Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Int J Mol Med ; 33(4): 795-808, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24424509

ABSTRACT

The (pro)renin receptor [(P)RR] is crucial for cardio-renal pathophysiology. The distinct molecular mechanisms of this receptor are still incompletely understood. The (P)RR is able to interact with different signalling proteins such as promyelocytic leukemia zinc finger protein (PLZF) and Wnt receptors. Moreover, domains of the (P)RR are essential for V-ATPase activity. V-ATPase- and Wnt-mediated effects imply constitutive, i.e., (pro)renin-independent functions of the (P)RR. Regarding ligand-dependent (P)RR signalling, the role of prorenin glycosylation is currently unknown. Therefore, the aim of this study was to analyse the contribution of constitutive (P)RR activity to its cellular effects and the relevance of prorenin glycosylation on its ligand activity. We were able to demonstrate that high glucose induces (P)RR signal transduction whereas deglycosylation of prorenin abolishes its intrinsic activity in neuronal and epithelial cells. By using siRNA against (P)RR or PLZF as well as the PLZF translocation blocker genistein and the specific V-ATPase inhibitor bafilomycin, we were able to dissect three distinct sub-pathways downstream of the (P)RR. The V-ATPase function is ligand-independently associated with strong pro-proliferative effects whereas prorenin causes moderate proliferation in vitro. In contrast, PLZF per se [i.e., in the absence of (pro)renin] does not interfere with cell number.


Subject(s)
Genistein/pharmacology , Kruppel-Like Transcription Factors/metabolism , Macrolides/pharmacology , Receptors, Cell Surface/metabolism , Vacuolar Proton-Translocating ATPases/metabolism , Animals , Cell Count , Cell Line, Tumor , Cell Proliferation/drug effects , Glucose/pharmacology , Glycosylation/drug effects , HEK293 Cells , Humans , Hydrogen-Ion Concentration/drug effects , Kruppel-Like Transcription Factors/genetics , Lysosomes/drug effects , Lysosomes/metabolism , Mice , Models, Biological , Peroxisomes/drug effects , Peroxisomes/metabolism , Promoter Regions, Genetic/genetics , Promyelocytic Leukemia Zinc Finger Protein , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Transport/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering , Receptors, Cell Surface/genetics , Signal Transduction/drug effects , Vacuolar Proton-Translocating ATPases/genetics
2.
Oncol Rep ; 30(1): 462-70, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23604472

ABSTRACT

Alternative splicing is a key regulatory mechanism for cellular metabolism controlling cell proliferation and angiogenesis, both of which are crucial processes for tumorigenesis under hypoxia. Human cells express two tissue factor (TF) isoforms, alternatively spliced TF (asTF) and 'full length' TF (flTF). flTF is the major source of thrombogenicity whereas, the function of soluble asTF, particularly in cancer, is widely unknown. In the present study, we examined the impact of alternative splicing on the pro-angiogenic potential and the TF expression pattern of A549 cells under hypoxia. We focused our efforts toward alternative splicing factors, such as Clk1, and pro-angiogenic proliferation-regulating factors, such as Cyr61. We further examined the influence of asTF overexpression on the expression of MCP-1, Cyr61 and VEGF, as well as on cell number and pro-angiogenic properties of A549 cells. Notably, we found hypoxia to induce the expression of alternative splicing factors (Clk1 and Clk4) as well as proliferation- and angiogenesis-promoting factors (Cyr61 and flTF). asTF overexpression in A549 cells also increased both cell number and tube formation. These effects were mediated by the induction of Cyr61, MCP-1 and VEGF, as well as by integrin α(v)ß(3). Taken together, our results suggest that the pro-angiogenic potential of A549 lung cancer cells is modulated under hypoxic conditions via modulation of TF isoform expression which in turn is controlled by alternative splicing.


Subject(s)
Alternative Splicing/genetics , Cell Hypoxia , Lung Neoplasms/metabolism , Neovascularization, Pathologic/metabolism , Thromboplastin/metabolism , Cell Line, Tumor , Cell Proliferation , Chemokine CCL2/biosynthesis , Chemokine CCL2/metabolism , Cysteine-Rich Protein 61/biosynthesis , Cysteine-Rich Protein 61/metabolism , Humans , Integrin alphaVbeta3/biosynthesis , Integrin alphaVbeta3/metabolism , Protein Isoforms/metabolism , Protein Serine-Threonine Kinases/biosynthesis , Protein-Tyrosine Kinases/biosynthesis , Vascular Endothelial Growth Factor A/biosynthesis , Vascular Endothelial Growth Factor A/metabolism
3.
PLoS One ; 8(3): e57674, 2013.
Article in English | MEDLINE | ID: mdl-23469216

