Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
IEEE Trans Biomed Eng ; 71(6): 1980-1992, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38498749

ABSTRACT

OBJECTIVE: This study aims to explore the potential of organic electrolytic photocapacitors (OEPCs), an innovative photovoltaic device, in mediating the activation of native voltage-gated Cav1.2 channels (ICa,L) in Guinea pig ventricular cardiomyocytes. METHODS: Whole-cell patch-clamp recordings were employed to examine light-triggered OEPC mediated ICa,L activation, integrating the channel's kinetic properties into a multicompartment cell model to take intracellular ion concentrations into account. A multidomain model was additionally incorporated to evaluate effects of OEPC-mediated stimulation. The final model combines external stimulation, multicompartmental cell simulation, and a patch-clamp amplifier equivalent circuit to assess the impact on achievable intracellular voltage changes. RESULTS: Light pulses activated ICa,L, with amplitudes similar to voltage-clamp activation and high sensitivity to the L-type Ca2+ channel blocker, nifedipine. Light-triggered ICa,L inactivation exhibited kinetic parameters comparable to voltage-induced inactivation. CONCLUSION: OEPC-mediated activation of ICa,L demonstrates their potential for nongenetic optical modulation of cellular physiology potentially paving the way for the development of innovative therapies in cardiovascular health. The integrated model proves the light-mediated activation of ICa,L and advances the understanding of the interplay between the patch-clamp amplifier and external stimulation devices. SIGNIFICANCE: Treating cardiac conduction disorders by minimal-invasive means without genetic modifications could advance therapeutic approaches increasing patients' quality of life compared with conventional methods employing electronic devices.


Subject(s)
Calcium Channels, L-Type , Computer Simulation , Myocytes, Cardiac , Animals , Guinea Pigs , Myocytes, Cardiac/physiology , Calcium Channels, L-Type/metabolism , Patch-Clamp Techniques , Models, Cardiovascular , Action Potentials/physiology , Action Potentials/radiation effects , Light
2.
Proc Natl Acad Sci U S A ; 120(28): e2210152120, 2023 07 11.
Article in English | MEDLINE | ID: mdl-37406102

ABSTRACT

Sepsis has emerged as a global health burden associated with multiple organ dysfunction and 20% mortality rate in patients. Numerous clinical studies over the past two decades have correlated the disease severity and mortality in septic patients with impaired heart rate variability (HRV), as a consequence of impaired chronotropic response of sinoatrial node (SAN) pacemaker activity to vagal/parasympathetic stimulation. However, the molecular mechanism(s) downstream to parasympathetic inputs have not been investigated yet in sepsis, particularly in the SAN. Based on electrocardiography, fluorescence Ca2+ imaging, electrophysiology, and protein assays from organ to subcellular level, we report that impaired muscarinic receptor subtype 2-G protein-activated inwardly-rectifying potassium channel (M2R-GIRK) signaling in a lipopolysaccharide-induced proxy septic mouse model plays a critical role in SAN pacemaking and HRV. The parasympathetic responses to a muscarinic agonist, namely IKACh activation in SAN cells, reduction in Ca2+ mobilization of SAN tissues, lowering of heart rate and increase in HRV, were profoundly attenuated upon lipopolysaccharide-induced sepsis. These functional alterations manifested as a direct consequence of reduced expression of key ion-channel components (GIRK1, GIRK4, and M2R) in the mouse SAN tissues and cells, which was further evident in the human right atrial appendages of septic patients and likely not mediated by the common proinflammatory cytokines elevated in sepsis.


