Network


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

Hotspot


Dive into the research topics where Paul Y. Kim is active.

Publication


Featured researches published by Paul Y. Kim.


Journal of Thrombosis and Haemostasis | 2013

Insights into thrombin activatable fibrinolysis inhibitor function and regulation.

Jonathan H. Foley; Paul Y. Kim; Nicola J. Mutch; Ann Gils

Fibrinolysis is initiated when the zymogen plasminogen is converted to plasmin via the action of plasminogen activators. Proteolytic cleavage of fibrin by plasmin generates C‐terminal lysine residues capable of binding both plasminogen and the plasminogen activator, thereby stimulating plasminogen activator‐mediated plasminogen activation and propagating fibrinolysis. This positive feedback mechanism is regulated by activated thrombin activatable fibrinolysis inhibitor (TAFIa), which cleaves C‐terminal lysine residues from the fibrin surface, thereby decreasing its cofactor activity. TAFI can be activated by thrombin alone, but the rate of activation is accelerated when in complex with thrombomodulin. Plasmin is also known to activate TAFI. TAFIa has no known physiologic inhibitors and consequently, its primary regulatory mechanism involves its intrinsic thermal instability. The rate of TAFI activation and stability of the active form, TAFIa, function in maintaining its concentration above the threshold value required to down‐regulate fibrinolysis. Although all methods to quantify TAFI or TAFIa have their limitations, epidemiologic studies have indicated that elevated TAFI levels are correlated with an increased risk of venous thrombosis. Major efforts have been made to develop TAFI inhibitors that can either directly interfere with TAFIa activity or impair its activation. However, the anti‐inflammatory properties of TAFIa might complicate the development and application of a TAFIa inhibitor that aims to increase the efficiency of thrombolytic therapy.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2015

Cell-Free DNA Modulates Clot Structure and Impairs Fibrinolysis in Sepsis

Travis J. Gould; Trang T. Vu; Alan R. Stafford; Dhruva J. Dwivedi; Paul Y. Kim; Alison E. Fox-Robichaud; Jeffrey I. Weitz; Patricia C. Liaw

Objectives—Sepsis is characterized by systemic activation of inflammation and coagulation in response to infection. In sepsis, activated neutrophils extrude neutrophil extracellular traps composed of cell-free DNA (CFDNA) that not only trap pathogens but also provide a stimulus for clot formation. Although the effect of CFDNA on coagulation has been extensively studied, much less is known about the impact of CFDNA on fibrinolysis. To address this, we (1) investigated the relationship between CFDNA levels and fibrinolytic activity in sepsis and (2) determined the mechanisms by which CFDNA modulates fibrinolysis. Approach and Results—Plasma was collected from healthy and septic individuals, and CFDNA was quantified. Clot lysis assays were performed in plasma and purified systems, and lysis times were determined by monitoring absorbance. Clot morphology was assessed using scanning electron microscopy. Clots formed in plasma from septic patients containing >5 µg/mL CFDNA were dense in structure and resistant to fibrinolysis, a phenomenon overcome by deoxyribonuclease addition. These effects were recapitulated in control plasma supplemented with CFDNA. In a purified system, CFDNA delayed fibrinolysis but did not alter tissue-type plasminogen activator–induced plasmin generation. Using surface plasmon resonance, CFDNA bound plasmin with a Kd value of 4.2±0.3 µmol/L, and increasing concentrations of CFDNA impaired plasmin-mediated degradation of fibrin clots via the formation of a nonproductive ternary complex between plasmin, CFDNA, and fibrin. Conclusions—Our studies suggest that the increased levels of CFDNA in sepsis impair fibrinolysis by inhibiting plasmin-mediated fibrin degradation, thereby identifying CFDNA as a potential therapeutic target for sepsis treatment.


Journal of Thrombosis and Haemostasis | 2016

Activation of protein C and thrombin activable fibrinolysis inhibitor on cultured human endothelial cells.

Chengliang Wu; Paul Y. Kim; Laura L. Swystun; Patricia C. Liaw; Jeffrey I. Weitz

Essentials It is unknown if thrombin activatable fibrinolysis inhibitor (TAFI) and protein C compete on cells. TAFI and protein C activation on endothelial cells was simultaneously quantified. TAFI and protein C do not compete for activation on endothelial cells. TAFI and protein C are independently recognized by the thrombin–thrombomodulin complex.


Journal of Thrombosis and Haemostasis | 2015

Evaluation of and recommendation for the nomenclature of the CPB2 gene product (also known as TAFI and proCPU): communication from the SSC of the ISTH

Jonathan H. Foley; Paul Y. Kim; Dirk Hendriks; John Morser; Ann Gils; Nicola J. Mutch

J . H . FOLEY ,* P . Y . K IM,† D. HENDRIKS ,‡ J . MORSER ,§ A. G ILS ¶ and N . J . MUTCH,** FOR THE SUBCOMMITTEE ON F IBR INOLYS I S *Research Department of Haematology, University College London and Katherine Dormandy Haemophilia Centre and Thrombosis Unit, London, UK; †Thrombosis and Atherosclerosis Research Institute, McMaster University, Hamilton, ON, Canada; ‡Laboratory for Medical Biochemistry, University of Antwerp, Antwerp, Belgium; §Division of Hematology, School of Medicine, Stanford University, Stanford, CA, USA; ¶Laboratory for Therapeutic and Diagnostic Antibodies, KU Leuven, Leuven, Belgium; and **Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK


Journal of Thrombosis and Thrombolysis | 2012

Thrombin-activatable fibrinolysis inhibitor is activated in vivo in a baboon model of Escherichia coli induced sepsis.