ABSTRACT

The (pro)renin receptor ((P)RR) signaling is involved in different pathophysiologies ranging from cardiorenal end-organ damage via diabetic retinopathy to tumorigenesis. We have previously shown that the transcription factor promyelocytic leukemia zinc finger (PLZF) is an adaptor protein of the (P)RR. Furthermore, recent publications suggest that major functions of the (P)RR are mediated ligand-independently by its transmembrane and intracellular part, which acts as an accessory protein of V-ATPases. The transcriptome and recruitmentome downstream of the V-ATPase function and PLZF in the context of the (P)RR are currently unknown. Therefore, we performed a set of microarray and chromatin-immunoprecipitation (ChIP)-chip experiments using siRNA against the (P)RR, stable overexpression of PLZF, the PLZF translocation inhibitor genistein and the specific V-ATPase inhibitor bafilomycin to dissect transcriptional pathways downstream of the (P)RR. We were able to identify distinct and overlapping genetic signatures as well as novel real-time PCR-validated target genes of the different molecular functions of the (P)RR. Moreover, bioinformatic analyses of our data confirm the role of (P)RRs signal transduction pathways in cardiovascular disease and tumorigenesis.


Subject(s)
Cardiovascular Diseases/genetics , Cell Transformation, Neoplastic/genetics , Kruppel-Like Transcription Factors/genetics , Receptors, Cell Surface/genetics , Transcriptome , Vacuolar Proton-Translocating ATPases/genetics , Cardiovascular Diseases/metabolism , Cell Line, Tumor , Cell Transformation, Neoplastic/metabolism , Chromatin Immunoprecipitation , Gene Expression Regulation/drug effects , Genistein/pharmacology , HEK293 Cells , Humans , Kruppel-Like Transcription Factors/antagonists & inhibitors , Kruppel-Like Transcription Factors/metabolism , Macrolides/pharmacology , Oligonucleotide Array Sequence Analysis , Promyelocytic Leukemia Zinc Finger Protein , Protein Kinase Inhibitors/pharmacology , RNA, Small Interfering/genetics , Receptors, Cell Surface/antagonists & inhibitors , Receptors, Cell Surface/metabolism , Signal Transduction/drug effects , Vacuolar Proton-Translocating ATPases/antagonists & inhibitors , Vacuolar Proton-Translocating ATPases/metabolism , Prorenin Receptor
4.
J Neurosci ; 32(47): 16807-20, 2012 Nov 21.
Article in English | MEDLINE | ID: mdl-23175834

ABSTRACT

Genetic factors strongly contribute to the pathogenesis of sporadic Alzheimer's disease (AD). Nevertheless, genome-wide association studies only yielded single nucleotide polymorphism loci of moderate importance. In contrast, microsatellite repeats are functionally less characterized structures within our genomes. Previous work has shown that endothelin-converting enzyme-1 (ECE-1) is able to reduce amyloid ß content. Here we demonstrate that a CpG-CA repeat within the human ECE-1c promoter is highly polymorphic, harbors transcriptional start sites, is able to recruit the transcription factors poly(ADP-ribose) polymerase-1 and splicing factor proline and glutamine-rich, and is functional regarding haplotype-specific promoter activity. Furthermore, genotyping of 403 AD patients and 444 controls for CpG-CA repeat length indicated shifted allelic frequency distributions. Sequencing of 245 haplotype clones demonstrated that the overall CpG-CA repeat composition of AD patients and controls is distinct. Finally, we show that human and chimpanzee [CpG](m)-[CA](n) ECE-1c promoter repeats are genetically and functionally distinct. Our data indicate that a short genomic repeat structure constitutes a novel core promoter element, coincides with human evolution, and contributes to the pathogenesis of AD.


Subject(s)
Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Aspartic Acid Endopeptidases/genetics , Biological Evolution , Metalloendopeptidases/genetics , Microsatellite Repeats/genetics , Promoter Regions, Genetic/genetics , Transcription, Genetic/genetics , Animals , Blotting, Western , Cardiovascular Diseases/genetics , Cardiovascular Diseases/physiopathology , Chromatography, Gel , DNA/genetics , DNA/isolation & purification , Electrophoretic Mobility Shift Assay , Endothelin-Converting Enzymes , Genotype , Humans , Nuclease Protection Assays , Pan troglodytes , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/genetics , RNA/genetics , RNA/isolation & purification , Real-Time Polymerase Chain Reaction
5.
Biochem Pharmacol ; 84(12): 1643-50, 2012 Dec 15.
Article in English | MEDLINE | ID: mdl-23022225

ABSTRACT

The (pro)renin receptor ((P)RR) and Wnt signalling are both involved in different diseases ranging from cardiac and renal end-organ damage to cancer. (P)RR function involves signalling via the transcription factor promyelocytic leukemia zinc finger protein (PLZF) as well as the furin-mediated generation of vacuolar proton-translocating ATPase (V-ATPase)-associated and soluble (P)RR isoforms. Recently, the (P)RR was described as adaptor protein of Wnt (co)receptors. The aim of this study was to analyse the contribution of these distinct (P)RR functions to Wnt signalling. Using Tcf/Lef reporter gene systems in HEK293T and HepG2 cells and quantification of endogenous axin2 mRNA and protein levels in HEK293T cells we were able to demonstrate that full-length (P)RR acts as a repressor of Wnt signalling in a system preactivated either by Wnt3a stimulation or by constitutively active ß-catenin. These repressive effects are mediated by Dvl but are independent of the mutation status of ß-catenin. Furthermore, the V-ATPase complex, but not PLZF translocation or renin enzymatic activity, is necessary for the induction of Tcf/Lef-responsive genes by Wnt3a. Our data indicate interference of (P)RR and Wnt cascades, a fact that has to be considered concerning pathophysiology of cardio-renal and oncological entities as well as in drug development programs targeting (P)RR or Wnt pathways.


Subject(s)
Receptors, Cell Surface/physiology , Signal Transduction/physiology , Wnt Proteins/metabolism , Animals , Base Sequence , Cell Line , DNA Primers , Genistein/pharmacology , HEK293 Cells , Hep G2 Cells , Humans , Macrolides/pharmacology , Mice , RNA, Small Interfering , Real-Time Polymerase Chain Reaction , Prorenin Receptor
6.
Pharmacology ; 90(5-6): 307-15, 2012.
Article in English | MEDLINE | ID: mdl-23037500

ABSTRACT

BACKGROUND/AIMS: Putative in vitro-in vivo correlations of pharmacokinetic (PK) parameters are regarded as a prerequisite to filter hits derived from high-throughput screening (HTS) approaches for subsequent murine in vivo PK studies. METHODS: In this study, we assessed stabilities in rat and human microsomes of 121 compounds from an early, academic drug discovery programme targeting the (pro)renin receptor and correlated the respective data with single-dose, in vivo PK parameters of 22 hits administered intravenously in rats. RESULTS: After transformation of in vitro half-lives to predicted in vivo hepatic clearances, r(2) regarding in vitro-in vivo clearance correlations were 0.31 and 0.27 for the rat and human species, respectively. CONCLUSIONS: Our data concerning structurally diverse real-world compounds indicate that microsomal stability testing is not a tool to triage early compounds for in vivo PK testing.


Subject(s)
Drug Evaluation, Preclinical/methods , Microsomes, Liver/metabolism , Pharmaceutical Preparations/metabolism , Pharmacokinetics , Animal Testing Alternatives/methods , Animals , Cells, Cultured , Half-Life , Humans , Male , Rats , Rats, Sprague-Dawley , Reproducibility of Results
7.
Cardiovasc Revasc Med ; 12(1): 47-55, 2011.
Article in English | MEDLINE | ID: mdl-21241972

ABSTRACT

BACKGROUND: Late thrombotic events are important complications associated with intracoronary brachytherapy (ICBT) using ionizing radiation (IR) or with antiproliferative treatment modalities such as drug-eluting stents (DES). The mechanism mediating these thrombotic events is not well understood. This study assessed the effect of prolonged clopidogrel treatment on tissue factor (TF) expression in coronary arteries and on the circulating TF level after percutaneous transluminal coronary angioplasty /ICBT in a porcine coronary model. METHODS: Pigs were treated with aspirin plus a 300 mg loading dose of clopidogrel one day before percutaneous coronary intervention (PCI), followed by a daily dose of clopidogrel and aspirin. During PCI one of the two balloon-injured arteries was treated by brachytherapy. Animals were sacrificed at different time points. The pigs, which were sacrificed 3 months post-PCI, were divided into two groups (Group I: clopidogrel for 3 months; Group II: clopidogrel for 1 month). Plasma TF was measured by enzyme-linked immunosorbent assay in blood samples taken from all pigs before and immediately after intervention and before sacrifice. Morphometric analysis was performed on digitalized images employing the software LUCIA G for TF staining. Vascular TF expression levels were assessed by quantitative real-time polymerase chain reaction. RESULTS: Prolonged clopidogrel application significantly reduced coronary TF at the protein (Group I vs. II, 8.975 ± 3.947% vs. 26.44 ± 5.375%, P = .007) and mRNA level [Group I vs. II, (0.3501 ± 0.0519) × 10(-3) vs. (0.7073 ± 0.0436) × 10(-3), P<.0005]. Circulating TF protein tended to be lower after 3 months than after 1 month clopidogrel treatment post-PCI (Group I vs. Group II, 488.3 ± 35.37 pg/ml vs. 572.3 ± 39.9 pg/ml, P = .130). CONCLUSIONS: Prolonged clopidogrel treatment reduced coronary TF expression and tended to reduce the blood TF level post-PCI, thus possibly modulating the risk of late thrombosis.


Subject(s)
Angioplasty, Balloon, Coronary , Coronary Thrombosis/prevention & control , Coronary Vessels/drug effects , Platelet Aggregation Inhibitors/administration & dosage , Thromboplastin/metabolism , Ticlopidine/analogs & derivatives , Angioplasty, Balloon, Coronary/adverse effects , Animals , Aspirin/administration & dosage , Brachytherapy/adverse effects , Clopidogrel , Coronary Thrombosis/etiology , Coronary Thrombosis/metabolism , Coronary Thrombosis/pathology , Coronary Vessels/metabolism , Coronary Vessels/pathology , Drug Administration Schedule , Drug Therapy, Combination , Enzyme-Linked Immunosorbent Assay , Fibrin/metabolism , Fibrinogen/metabolism , Immunohistochemistry , Models, Animal , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sus scrofa , Thromboplastin/genetics , Ticlopidine/administration & dosage , Time Factors , Up-Regulation
8.
Brain Pathol ; 21(1): 31-43, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20731660

ABSTRACT

Stroke is one of the major medical burdens in industrialized countries. Animal experiments indicate that blockade of the angiotensin AT1 receptor (AT1R) improves neurological outcome after cerebral ischemia. These protective effects are partially mediated by the angiotensin AT2 receptor (AT2R). The transcription factor promyelocytic leukemia zinc finger (PLZF) was identified as a direct adapter protein of the AT2R. Furthermore, our group was able to demonstrate that PLZF also directly binds and mediates the effects of the human (pro)renin receptor [(P)RR] which is involved in brain development. Therefore, we hypothesized that PLZF is involved in neuroprotection. Here we show that PLZF and its receptors (P)RR and AT2R exhibited an ubiquitous expression pattern in different brain regions. Furthermore, stable PLZF overexpression in human neuronal cells was able to mediate neuroprotection in a glutamate toxicity model in vitro. Consistently, PLZF mRNA and protein were downregulated on the ipsilateral side in a stroke model in vivo, whereas the neurodetrimental PLZF target genes cyclin A2 and BID were upregulated under this condition. Further analyses indicated that the neuroprotective AT2R is upregulated upon stable PLZF overexpression in cultured neuronal cells. Finally, reporter gene assays demonstrated the functionality of (P)RR promoter polymorphisms regarding basal and PLZF-induced activity.


Subject(s)
Cerebral Cortex/metabolism , Infarction, Middle Cerebral Artery/metabolism , Kruppel-Like Transcription Factors/metabolism , Neurons/metabolism , Receptor, Angiotensin, Type 2/metabolism , Zinc Fingers/genetics , Animals , BH3 Interacting Domain Death Agonist Protein/genetics , BH3 Interacting Domain Death Agonist Protein/metabolism , Blotting, Western , Cell Line, Tumor , Cells, Cultured , Cerebral Cortex/pathology , Cyclin A2/genetics , Cyclin A2/metabolism , Down-Regulation/genetics , Humans , Infarction, Middle Cerebral Artery/genetics , Infarction, Middle Cerebral Artery/pathology , Kruppel-Like Transcription Factors/genetics , Magnetic Resonance Imaging , Male , Neurons/pathology , Promyelocytic Leukemia Zinc Finger Protein , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Wistar , Receptor, Angiotensin, Type 1/genetics , Receptor, Angiotensin, Type 1/metabolism , Receptor, Angiotensin, Type 2/genetics , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Prorenin Receptor
9.
Biochem Pharmacol ; 77(12): 1795-805, 2009 Jun 15.
Article in English | MEDLINE | ID: mdl-19344625

ABSTRACT

The renin-angiotensin system (RAS) plays a crucial role in cardiovascular and neuronal (patho-)physiology. The angiotensin AT2 receptor (AT2R) seems to counteract the proinflammatory, prohypertrophic and profibrotic actions of the AT1 receptor. Recently, we identified a novel protein, termed "AT2R binding protein" (ATBP/ATIP) which seems essential for AT2R-mediated growth inhibition. Poly(ADP-ribose) polymerase-1 (PARP-1) can act as a nuclear integrator of angiotensin II-mediated cell signalling, and has been implicated in the pathogenesis of cardiovascular and neuronal disease. In this study, promoters of human AT2R and ATIP1 were cloned and two transcriptional start sites in the ATIP1 promoter were identified whereas only one was detected in the AT2R promoter. Promoter assays indicated that the exon 1-intron 1 region of AT2R is necessary and sufficient for AT2R promoter activity. Inverse cloning experiments indicated that this regulatory region is a promoter but not an enhancer element implicating (a) further start site(s) in this region. Consistently, the exon 1-intron 1 region of AT2R was shown to tether the basal transcriptional machinery. Overexpression, pharmacological inhibition and ablation of PARP demonstrated that PARP-1 activates the ATIP1 gene but represses the AT2R on promoter and mRNA levels in vitro, and in brain tissue in vivo. Additional experiments indicated that AT2R activation does not modulate PARP-1 transcript levels but increases AT2R promoter activity, thereby creating a positive feedback mechanism. Our results demonstrate that PARP-1 acts as novel node within the RAS network based on its ability to regulate downstream targets such as AT2R and its adapter protein ATBP.


