs inherent and acquired gemcitabine resistance in human pancreatic adenocarcinoma cells. Clin Cancer Res. 2004;10(7):2307–18.PubMedCrossRef Duxbury MS, Ito H, Zinner MJ, Ashley SW, Whang EE. Inhibition of SRC tyrosine kinase impairs inherent and acquired gemcitabine resistance in human pancreatic adenocarcinoma cells. Clin Cancer Res. 2004;10(7):2307–18.PubMedCrossRef
41.
Zurück zum Zitat Wilson JS, Pirola RC, Apte MV. Stars and stripes in pancreatic cancer: role of stellate cells and stroma in cancer progression. Front Physiol. 2014;5:52.PubMedPubMedCentralCrossRef Wilson JS, Pirola RC, Apte MV. Stars and stripes in pancreatic cancer: role of stellate cells and stroma in cancer progression. Front Physiol. 2014;5:52.PubMedPubMedCentralCrossRef
42.
Zurück zum Zitat Masamune A, Watanabe T, Kikuta K, Shimosegawa T. Roles of pancreatic stellate cells in pancreatic inflammation and fibrosis. Clin Gastroenterol Hepatol. 2009;7(11 Suppl):S48–54.PubMedCrossRef Masamune A, Watanabe T, Kikuta K, Shimosegawa T. Roles of pancreatic stellate cells in pancreatic inflammation and fibrosis. Clin Gastroenterol Hepatol. 2009;7(11 Suppl):S48–54.PubMedCrossRef
43.
Zurück zum Zitat Vonlaufen A, Joshi S, Qu C, Phillips PA, Xu Z, Parker NR, et al. Pancreatic stellate cells: partners in crime with pancreatic cancer cells. Cancer Res. 2008;68(7):2085–93.PubMedCrossRef Vonlaufen A, Joshi S, Qu C, Phillips PA, Xu Z, Parker NR, et al. Pancreatic stellate cells: partners in crime with pancreatic cancer cells. Cancer Res. 2008;68(7):2085–93.PubMedCrossRef
44.
Zurück zum Zitat Zhang H, Wu H, Guan J, Wang L, Ren X, Shi X, et al. Paracrine SDF-1alpha signaling mediates the effects of PSCs on GEM chemoresistance through an IL-6 autocrine loop in pancreatic cancer cells. Oncotarget. 2015;6(5):3085–97.PubMed Zhang H, Wu H, Guan J, Wang L, Ren X, Shi X, et al. Paracrine SDF-1alpha signaling mediates the effects of PSCs on GEM chemoresistance through an IL-6 autocrine loop in pancreatic cancer cells. Oncotarget. 2015;6(5):3085–97.PubMed
45.
Zurück zum Zitat Sipos B, Moser S, Kalthoff H, Torok V, Lohr M, Kloppel G. A comprehensive characterization of pancreatic ductal carcinoma cell lines: towards the establishment of an in vitro research platform. Virchows Arch. 2003;442(5):444–52.PubMed Sipos B, Moser S, Kalthoff H, Torok V, Lohr M, Kloppel G. A comprehensive characterization of pancreatic ductal carcinoma cell lines: towards the establishment of an in vitro research platform. Virchows Arch. 2003;442(5):444–52.PubMed
46.
Zurück zum Zitat de Sousa Cavalcante L, Monteiro G. Gemcitabine: metabolism and molecular mechanisms of action, sensitivity and chemoresistance in pancreatic cancer. Eur J Pharmacol. 2014;741:8–16.PubMedCrossRef de Sousa Cavalcante L, Monteiro G. Gemcitabine: metabolism and molecular mechanisms of action, sensitivity and chemoresistance in pancreatic cancer. Eur J Pharmacol. 2014;741:8–16.PubMedCrossRef
47.
Zurück zum Zitat Achiwa H, Oguri T, Sato S, Maeda H, Niimi T, Ueda R. Determinants of sensitivity and resistance to gemcitabine: the roles of human equilibrative nucleoside transporter 1 and deoxycytidine kinase in non-small cell lung cancer. Cancer Sci. 2004;95(9):753–7.PubMedCrossRef Achiwa H, Oguri T, Sato S, Maeda H, Niimi T, Ueda R. Determinants of sensitivity and resistance to gemcitabine: the roles of human equilibrative nucleoside transporter 1 and deoxycytidine kinase in non-small cell lung cancer. Cancer Sci. 2004;95(9):753–7.PubMedCrossRef
48.
49.
Zurück zum Zitat Erdogan B, Ao M, White LM, Means AL, Brewer BM, Yang L, et al. Cancer-associated fibroblasts promote directional cancer cell migration by aligning fibronectin. J Cell Biol. 2017;216(11):3799–816.PubMedPubMedCentralCrossRef Erdogan B, Ao M, White LM, Means AL, Brewer BM, Yang L, et al. Cancer-associated fibroblasts promote directional cancer cell migration by aligning fibronectin. J Cell Biol. 2017;216(11):3799–816.PubMedPubMedCentralCrossRef
50.
51.
