Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Thromb Haemost ; 117(9): 1700-1704, 2017 08 30.
Article in English | MEDLINE | ID: mdl-28640321

ABSTRACT

Laboratory diagnosis of lupus anticoagulant (LA) is based on prolongation in at least one coagulation assay (diluted Russell's viper venom time - dRVVT or activated partial thromboplastin time - aPTT), which normalises after addition of phospholipids. Both assays may be influenced by anticoagulants and therefore LA should not be tested during warfarin or heparin treatment. It has been shown (primarily in vitro) that direct oral anticoagulants (DOACs - dabigatran [DAB], rivaroxaban [RIV] and apixaban [API]) may also influence LA testing. We tested the effects of DOACs on assays routinely used for the diagnosis of LA in patients treated with these drugs in a real-life setting. Plasma from patients with atrial fibrillation treated with DAB (n=30), RIV (n=20) and API (n=17) and not known to have LA were tested using dRVVT (LA-screen and LA-confirm, Life Diagnostics) and aPTT (PTT-LA, Diagnostica Stago and aPTT Actin FS, Siemens Healthcare Diagnostics) assays. According to the diagnostics algorithm, dRVVT and aPTT ratios of <1.2 were considered negative, ratios of >1.4 positive, while if the ratios were 1.2-1.4 LA could not be ruled out. Plasma concentrations varied between 8-172 µg/l for DAB, 8-437 µg/l for RIV and 36-178 µg/l for API. LA diagnosis was negative in only eight (27 %) plasma samples from patients treated with DAB, and in five (25 %) and four samples (24 %) from patients treated with RIV and API, respectively. LA Positivity (dRVVT and aPTT ratios >1.4) was found in 5 cases (17 %) among patients treated with DAB, in 10 cases (50 %) treated with RIV and in 7 cases (41 %) treated with API. A concentration-dependent effect of DOACs on dRVVT-based parameters was observed, particularly as regards DAB. At lower concentrations, RIV and API had only minor effects on the confirmatory tests (below 100 µg/l and 70 µg/l, respectively). Our results suggest that a risk of overestimation of LA detection is present in samples from patients treated with DOACs. Therefore, LA testing should not be performed during treatment with DOACs. Prolongation in confirmatory assays may be helpful for the recognition of false positivity, especially as regards DAB.


Subject(s)
Anticoagulants/administration & dosage , Atrial Fibrillation/drug therapy , Blood Coagulation/drug effects , Dabigatran/administration & dosage , Lupus Coagulation Inhibitor/blood , Partial Thromboplastin Time , Prothrombin Time , Pyrazoles/administration & dosage , Pyridones/administration & dosage , Rivaroxaban/administration & dosage , Administration, Oral , Anticoagulants/adverse effects , Anticoagulants/blood , Atrial Fibrillation/blood , Atrial Fibrillation/diagnosis , Biomarkers/blood , Dabigatran/adverse effects , Dabigatran/blood , Dose-Response Relationship, Drug , Drug Monitoring , False Positive Reactions , Humans , Predictive Value of Tests , Pyrazoles/adverse effects , Pyrazoles/blood , Pyridones/adverse effects , Pyridones/blood , Reproducibility of Results , Rivaroxaban/adverse effects , Rivaroxaban/blood
2.
Int J Oncol ; 42(4): 1239-48, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23443741

ABSTRACT

Tumor necrosis factor-like weak inducer of apoptosis (TWEAK), a member of the tumor necrosis factor (TNF) family of cytokines, acts on responsive cells via binding to a cell surface receptor called Fn14. TWEAK binding to an Fn14 receptor or constitutive Fn14 overexpression has been shown to activate nuclear factor κB signaling which is important in tumorigenesis and cancer therapy resistance. In the present study, we demonstrate that TWEAK and Fn14 are expressed in neuroblastoma cell lines and primary tumors, and both are observed at increased levels in high-stage tumors. The treatment of neuroblastoma cell lines with recombinant TWEAK in vitro causes increased survival, and this effect is partially due to the activation of NF-κB signaling. Moreover, TWEAK induces the release of matrix metalloprotease-9 (MMP-9) in neuroblastoma cells, suggesting that TWEAK may play a role in the invasive phase of neuroblastoma tumorigenesis. TWEAK-induced cell survival was significantly reduced by silencing the TWEAK and Fn14 gene functions by siRNA. Thus, the expression of TWEAK and Fn14 in neuroblastoma suggests that TWEAK functions as an important regulator of primary neuroblastoma growth, invasion and survival and that the therapeutic intervention of the TWEAK/Fn14 pathway may be an important clinical strategy in neuroblastoma therapy.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Neuroblastoma/metabolism , Receptors, Tumor Necrosis Factor/metabolism , Tumor Necrosis Factors/metabolism , Cell Line, Tumor , Cell Nucleus/metabolism , Cell Survival , Child , Child, Preschool , Cytokine TWEAK , Enzyme Precursors/metabolism , Female , Gene Expression , Humans , Infant , Infant, Newborn , Male , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9 , NF-kappa B/metabolism , Neuroblastoma/pathology , Protein Transport , TWEAK Receptor
3.
Carcinogenesis ; 34(5): 1081-8, 2013 May.
Article in English | MEDLINE | ID: mdl-23349014