Paul Y. Kim; Paula Y.G. Kim; Fletcher B. Taylor; Michael E. Nesheim

Activated thrombin-activatable fibrinolysis inhibitor (TAFIa or CPU) is a carboxypeptidase that is able to attenuate fibrinolysis. Although its role in fibrinolysis and inflammation has been studied extensively in vitro, its levels and subsequent effect in vivo has not been studied to the same extent. Using our recently developed assay that is specific for TAFIa, we were able to quantify its levels in plasma samples obtained from an Escherichia coli (E. coli) challenged baboon sepsis model. TAFIa levels accumulated appeared to be E. coli dose dependent, where the lethal dose of 1010xa0CFU/kg generated a peak TAFIa level of 24xa0nM by 2xa0h, which represents almost 32% of total plasma level of its precursor, thrombin-activatable fibrinolysis inhibitor (TAFI or proCPU). Furthermore, our data suggest that there is continual TAFI activation under lethal level of E. coli as the apparent half-life of TAFIa is increased from 8xa0min to 2.2xa0h. Two sublethal doses of 108 and 106xa0CFU/kg generated peak TAFIa levels of 1.1 and 0.4xa0nM, respectively, both by 6xa0h. Taken together, our data show that TAFIa is generated at systemic levels, in a dose-dependent manner, that can substantially affect both fibrinolysis and inflammatory response in the E. coli challenged baboon sepsis model.


Blood | 2015

Platelets: connecting clotting and lysis.

Paul Y. Kim

In this issue of Blood, Whyte et al describe how under flow conditions, phosphatidylserine-expressing platelets modulate the lysis of whole blood clots by providing direct and indirect binding sites for plasminogen.


Thrombosis and Haemostasis | 2013

Interactions of heparin and a covalently-linked antithrombin-heparin complex with components of the fibrinolytic system.

Ankush Chander; Helen M. Atkinson; Ivan Stevic; Leslie R. Berry; Paul Y. Kim; Anthony K.C. Chan

Unfractionated heparin (UFH) is used as an adjunct during thrombolytic therapy. However, its use is associated with limitations, such as the inability to inhibit surface bound coagulation factors. We have developed a covalent conjugate of antithrombin (AT) and heparin (ATH) with superior anticoagulant properties compared with UFH. Advantages of ATH include enhanced inhibition of surface-bound coagulation enzymes and the ability to reduce the overall size and mass of clots in vivo. The interactions of UFH or ATH with the components of the fibrinolytic pathway are not well understood. Our study utilised discontinuous second order rate constant (k₂) assays to compare the rates of inhibition of free and fibrin-associated plasmin by AT+UFH vs ATH. Additionally, we evaluated the effects of AT+UFH and ATH on plasmin generation in the presence of fibrin. The k₂ values for inhibition of plasmin were 5.74 ± 0.28 x 10⁶ M⁻¹ min⁻¹ and 6.39 ± 0.59 x 10⁶ M⁻¹ min⁻¹ for AT+UFH and ATH, respectively. In the presence of fibrin, the k₂ values decreased to 1.45 ± 0.10 x 10⁶ M⁻¹ min⁻¹ and 3.07 ± 0.19 x 10⁶ M⁻¹ min⁻¹ for AT+UFH and ATH, respectively. Therefore, protection of plasmin by fibrin was observed for both inhibitors; however, ATH demonstrated superior inhibition of fibrin-associated plasmin. Rates of plasmin generation were also decreased by both inhibitors, with ATH causing the greatest reduction (approx. 38-fold). Nonetheless, rates of plasmin inhibition were 2-3 orders of magnitude lower than for thrombin, and in a plasma-based clot lysis assay ATH significantly inhibited clot formation but had little impact on clot lysis. Cumulatively, these data may indicate that, relative to coagulant enzymes, the fibrinolytic system is spared from inhibition by both AT+UFH and ATH, limiting reduction in fibrinolytic potential during anticoagulant therapy.


