Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Ute Klinkhardt is active.

Publication


Featured researches published by Ute Klinkhardt.


Clinical Pharmacology & Therapeutics | 2003

Clopidogrel but not aspirin reduces P‐selectin expression and formation of platelet‐leukocyte aggregates in patients with atherosclerotic vascular disease*

Ute Klinkhardt; Rupert Bauersachs; Jan Adams; Jochen Graff; Edeltraud Lindhoff‐Last; Sebastian Harder

Formation of platelet‐leukocyte aggregates via the CD62 ligand represents an important mechanism by which leukocytes contribute to thrombotic events. In a cross‐sectional study, we investigated platelet‐leukocyte aggregate formation and markers indicative for platelet, leukocyte, and endothelial activation (CD62, activated fibrinogin receptor glycoprotein IIb/IIIA [PAC‐1], CD11b/CD18 [MAC‐1], and soluble intercellular adhesion molecule 1) in 44 patients with atherosclerotic vascular disease and peripheral occlusions receiving clopidogrel (n = 12), aspirin (n = 17), their combination (n = 8), or no treatment (n = 7), as well as in a group of healthy subjects (n = 9). Whole‐blood flow cytometry was performed before (baseline) and after stimulation with thrombin receptor‐activating peptide or adenosine diphosphate. Both at baseline and after stimulation, untreated patients and those receiving aspirin monotherapy exhibited significantly higher levels of platelet CD62 expression (baseline CD62: untreated, 22% [median]; with aspirin, 16%) and had higher rates of platelet‐leukocyte aggregate formation (monocyte‐platelet‐leukocyte aggregates at baseline: untreated, 27%; with aspirin, 16%) when compared with patients receiving clopidogrel alone (baseline CD62: 10% [P < .05]; monocyte‐platelet‐leukocyte aggregates: 13% [P < .05]) or combined with aspirin (baseline CD62: 5% [P < .05]; monocyte‐platelet‐leukocyte aggregates: 7% [P < .05]). Up‐regulation of MAC‐1 on monocytes after stimulation with thrombin receptor‐activating peptide and adenosine diphosphate was significantly lower in patients treated with clopidogrel and aspirin. Plasma levels of soluble intercellular adhesion molecule 1 were significantly lower in the group of healthy subjects (median, 186 ng/mL) when compared with those in untreated patients (median, 352 ng/mL) (P < .05), whereas intercellular adhesion molecule 1 levels in treated patients were similar for any antiplatelet regimen (aspirin, 262 ng/mL; clopidogrel, 274 ng/mL; combination therapy, 273 ng/mL) but significantly lower than those in untreated patients. This is the first report showing that platelet‐leukocyte aggregate formation is enhanced in atherosclerotic vascular disease but was found to be reduced in patients receiving clopidogrel.


Clinical Pharmacokinectics | 2004

Avoidance of Bleeding During Surgery in Patients Receiving Anticoagulant and/or Antiplatelet Therapy: Pharmacokinetic and Pharmacodynamic Considerations

Sebastian Harder; Ute Klinkhardt; John M. Alvarez

Perioperative management of chronically anticoagulated patients and/or patients treated with antiplatelet therapy is a complex medical problem. This review considers the pharmacokinetic and pharmacodynamic properties of commonly used antiplatelet and anticoagulant drugs with special emphasis on loss of effects after discontinuation and possible counteracting (or antidote) strategies. These drugs are aspirin (acetylsalicylic acid), ticlopidine/clopidogrel, abciximab, tirofiban and eptifibatide, heparin (unfractionated and low-molecular-weight), warfarin and direct thrombin inhibitors. Since the pharmacological mechanisms of some of these drugs are based on irreversible or slowly reversible effects, their pharmacokinetic profiles are not necessarily predictive for their pharmacodynamic profiles. A close and direct relationship between plasma concentrations and effects is seen only for the glycoprotein (GP) IIb/IIIa inhibitors tirofiban and eptifibatide with a fast off-rate for dissociation from the GPIIb/IIIa receptor, and for direct thrombin inhibitors (hirudin and argatroban). For other compounds, drug concentrations in plasma and pharmacodynamic effects are not closely correlated because of, for example, irreversible binding to their target (aspirin, Clopidogrel and abciximab), inhibition of the generation of a subset of clotting factors with differing regeneration and degradation rates (coumarins) or sustained binding to the vascular wall (heparins).Surgery in patients on anticoagulant and/or antiplatelet therapy may be categorised as: (i) elective versus urgent; and (ii) cardiopulmonary bypass (CPB) versus non-CPB. Monotherapy with Clopidogrel or aspirin need not be discontinued in elective non-CPB surgery, and temporary discontinuation of warfarin should be accompanied by preoperative intravenous heparin only in selected high-risk patients. Vitamin K as an antidote for warfarin should only be used subcutaneously and solely in urgent/emergency surgery. In elective surgery requiring CPB (coronary artery bypass grafting), it is recommended to discontinue aspirin 7 days preoperatively in patients with a low risk profile. Patients requiring urgent CPB surgery (e.g. after failure of a percutaneous coronary angioplasty with or without coronary stent deployment) are usually pretreated with several antiplatelet agents (e.g. aspirin and Clopidogrel, together with a GPIIb/IIIa inhibitor) together with unfractionated or low-molecular-weight heparin. With judicious planning, urgent/emergency cardiac surgery can be safely performed on these patients. Delaying surgery (e.g. for 12 hours in patients treated with abciximab) should be considered if possible. Standard heparin doses should be given to achieve optimal anticoagulation for CPB. Prophylactic use of aprotinin (intraand/or postoperatively), aminocaproic acid or tranexamic acid should be considered. Early (in the operating theatre prior to chest closure) and judicious use of replacement blood products (platelets) should be commenced when clinically indicated.


