Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
J Pathol ; 246(3): 311-322, 2018 11.
Article in English | MEDLINE | ID: mdl-30027561

ABSTRACT

Unstable and dysfunctional tumor vasculature promotes cancer progression and spread. Signal transduction by the pro-angiogenic vascular endothelial growth factor (VEGF) receptor-2 (VEGFR2) is modulated by VEGFA-dependent complex formation with neuropilin 1 (NRP1). NRP1 expressed on tumor cells can form VEGFR2/NRP1 trans-complexes between tumor cells and endothelial cells which arrests VEGFR2 on the endothelial surface, thus interfering with productive VEGFR2 signaling. In mouse fibrosarcoma, VEGFR2/NRP1 trans-complexes correlated with reduced tumor vessel branching and reduced tumor cell proliferation. Pancreatic ductal adenocarcinoma (PDAC) strongly expressed NRP1 on both tumor cells and endothelial cells, in contrast to other common cancer forms. Using proximity ligation assay, VEGFR2/NRP1 trans-complexes were identified in human PDAC tumor tissue, and its presence was associated with reduced tumor vessel branching, reduced tumor cell proliferation, and improved patient survival after adjusting for other known survival predictors. We conclude that VEGFR2/NRP1 trans-complex formation is an independent predictor of PDAC patient survival. © 2018 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.


Subject(s)
Carcinoma, Pancreatic Ductal/blood supply , Carcinoma, Pancreatic Ductal/metabolism , Endothelial Cells/metabolism , Neovascularization, Pathologic , Neuropilin-1/metabolism , Pancreatic Neoplasms/blood supply , Pancreatic Neoplasms/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Aged , Animals , Carcinoma, Pancreatic Ductal/mortality , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cell Proliferation , Endothelial Cells/pathology , Fibrosarcoma/blood supply , Fibrosarcoma/metabolism , Fibrosarcoma/pathology , Humans , Mice, Inbred C57BL , Middle Aged , Neuropilin-1/genetics , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/pathology , Prognosis , Protein Binding , Risk Factors , Signal Transduction , Stomach Neoplasms/blood supply , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Sus scrofa , Tumor Burden , Vascular Endothelial Growth Factor Receptor-2/genetics
2.
Oncotarget ; 8(42): 71672-71684, 2017 Sep 22.
Article in English | MEDLINE | ID: mdl-29069737

ABSTRACT

The pancreatic stellate cell (PSC) is the principal cell type of the desmoplastic stroma of pancreatic ductal adenocarcinoma (PDAC). PSCs interact with cancer cells and influence the progression of the disease through a complex network of signaling molecules including hepatocyte growth factor (HGF). Functional heterogeneity of PSCs within a tumor might conceivably influence tumor progression. We investigated PSC populations isolated from different human PDACs and examined the effects of PSC-conditioned medium on BxPC-3 and AsPC-1 pancreatic cancer cells. The different PSC populations exhibited a wide range of variation (120-3,000 pg/ml) in their ability to secrete HGF. Media from high-HGF-producing PSCs stimulated phosphorylation of Met, Gab1, and ERK in the cancer cells and induced increases in DNA synthesis and migration which were blocked by the Met inhibitor SU11274, indicating a role of HGF as a mediator. HGF levels produced by PSCs and the effects of PSC media on the cancer cells were increased by IL-1α and inhibited by TGFß. The functional heterogeneity of PSCs in terms of HGF-mediated tumor-stroma interactions suggests that inhibition of the HGF pathway as a novel treatment approach in PDAC might have different effects in different subsets of patients.

3.
Neoplasia ; 18(7): 447-56, 2016 07.
Article in English | MEDLINE | ID: mdl-27435927

ABSTRACT

Pancreatic ductal adenocarcinoma is characterized by a prominent fibroinflammatory stroma with both tumor-promoting and tumor-suppressive functions. The pancreatic stellate cell (PSC) is the major cellular stromal component and the main producer of extracellular matrix proteins, including collagens, which are degraded by metalloproteinases (MMPs). PSCs interact with cancer cells through various factors, including transforming growth factor (TGF)ß and interleukin (IL)-1α. The role of TGFß in the dual nature of tumor stroma, i.e., protumorigenic or tumor suppressive, is not clear. We aimed to investigate the roles of TGFß and IL-1α in the regulation of MMP profiles in PSCs and the subsequent effects on cancer cell migration. Human PSCs isolated from surgically resected specimens were cultured in the presence of pancreatic cancer cell lines, as well as IL-1α or TGFß. MMP production and activities in PSCs were quantified by gene array transcripts, mRNA measurements, fluorescence resonance energy transfer-based activity assay, and zymography. PSC-conditioned media and pancreatic cancer cells were included in a collagen matrix cell migration model. We found that production of IL-1α by pancreatic cancer cells induced alterations in MMP and tissue inhibitors of matrix metalloproteinase (TIMP) profiles and activities in PSCs, upregulated expression and activation of MMP1 and MMP3, and enhanced migration of pancreatic cancer cells in the collagen matrix model. TGFß counteracted the effects of IL-1α on PSCs, reestablished PSC MMP and TIMP profiles and activities, and inhibited migration of cancer cells. This suggests that tumor TGFß has a role as a suppressor of stromal promotion of tumor progression through alterations in PSC MMP profiles with subsequent inhibition of pancreatic cancer cell migration.


Subject(s)
Carcinoma, Pancreatic Ductal/pathology , Interleukin-1alpha/pharmacology , Matrix Metalloproteinase 1/metabolism , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 3/metabolism , Pancreatic Neoplasms/pathology , Pancreatic Stellate Cells/metabolism , Tissue Inhibitor of Metalloproteinase-1/metabolism , Tissue Inhibitor of Metalloproteinase-2/metabolism , Tissue Inhibitor of Metalloproteinase-3/metabolism , Transforming Growth Factor beta/pharmacology , Cell Line, Tumor , Cell Movement , Enzyme Activation/drug effects , Fluorescence Resonance Energy Transfer , Humans , Interleukin-1alpha/metabolism , Pancreatic Ducts/pathology , Tumor Suppressor Proteins/metabolism
4.
J Exp Clin Cancer Res ; 35(1): 122, 2016 07 29.
Article in English | MEDLINE | ID: mdl-27473228

ABSTRACT

BACKGROUND: The most abundant cells in the extensive desmoplastic stroma of pancreatic adenocarcinomas are the pancreatic stellate cells, which interact with the carcinoma cells and strongly influence the progression of the cancer. Tumor stroma interactions induced by IL-1α/IL-1R1 signaling have been shown to be involved in pancreatic cancer cell migration. TGFß and its receptors are overexpressed in pancreatic adenocarcinomas. We aimed at exploring TGFß and IL-1α signaling and cross-talk in the stellate cell cancer cell interactions regulating pancreatic adenocarcinoma cell migration. METHODS: Human pancreatic stellate cells were isolated from surgically resected pancreatic adenocarcinomas and cultured in the presence of TGFß or pancreatic adenocarcinoma cell lines. The effects of TGFß were blocked by inhibitors or amplified by silencing the endogenous inhibitor of SMAD signaling, SMAD7. Pancreatic stellate cell responses to IL-1α or to IL-1α-expressing pancreatic adenocarcinoma cells (BxPC-3) were characterized by their ability to stimulate migration of cancer cells in a 2D migration model. RESULTS: In pancreatic stellate cells, IL-1R1 expression was found to be down-regulated by TGFß and blocking of TGFß signaling re-established the expression. Endogenous inhibition of TGFß signaling by SMAD7 was found to correlate with the levels of IL-1R1, indicating a regulatory role of SMAD7 in IL-1R1 expression. Pancreatic stellate cells cultured in the presence of IL-1α or in co-cultures with BxPC-3 cells enhanced the migration of cancer cells. This effect was blocked after treatment of the pancreatic stellate cells with TGFß. Silencing of stellate cell expression of SMAD7 was found to suppress the levels of IL-1R1 and reduce the stimulatory effects of IL-1α, thus inhibiting the capacity of pancreatic stellate cells to induce cancer cell migration. CONCLUSIONS: TGFß signaling suppressed IL-1α mediated pancreatic stellate cell induced carcinoma cell migration. Depletion of SMAD7 upregulated the effects of TGFß and reduced the expression of IL-1R1, leading to inhibition of IL-1α induced stellate cell enhancement of carcinoma cell migration. SMAD7 might represent a target for inhibition of IL-1α induced tumor stroma interactions.


Subject(s)
Interleukin-1alpha/metabolism , Pancreatic Neoplasms/metabolism , Pancreatic Stellate Cells/cytology , Smad3 Protein/metabolism , Transforming Growth Factor beta/metabolism , Cell Communication , Cell Line, Tumor , Cell Movement , Cells, Cultured , Coculture Techniques , Gene Expression Regulation, Neoplastic , Humans , Interleukin-1alpha/genetics , Pancreatic Neoplasms/pathology , Pancreatic Stellate Cells/metabolism , Receptors, Interleukin-1 Type I/genetics , Receptors, Interleukin-1 Type I/metabolism , Signal Transduction , Smad3 Protein/genetics , Smad7 Protein/genetics , Smad7 Protein/metabolism , Pancreatic Neoplasms
5.
Oncotarget ; 7(13): 16396-408, 2016 Mar 29.
Article in English | MEDLINE | ID: mdl-26918939

ABSTRACT

Pancreatic stellate cells (PSCs) are the key precursor cells for cancer-associated fibroblasts (CAFs) in pancreatic tumor stroma. In this study, we explored miRNA as therapeutic targets in tumor stroma and found miR-199a-3p and miR-214-3p induced in patient-derived pancreatic CAFs and TGF-ß-activated human PSCs (hPSCs). Inhibition of miR-199a/-214 using hairpin inhibitors significantly inhibited TGFß-induced differentiation markers (e.g. α-SMA, collagen, PDGFßR), migration and proliferation. Furthermore, heterospheroids of Panc-1 and hPSCs attained smaller size with hPSCs transfected with anti-miR-199a/-214 compared to control anti-miR. The conditioned medium obtained from TGFß-activated hPSCs induced tumor cell growth and endothelial cell tube formation. Interestingly, these inductions were abrogated in hPSCs transfected with anti-miR-199a or miR-214. Moreover, IPA analyses revealed signaling pathways related to miR-199a (TP53, mTOR, Smad1) and miR-214 (PTEN, Bax, ING4). Taken together, this study reveals miR-199a-3p and miR-214-3p as major regulators of PSC activation and PSC-induced pro-tumoral effects, representing them as key therapeutic targets in pancreatic cancer.


Subject(s)
MicroRNAs/genetics , Pancreatic Neoplasms/genetics , Pancreatic Stellate Cells/metabolism , Antagomirs/genetics , Apoptosis/genetics , Cancer-Associated Fibroblasts/drug effects , Cancer-Associated Fibroblasts/metabolism , Cell Differentiation/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Cells, Cultured , Gene Expression Regulation, Neoplastic/drug effects , Humans , In Situ Hybridization , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Pancreatic Stellate Cells/drug effects , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/genetics , Transforming Growth Factor beta1/pharmacology
6.
J Oral Pathol Med ; 45(6): 425-32, 2016 Jul.
Article in English | MEDLINE | ID: mdl-26602326

ABSTRACT

BACKGROUND: The glycerophospholipid lysophosphatidic acid (LPA), which is present in most tissues and in high concentrations in saliva, may exert profound effects on oral cancer cells. We have investigated mitogenic signalling induced by LPA in the two oral carcinoma cell lines, D2 and E10, focusing on the role of EGFR transactivation and downstream pathways. METHODS: Two oral squamous carcinoma cell lines, D2 and E10, were analysed for effects of LPA on signalling pathways and induction of DNA synthesis. Pathway activation was investigated by examining phosphorylation of signalling proteins and by the use of specific pathway inhibitors. RESULTS: The D2 cells had higher levels of activated signalling proteins and higher DNA synthesis activity in the basal condition than E10 cells. EGF did not induce proliferation in D2 cells, whereas LPA induced proliferation in both cell lines, by mechanisms depending on EGFR transactivation. Release of EGFR ligands was involved in basal and LPA-induced proliferation in both D2 and E10 cells. The proliferation in D2 cells was dependent on the PI3K/Akt pathway, but not the MEK/ERK pathway. In E10 cells, the PI3K/Akt, MEK/ERK and p38 pathways were all involved in the proliferation. CONCLUSION: Transactivation of EGFR is required for LPA-induced DNA synthesis in D2 and E10 cells. Our results also show that although proliferation of oral carcinoma cells is regulated by several pathways, and differentially in E10 and D2 cells, the PI3K pathway has a crucial role in both cell lines.


Subject(s)
Carcinoma, Squamous Cell/metabolism , DNA, Neoplasm/biosynthesis , Head and Neck Neoplasms/metabolism , Lysophospholipids/pharmacology , Mouth Neoplasms/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cell Movement/drug effects , DNA Replication/drug effects , DNA Replication/physiology , DNA, Neoplasm/genetics , Enzyme Activation , ErbB Receptors/genetics , ErbB Receptors/metabolism , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/pathology , Humans , MAP Kinase Signaling System/drug effects , Metabolic Networks and Pathways/drug effects , Mouth Neoplasms/genetics , Mouth Neoplasms/pathology , Phosphorylation , Squamous Cell Carcinoma of Head and Neck , Transcriptional Activation/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism
7.
BMC Cancer ; 14: 413, 2014 Jun 09.
Article in English | MEDLINE | ID: mdl-24912820

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 [(3)H]-proline incorporation, or by enzyme immunoassay measurement of collagen C-peptide. DNA synthesis was measured by incorporation of [(3)H]-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-1ß (IL-1ß), epidermal growth factor (EGF), thrombin, and PGE2, but not by transforming growth factor-ß1 (TGFß). 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 TGFß-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.


Subject(s)
Collagen/biosynthesis , Dinoprostone/pharmacology , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Pancreatic Stellate Cells/drug effects , Pancreatic Stellate Cells/metabolism , Cell Line, Tumor , Cells, Cultured , Coculture Techniques , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , DNA Replication/drug effects , Gene Expression , Humans , Immunohistochemistry , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms
8.
PLoS One ; 8(8): e70874, 2013.
Article in English | MEDLINE | ID: mdl-23951028

ABSTRACT

The interplay between the tumor cells and the surrounding stroma creates inflammation, which promotes tumor growth and spread. The inflammation is a hallmark for pancreatic adenocarcinoma (PDAC) and is to high extent driven by IL-1α. IL-1α is expressed and secreted by the tumor cells and exerting its effect on the stroma, i.e. cancer associated fibroblasts (CAF), which in turn produce massive amount of inflammatory and immune regulatory factors. IL-1 induces activation of transcription factors such as nuclear factor-κß (NF-κß), but also activator protein 1 (AP-1) via the small G-protein Ras. Dysregulation of Ras pathways are common in cancer as this oncogene is the most frequently mutated in many cancers. In contrast, the signaling events leading up to the expression of IL-1α by tumor cells are not well elucidated. Our aim was to examine the signaling cascade involved in the induction of IL-1α expression in PDAC. We found p38MAPK, activated by the K-Ras signaling pathway, to be involved in the expression of IL-1α by PDAC as blocking this pathway decreased both the gene and protein expression of IL-1α. Blockage of the P38MAPK signaling in PDAC also dampened the ability of the tumor cell to induce inflammation in CAFs. In addition, the IL-1α autocrine signaling regulated the migratory capacity of PDAC cells. Taken together, the blockage of signaling pathways leading to IL-1α expression and/or neutralization of IL-1α in the PDAC microenvironment should be taken into consideration as possible treatment or complement to existing treatment of this cancer.


Subject(s)
Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Cell Movement/genetics , Interleukin-1alpha/genetics , MAP Kinase Signaling System , Cell Communication/drug effects , Cell Line, Tumor , Extracellular Signal-Regulated MAP Kinases/metabolism , Fibroblasts/metabolism , Gene Expression Regulation, Neoplastic , Humans , Inflammation/genetics , Inflammation/metabolism , Interleukin 1 Receptor Antagonist Protein/pharmacology , MAP Kinase Signaling System/drug effects , Protein Kinase Inhibitors/pharmacology , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/metabolism
9.
Clin Dev Immunol ; 2011: 212810, 2011.
Article in English | MEDLINE | ID: mdl-22190968

ABSTRACT

Tumor microenvironment is composed of tumor cells, fibroblasts, and infiltrating immune cells, which all work together and create an inflammatory environment favoring tumor progression. The present study aimed to investigate the role of the desmoplastic stroma in pancreatic ductal adenocarcinoma (PDAC) regarding expression of inflammatory factors and infiltration of immune cells and their impact on the clinical outcome. The PDAC tissues examined expressed significantly increased levels of immunomodulatory and chemotactic factors (IL-6, TGFß, IDO, COX-2, CCL2, and CCL20) and immune cell-specific markers corresponding to macrophages, myeloid, and plasmacytoid dendritic cells (DCs) as compared to controls. Furthermore, short-time survivors had the lowest levels of DC markers. Immunostainings indicated that the different immune cells and inflammatory factors are mainly localized to the desmoplastic stroma. Therapies modulating the inflammatory tumor microenvironment to promote the attraction of DCs and differentiation of monocytes into functional DCs might improve the survival of PDAC patients.


Subject(s)
Carcinoma, Pancreatic Ductal/immunology , Pancreatic Neoplasms/immunology , Tumor Microenvironment/immunology , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/mortality , Case-Control Studies , Cytokines/immunology , Cytokines/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Humans , Immunophenotyping , Inflammation Mediators/immunology , Inflammation Mediators/metabolism , Macrophages/immunology , Macrophages/metabolism , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/mortality , Receptors, Cell Surface/metabolism , Stromal Cells , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism
10.
Neoplasia ; 13(8): 664-75, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21847358

ABSTRACT

The tumor microenvironment in pancreatic ductal adenocarcinoma (PDAC) is dynamic, with an extensive interaction between the stroma and tumor cells. The aim of this study was to delineate the cross talk between PDAC and cancer-associated fibroblasts (CAFs), with a focus on the mechanism creating the chronic inflammatory tumor milieu. We assessed the effects of the cross talk between PDAC and CAF cell lines on the creation and sustenance of the inflammatory tumor microenvironment in pancreatic cancer. The coculture of PDAC and CAF cell lines enhanced the levels of inflammatory factors including IL-1α, IL-6, CXCL8, VEGF-A, CCL20, and COX-2. CAFs were superior to tumor cells regarding the production of most inflammatory factors, and tumor cell-associated IL-1α was established as the initiator of the enhanced production of inflammatory factors through the binding of IL-1α to IL-1 receptor 1 (IL-1R1) expressed predominantly by CAFs. Furthermore, we found a correlation between IL-1α and CXCL8 expression levels in PDAC tissues and correlation between IL-1α expression and the clinical outcome of the patients. This confirmed an important role for the IL-1 signaling cascade in the creation and sustenance of a tumor favorable microenvironment. Neutralization of the IL-1α signaling efficiently diminished the cross talk-induced production of inflammatory factors. These data suggest that the cross talk between PDAC cells and the main stroma cell type, i.e. CAFs, is one essential factor in the formation of the inflammatory tumor environment, and we propose that neutralization of the IL-1α signaling might be a potential therapy for this cancer.


Subject(s)
Carcinoma, Pancreatic Ductal/metabolism , Fibroblasts/metabolism , Inflammation Mediators/metabolism , Interleukin-1alpha/metabolism , Pancreatic Neoplasms/metabolism , Tumor Microenvironment , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/metabolism , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/mortality , Carcinoma, Pancreatic Ductal/pathology , Cell Communication , Cell Line , Coculture Techniques , Fibroblasts/pathology , Gene Expression Profiling , Humans , Interleukin-1alpha/genetics , Interleukin-1alpha/immunology , Neoplasm Staging , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/pathology , Receptors, Interleukin-1/genetics , Receptors, Interleukin-1/immunology , Receptors, Interleukin-1/metabolism , Survival Analysis , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
11.
PLoS One ; 5(10): e13441, 2010 Oct 15.
Article in English | MEDLINE | ID: mdl-20976171