ABSTRACT

Tumor-associated inflammation is a driving force in several adult cancers and intake of low-dose aspirin has proven to reduce cancer incidence. Little is known about tumor-associated inflammation in pediatric neoplasms and no in vivo data exists on the effectiveness of low-dose aspirin on established tumors. The present study employs the transgenic TH-MYCN mouse model for neuroblastoma (NB) to evaluate inflammatory patterns paralleling tumor growth in vivo and low-dose aspirin as a therapeutic option for high-risk NB. Spontaneously arising abdominal tumors were monitored for tumor-associated inflammation ex vivo at various stages of disease and homozygous mice received daily low-dose aspirin (10mg/kg) using oral gavage or no treatment, from 4.5 to 6 weeks of age. Using flow cytometry, a transition from an adaptive immune response predominated by CD8(+) T cell in early neoplastic lesions, towards enrichment in immature cells of the innate immune system, including myeloid-derived suppressor cells, dendritic cells and tumor-associated macrophages, was detected during tumor progression. An M1 to M2 transition of tumor-associated macrophages was demonstrated, paralleled by a deterioration of dendritic cell status. Treatment with low-dose aspirin to mice homozygous for the TH-MYCN transgene significantly reduced the tumor burden (P < 0.01), the presence of tumor-associated cells of the innate immune system (P < 0.01), as well as the intratumoral expression of transforming growth factor-ß, thromboxane A2 (P < 0.05) and prostaglandin D2 (P < 0.01). In conclusion, tumor-associated inflammation appears as a potential therapeutic target in NB and low-dose aspirin reduces tumor burden in the TH-MYCN transgenic mouse model of NB, hence warranting further studies on aspirin in high-risk NB.


Subject(s)
Aspirin/pharmacology , Inflammation/drug therapy , Neuroblastoma/drug therapy , Neuroblastoma/pathology , Animals , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cytokines/genetics , Cytokines/immunology , Cytokines/metabolism , Dendritic Cells/drug effects , Dendritic Cells/immunology , Disease Models, Animal , Disease Progression , Homozygote , Immunity, Innate/drug effects , Immunity, Innate/genetics , Immunity, Innate/immunology , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Macrophages/drug effects , Macrophages/immunology , Macrophages/pathology , Mice , Mice, Transgenic , Neuroblastoma/genetics , Neuroblastoma/immunology , Prostaglandin D2/genetics , Prostaglandin D2/immunology , Prostaglandin D2/metabolism , Th2 Cells/drug effects , Th2 Cells/immunology , Th2 Cells/metabolism , Thromboxane A2/genetics , Thromboxane A2/immunology , Thromboxane A2/metabolism , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/immunology , Transforming Growth Factor beta/metabolism
4.
PLoS One ; 7(1): e29331, 2012.
Article in English | MEDLINE | ID: mdl-22276108

ABSTRACT

BACKGROUND: Prostaglandin E(2) (PGE(2)) is an important mediator in tumor-promoting inflammation. High expression of cyclooxygenase-2 (COX-2) has been detected in the embryonic childhood tumor neuroblastoma, and treatment with COX inhibitors significantly reduces tumor growth. Here, we have investigated the significance of a high COX-2 expression in neuroblastoma by analysis of PGE(2) production, the expression pattern and localization of PGE(2) receptors and intracellular signal transduction pathways activated by PGE(2). PRINCIPAL FINDINGS: A high expression of the PGE(2) receptors, EP1, EP2, EP3 and EP4 in primary neuroblastomas, independent of biological and clinical characteristics, was detected using immunohistochemistry. In addition, mRNA and protein corresponding to each of the receptors were detected in neuroblastoma cell lines. Immunofluorescent staining revealed localization of the receptors to the cellular membrane, in the cytoplasm, and in the nuclear compartment. Neuroblastoma cells produced PGE(2) and stimulation of serum-starved neuroblastoma cells with PGE(2) increased the intracellular concentration of calcium and cyclic AMP with subsequent phosphorylation of Akt. Addition of 16,16-dimethyl PGE(2) (dmPGE(2)) increased cell viability in a time, dose- and cell line-dependent manner. Treatment of neuroblastoma cells with a COX-2 inhibitor resulted in a diminished cell growth and viability that was reversed by the addition of dmPGE(2). Similarly, PGE(2) receptor antagonists caused a decrease in neuroblastoma cell viability in a dose-dependent manner. CONCLUSIONS: These findings demonstrate that PGE(2) acts as an autocrine and/or paracrine survival factor for neuroblastoma cells. Hence, specific targeting of PGE(2) signaling provides a novel strategy for the treatment of childhood neuroblastoma through the inhibition of important mediators of tumor-promoting inflammation.