Subject(s)
Gene Expression Regulation , Poly(ADP-ribose) Polymerases/physiology , Receptor, Angiotensin, Type 2/genetics , Cell Line, Tumor , Humans , Membrane Transport Proteins , RNA, Messenger , Transcription, Genetic
10.
Oncol Rep ; 20(1): 123-8, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18575726

ABSTRACT

Non-small cell lung cancer (NSCLC) comprises of 75% of all lung cancers. Human full length tissue factor (flHTF), the physiological initiator of blood coagulation, is aberrantly expressed in certain solid tumors. FlHTF and its soluble isoform, alternatively spliced human tissue factor (asHTF), have been shown to contribute to thrombogenicity of the blood of healthy individuals. The aim of this study was to quantify flHTF and asHTF on mRNA and protein levels (using immunohistochemistry, immunoblotting, and ELISA) on a panel of human NSCLC tissue and plasma specimens. The tissue factor (TF) expression of 21 pulmonary adenomatous (AC) and 12 normal healthy tissues was assessed by real-time qRT-PCR. The TF protein concentration was quantified by ELISA in a subset of 11 AC and 9 normal tissue specimens as well as in the plasma of 13 lung cancer patients and 15 healthy controls. We found a significant increase in the ratio of flHTF/HGAPDH mRNA in AC (0.24+/-0.06 vs. 0.07+/-0.01; p=0.02 vs. controls) and in asHTF/HGAPDH mRNA (0.027+/-0.01 vs. 0.004+/-0.001; p=0.03 AC vs. controls). AsHTF mRNA expression was significantly lower in patients with stage IA disease compared to patients with higher grade stages, pointing to TF as being a marker of malignancy and metastases. TF protein of lung tumors was significantly increased in AC (p=0.004 vs. controls). TF in plasma was up-regulated in lung cancer patients (334.9+/-95.4 vs. 124.1+/-14.8 pg/ml; p=0.02 vs. controls). Immunohistochemical and immunoblotting data are in line with the increased TF expression, showing elevated blood thrombogenicity of NSCLC patients. The up-regulation of flHTF and, especially, asHTF in AC suggests not only a raised risk of thrombosis, but also of tumor progression, thereby, indicating a poor prognosis in these patients.


Subject(s)
Carcinoma, Non-Small-Cell Lung/chemistry , Lung Neoplasms/chemistry , Thromboplastin/analysis , Thrombosis/etiology , Adenocarcinoma/chemistry , Blotting, Western , Carcinoma, Non-Small-Cell Lung/pathology , Humans , Immunohistochemistry , Lung Neoplasms/pathology , Neoplasm Metastasis , RNA, Messenger/analysis , Thromboplastin/genetics , Thromboplastin/physiology
11.
Int Immunopharmacol ; 8(2): 307-11, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18182245

ABSTRACT

BACKGROUND: Intracoronary brachytherapy as well as rapamycin or paclitaxel coated stents reduce restenosis rates after stenting, but are associated with an increased risk of late stent thrombosis. Tissue factor (TF) may contribute to late thrombosis. This study examined the impact of rapamycin and paclitaxel on TF expression and compares the TF activity induction of both drugs and irradiation in SMCs. METHODS: SMCs were stimulated with rapamycin, paclitaxel or irradiation. Real-time PCR, a chromogenic TF activity assay and Western blotting were done. RESULTS: Rapamycin or paclitaxel increased the TF mRNA expression 5-fold and the TF activity 4- to 6-fold with a maximum at 5 h. Irradiation induced TF activity 2- to 4-fold with a maximum at 7 days. CONCLUSION: The fast increase of TF expression in SMCs post rapamycin and paclitaxel treatment may explain acute stent thrombosis when anti-thrombotic therapies are withdrawn, whereas the irradiation induced long term increase of cellular thrombogenicity may contribute to late thrombosis post intracoronary brachytherapy. Therapies counteracting these side effects may reduce the risk of thrombotic complications after the coronary application of anti-proliferative therapies.


Subject(s)
Muscle, Smooth, Vascular/drug effects , Paclitaxel/pharmacology , Sirolimus/pharmacology , Thromboplastin/genetics , Thrombosis/etiology , Cells, Cultured , Humans , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/radiation effects , Thromboplastin/analysis , Up-Regulation
12.
Cardiovasc Revasc Med ; 8(3): 183-8, 2007.
Article in English | MEDLINE | ID: mdl-17765648

ABSTRACT

OBJECTIVE: We determined the effect of prolonged treatment with clopidogrel on C-reactive protein (CRP) concentrations and blood thrombogenicity after percutaneous transluminal coronary angioplasty followed by intracoronary brachytherapy in the porcine model. ANIMAL MODEL: All 48 pigs received antiplatelet therapy, including aspirin (325 mg, daily) and clopidogrel (300 mg, loading dose) 1 day before PCI, followed by a daily dose of clopidogrel (75 mg/day) in addition to aspirin. During PCI, one of two balloon-injured arteries was randomly assigned to receive immediate radiation treatment. Animals were sacrificed after 24 h, 1 month, and 3 months post-PCI. The pigs, which were sacrificed 3 months post-PCI, were divided into two groups. The first group received clopidogrel in addition to aspirin for 3 months, and the second group received clopidogrel in addition to aspirin for only 1 month after PCI and then aspirin alone. METHODS: Blood was taken from all pigs before intervention, immediately after intervention, and before sacrifice. Serum CRP was measured by enzyme-linked immunosorbent assay. To analyze the procoagulant effects of PCI on blood thrombogenicity, a one-stage clotting assay was performed. RESULTS: Clopidogrel treatment for 3 months reduced CRP levels more than did clopidogrel therapy for 1 month only at 3 months post-PCI (27.9+/-3.9 vs. 56.6+/-11.3 microg/ml; P=.019). Baseline CRP levels were found to be 50.4+/-4.8 microg/ml. Plasma clotting was not affected by prolonged clopidogrel therapy (322.8+/-59.3 s vs. 295.2+/-52.5 s; P=ns). CONCLUSIONS: Prolonged treatment with clopidogrel reduced CRP levels post-PCI.


Subject(s)
Angioplasty, Balloon, Coronary/adverse effects , Anti-Inflammatory Agents/pharmacology , Blood Coagulation/drug effects , Brachytherapy/adverse effects , C-Reactive Protein/metabolism , Inflammation/prevention & control , Platelet Aggregation Inhibitors/pharmacology , Ticlopidine/analogs & derivatives , Animals , Anti-Inflammatory Agents/therapeutic use , Aspirin/pharmacology , Clopidogrel , Inflammation/blood , Inflammation/etiology , Models, Animal , Platelet Aggregation Inhibitors/therapeutic use , Sus scrofa , Ticlopidine/pharmacology , Ticlopidine/therapeutic use , Time Factors , Treatment Outcome
13.
Haematologica ; 92(8): 1091-8, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17640852

ABSTRACT

BACKGROUND AND OBJECTIVES: Ionizing radiation (IR) is associated with thrombotic vascular occlusion predicting a poor clinical outcome. Our study examined whether IR induced tissue factor (TF) expression and procoagulability. We further investigated coordinated gene alterations associated with TF upregulation in the myelomonocytic leukemia THP-1 cells. DESIGN AND METHODS: TF expression was determined by quantitative Reverse Transcriptase (TaqMan) PCR, TF ELISA and TF activity by a two stage chromogenic assay in the time course of days 1, 3, 7, 10, and 17 post IR. To detect IR-induced alterations in gene expression, Affymetrix HG U133 Plus 2.0 microarrays were used. RESULTS IR induced a significant increase in TF/GAPDH mRNA ratios and cellular TF protein on days 3 and 7 post IR (20 Gy [p>or=0.01] and 40 Gy [p or=0.001] vs. control respectively), suggesting IR immediately alters the cellular thrombogenicity. TF upregulation post IR was confirmed in PBMNCs. Gene expression profiling showed IR increased the expression of inflammatory and apoptosis-related pathways known to be involved in the regulation of TF expression. INTERPRETATION AND CONCLUSIONS: TF upregulation together with inflammation and apoptosis may increase the thrombogenicity of tissues. The demonstrated upregulation of TF might play a pivotal role in radiation associated thrombosis.


Subject(s)
Blood Coagulation Factors/biosynthesis , Gene Expression Regulation, Leukemic/radiation effects , Leukemia, Myelomonocytic, Acute/pathology , Neoplasm Proteins/biosynthesis , Thrombophilia/etiology , Thromboplastin/biosynthesis , Apoptosis/genetics , Apoptosis/radiation effects , Blood Coagulation Factors/genetics , Blood Coagulation Factors/radiation effects , Cell Line, Tumor/metabolism , Cell Line, Tumor/radiation effects , Enzyme-Linked Immunosorbent Assay , Factor Xa/biosynthesis , Gene Expression Profiling , Humans , Inflammation , Leukemia, Myelomonocytic, Acute/blood , Leukemia, Myelomonocytic, Acute/complications , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Nitriles/pharmacology , Oligonucleotide Array Sequence Analysis , Particle Accelerators , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Neoplasm/biosynthesis , RNA, Neoplasm/genetics , Radiation, Ionizing , Reverse Transcriptase Polymerase Chain Reaction , Sulfones/pharmacology , Thromboplastin/genetics
14.
Thromb Res ; 120(6): 857-64, 2007.
Article in English | MEDLINE | ID: mdl-17337288

ABSTRACT

INTRODUCTION: The therapeutic application of ionizing radiation is associated with thrombotic events, but the exact underlying molecular mechanisms are still unclear. Tissue factor (TF), the primary initiator of blood coagulation, is essentially involved in the pathophysiology of thrombosis. Circulating monocytes have been identified to upregulate TF under inflammatory conditions and, thereby, enhance blood thrombogenicity. The study examines the effect of irradiation on the cellular procoagulability and TF protein expression of human peripheral blood mononuclear cells (PBMNCs) in a time period of 7 days. MATERIALS AND METHODS: Human PBMNCs were irradiated with 20 Gy. Procoagulability of PBMNCs, released microparticles and microparticle-free cell supernatant was analyzed by a chromogenic assay and TF protein expression quantified by TF ELISA. To determine whether irradiated PBMNCs and shed microparticles initiate plasma clotting, a one stage clotting assay was performed. RESULTS: We found a significant increase of PBMNC-associated procoagulant activity over a time period of 7 days post irradiation. Moreover, 3 days post irradiation PBMNCs initiated the plasma clotting faster than non-irradiated cells. An enhanced cellular TF protein concentration was persistently observed throughout the investigated time up to 7 days post irradiation. Microparticle-associated TF activity significantly increased 3 days post irradiation compared with the non-irradiated controls. PBMNC-derived microparticles post irradiation also initiated the plasma clotting faster than microparticles derived from controls. CONCLUSIONS: The results show irradiation to induce TF expression and to increase procoagulability of PBMNCs and cell-derived microparticles. This could be a possible mechanism by which ionizing radiation enhances blood thrombogenicity.


Subject(s)
Blood Coagulation/immunology , Blood Coagulation/radiation effects , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/radiation effects , Thromboplastin/metabolism , Cells, Cultured , Humans , Leukocytes, Mononuclear/cytology , Particle Size , Radiation, Ionizing , Thrombosis/etiology , Thrombosis/immunology , Time Factors , Up-Regulation/immunology , Up-Regulation/radiation effects
15.
Cardiovasc Res ; 73(4): 806-12, 2007 Mar 01.
Article in English | MEDLINE | ID: mdl-17266944

ABSTRACT

OBJECTIVES: The aim of this study was to investigate the effect of the antioxidants pyrrolidine dithiocarbamate (PDTC) and N-acetylcysteine (NAC) on the ionizing radiation (IR)- and tumor necrosis factor-alpha (TNF-alpha) induced tissue factor (TF) expression and its release from human umbilical vein endothelial cells (HUVECs). METHODS: HUVECs were irradiated with a single dose of either 5 Gy or 10 Gy and stimulated with TNF-alpha (10 ng/mL) in the presence or absence of PDTC and NAC, respectively. Quantitative real-time PCR, ELISA, and TF activity measurements were performed, including TF activity in the supernatant. Apoptosis was detected by flow cytometric active caspase-3 measurement and formation of reactive oxygen species (ROS) by chemiluminescence. RESULTS: We demonstrated a thus far uninvestigated persistent induction of TF expression in HUVECs after treatment with IR and TNF-alpha. Combined stimulation with IR and TNF-alpha led to an immense shedding of microparticle-associated TF which was positively correlated with apoptosis and ROS formation. Antioxidative pre-treatment reduced not only apoptosis and ROS formation, but also the release of thrombogenic microparticles. CONCLUSIONS: Antioxidative treatment inhibited apoptosis and shedding of microparticles, thereby reducing thrombogenicity. Thus, antioxidants may help to prevent late thrombosis after antiproliferative treatment when used in combination with anticoagulants.


Subject(s)
Acetylcysteine/pharmacology , Antioxidants/pharmacology , Endothelial Cells/metabolism , Pyrrolidines/pharmacology , Thiocarbamates/pharmacology , Thromboplastin/metabolism , Analysis of Variance , Apoptosis , Caspase 3/analysis , Cells, Cultured , Endothelial Cells/drug effects , Endothelial Cells/radiation effects , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Luminescent Measurements , Radiation, Ionizing , Reactive Oxygen Species/analysis , Reactive Oxygen Species/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Thromboplastin/analysis , Tumor Necrosis Factor-alpha/pharmacology , Umbilical Veins
17.
J Am Coll Cardiol ; 45(7): 1081-9, 2005 Apr 05.
Article in English | MEDLINE | ID: mdl-15808768

ABSTRACT

OBJECTIVES: We investigated the myocardial localization and expression of tissue factor (TF) and alternatively spliced human tissue factor (asHTF) in patients with dilated cardiomyopathy (DCM). BACKGROUND: Tissue factor is expressed in cardiac muscle and may play a role in maintaining myocardial structure. METHODS: Myocardial biopsies were obtained from patients with a normal or mildly impaired ejection fraction (EF) (> or =50%) and moderate to severely reduced EF (<50%). Explanted DCM hearts were also examined. Myocardial TF expression level was assessed by real-time polymerase chain reaction, TF protein by enzyme-linked immunosorbent assay, and localization by immunohistochemistry. RESULTS: We report the identification of asHTF in the human myocardium: it was located in cardiomyocytes and endothelial cells. Quantification of myocardial TF messenger ribonucleic acid in DCM revealed a decrease in the TF/glyceraldehyde-3-phosphate dehydrogenase (GAPDH) ratio (1.76 x 10(-1) +/- 6.08 x 10(-2) for EF > or =50% [n = 19] vs. 1.06 x 10(-1) +/- 5.26 x 10(-2) for EF <50% [n = 27]; p < 0.001) and asHTF/GAPDH ratio (13.91 x 10(-5) +/- 11.20 x 10(-5) for EF > or =50% vs. 7.17 x 10(-5) +/- 3.82 x 10(-5) for EF <50%; p = 0.014). Tissue factor isoform expression level was also decreased in explanted DCM hearts (p < 0.01; n = 12). Total TF protein was reduced by 26% in DCM (p < 0.05). The TF/GAPDH ratio correlated positively with the EF (r = 0.504, p < 0.0001). Immunohistochemistry showed TF localized to the sarcolemma and Z-bands of the cardiomyocytes in patients with normal EF, whereas TF was found in the cardiomyocytic cytosol around the nucleus in DCM. CONCLUSIONS: Tissue factor was down-regulated in the myocardium of DCM patients. The reduction in TF expression and change in localization may influence cell-to-cell contact stability and contractility, thereby contributing to cardiac dysfunction in DCM.


Subject(s)
Cardiomyopathy, Dilated/metabolism , Thromboplastin/metabolism , Blotting, Western , Cardiomyopathy, Dilated/pathology , Case-Control Studies , DNA Primers , Female , Gene Expression , Humans , Immunohistochemistry , Male , Middle Aged , Myocardium/metabolism , Myocardium/pathology , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction , Severity of Illness Index , Thromboplastin/genetics
18.
Clin Cancer Res ; 10(5): 1605-12, 2004 Mar 01.
Article in English | MEDLINE | ID: mdl-15014010

ABSTRACT

PURPOSE: Inconsistent reports on the detection of melanoma cells in peripheral blood by reverse transcriptase-PCR (RT-PCR) have resulted in uncertainty on the prognostic value of circulating melanoma cells. EXPERIMENTAL DESIGN: We developed real-time RT-PCR assays for quantitation of tyrosinase, MelanA/MART1, and gp100 and for porphobilinogen deaminase housekeeping gene. Melanoma tissue (n = 18), peripheral blood samples from healthy donors (n = 21), and patients with cutaneous (n = 122) and uveal (n = 64) melanoma from our institution were analyzed. For quality control, an additional 251 samples from ongoing multicenter studies were compared with in-house samples. RESULTS: Tyrosinase was not detected in healthy donor blood samples. For the two other markers, cutoff values had to be defined to distinct patient samples from controls. Patients with stage IV uveal and cutaneous melanoma expressed all three markers more frequently and at higher levels in peripheral blood as compared with earlier stages. The variation of expression was 4 logs and correlated with tumor load and serum lactate dehydrogenase. In 2 of 3 uveal melanoma patients, detection of circulating tumor cells preceded the development of liver metastases. The diagnostic sensitivity was optimal in blood samples containing >0.1pg/ microl porphobilinogen deaminase (95.7% of in-house samples and 57.4% of multicenter samples). CONCLUSIONS: Real-time RT-PCR is able to quantitatively define the quality of a sample and provides quantitative data for melanoma markers. Disparities in the results of previous studies may be attributable to undetected differences in sample quality. The prognostic relevance of this assay is currently under evaluation in several prospective randomized trials.


Subject(s)
Eye Neoplasms/pathology , Melanoma/pathology , Antigens, Neoplasm/genetics , Base Sequence , DNA Primers , Eye Neoplasms/blood , Eye Neoplasms/genetics , Humans , Hydroxymethylbilane Synthase/genetics , MART-1 Antigen , Melanoma/blood , Melanoma/genetics , Membrane Glycoproteins/blood , Membrane Glycoproteins/genetics , Monophenol Monooxygenase/genetics , Neoplasm Proteins/blood , Neoplasm Proteins/genetics , Plasmids/genetics , Reference Values , Reverse Transcriptase Polymerase Chain Reaction/methods , Transcription, Genetic , gp100 Melanoma Antigen
SELECTION OF CITATIONS
SEARCH DETAIL