Clinical Pharmacology & Therapeutics | 2002

Clopidogrel, but not abciximab, reduces platelet leukocyte conjugates and P‐selectin expression in a human ex vivo in vitro model

Ute Klinkhardt; Jochen Graff; Sebastian Harder

Formation of platelet‐leukocyte aggregates (PLA) through the CD62 ligand is an important mechanism by which leukocytes contribute to thrombosis and inflammation. We investigated the formation of PLA in human subjects after stimulation with thrombin receptor activating peptide and adenosine diphosphate (ADP) after treatment with clopidogrel and after in vitro application of the platelet glycoprotein IIb/IIIa complex antagonist abciximab. Expression of CD62 was significantly reduced 30% to 50% with clopidogrel, depending on the type and concentration of the inducer, but addition of abciximab led to a significant approximately 30% increase in CD62 expression whenplatelets were stimulated by ADP. Formation of PLA decreased significantly with clopidogrel to 55% to 75% of the baseline value, whereas addition of abciximab caused a significant increase in PLA in ADP‐stimulated samples before but not after administration of clopidogrel. The increase in formation of PLA after in vitro addition of abciximab was not paralleled by a decrease in platelet microaggregates and is therefore presumed not caused by enhanced availability of platelets. To our knowledge, this is the first report showing that clopidogrel reduces formation of PLA. The findings also suggest intersection between an “outside‐in” signal generated by abciximab and stimulation of platelet P2T12 purinergic receptors that augments degranulation and increases formation of PLA but is inhibited by clopidogrel.


Platelets | 2010

Using ImageJ for the quantitative analysis of flow-based adhesion assays in real-time under physiologic flow conditions

Sascha Meyer dos Santos; Ute Klinkhardt; Reinhard Schneppenheim; Sebastian Harder

This article intends to close the gap between the abundance of regular articles focusing on adhesive mechanisms of cells in a flow field and purely technical reports confined to the description of newly developed algorithms, not yet ready to be used by users without programming skills. A simple and robust method is presented for analysing raw videomicroscopic data of flow-based adhesion assays using the freely available public domain software ImageJ. We describe in detail the image processing routines used to rapidly and reliably evaluate the number of adherent and translocating platelets in videomicroscopic recordings. The depicted procedures were exemplified by analysing platelet interaction with immobilized von Willebrand factor and fibrinogen in flowing blood under physiological wall shear rates. Neutralizing GPIbα function reduced shear-dependent platelet translocation on von Willebrand factor and abolished firm platelet adhesion. Abciximab, Tirofiban and Eptifibatide completely inhibited GPIIb/IIIa-dependent stable platelet deposition on fibrinogen. The presented method to analyse videomicroscopic recordings from flow-based adhesion assays offers the advantage of providing a simple and reliable way to quantify flow-based adhesion assays, which is completely based on ImageJ and can easily be applied to study adhesion mechanisms of cells in non-fluorescent modes without the need to deviate from the presented protocol.


Biomicrofluidics | 2013

A novel μ-fluidic whole blood coagulation assay based on Rayleigh surface-acoustic waves as a point-of-care method to detect anticoagulants

Sascha Meyer dos Santos; Anita Zorn; Zeno v. Guttenberg; Bettina Picard-Willems; Christina Kläffling; Karen Nelson; Ute Klinkhardt; Sebastian Harder

A universal coagulation test that reliably detects prolonged coagulation time in patients, irrespective of the anticoagulant administered, has not been available to date. An easily miniaturised, novel μ-fluidic universal coagulation test employing surface acoustic waves (SAW) is presented here. SAW was employed to instantly mix and recalcify 6 μl citrated whole blood and image correlation analysis was used to quantify clot formation kinetics. The detection of clinically relevant anticoagulant dosing with old anticoagulants (unfractionated heparin, argatroban) and new anticoagulants (dabigatran, rivaroxaban) has been tested and compared to standard plasma coagulation assays. The applicability of this novel method has been confirmed in a small patient population. Coagulation was dose-proportionally prolonged with heparin, argatroban, dabigatran, and rivaroxaban, comparable to standard tests. Aspirin and clopidogrel did not interfere with the SAW-induced clotting time (SAW-CT), whereas the strong GPIIb/IIIa-inhibitor abciximab did interfere. Preliminary clinical data prove the suitability of the SAW-CT in patients being treated with warfarin, rivaroxaban, or dabigatran. The system principally allows assessment of whole blood coagulation in humans in a point-of-care setting. This method could be used in stroke units, emergency vehicles, general and intensive care wards, as well as for laboratory and home testing of coagulation.


Blood | 2011

The CX3C chemokine fractalkine mediates platelet adhesion via the von Willebrand receptor glycoprotein Ib

Sascha Meyer dos Santos; Ute Klinkhardt; Klaus Scholich; Karen Nelson; Nadejda Monsefi; Hans Deckmyn; Karina Kuczka; Anita Zorn; Sebastian Harder

The membrane-anchored CX3C chemokine fractalkine (FKN) is expressed on activated endothelium and is associated with the development of atherosclerosis. The potential of FKN in mediating platelet adhesion beyond platelet activation remains unexplored to date. A flow-based adhesion assay was used to study the adhesion of platelets to immobilized FKN under physiologic flow conditions. Platelet adhesion to von Willebrand factor (VWF) was increased in the presence of FKN at 600 inverse seconds. Additional platelet adhesion to FKN coimmobilized with VWF was dependent on the FKN receptor CX3CR1 and activation of glycoprotein (GP) IIb/IIIa. The number of platelets rolling on VWF was likewise enhanced in the presence of FKN. The enhancement of rolling on FKN and VWF was insensitive to anti-CX3CR1 antibody but was fully inhibited by neutralizing GPIbα function. The extracellular domain of GPIbα was covalently coupled to fluorescent microspheres, and microsphere binding was significantly higher in the presence of FKN. Platelet adhesion to activated endothelium in vitro and to intact human arteries was substantially increased in an FKN-dependent manner. These data demonstrate that endothelial expressed FKN activates platelets via its cognate receptor CX3CR1, whereas platelet adhesion is predominantly mediated by GPIbα and independent of CX3CR1.


Thrombosis Research | 2000

Differential in Vitro Effects of the Platelet Glycoprotein IIb/IIIa Inhibitors abixicimab or SR121566A on Platelet Aggregation, Fibrinogen Binding and Platelet Secretory Parameters

Ute Klinkhardt; C.M. Kirchmaier; Dagmar Westrup; Hans Klaus Breddin; R. Mahnel; Jochen Graff; Monika Hild; Sebastian Harder

The aim of this study was to compare fibrinogen binding, inhibition of platelet aggregation and secretory potential of the MAb abciximab (0.5-5 microg/mL) and the peptidomimetic compound SR121566A (15-250 ng/mL) in vitro in whole blood. Fibrinogen binding was followed by flow cytometry; platelet function was evaluated by light transmittance and by impedance aggregometry. Secretory functions of platelets were evaluated using ATP as marker for early secretion by dense granulae and P-selectin (CD62) for alpha-granular secretion as well as CD63 for lysosomal degranulation. Results showed that fibrinogen binding induced by 5 microM TRAP was maximally inhibited greater than 80% at 3 microg/mL abciximab or at 250 ng/mL SR121566A. At these concentrations of antagonists, platelet aggregation induced by 5 microM ADP or 2 microg/mL collagen was inhibited completely. Expression of CD62 was reduced 34% with abciximab or 15% with SR121566A; CD63 expression was reduced 22% with both agents. With both agents, the EC50 for inhibition of CD62 and CD63 expressions was in similar magnitudes than the EC50 for fibrinogen binding inhibition. With 3 microg/mL abciximab, ATP secretion was maximally reduced to 50% of the control, whereas SR121566A at 250 ng/mL had no inhibitory effect on this parameter. A slight increase in ATP secretion was seen with 0.5 microg/mL abciximab and with SR121566A in concentrations of less than 45 ng/mL. The data suggest a discoupling between the anti-aggregatory and the antisecretory effects of IIb/IIIa antagonists. Because it is not established to what extend CD62 or CD63 expression can be reduced by any means, the reduction by 20-30% obtained by 3 microg/mL abciximab or 250 ng/mL SR121566A might already be the maximum possible inhibition by these agents.


Clinical Pharmacology & Therapeutics | 2002

Immunosuppressive therapy regimen and platelet activation in renal transplant patients

Jochen Graff; Ute Klinkhardt; Sebastian Harder; Wolfgang Wegert; Tomas Lenz; Ernst-Heinrich Scheuermann; Jan Gossmann

Increased platelet activation caused by an immunosuppressive therapy regimen may contribute to the high incidence of death from cardiovascular disease in renal transplant patients. Cyclosporine (INN, ciclosporin) and azathioprine are reported to activate platelets, but data are rare and controversial for tacrolimus and mycophenolate mofetil.


British Journal of Pharmacology | 2005

Induction of central signalling pathways and select functional effects in human platelets by β-boswellic acid

Daniel Poeckel; Lars Tausch; Anja Altmann; Christian Feißt; Ute Klinkhardt; Jochen Graff; Sebastian Harder; Oliver Werz

We have recently shown that in polymorphonuclear leukocytes, 11‐keto boswellic acids (KBAs) induce Ca2+ mobilisation and activation of mitogen‐activated protein kinases (MAPK). Here we addressed the effects of BAs on central signalling pathways in human platelets and on various platelet functions. We found that β‐BA (10 μM), the 11‐methylene analogue of KBA, caused a pronounced mobilisation of Ca2+ from internal stores and induced the phosphorylation of p38 MAPK, extracellular signal‐regulated kinase (ERK)2, and Akt. These effects of β‐BA were concentration dependent, and the magnitude of the responses was comparable to those obtained after platelet stimulation with thrombin or collagen. Based on inhibitor studies, β‐BA triggers Ca2+ mobilisation via the phospholipase (PL)C/inositol‐1,4,5‐trisphosphate pathway, and involves Src family kinase signalling. Investigation of platelet functions revealed that β‐BA (10 μM) strongly stimulates the platelet‐induced generation of thrombin in an ex‐vivo in‐vitro model, the liberation of arachidonic acid (AA), and induces platelet aggregation in a Ca2+‐dependent manner. In contrast to β‐BA, the 11‐keto‐BAs (KBA or AKBA) evoke only moderate Ca2+ mobilisation and activate p38 MAPK, but fail to induce phosphorylation of ERK2 or Akt, and do not cause aggregation or significant generation of thrombin. In summary, β‐BA potently induces Ca2+ mobilisation as well as the activation of pivotal protein kinases, and elicits functional platelet responses such as thrombin generation, liberation of AA, and aggregation.


The Journal of Clinical Pharmacology | 2011

Anticoagulation With Argatroban for Elective Percutaneous Coronary Intervention: Population Pharmacokinetics and Pharmacokinetic-Pharmacodynamic Relationship of Coagulation Parameters

Kei Akimoto; Ute Klinkhardt; Andreas Zeiher; Margit Niethammer; Sebastian Harder

The synthetic direct thrombin inhibitor argatroban has a rapid onset and offset of anticoagulation. However, there are no data about the pharmacokinetic‐pharmacodynamic (PK‐PD) relationship of argatroban in patients undergoing contemporary percutaneous coronary intervention (PCI) and no data about other coagulation parameters than activated clotting time (ACT) in this setting. In the ARG‐E04‐trial, 140 patients were randomly assigned to argatroban (250, 300, or 350 μg/kg as bolus before PCI, followed by 15, 20, or 25 μg/kg/min infusion) or unfractionated heparin (70–100 IU/kg bolus). A 2‐compartment model with first‐order elimination adequately described the pharmacokinetic profile of argatroban over all 3 dosing groups. Clearance (CL) and distribution volumes (V1 and V2) were 21 L/h, 9.2 L, and 6.6 L, respectively. A significant sigmoidal Emax relationship was established between the argatroban plasma concentration and the response in ACT and the endogenous thrombin potential (ETP), whereas the response in activated partial thromoplastin time (aPTT), ecarin time (ECA‐T), and prothrombinase‐induced clotting time (PiCT) could be described by a nonsigmoidal Emax model. This study proves a relatively small interindividual variability of both PK and PK‐PD properties of argatroban even at high doses and supports the profile of argatroban as a drug with a predictive dose‐effect relationship and therefore good controllability.

Collaboration


Dive into the Ute Klinkhardt's collaboration.

Top Co-Authors

Avatar

Sebastian Harder

Goethe University Frankfurt

View shared research outputs
Top Co-Authors

Avatar

Jochen Graff

Goethe University Frankfurt

View shared research outputs
Top Co-Authors

Avatar

Karina Kuczka

Goethe University Frankfurt

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Anita Zorn

Goethe University Frankfurt

View shared research outputs
Top Co-Authors

Avatar

C.M. Kirchmaier

Goethe University Frankfurt

View shared research outputs
Top Co-Authors

Avatar

Jan Gossmann

Goethe University Frankfurt

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Tomas Lenz

Goethe University Frankfurt

View shared research outputs
Top Co-Authors

Avatar

Annette Sedlmeyer

Goethe University Frankfurt

View shared research outputs
Researchain Logo
Decentralizing Knowledge