Zurück zum Zitat To WS, Midwood KS. Plasma and cellular fibronectin: distinct and independent functions during tissue repair. Fibrogenesis Tissue Repair. 2011;4:21.PubMedPubMedCentralCrossRef To WS, Midwood KS. Plasma and cellular fibronectin: distinct and independent functions during tissue repair. Fibrogenesis Tissue Repair. 2011;4:21.PubMedPubMedCentralCrossRef
52.
53.
Zurück zum Zitat Crawford DC, Chobanian AV, Brecher P. Angiotensin II induces fibronectin expression associated with cardiac fibrosis in the rat. Circ Res. 1994;74(4):727–39.PubMedCrossRef Crawford DC, Chobanian AV, Brecher P. Angiotensin II induces fibronectin expression associated with cardiac fibrosis in the rat. Circ Res. 1994;74(4):727–39.PubMedCrossRef
54.
Zurück zum Zitat Lee JW, Juliano R. Mitogenic signal transduction by integrin- and growth factor receptor-mediated pathways. Mol Cells. 2004;17(2):188–202.PubMed Lee JW, Juliano R. Mitogenic signal transduction by integrin- and growth factor receptor-mediated pathways. Mol Cells. 2004;17(2):188–202.PubMed
55.
Zurück zum Zitat Legate KR, Wickstrom SA, Fassler R. Genetic and cell biological analysis of integrin outside-in signaling. Genes Dev. 2009;23(4):397–418.PubMedCrossRef Legate KR, Wickstrom SA, Fassler R. Genetic and cell biological analysis of integrin outside-in signaling. Genes Dev. 2009;23(4):397–418.PubMedCrossRef
56.
Zurück zum Zitat Chadha KS, Khoury T, Yu J, Black JD, Gibbs JF, Kuvshinoff BW, et al. Activated Akt and Erk expression and survival after surgery in pancreatic carcinoma. Ann Surg Oncol. 2006;13(7):933–9.PubMedCrossRef Chadha KS, Khoury T, Yu J, Black JD, Gibbs JF, Kuvshinoff BW, et al. Activated Akt and Erk expression and survival after surgery in pancreatic carcinoma. Ann Surg Oncol. 2006;13(7):933–9.PubMedCrossRef
57.
Zurück zum Zitat Yamamoto S, Tomita Y, Hoshida Y, Morooka T, Nagano H, Dono K, et al. Prognostic significance of activated Akt expression in pancreatic ductal adenocarcinoma. Clin Cancer Res. 2004;10(8):2846–50.PubMedCrossRef Yamamoto S, Tomita Y, Hoshida Y, Morooka T, Nagano H, Dono K, et al. Prognostic significance of activated Akt expression in pancreatic ductal adenocarcinoma. Clin Cancer Res. 2004;10(8):2846–50.PubMedCrossRef
58.
Zurück zum Zitat Zheng C, Jiao X, Jiang Y, Sun S. ERK1/2 activity contributes to gemcitabine resistance in pancreatic cancer cells. J Int Med Res. 2013;41(2):300–6.PubMedCrossRef Zheng C, Jiao X, Jiang Y, Sun S. ERK1/2 activity contributes to gemcitabine resistance in pancreatic cancer cells. J Int Med Res. 2013;41(2):300–6.PubMedCrossRef
59.
Zurück zum Zitat Wang M, Lu X, Dong X, Hao F, Liu Z, Ni G, et al. pERK1/2 silencing sensitizes pancreatic cancer BXPC-3 cell to gemcitabine-induced apoptosis via regulating Bax and Bcl-2 expression. World J Surg Oncol. 2015;13:66.PubMedPubMedCentralCrossRef Wang M, Lu X, Dong X, Hao F, Liu Z, Ni G, et al. pERK1/2 silencing sensitizes pancreatic cancer BXPC-3 cell to gemcitabine-induced apoptosis via regulating Bax and Bcl-2 expression. World J Surg Oncol. 2015;13:66.PubMedPubMedCentralCrossRef
60.
Zurück zum Zitat Ng SSW, Tsao MS, Chow S, Hedley DW. Inhibition of phosphatidylinositide 3-kinase enhances gemcitabine-induced apoptosis in human pancreatic cancer cells. Cancer Res. 2000;60(19):5451–5.PubMed Ng SSW, Tsao MS, Chow S, Hedley DW. Inhibition of phosphatidylinositide 3-kinase enhances gemcitabine-induced apoptosis in human pancreatic cancer cells. Cancer Res. 2000;60(19):5451–5.PubMed
61.
Zurück zum Zitat Ng SS, Tsao MS, Nicklee T, Hedley DW. Wortmannin inhibits pkb/akt phosphorylation and promotes gemcitabine antitumor activity in orthotopic human pancreatic cancer xenografts in immunodeficient mice. Clin Cancer Res. 2001;7(10):3269–75.PubMed Ng SS, Tsao MS, Nicklee T, Hedley DW. Wortmannin inhibits pkb/akt phosphorylation and promotes gemcitabine antitumor activity in orthotopic human pancreatic cancer xenografts in immunodeficient mice. Clin Cancer Res. 2001;7(10):3269–75.PubMed
62.
Zurück zum Zitat Arlt A, Gehrz A, Muerkoster S, Vorndamm J, Kruse ML, Folsch UR, et al. Role of NF-kappaB and Akt/PI3K in the resistance of pancreatic carcinoma cell lines against gemcitabine-induced cell death. Oncogene. 2003;22(21):3243–51.PubMedCrossRef Arlt A, Gehrz A, Muerkoster S, Vorndamm J, Kruse ML, Folsch UR, et al. Role of NF-kappaB and Akt/PI3K in the resistance of pancreatic carcinoma cell lines against gemcitabine-induced cell death. Oncogene. 2003;22(21):3243–51.PubMedCrossRef
63.
Zurück zum Zitat Sawai H, Okada Y, Funahashi H, Takahashi H, Matsuo Y, Yasuda A, et al. Basement membrane proteins play an important role in the invasive processes of human pancreatic cancer cells. J Surg Res. 2008;144(1):117–23.PubMedCrossRef Sawai H, Okada Y, Funahashi H, Takahashi H, Matsuo Y, Yasuda A, et al. Basement membrane proteins play an important role in the invasive processes of human pancreatic cancer cells. J Surg Res. 2008;144(1):117–23.PubMedCrossRef
64.
Zurück zum Zitat Shields MA, Dangi-Garimella S, Redig AJ, Munshi HG. Biochemical role of the collagen-rich tumour microenvironment in pancreatic cancer progression. Biochem J. 2012;441(2):541–52.PubMedCrossRef Shields MA, Dangi-Garimella S, Redig AJ, Munshi HG. Biochemical role of the collagen-rich tumour microenvironment in pancreatic cancer progression. Biochem J. 2012;441(2):541–52.PubMedCrossRef
65.
Zurück zum Zitat Januchowski R, Swierczewska M, Sterzynska K, Wojtowicz K, Nowicki M, Zabel M. Increased expression of several collagen genes is associated with drug resistance in ovarian Cancer cell lines. J Cancer. 2016;7(10):1295–310.PubMedPubMedCentralCrossRef Januchowski R, Swierczewska M, Sterzynska K, Wojtowicz K, Nowicki M, Zabel M. Increased expression of several collagen genes is associated with drug resistance in ovarian Cancer cell lines. J Cancer. 2016;7(10):1295–310.PubMedPubMedCentralCrossRef
66.
Zurück zum Zitat Iseri OD, Kars MD, Arpaci F, Gunduz U. Gene expression analysis of drug-resistant MCF-7 cells: implications for relation to extracellular matrix proteins. Cancer Chemother Pharmacol. 2010;65(3):447–55.PubMedCrossRef Iseri OD, Kars MD, Arpaci F, Gunduz U. Gene expression analysis of drug-resistant MCF-7 cells: implications for relation to extracellular matrix proteins. Cancer Chemother Pharmacol. 2010;65(3):447–55.PubMedCrossRef
67.
Zurück zum Zitat Dangi-Garimella S, Krantz SB, Barron MR, Shields MA, Heiferman MJ, Grippo PJ, et al. Three-dimensional collagen I promotes gemcitabine resistance in pancreatic cancer through MT1-MMP-mediated expression of HMGA2. Cancer Res. 2011;71(3):1019–28.PubMedCrossRef Dangi-Garimella S, Krantz SB, Barron MR, Shields MA, Heiferman MJ, Grippo PJ, et al. Three-dimensional collagen I promotes gemcitabine resistance in pancreatic cancer through MT1-MMP-mediated expression of HMGA2. Cancer Res. 2011;71(3):1019–28.PubMedCrossRef
68.
Zurück zum Zitat Ruess DA, Gorgulu K, Wormann SM, Algul H. Pharmacotherapeutic Management of Pancreatic Ductal Adenocarcinoma: current and emerging concepts. Drugs Aging. 2017;34(5):331–57.PubMedCrossRef Ruess DA, Gorgulu K, Wormann SM, Algul H. Pharmacotherapeutic Management of Pancreatic Ductal Adenocarcinoma: current and emerging concepts. Drugs Aging. 2017;34(5):331–57.PubMedCrossRef
69.
Zurück zum Zitat Whatcott CJ, Han H, Von Hoff DD. Orchestrating the tumor microenvironment to improve survival for patients with pancreatic Cancer: normalization, not destruction. Cancer J. 2015;21(4):299–306.PubMedPubMedCentralCrossRef Whatcott CJ, Han H, Von Hoff DD. Orchestrating the tumor microenvironment to improve survival for patients with pancreatic Cancer: normalization, not destruction. Cancer J. 2015;21(4):299–306.PubMedPubMedCentralCrossRef
Metadaten
Titel
Secretion of fibronectin by human pancreatic stellate cells promotes chemoresistance to gemcitabine in pancreatic cancer cells
Publikationsdatum
01.12.2019
Erschienen in
BMC Cancer / Ausgabe 1/2019
Elektronische ISSN: 1471-2407
DOI
https://doi.org/10.1186/s12885-019-5803-1

Weitere Artikel der Ausgabe 1/2019

BMC Cancer 1/2019 Zur Ausgabe

Update Onkologie

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