Subject(s)
Lipopolysaccharides , Sepsis , Humans , Animals , Mice , Lipopolysaccharides/toxicity , Lipopolysaccharides/metabolism , Sinoatrial Node/physiology , G Protein-Coupled Inwardly-Rectifying Potassium Channels/genetics , G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , Signal Transduction/physiology , Sepsis/chemically induced , Sepsis/metabolism
3.
Front Physiol ; 9: 1647, 2018.
Article in English | MEDLINE | ID: mdl-30534078

ABSTRACT

The Antarctic continent is an environment of extreme conditions. Only few research stations exist that are occupied throughout the year. The German station Neumayer III and the French-Italian Concordia station are such research platforms and human outposts. The seasonal shifts of complete daylight (summer) to complete darkness (winter) as well as massive changes in outside temperatures (down to -80°C at Concordia) during winter result in complete confinement of the crews from the outside world. In addition, the crew at Concordia is subjected to hypobaric hypoxia of ∼650 hPa as the station is situated at high altitude (3,233 m). We studied three expedition crews at Neumayer III (sea level) (n = 16) and two at Concordia (high altitude) (n = 15) to determine the effects of hypobaric hypoxia on hormonal/metabolic stress parameters [endocannabinoids (ECs), catecholamines, and glucocorticoids] and evaluated the psychological stress over a period of 11 months including winter confinement. In the Neumayer III (sea level) crew, EC and n-acylethanolamide (NAE) concentrations increased significantly already at the beginning of the deployment (p < 0.001) whereas catecholamines and cortisol remained unaffected. Over the year, ECs and NAEs stayed elevated and fluctuated before slowly decreasing till the end of the deployment. The classical stress hormones showed small increases in the last third of deployment. By contrast, at Concordia (high altitude), norepinephrine concentrations increased significantly at the beginning (p < 0.001) which was paralleled by low EC levels. Prior to the second half of deployment, norepinephrine declined constantly to end on a low plateau level, whereas then the EC concentrations increased significantly in this second period during the overwintering (p < 0.001). Psychometric data showed no significant changes in the crews at either station. These findings demonstrate that exposition of healthy humans to the physically challenging extreme environment of Antarctica (i) has a distinct modulating effect on stress responses. Additionally, (ii) acute high altitude/hypobaric hypoxia at the beginning seem to trigger catecholamine release that downregulates the EC response. These results (iii) are not associated with psychological stress.

4.
Front Physiol ; 9: 546, 2018.
Article in English | MEDLINE | ID: mdl-29867582

ABSTRACT

Beat to beat variability of cardiac tissue or isolated cells is frequently investigated by determining time intervals from electrode measurements in order to compute scale dependent or scale independent parameters. In this study, we utilize high-speed video camera recordings to investigate the variability of intervals as well as mechanical contraction strengths and relative contraction strengths with nonlinear analyses. Additionally, the video setup allowed us simultaneous electrode registrations of extracellular potentials. Sinoatrial node tissue under control and acetylcholine treated conditions was used to perform variability analyses by computing sample entropies and Higuchi dimensions. Beat to beat interval variabilities measured by the two recording techniques correlated very well, and therefore, validated the video analyses for this purpose. Acetylcholine treatment induced a reduction of beating rate and contraction strength, but the impact on interval variability was negligible. Nevertheless, the variability analyses of contraction strengths revealed significant differences in sample entropies and Higuchi dimensions between control and acetylcholine treated tissue. Therefore, the proposed high-speed video camera technique might represent a non-invasive tool that allows long-lasting recordings for detecting variations in beating behavior over a large range of scales.