Thrombosis Research | 2017

The impact of the endothelial protein C receptor on thrombin generation and clot lysis

Laura Pepler; Chengliang Wu; Dhruva J. Dwivedi; Cynthia Wu; Paul Y. Kim; Patricia C. Liaw

INTRODUCTIONnWhen thrombin is bound to thrombomodulin (TM), it becomes a potent activator of protein C (PC) and thrombin-activable fibrinolysis inhibitor (TAFI). Activation of PC is enhanced when PC is bound to the endothelial protein C receptor (EPCR). Activated protein C (APC) inhibits thrombin generation while activated TAFI (TAFIa) attenuates fibrinolysis. To determine the impact of diminished EPCR function on thrombin generation and fibrinolysis we generated cells that expressed TM and a variant of EPCR (R96C) that does not bind PC.nnnMETHODSnTo determine the impact of EPCR on the generation of APC and TAFIa and how this affects thrombin generation and fibrinolysis we performed thrombin generation and clot lysis assays in the presence of cells expressing wild-type TM and EPCR (WT cells) or wild-type TM and the R96C variant of EPCR (R96C cells).nnnRESULTSnIn the presence of R96C cells, thrombin generation in normal plasma is increased, as a result of impaired PC activation when compared to WT cells. In addition, clot lysis is delayed in normal plasma in the presence of R96C cells, despite no increase in TAFI activation. In PC deficient plasma, clot lysis is delayed in the presence of WT and R96C cells as a result of increased TAFI activation.nnnCONCLUSIONSnWe demonstrate that impaired EPCR function can be detected by thrombin generation and clot lysis assays on cells expressing TM and EPCR. We also demonstrated that deficiency in EPCR has procoagulant effects that lead to a delay in clot lysis.


TH Open | 2018

Mechanistic Basis for the Differential Effects of Rivaroxaban and Apixaban on Global Tests of Coagulation

Paul Y. Kim; Calvin H. Yeh; Brian Dale; Beverly A. Leslie; Alan R. Stafford; James C. Fredenburgh; Jack Hirsh; Jeffrey I. Weitz

Rivaroxaban and apixaban are both small molecules that reversibly inhibit factor Xa. Compared with rivaroxaban, apixaban has minimal effects on the prothrombin time and activated partial thromboplastin time. To investigate this phenomenon, we used a factor Xa-directed substrate in a buffer system. Although rivaroxaban and apixaban inhibited factor Xa with similar K i values at equilibrium, kinetic measurements revealed that rivaroxaban inhibited factor Xa up to 4-fold faster than apixaban ( p u2009<u20090.001). Using a discontinuous chromogenic assay to monitor thrombin production by prothrombinase in a purified system, rivaroxaban was 4-fold more potent than apixaban (K i values of 0.7u2009±u20090.3 and 2.9u2009±u20090.5u2009nM, respectively; p u2009=u20090.02). Likewise, in thrombin generation assays in plasma, rivaroxaban prolonged the lag time and suppressed endogenous thrombin potential to a greater extent than apixaban. To characterize how the two inhibitors differ in recognizing factor Xa, inhibition of prothrombinase was monitored in real-time using a fluorescent probe for thrombin. The data were fit using a mixed-inhibition model and the individual association and dissociation rate constants were determined. The association rates for the binding of rivaroxaban to either free factor Xa or factor Xa incorporated into the prothrombinase complex were 10- and 1,193-fold faster than those for apixaban, respectively, whereas dissociation rates were about 3-fold faster. Collectively, these findings suggest that rivaroxaban and apixaban differ in their capacity to inhibit factor Xa and provide a plausible explanation for the observation that rivaroxaban has a greater effect on global tests of coagulation than apixaban.


Journal of Thrombosis and Haemostasis | 2018

Fibrinolysis: Strategies to enhance the treatment of acute ischemic stroke.

Sara J. Henderson; Jeffrey I. Weitz; Paul Y. Kim

Stroke is a major cause of disability worldwide, and is the second leading cause of death after ischemic heart disease. Until recently, tissue‐type plasminogen activator (t‐PA) was the only treatment for acute ischemic stroke. If administered within 4.5 h of symptom onset, t‐PA improves the outcome in stroke patients. Mechanical thrombectomy is now the preferred treatment for patients with acute ischemic stroke resulting from a large‐artery occlusion in the anterior circulation. However, the widespread use of mechanical thrombectomy is limited by two factors. First, only ⁓ 10% of patients with acute ischemic stroke have a proximal large‐artery occlusion in the anterior circulation and present early enough to undergo mechanical thrombectomy within 6 h; an additional 9–10% of patients presenting within the 6–24‐h time window may also qualify for the procedure. Second, not all stroke centers have the resources or expertise to perform mechanical thrombectomy. Nonetheless, patients who present to hospitals where thrombectomy is not an option can receive intravenous t‐PA, and those with qualifying anterior circulation strokes can then be transferred to tertiary stroke centers where thrombectomy is available. Therefore, despite the advances afforded by mechanical thrombectomy, there remains a need for treatments that improve the efficacy and safety of thrombolytic therapy. In this review, we discuss: (i) current treatment options for acute ischemic stroke; (ii) the mechanism of action of fibrinolytic agents; and (iii) potential strategies to manipulate the fibrinolytic system to promote endogenous fibrinolysis or to enhance the efficacy of fibrinolytic therapy.

Collaboration


Dive into the Paul Y. Kim's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ann Gils

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge