Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
Methods Appl Fluoresc ; 11(4)2023 Aug 23.
Article in English | MEDLINE | ID: mdl-37612784

ABSTRACT

Antibodies have gained considerable importance in laboratory and clinical settings. Currently, antibodies are extensively employed for the diagnosis and treatment of several human diseases. Herein, using targeted and cell immunisation approaches, we developed and characterised an antibody clone, DWH24. We found that DWH24 is an IgMκtype antibody that enables excellent visualisation and quantification of dead cells using immunofluorescence, fluorescence microscopy, and flow cytometry. This property was proved by the spontaneous cell death of several tumour cell lines and stimulated T cells, as well as after chemo- and photodynamic therapy. Unlike conventional apoptosis and cell death markers, DWH24 binding occurred in a Ca2+- and protein-independent manner and enabled live imaging of cell death progress, as shown using time-lapse microscopy. The binding specificity of DWH24 was analysed using a human proteome microarray, which revealed a complex response profile with very high spot intensities against various proteins, such as tropomyosin variants and FAM131C. Accordingly, DWH24 can be employed as a suitable tool for the cost-effective and universal analysis of cell death using fluorescence imaging and flow cytometry.


Subject(s)
Apoptosis , Humans , Cell Death , Microscopy, Fluorescence , Cell Line, Tumor , Flow Cytometry
3.
Oncoimmunology ; 11(1): 2043037, 2022.
Article in English | MEDLINE | ID: mdl-35251770

ABSTRACT

Immune checkpoint molecule B7-H1 plays a decisive immune regulatory role in different pathologies including cancer, and manipulation of B7-H1 expression became an attractive approach in cancer immunotherapy. Pancreatic cancer (PDAC) is characterized by pronounced immunosuppressive environment and B7-H1 expression correlates with PDAC prognosis. However, the first attempts to diminish B7-H1 expression in patients were not so successful. This points the complicity of PDAC immunosuppressive network and requires further examinations. We investigated the effect of B7-H1 deficiency in PDAC. Our results clearly show that partial or complete B7-H1 inhibition in vivo let to reduced tumor volume and improved survival of PDAC-bearing mice. This oncological benefit is due to the abrogation of immunosuppression provided by MDSC, macrophages, DC and Treg, which resulted in simultaneous restoration of anti-tumor immune response, namely improved accumulation and functionality of effector-memory CD4 and CD8 T cells. Our results underline the potential of B7-H1 molecule to control immunosuppressive network in PDAC and provide new issues for further clinical investigations.


Subject(s)
B7-H1 Antigen , Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Animals , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/immunology , Carcinoma, Pancreatic Ductal/immunology , Carcinoma, Pancreatic Ductal/therapy , Humans , Immune Checkpoint Inhibitors/pharmacology , Immunotherapy/methods , Mice , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/immunology , Programmed Cell Death 1 Receptor
4.
Front Immunol ; 11: 589818, 2020.
Article in English | MEDLINE | ID: mdl-33193420

ABSTRACT

Introduction: The broccoli isothiocyanate sulforaphane was shown to inhibit inflammation and tumor progression, also in pancreatic cancer, while its effect on tumor immunity is poorly understood. We investigated the immunoregulatory effect of sulforaphane on human dendritic cells alone and in presence of pancreatic tumor antigens, as well as underlying molecular mechanisms. Methods: Sulforaphane-treated human dendritic cells were matured in vitro with a cytokine cocktail, and the expression of regulatory molecules was examined by flow cytometry. The subsequent T-cell response was analyzed by T-cell proliferation assay and CD25 expression. To confirm the findings, dendritic cells pulsed with pancreatic cancer-derived tumor antigens were used. To identify the involved pathway- and microRNA-signaling in sulforaphane-treated dendritic cells, inhibitors of various signaling pathways, western blot analysis, microRNA array, and bioinformatic analysis were applied. Results: Sulforaphane modulated the expression of the costimulatory CD80, CD83 and the suppressive B7-H1 molecules on dendritic cells and thereby promoted activation of T cells. The effect was verified in presence of pancreatic tumor antigens. Phosphorylation of STAT3 in dendritic cells was diminished by sulforaphane, and the inhibition of JAK/STAT3 led to downregulation of B7-H1 expression. Among the identified top 100 significant microRNA candidates, the inhibition of miR-155-5p, important for the expression of costimulatory molecules, and the induction of miR-194-5p, targeting the B7-H1 gene, were induced by sulforaphane. Conclusion: Our findings demonstrate that sulforaphane promotes T-cell activation by dendritic cells through the modulation of regulatory molecules, JAK/STAT3- and microRNA-signaling in healthy conditions and in context of pancreatic cancer-derived antigens. They explore the immunoregulatory properties of sulforaphane and justify further research on nutritional strategies in the co-treatment of cancer.


Subject(s)
Dendritic Cells/drug effects , Immunologic Factors/pharmacology , Isothiocyanates/pharmacology , Janus Kinases/metabolism , STAT3 Transcription Factor/metabolism , Sulfoxides/pharmacology , Antigens, CD/metabolism , B7-1 Antigen/metabolism , B7-H1 Antigen/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Dendritic Cells/metabolism , Humans , Immunoglobulins/metabolism , Membrane Glycoproteins/metabolism , MicroRNAs/metabolism , Signal Transduction/drug effects , CD83 Antigen
5.
Oncotarget ; 11(14): 1203-1217, 2020 Apr 07.
Article in English | MEDLINE | ID: mdl-32292571

ABSTRACT

Food-derived plant microRNAs are suggested to control human genes by "cross-kingdom" regulation. We examined microRNAs in sprouts from Brassica rapa sylvestris, known as broccoletti, which are widely used as sulforaphane supplements, and assessed their influence on pancreatic cancer. RNA was isolated from 4-day-old sprouts, followed by deep sequencing and bioinformatic analysis. We identified 2 new and 745 known plant microRNA sequences in the miRbase database and predicted 15,494 human target genes and 76,747 putative 3'-UTR binding sites in these target genes. The most promising candidates were the already known microRNA sequence bra-miR156g-5p and the new sequence Myseq-330, both with predicted human target genes related to apoptosis. The overexpression of the respective oligonucleotides by lipofection did not alter the viability, apoptosis, clonogenicity, migration or associated protein expression patterns in pancreatic cancer cells. These data demonstrate that broccoletti sprouts contain microRNA sequences with putative binding sites in human genes, but the sequences evaluated here did not affect cancer growth. Our database of broccoletti-derived microRNA sequences provides a valuable tool for future analysis.

6.
Oncoimmunology ; 9(1): 1744946, 2020 04 09.
Article in English | MEDLINE | ID: mdl-33457090

ABSTRACT

Synthesis of extracellular adenosine by the ectonucleotidases CD39 and CD73 represents an important pathway of immune suppression in the tumor microenvironment. Using two mouse models (RET transgenic melanoma and Panc02 orthotopic pancreatic adenocarcinoma), we identified an elevated frequency of ectonucleotidase-expressing T cells in tumors and spleens. Importantly, these ectonucleotidase-positive T cells also showed a pronounced expression of PD-1. Conversely, the PD-1+ T cell subsets in tumors contained substantially larger proportions of ectonucleotidase-expressing cells compared to their counterparts lacking PD-1 expression. Our in vitro experiments showed that the activation of normal T cells resulted in an increase in the CD39 expression. CD39+ and CD73+ T cells displayed effector or memory phenotypes and produced IFN-γ, thereby linking ectonucleotidase expression to T cell effector functions. An accumulation of conventional and regulatory T cells expressing CD39 and/or CD73 was also detected in the peripheral blood of patients with melanoma and pancreatic cancer. Moreover, we demonstrated a significant association between low frequencies of circulating CD73+CD8+ T cells and CD73+CD4+ regulatory T cells and better overall survival of melanoma patients. Tumor-derived soluble factors (in particular, TGF-ß) significantly enhanced the frequencies of ectonucleotidase-expressing cells in mice. Our findings suggest that the upregulation of ectonucleotidase expression in T cells promotes extracellular adenosine accumulation and represents an important mechanism of homeostatic immune auto-regulation, which could be hijacked by tumors to evade anti-cancer immunity. Targeting CD39 and CD73 can open new avenues for cancer immunotherapy.


Subject(s)
Adenocarcinoma , Pancreatic Neoplasms , Animals , CD8-Positive T-Lymphocytes , Humans , Immunity , Mice , Pancreatic Neoplasms/genetics , T-Lymphocyte Subsets , Tumor Microenvironment
7.
Cancer Lett ; 469: 238-245, 2020 01 28.
Article in English | MEDLINE | ID: mdl-31678166

ABSTRACT

The therapy resistance of pancreatic cancer is associated with the loss of gap junction intercellular communication and connexin 43 expression. The broccoli isothiocyanate sulforaphane restored these features and therapy sensitivity. We investigated whether microRNA signaling is involved. Established cell lines and a patient tissue array (n = 96) were evaluated by miRNA and gene array, bioinformatics, expression studies, in situ hybridization and immunohistochemistry. Sulforaphane inhibited the expression of our top candidate miR30a-3p. Upon transfection of miR30a-3p inhibitors, the gemcitabine bystander effect, Cx43 expression and intercellular communication increased, whereas miR30a-3p mimics inhibited the luciferase activity of a Cx43 3'-UTR construct. miR30a-3p-overexpressing tumor xenografts had a decreased tumor volume and increased gemcitabine sensitivity. In patient tissues, a higher expression of miR30a-3p and a lower expression of Cx43 correlated with malignancy. These findings provide new knowledge and suggest sulforaphane as cotreatment for pancreatic cancer.


Subject(s)
Connexin 43/genetics , Isothiocyanates/pharmacology , MicroRNAs/genetics , Pancreatic Neoplasms/drug therapy , Adult , Aged , Animals , Cell Communication/drug effects , Cell Line, Tumor , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Female , Gap Junctions/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Heterografts , Humans , In Situ Hybridization , Male , Mice , Middle Aged , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Signal Transduction/drug effects , Sulfoxides , Gemcitabine
8.
Cell Physiol Biochem ; 52(5): 1192-1202, 2019.
Article in English | MEDLINE | ID: mdl-31001959

ABSTRACT

BACKGROUND/AIMS: The extracellular ecto-5'-nucleotidase (CD73) is involved in the production of immunosuppressive adenosin (Ado), which can influence different immune cells through the specific adenosine receptors. The main aim of this work was to characterize immune cell populations as well as serum cytokine level in healthy CD73-deficient mice compared to healthy wild-type animals. METHODS: Profound immnophenotyping of splenocytes from healthy CD73-deficient and wild-type mice was done using flow cytometry (FACS analysis). Cytokine measurement in the serum of the animals was carried out with a Bio-Plex assay. RESULTS: The CD73-deficience leads to an increase in a percentage of NK cells and pDC, as well as influences expression of the costimulatory molecules CD80 and CD86. The knockout mice in opposite to wild-type animals show high amount of effector CD4+ T-cells in the spleens. No changes have been found in the subpopulations of CD8+ T-cells. Besides, CD73-deficience leads to a decrease in the percentage of regulatory T cells. Compared with the wild-type animals we found that CD73 knockout mice possess low serum concentration of IL-6. CONCLUSION: This in vivo study clear demonstrated certain immunological changes in the CD73-deficient mice and thus immunoregulatory potential of CD73 molecule. This makes this extracellular enzyme to a real immune check point molecule, attractive for further investigations and clinical studies.


Subject(s)
5'-Nucleotidase/deficiency , CD4-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Killer Cells, Natural/immunology , Spleen/immunology , 5'-Nucleotidase/immunology , Animals , B7-1 Antigen/genetics , B7-1 Antigen/immunology , B7-2 Antigen/genetics , B7-2 Antigen/immunology , Dendritic Cells/pathology , Interleukin-6/genetics , Interleukin-6/immunology , Killer Cells, Natural/pathology , Mice , Mice, Knockout , Spleen/pathology
9.
Cancer Lett ; 452: 203-212, 2019 06 28.
Article in English | MEDLINE | ID: mdl-30910589

ABSTRACT

NF-κB contributes to the aggressiveness of pancreatic ductal adenocarcinoma (PDA), which is counteracted by the bioactive agent sulforaphane. We investigated sulforaphane-induced microRNA signaling and its influence on progression features. Using established cell lines, microRNA and gene arrays, we predicted miR-365a as the top candidate for the sulforaphane-induced inhibition of the NF-κB subunit c-Rel. The lipofection of miR-365a-3p mimics inhibited the luciferase activity of a c-Rel 3'-UTR construct, as well as c-Rel expression, NF-κB activity, and tumor viability, migration, and clonogenicity, whereas apoptosis was induced. In vivo, miR-365a-3p reduced the volume of tumor xenografts and the expression of progression markers. In a tissue array, the expression of miR-365a-3p was absent in almost all 91 malignant tissues but not in 5 normal tissues, thus confirming the previous results. Our observations suggest that sulforaphane-induced miR-365a-3p expression inhibits NF-κB activity by downregulating c-Rel, which prevents the progression of PDA.


Subject(s)
Carcinoma, Pancreatic Ductal/pathology , MicroRNAs/genetics , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins c-rel/metabolism , Transcription Factor RelA/metabolism , Animals , Anticarcinogenic Agents/pharmacology , Apoptosis/drug effects , Carcinoma, Pancreatic Ductal/genetics , Cell Line, Tumor , Cell Movement/drug effects , Chick Embryo , Disease Progression , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Humans , Isothiocyanates/pharmacology , MicroRNAs/drug effects , Pancreatic Neoplasms/genetics , Signal Transduction , Sulfoxides , Xenograft Model Antitumor Assays
11.
Front Immunol ; 9: 2129, 2018.
Article in English | MEDLINE | ID: mdl-30356906

ABSTRACT

Interferon-α (IFNα) has one of the longest histories of use amongst cytokines in clinical oncology and has been applied for the treatment of many types of cancers. Due to its immune-activating properties, IFNα is also an attractive candidate for combinatory anti-cancer therapies. Despite its extensive use in animal tumor models as well as in several clinical trials, the different mechanisms underlying patient responses and affecting desirable clinical benefits are still under investigation. Here we show that in addition to its immune-activating properties, IFNα induces the expression of a key negative regulator, immunosuppressive PD-L1 molecule, in the majority of the specific immune cell populations, particularly in the dendritic cells (DC). DC can modulate immune responses by a variety of mechanisms, including expression of T-cell regulatory molecules and cytokines. Our results showed that treatment of DC with IFNα-2b led to pronounced up-regulation of surface expression of PD-L1 molecules, increased IL-6 and decreased IL-12 production. Moreover, we present evidence that IFNα-treated DC exhibited a reduced capacity to stimulate interferon-γ production in T cells compared to control DC. This T-cell response after treatment of DC with IFNα was recovered by a pre-treatment with an anti-PD-L1 blocking antibody. Further analyses revealed that IFNα regulated PD-L1 expression through the STAT3 and p38 signaling pathways, since blocking of STAT3 and p38 activation with specific inhibitors prevented PD-L1 up-regulation. Our findings underline the important roles of p38 and STAT3 in the regulation of PD-L1 expression and prove that IFNα induces STAT3/p38-mediated expression of PD-L1 and thereby a reduced stimulatory ability of DC. The augmentation of PD-L1 expression in immune cells through IFNα treatment should be considered by use of IFNα in an anti-cancer therapy.


Subject(s)
B7-H1 Antigen/metabolism , Dendritic Cells/immunology , Interferon-alpha/metabolism , MAP Kinase Signaling System/immunology , Animals , B7-H1 Antigen/immunology , Blood Buffy Coat/cytology , Cells, Cultured , Coculture Techniques , Dendritic Cells/metabolism , Healthy Volunteers , Humans , Interferon-alpha/immunology , MAP Kinase Signaling System/drug effects , Mice , Mice, Inbred C57BL , Primary Cell Culture , STAT3 Transcription Factor/antagonists & inhibitors , STAT3 Transcription Factor/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Up-Regulation/immunology , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/metabolism
12.
Naunyn Schmiedebergs Arch Pharmacol ; 390(11): 1125-1134, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28779210

ABSTRACT

Activation of receptor tyrosine kinases is recognized as a hallmark of cancer. Vascular endothelial growth factor (VEGF) and its receptor VEGFR are the prominent players in the induction of tumor neoangiogenesis. Strategies to inhibit VEGF and VEGFR are under intensive investigation in preclinical and clinical settings. Regorafenib is a multikinase inhibitor targeting some VEGFR and other receptor kinases. Preclinical results led to the FDA approval of regorafenib for treatment of metastatic colorectal cancer patients. Effects of this drug in pancreatic ductal adenocarcinoma (PDAC) have not been investigated yet. Gene expression was assessed with real-time PCR analysis. In vitro cell viability, proliferation, apoptosis, necrosis, migration, and invasion of the PDAC cells were assessed after regorafenib treatment. Ex vivo anti-tumor effects of regorafenib were investigated in a spheroid model of PDAC. In vivo anti-tumor effects of the drug were evaluated in a fertilized chicken egg model. In this work, we have demonstrated only a marginal anticancer effect of regorafenib in PDAC in vitro and ex vivo. However, in the egg model of PDAC, this drug reduced tumor volume. Besides, regorafenib is capable of modulating the expression of cancer stem cell (CSC) markers and epithelial-to-mesenchymal transition (EMT) markers on PDAC cells. We found out that effects of regorafenib on the expression of CSC and EMT markers are very heterogeneous and depend obviously on original expression of these markers. We concluded that regorafenib might be a potential drug for PDAC and it should be investigated in future clinical trials.


Subject(s)
Antineoplastic Agents/pharmacology , Phenylurea Compounds/pharmacology , Pyridines/pharmacology , Animals , Cell Line, Tumor , Cell Movement/drug effects , Cell Survival/drug effects , Chickens , Humans , Mice , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , Spheroids, Cellular/drug effects , Tumor Burden/drug effects , Tumor Cells, Cultured , Zygote , Pancreatic Neoplasms
14.
Int J Cancer ; 139(1): 130-9, 2016 Jul 01.
Article in English | MEDLINE | ID: mdl-26914404

ABSTRACT

Our understanding in the last few years about reactive oxygen species (ROS) has changed from being harmful substances to crucial intra- and extracellular messengers as well as important regulators controlling a wide spectrum of signaling pathways, including those in cancer immunology. Therefore, these multiple essential roles of ROS and especially of mitochondria-derived ROS in malignant transformation and cancer progression make them a promising target for anticancer therapy. Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest cancers in the world. A link between ROS, antioxidants and the PDAC development and progression has been recently established. Therefore, usage of specific highly efficient antioxidants could bring an option for treatment and/or prevention of PDAC. 10-(6'-plastoquinonyl) decyltriphenylphosphonium (SkQ1) is a new antioxidant with the highest mitochondrion membrane penetrating ability and potent antioxidant capability. In this work, we investigated an impact of SkQ1 on tumor angiogenesis, immune micromilieu, and oncological parameters in the orthotopic Panc02 murine model of PDAC. We showed that in this model SkQ1 treatment leads to the elevation of pro-angiogenic factors and to building of mainly an anti-inflammatory cytokine milieu. On the cellular level we showed an increase in a percentage of memory T cells and a decrease in frequency on natural killer T (NKT) cells. At the same time, SkQ1 was ineffective in the improvement of oncological parameters of PDAC-bearing mice. New studies are needed to clarify the absence of therapeutic and/or prophylactic benefits of the antioxidant.


Subject(s)
Adenocarcinoma/drug therapy , Carcinoma, Pancreatic Ductal/drug therapy , Inflammation/drug therapy , Neovascularization, Pathologic/drug therapy , Plastoquinone/analogs & derivatives , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Antioxidants/administration & dosage , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Humans , Inflammation/metabolism , Inflammation/pathology , Mice , Mitochondria/drug effects , Mitochondria/pathology , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Plastoquinone/administration & dosage , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects
15.
J Cell Physiol ; 231(12): 2570-81, 2016 12.
Article in English | MEDLINE | ID: mdl-26895995

ABSTRACT

Mitochondria are indispensable for energy metabolism, apoptosis regulation, and cell signaling. Mitochondria in malignant cells differ structurally and functionally from those in normal cells and participate actively in metabolic reprogramming. Mitochondria in cancer cells are characterized by reactive oxygen species (ROS) overproduction, which promotes cancer development by inducing genomic instability, modifying gene expression, and participating in signaling pathways. Mitochondrial and nuclear DNA mutations caused by oxidative damage that impair the oxidative phosphorylation process will result in further mitochondrial ROS production, completing the "vicious cycle" between mitochondria, ROS, genomic instability, and cancer development. The multiple essential roles of mitochondria have been utilized for designing novel mitochondria-targeted anticancer agents. Selective drug delivery to mitochondria helps to increase specificity and reduce toxicity of these agents. In order to reduce mitochondrial ROS production, mitochondria-targeted antioxidants can specifically accumulate in mitochondria by affiliating to a lipophilic penetrating cation and prevent mitochondria from oxidative damage. In consistence with the oncogenic role of ROS, mitochondria-targeted antioxidants are found to be effective in cancer prevention and anticancer therapy. A better understanding of the role played by mitochondria in cancer development will help to reveal more therapeutic targets, and will help to increase the activity and selectivity of mitochondria-targeted anticancer drugs. In this review we summarized the impact of mitochondria on cancer and gave summary about the possibilities to target mitochondria for anticancer therapies. J. Cell. Physiol. 231: 2570-2581, 2016. © 2016 Wiley Periodicals, Inc.


Subject(s)
Antineoplastic Agents/therapeutic use , Mitochondria/metabolism , Neoplasms/drug therapy , Neoplasms/metabolism , Reactive Oxygen Species/metabolism , Antineoplastic Agents/pharmacology , DNA, Mitochondrial/genetics , Drug Delivery Systems , Humans , Mitochondria/drug effects
16.
Transpl Immunol ; 34: 14-24, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26658573

ABSTRACT

Orthotopic liver transplantation (LTP) is nowadays a standard procedure, and provides the chance of survival of patients with end-stage non-treatable chronic liver disease or acute liver failure. Despite long-term survival with a good quality of life in the majority of patients, about 20% develop early allograft dysfunction (EAD), which leads to death or the need for re-transplantation. Therefore, the early diagnosis of EAD and evaluation of its risk factors are very important. Many primary pathological processes leading to EAD are accompanied by the release of different mediators and by a change of biochemical parameters detectable in the peripheral blood. The aim of this study was to investigate cytokines as well as soluble mediators in the serum of patients with and without EAD from our LTP bank, and to evaluate their predictive and prognostic values for EAD. We demonstrated for the first time that the level of IFNγ during the nearest preoperative period may serve as a predictive parameter for EAD. We additionally found that IL-10 and CXCL10 (IP-10) levels in the early postoperative period can be prognostic for EAD. We believe our data expand the spectrum of predictive and prognostic parameters for EAD in LTP.


Subject(s)
Biomarkers/blood , Chemokine CXCL10/blood , Graft Rejection/diagnosis , Interferon-gamma/blood , Interleukin-10/blood , Liver Transplantation , Adult , Aged , Early Diagnosis , Female , Humans , Male , Middle Aged , Predictive Value of Tests , Preoperative Period , Prognosis
17.
Immunol Lett ; 168(1): 111-9, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26450014

ABSTRACT

In general, conventional chemotherapy is associated with significant toxicity leading to immunosuppression manifesting mainly in the lymphocyte depletion. This immunosuppression promotes tumor growth and elicits the tumor cell dissemination. However, chemotherapy can be immune stimulative especially in combination with an immunotherapy. In this work, we investigated in vitro effects of gemcitabine alone and in combination with interferon-alpha on splenocytes obtained from healthy and pancreatic carcinoma bearing mice. We showed that gemcitabine alone depletes the regulatory T cells in the splenocyte culture. Gemcitabine in combination with interferon-alpha demonstrated some immunomodulatory features, but these effects were interferon-alpha dependent. We concluded that combination of both drugs induces rather cumulative effects, supposing that these therapeutic could be applied together for a chemo-immunotherapy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/immunology , Deoxycytidine/analogs & derivatives , Interferon-alpha/immunology , Pancreatic Neoplasms/immunology , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis/drug effects , Apoptosis/immunology , B7-1 Antigen/immunology , B7-1 Antigen/metabolism , B7-2 Antigen/immunology , B7-2 Antigen/metabolism , CD4-CD8 Ratio , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/immunology , Cells, Cultured , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dendritic Cells/metabolism , Deoxycytidine/administration & dosage , Deoxycytidine/immunology , Flow Cytometry , Interferon-alpha/administration & dosage , Mice, Inbred C57BL , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , Spleen/cytology , Spleen/drug effects , Spleen/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Gemcitabine
18.
Oncoimmunology ; 4(4): e998519, 2015 Apr.
Article in English | MEDLINE | ID: mdl-26137414

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) represents one of the deadliest cancers in the world. PDAC cells activate tumor-specific immune responses but simultaneously trigger a strong immunosuppression. We showed that PDAC cells produce high amount of chronic inflammatory mediators and PDAC tumors build an immunosuppressive cytokine milieu, which correlates with tumor progression. We observed a low frequency of dendritic cells (DC) and a pronounced accumulation of macrophages and myeloid-derived suppressor cells (MDSC) in murine PDAC tumors. A strong accumulation of MDSC has also been demonstrated in the peripheral blood of resected PDAC patients. While DC and macrophages seem not to play a significant role in this PDAC model in the context of immunosuppression, MDSC are highly suppressive, and their accumulation is associated with an increase in intratumoral VEGF concentration during the PDAC progression. Application of the phosphodiesterase-5 inhibitor sildenafil led to a prolonged survival of PDAC-bearing female mice, which was due to the decrease in MDSC frequencies and in the systemic VEGF level. This led to a restoration of anticancer immune responses, manifested in the recovery of T lymphocyte functions and in an increase in the frequency of conventional CD4+ T cells in tumors and IFNγ level in serum of PDAC-bearing mice. Thus, MDSC are strongly involved in the PDAC-associated immunosuppression and that their depletion could create new approaches for therapy of PDAC.

19.
Naunyn Schmiedebergs Arch Pharmacol ; 388(11): 1121-8, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26105003

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest cancers in the world. Therefore, new therapeutic options are urgently needed to improve the survival of PDAC patients. Protein kinase G (PKG) conducts the interlude of cGMP signaling which is important for healthy as well as for cancer cells. DT3 is a specific inhibitor of PKG, and it has been shown to possess an anti-tumor cytotoxic activity in vitro. The main aim of this work was to investigate anti-tumor effects of DT3 upon PDAC in vivo.Expression of PKG was assessed with real-time PCR analysis in the normal and tumor pancreatic cells. In vitro cell viability, proliferation, apoptosis, necrosis, migration, and invasion of the murine PDAC cell line Panc02 were assessed after DT3 treatment. In vivo anti-tumor effects of DT3 were investigated in the murine Panc02 orthotopic model of PDAC. Western blot analysis was used to determine the phosphorylation state of the proteins of interest.Functional PKGI is preferentially expressed in PDAC cells. DT3 was capable to reduce viability, proliferation, and migration of murine PDAC cells in vitro. At the same time, DT3 treatment did not change the viability of normal epithelial cells of murine liver. In vivo, DT3 treatment reduced the tumor volume and metastases in PDAC-bearing mice, but it was ineffective to prolong the survival of the tumor-bearing animals. In addition, DT3 treatment decreased phosphorylation of GSK-3, P38, and CREB in murine PDAC.Inhibition of PKG could be a potential therapeutic strategy for PDAC treatment which should be carefully validated in future pre-clinical studies.


Subject(s)
Antineoplastic Agents/therapeutic use , Cyclic GMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic GMP/antagonists & inhibitors , Pancreatic Neoplasms/drug therapy , Peptides/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Line , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Cyclic AMP Response Element-Binding Protein/metabolism , Cyclic GMP-Dependent Protein Kinases/genetics , Cyclic GMP-Dependent Protein Kinases/metabolism , Epithelial Cells/drug effects , Glycogen Synthase Kinase 3/metabolism , Liver/cytology , Mice , Mice, Inbred C57BL , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Peptides/pharmacology , Protein Kinase Inhibitors/pharmacology , Tumor Burden/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism , Pancreatic Neoplasms
20.
Mol Carcinog ; 54(9): 870-9, 2015 Sep.
Article in English | MEDLINE | ID: mdl-24729540

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) represents one of the deadliest cancers in the world. All-trans retinoic acid (ATRA) is the major physiologically active form of vitamin A, regulating expression of many genes. Disturbances of vitamin A metabolism are prevalent in some cancer cells. The main aim of this work was to investigate deeply the components of retinoid signaling in PDAC compared to in the normal pancreas and to prove the clinical importance of retinoid receptor expression. For the study, human tumor tissues obtained from PDAC patients and murine tumors from the orthotopic Panc02 model were used for the analysis of retinoids, using high performance liquid chromatography mass spectrometry and real-time RT-PCR gene expression analysis. Survival probabilities in univariate analysis were estimated using the Kaplan-Meier method and the Cox proportional hazards model was used for the multivariate analysis. In this work, we showed for the first time that the ATRA and all-trans retinol concentration is reduced in PDAC tissue compared to their normal counterparts. The expression of RARα and ß as well as RXRα and ß are down-regulated in PDAC tissue. This reduced expression of retinoid receptors correlates with the expression of some markers of differentiation and epithelial-to-mesenchymal transition as well as of cancer stem cell markers. Importantly, the expression of RARα and RXRß is associated with better overall survival of PDAC patients. Thus, reduction of retinoids and their receptors is an important feature of PDAC and is associated with worse patient survival outcomes.


Subject(s)
Carcinoma, Pancreatic Ductal/genetics , Down-Regulation , Pancreas/pathology , Pancreatic Neoplasms/genetics , Receptors, Retinoic Acid/genetics , Retinoids/genetics , Animals , Carcinoma, Pancreatic Ductal/diagnosis , Carcinoma, Pancreatic Ductal/pathology , Cell Differentiation , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , Humans , Kaplan-Meier Estimate , Mice, Inbred C57BL , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Pancreas/metabolism , Pancreatic Neoplasms/diagnosis , Pancreatic Neoplasms/pathology , Prognosis , Proportional Hazards Models , Receptors, Retinoic Acid/analysis , Retinoids/analysis , Survival Analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...