5.
Biochem Pharmacol ; 145: 64-80, 2017 12 01.
Article in English | MEDLINE | ID: mdl-28859968

ABSTRACT

Saxagliptin treatment has been associated with increased rate of hospitalization for heart failure in type 2 diabetic patients, though the underlying mechanism(s) remain elusive. To address this, we assessed the effects of saxagliptin on human atrial trabeculae, guinea pig hearts and cardiomyocytes. We found that the primary target of saxagliptin, dipeptidyl peptidase-4, is absent in cardiomyocytes, yet saxagliptin internalized into cardiomyocytes and impaired cardiac contractility via inhibition of the Ca2+/calmodulin-dependent protein kinase II-phospholamban-sarcoplasmic reticulum Ca2+-ATPase 2a axis and Na+-Ca2+ exchanger function in Ca2+ extrusion. This resulted in reduced sarcoplasmic reticulum Ca2+ content, diastolic Ca2+ overload, systolic dysfunction and impaired contractile force. Furthermore, saxagliptin reduced protein kinase C-mediated delayed rectifier K+ current that prolonged action potential duration and consequently QTc interval. Importantly, saxagliptin aggravated pre-existing cardiac dysfunction induced by ischemia/reperfusion injury. In conclusion, our novel results provide mechanisms for the off-target deleterious effects of saxagliptin on cardiac function and support the outcome of SAVOR-TIMI 53 trial that linked saxagliptin with the risk of heart failure.


Subject(s)
Adamantane/analogs & derivatives , Dipeptides/toxicity , Dipeptidyl Peptidase 4/metabolism , Gene Expression Regulation, Enzymologic/drug effects , Heart Atria/cytology , Myocytes, Cardiac/drug effects , Adamantane/toxicity , Aged , Animals , Cell Line , Dipeptidyl Peptidase 4/genetics , Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Female , Gene Expression Regulation, Enzymologic/physiology , Heart Diseases/metabolism , Humans , Male , Mice , Middle Aged , Myocardial Contraction/drug effects , Myocytes, Cardiac/enzymology
6.
J Mol Cell Cardiol ; 72: 64-73, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24583250

ABSTRACT

Lower heart rate is associated with better survival in patients with multiple organ dysfunction syndrome (MODS), a disease mostly caused by sepsis. The benefits of heart rate reduction by ivabradine during MODS are currently being investigated in the MODIfY clinical trial. Ivabradine is a selective inhibitor of the pacemaker current If and since If is impaired by lipopolysaccharide (LPS, endotoxin), a trigger of sepsis, we aimed to explore If blocking potency of ivabradine under elevated endotoxin levels in human atrial cardiomyocytes. Treatment of myocytes with S-LPS (containing the lipid A moiety, a core oligosaccharide and an O-polysaccharide chain) but not R595 (an O-chain lacking LPS-form) caused If inhibition under acute and chronic septic conditions. The specific interaction of S-LPS but not R595 to pacemaker channels HCN2 and HCN4 proves the necessity of O-chain for S-LPS-HCN interaction. The efficacy of ivabradine to block If was reduced under septic conditions, an observation that correlated with lower intracellular ivabradine concentrations in S-LPS- but not R595-treated cardiomyocytes. Computational analysis using a sinoatrial pacemaker cell model revealed that despite a reduction of If under septic conditions, ivabradine further decelerated pacemaking activity. This novel finding, i.e. If inhibition by ivabradine under elevated endotoxin levels in vitro, may provide a molecular understanding for the efficacy of this drug on heart rate reduction under septic conditions in vivo, e.g. the MODIfY clinical trial.


Subject(s)
Action Potentials/drug effects , Benzazepines/pharmacology , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/antagonists & inhibitors , Lipopolysaccharides/pharmacology , Muscle Proteins/antagonists & inhibitors , Myocytes, Cardiac/drug effects , Sinoatrial Node/drug effects , Clinical Trials as Topic , Heart Atria/cytology , Heart Atria/drug effects , Heart Atria/metabolism , Heart Rate/drug effects , Humans , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/metabolism , Ivabradine , Models, Biological , Muscle Proteins/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Patch-Clamp Techniques , Potassium Channels/metabolism , Primary Cell Culture , Sinoatrial Node/cytology , Sinoatrial Node/metabolism
7.
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
8.
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
9.
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
10.
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
11.
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
12.
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
13.
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
14.
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
15.
Naunyn Schmiedebergs Arch Pharmacol ; 382(5-6): 433-40, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20838990

ABSTRACT

Thienopyridines (ticlopidine, clopidogrel) are frequently used drugs in antiplatelet therapy and have been shown to exert a more pronounced negative inotropic effect than thienopyrimidinones. We hypothesized that these differences are due to a differential impact of thienopyridines and thienopyrimidinones on L-type calcium current at the single-cell level. The effects of thienopyridines and thienopyrimidinones were studied on L-type calcium current and action potential parameters with the whole-cell patch-clamp technique in isolated myocytes from guinea pig ventricle and human atrial appendage. Ticlopidine showed the greatest impact on the L-type calcium current in guinea pig myocytes. It significantly reduced L-type calcium current density as well as shifted half maximal inactivation potential to more negative potentials compared to clopidogrel (at 30 µmol/L) and to all thienopyrimidinones (30 and 100 µmol/L). Clopidogrel significantly reduced the L-type calcium current density as well as shifted the half maximal inactivation potential to more negative potentials compared to all thienopyrimidinones at 100 µmol/L only. In contrast, thienopyrimidinones did not affect L-type calcium current properties. The significant different effects of thienopyridines and thienopyrimidinones could also be demonstrated in human atrial myocytes. The more pronounced negative inotropic effect of thienopyridines is well explained by our results demonstrating a differential impairment of L-type calcium current by thienopyridines and thienopyrimidinones. L-type calcium current impairment by thienopyridines may be of special relevance for patients with cardiac diseases characterized by ionic remodelling.


Subject(s)
Action Potentials/drug effects , Calcium Channels, L-Type/physiology , Myocytes, Cardiac/drug effects , Platelet Aggregation Inhibitors/pharmacology , Pyrimidines/pharmacology , Thienopyridines/pharmacology , Thiophenes/pharmacology , Animals , Atrial Appendage/cytology , Cells, Cultured , Clopidogrel , Guinea Pigs , Heart Ventricles/cytology , Humans , In Vitro Techniques , Myocytes, Cardiac/physiology , Patch-Clamp Techniques , Ticlopidine/analogs & derivatives , Ticlopidine/pharmacology
16.
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
17.
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
18.
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
19.
Shock ; 28(6): 655-661, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18092381

ABSTRACT

LPSs trigger the development of sepsis by gram-negative bacteria and cause a variety of biological effects on host cells, including alterations on ionic channels. Because heart rate variability is reduced in human sepsis and endotoxemia, we hypothesized that LPS affects the pacemaker current I(f) in human heart, which might--at least in part--explain this phenomenon. Isolated human myocytes from right atrial appendages were incubated for 6 to 10 h with LPS (1 and 10 microg/mL) and afterwards used to investigate the pacemaker current I(f). I(f) was measured with the whole-cell patch-clamp technique (at 37 degrees C). Incubation of atrial myocytes with 10 microg/mL LPS was found to significantly impair I(f) by suppressing the current at membrane potentials positive to -80 mV and slowing down current activation, but without effecting maximal current conductance. Furthermore, in incubated cells (10 microg/mL), the response of I(f) to [beta]-adrenergic stimulation (1 microM isoproterenol) was significantly larger compared with control cells (shift of half-maximal activation voltage to more positive potentials amounted to -10 and -14 mV in untreated and treated cells, respectively). Simulations using a spontaneously active sinoatrial cell model demonstrated that LPS-induced I(f) impairment reduced the responsiveness of the model cell to fluctuations of autonomic input. This study showed a direct impact of LPS on the cardiac pacemaker current I(f). The LPS-induced I(f) impairment may contribute to the clinically observed reduction in heart rate variability under septic conditions and in cardiac diseases such as heart failure, where endotoxin can be of pathophysiological relevance.


Subject(s)
Lipopolysaccharides/pharmacology , Myocytes, Cardiac/drug effects , Adult , Aged , Cells, Cultured , Electrophysiology , Female , Heart Rate/drug effects , Humans , Ion Channels/physiology , Isoproterenol/pharmacology , Male , Membrane Potentials/drug effects , Middle Aged , Myocytes, Cardiac/cytology , Myocytes, Cardiac/physiology , Patch-Clamp Techniques , Sepsis/pathology , Sepsis/physiopathology
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...