Subject(s)
16,16-Dimethylprostaglandin E2/pharmacology , Autocrine Communication/drug effects , Dinoprostone/metabolism , Neuroblastoma/metabolism , Biphenyl Compounds/pharmacology , Blotting, Western , Calcium/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cyclic AMP/metabolism , Cyclooxygenase 2 Inhibitors/pharmacology , Humans , Immunohistochemistry , In Vitro Techniques , Phosphorylation/drug effects , Receptors, Prostaglandin E, EP1 Subtype/antagonists & inhibitors , Receptors, Prostaglandin E, EP1 Subtype/metabolism , Receptors, Prostaglandin E, EP2 Subtype/antagonists & inhibitors , Receptors, Prostaglandin E, EP2 Subtype/metabolism , Receptors, Prostaglandin E, EP3 Subtype/antagonists & inhibitors , Receptors, Prostaglandin E, EP3 Subtype/metabolism , Receptors, Prostaglandin E, EP4 Subtype/antagonists & inhibitors , Receptors, Prostaglandin E, EP4 Subtype/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tandem Mass Spectrometry , Thiophenes/pharmacology , Triazoles/pharmacology
5.
PLoS One ; 7(12): e51297, 2012.
Article in English | MEDLINE | ID: mdl-23284678

ABSTRACT

BACKGROUND: The TH-MYCN transgenic neuroblastoma model, with targeted MYCN expression to the developing neural crest, has been used to study neuroblastoma development and evaluate novel targeted tumor therapies. METHODS: We followed tumor development in 395 TH-MYCN (129X1/SvJ) mice (125 negative, 206 hemizygous and 64 homozygous mice) by abdominal palpations up to 40 weeks of age. DNA sequencing of MYCN in the original plasmid construct and mouse genomic DNA was done to verify the accuracy. Copy number analysis with Affymetrix® Mouse Diversity Genotyping Arrays was used to characterize acquired genetic aberrations. RESULTS: DNA sequencing confirmed presence of human MYCN cDNA in genomic TH-MYCN DNA corresponding to the original plasmid construct. Tumor incidence and growth correlated significantly to transgene status with event-free survival for hemizygous mice at 50%, and 0% for homozygous mice. Hemizygous mice developed tumors at 5.6-19 weeks (median 9.1) and homozygous mice at 4.0-6.9 weeks (5.4). The mean treatment window, time from palpable tumor to sacrifice, for hemizygous and homozygous mice was 15 and 5.2 days, respectively. Hemizygous mice developing tumors as early as homozygous mice had a longer treatment window. Age at tumor development did not influence treatment window for hemizygous mice, whereas treatment window in homozygous mice decreased significantly with increasing age. Seven out of 10 analysed tumors had a flat DNA profile with neither segmental nor numerical chromosomal aberrations. Only three tumors from hemizygous mice showed acquired genetic features with one or more numerical aberrations. Of these, one event corresponded to gain on the mouse equivalent of human chromosome 17. CONCLUSION: Hemizygous and homozygous TH-MYCN mice have significantly different neuroblastoma incidence, tumor growth characteristics and treatment windows but overlap in age at tumor development making correct early genotyping essential to evaluate therapeutic interventions. Contrasting previous studies, our data show that TH-MYCN tumors have few genetic aberrations.


Subject(s)
DNA Copy Number Variations , Neuroblastoma/genetics , Neuroblastoma/pathology , Nuclear Proteins/genetics , Oncogene Proteins/genetics , Animals , Breeding , Cell Proliferation , Disease Models, Animal , Female , Hemizygote , Homozygote , Humans , Male , Mice , Mice, Transgenic , N-Myc Proto-Oncogene Protein , Neuroblastoma/diagnostic imaging , Neuroblastoma/therapy , Sequence Analysis, DNA , Transgenes/genetics , Ultrasonography
6.
Proc Natl Acad Sci U S A ; 106(44): 18757-62, 2009 Nov 03.
Article in English | MEDLINE | ID: mdl-19846775

ABSTRACT

There is strong evidence for a role of prostaglandin E(2) (PGE(2)) in cancer cell proliferation and tumor development. In PGE(2) biosynthesis, cyclooxygenases (COX-1/COX-2) convert arachidonic acid to PGH(2), which can be isomerized to PGE(2) by microsomal PGE-synthase-1 (MPGES-1). The human prostate cancer cell line DU145 expressed high amounts of MPGES-1 in a constitutive manner. MPGES-1 expression also was detectable in human prostate cancer tissues, where it appeared more abundant compared with benign hyperplasia. By using shRNA, we established stable and practically complete knockdown of MPGES-1, both in DU145 cells with high constitutive expression and in the non-small cell lung cancer cell line A549, where MPGES-1 is inducible. For microsomes prepared from knockdown clones, conversion of PGH(2) to PGE(2) was reduced by 85-90%. This resulted in clear phenotypic changes: MPGES-1 knockdown conferred decreased clonogenic capacity and slower growth of xenograft tumors (with disintegrated tissue structure) in nude mice. For DU145 cells, MPGES-1 knockdown gave increased apoptosis in response to genotoxic stress (adriamycin), which could be rescued by exogenous PGE(2). The results suggest that MPGES-1 is an alternative therapeutic target in cancer cells expressing this enzyme.


Subject(s)
Intramolecular Oxidoreductases/metabolism , Lung Neoplasms/enzymology , Lung Neoplasms/pathology , Microsomes/enzymology , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/pathology , Animals , Apoptosis/drug effects , Blotting, Western , Cell Line, Tumor , Cell Proliferation/drug effects , Clone Cells , Cyclooxygenase 2/metabolism , Doxorubicin/pharmacology , Gene Knockdown Techniques , Humans , Intramolecular Oxidoreductases/antagonists & inhibitors , Male , Mice , Microsomes/drug effects , Prostaglandin-E Synthases , Protein Transport/drug effects , Receptors, Androgen/metabolism , Subcellular Fractions/drug effects , Subcellular Fractions/enzymology , Tumor Stem Cell Assay , Xenograft Model Antitumor Assays
7.
FASEB J ; 22(10): 3525-36, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18591367

ABSTRACT

The metabolism of arachidonic acid by the cyclooxygenase (COX) or lipoxygenase (LO) pathways generates eicosanoids that have been implicated in the pathogenesis of a variety of human diseases, including cancer. In this study, we examined the expression and significance of components within the 5-LO pathway in human neuroblastoma, an embryonal tumor of the sympathetic nervous system. High expression of 5-LO, 5-LO-activating protein (FLAP), leukotriene A(4) hydrolase, leukotriene C(4) synthase, and leukotriene receptors was detected in a majority of primary neuroblastoma tumors and all cell lines investigated. Expression of 5-LO and FLAP was evident in tumor cells but not in nonmalignant adrenal medulla where neuroblastomas typically arise. Moreover, neuroblastoma cells produce leukotrienes, and stimulation of neuroblastoma cells with leukotrienes increased neuroblastoma cell viability. Inhibitors of 5-LO (AA-861), FLAP (MK-886), or the leukotriene receptor antagonist montelukast inhibited neuroblastoma cell growth by induction of G(1)-cell cycle arrest and apoptosis. Similarly, specific 5-LO and leukotriene receptor silencing by small interfering RNA decreased neuroblastoma cell growth. These findings provide new insights into the pathobiology of neuroblastoma, and the use of leukotriene pathway inhibitors as a novel adjuvant therapy for children with neuroblastoma warrants further consideration.


Subject(s)
Leukotrienes/biosynthesis , Neuroblastoma/metabolism , Neuroblastoma/pathology , 5-Lipoxygenase-Activating Proteins , Apoptosis , Arachidonate 5-Lipoxygenase/biosynthesis , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/biosynthesis , Cell Cycle , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Enzyme Inhibitors/pharmacology , Epoxide Hydrolases/biosynthesis , Glutathione Transferase/biosynthesis , Humans , Leukotriene Antagonists/pharmacology , Lipoxygenase Inhibitors , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/biosynthesis , Neuroblastoma/drug therapy , Receptors, Leukotriene/biosynthesis , Receptors, Leukotriene/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...