ABSTRACT

BACKGROUND: Much evidence exists regarding the fact that blood DCs, both myeloid DCs (MDCs) and plasmacytoid DCs (PDCs), are negatively affected in different types of cancer, with both reduced numbers and impaired functionality. Functional impairment of DCs in patients with pancreatic ductal adenocarcinoma (PDAC), may contribute to the poor clinical outcome. The aim of this study was to examine the effects PDAC had on blood DCs and elucidate the underlying mechanism responsible for the DC impairment. METHODOLOGY/PRINCIPAL FINDINGS: We examined the systemic influence PDAC exerted on blood DCs by ex vivo measuring numerous activation and maturation markers expressed on these cells. Furthermore, the effect patient plasma and the inflammatory factors CXCL8 and PGE(2) had on purified MDCs and PDCs from healthy donors was assessed and compared to the DCs existing in PDAC patients. We found a partial maturation of the blood MDCs and PDCs in PDAC patients with significantly enhanced expression of CD83, CD40, B7H3, PDL-1, CCR6, and CCR7 and decreased expression of ICOSL, and DCIR. These changes lead to impairment in their immunostimulatory function. Furthermore, chronic pancreatitis gave rise to DCs with similar semi-mature phenotype as seen in PDAC. Low expression of ICOSL was associated with poor prognosis. We found that the mechanism underlying this semi-maturation of DCs was inflammatory factors existing in the PDAC patients' plasma. Of note, PGE(2), which is elevated PDAC patient plasma, was one contributing factor to the changes seen in MDCs and PDCs phenotype. CONCLUSION/SIGNIFICANCE: Our findings point to a role for the systemic inflammation in transforming blood MDCs and PDCs into semi-mature cells in PDAC patients and we show a correlation between maturation status and clinical outcome. Thus, means to preserve a functional blood DC compartment in PDAC patients by diminishing the inflammation could facilitate their ability to control the disease and improve survival.


Subject(s)
Carcinoma, Pancreatic Ductal/blood , Dendritic Cells/pathology , Pancreatic Neoplasms/blood , Carcinoma, Pancreatic Ductal/pathology , Humans , Pancreatic Neoplasms/pathology
12.
BMC Cancer ; 10: 87, 2010 Mar 09.
Article in English | MEDLINE | ID: mdl-20214814

ABSTRACT

BACKGROUND: Dendritic cells (DCs) isolated from tumor bearing animals or from individuals with solid tumors display functional abnormalities and the DC impairment has emerged as one mechanism for tumor evasion from the control of the immune system. Ductal pancreatic adenocarcinoma (PDAC), the most common pancreatic cancer, is recognized as a very aggressive cancer type with a mortality that almost matches the rate of incidence. METHODS: We examined the systemic influence ductal pancreatic adenocarcinoma (PDAC) exerted on levels of peripheral blood DCs and inflammatory mediators in comparison to the effects exerted by other pancreatic tumors, chronic pancreatitis, and age-matched controls. RESULTS: All groups examined, including PDAC, had decreased levels of myeloid DCs (MDC) and plasmacytoid DCs (PDC) and enhanced apoptosis in these cells as compared to controls. We found elevated levels of PGE2 and CXCL8 in subjects with PDAC, and chronic pancreatitis. Levels of these inflammatory factors were in part restored in PDAC after tumor resection, whereas the levels of DCs were impaired in the majority of these patients approximately 12 weeks after tumor removal. Our results prove that solid pancreatic tumors, including PDAC, systemically affect blood DCs. The impairments do not seem to be tumor-specific, since similar results were obtained in subjects with chronic pancreatitis. Furthermore, we found that PDAC patients with a survival over 2 years had significant higher levels of blood DCs compared to patients with less than one year survival. CONCLUSIONS: Our findings points to the involvement of inflammation in the destruction of the blood MDCs and PDCs. Furthermore, the preservation of the blood DCs compartment in PDAC patients seems to benefit their ability to control the disease and survival.


Subject(s)
Adenocarcinoma/therapy , Carcinoma, Pancreatic Ductal/pathology , Dendritic Cells/cytology , Myeloid Cells/cytology , Pancreatic Neoplasms/therapy , Adenocarcinoma/diagnosis , Aged , Aged, 80 and over , Antibodies, Monoclonal/chemistry , Carcinoma, Pancreatic Ductal/metabolism , Case-Control Studies , Female , Flow Cytometry/methods , Humans , Immune System , Leukocytes, Mononuclear/cytology , Male , Middle Aged , Pancreatic Neoplasms/diagnosis
SELECTION OF CITATIONS
SEARCH DETAIL
...