Network


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

Hotspot


Dive into the research topics where Kathy Koenig is active.

Publication


Featured researches published by Kathy Koenig.


American Journal of Physiology-lung Cellular and Molecular Physiology | 2009

Regulation of intrapleural fibrinolysis by urokinase-α-macroglobulin complexes in tetracycline-induced pleural injury in rabbits

Andrey A. Komissarov; Andrew P. Mazar; Kathy Koenig; Anna Kurdowska; Steven Idell

The proenzyme single-chain urokinase plasminogen activator (scuPA) more effectively resolved intrapleural loculations in rabbits with tetracycline (TCN)-induced loculation than a range of clinical doses of two-chain uPA (Abbokinase) and demonstrated a trend toward greater efficacy than single-chain tPA (Activase) (Idell S et al., Exp Lung Res 33: 419, 2007.). scuPA more slowly generates durable intrapleural fibrinolytic activity than Abbokinase or Activase, but the interactions of these agents with inhibitors in pleural fluids (PFs) have been poorly understood. PFs from rabbits with TCN-induced pleural injury treated with intrapleural scuPA, its inactive Ser195Ala mutant, Abbokinase, Activase, or vehicle, were analyzed to define the mechanism by which scuPA induces durable fibrinolysis. uPA activity was elevated in PFs of animals treated with scuPA, correlated with the ability to clear pleural loculations, and resisted (70-80%) inhibition by PAI-1. Alpha-macroglobulin (alphaM) but not urokinase receptor complexes immunoprecipitated from PFs of scuPA-treated rabbits retained uPA activity that resists PAI-1 and activates plasminogen. Conversely, little plasminogen activating or enzymatic activity resistant to PAI-1 was detectable in PFs of rabbits treated with Abbokinase or Activase. Consistent with these findings, PAI-1 interacts with scuPA much slower than with Activase or Abbokinase in vitro. An equilibrium between active and inactive scuPA (k(on) = 4.3 h(-1)) limits the rate of its inactivation by PAI-1, favoring formation of complexes with alphaM. These observations define a newly recognized mechanism that promotes durable intrapleural fibrinolysis via formation of alphaM/uPA complexes. These complexes promote uPA-mediated plasminogen activation in scuPA-treated rabbits with TCN-induced pleural injury.


American Journal of Respiratory Cell and Molecular Biology | 2013

Intrapleural Adenoviral Delivery of Human Plasminogen Activator Inhibitor–1 Exacerbates Tetracycline-Induced Pleural Injury in Rabbits

Sophia Karandashova; Galina Florova; Ali O. Azghani; Andrey A. Komissarov; Kathy Koenig; Torry A. Tucker; Timothy Craig Allen; Kris Stewart; Amy Tvinnereim; Steven Idell

Elevated concentrations of plasminogen activator inhibitor-1 (PAI-1) are associated with pleural injury, but its effects on pleural organization remain unclear. A method of adenovirus-mediated delivery of genes of interest (expressed under a cytomegalovirus promoter) to rabbit pleura was developed and used with lacZ and human (h) PAI-1. Histology, β-galactosidase staining, Western blotting, enzymatic and immunohistochemical analyses of pleural fluids (PFs), lavages, and pleural mesothelial cells were used to evaluate the efficiency and effects of transduction. Transduction was selective and limited to the pleural mesothelial monolayer. The intrapleural expression of both genes was transient, with their peak expression at 4 to 5 days. On Day 5, hPAI-1 (40-80 and 200-400 nM of active and total hPAI-1 in lavages, respectively) caused no overt pleural injury, effusions, or fibrosis. The adenovirus-mediated delivery of hPAI-1 with subsequent tetracycline-induced pleural injury resulted in a significant exacerbation of the pleural fibrosis observed on Day 5 (P = 0.029 and P = 0.021 versus vehicle and adenoviral control samples, respectively). Intrapleural fibrinolytic therapy (IPFT) with plasminogen activators was effective in both animals overexpressing hPAI-1 and control animals with tetracycline injury alone. An increase in intrapleural active PAI-1 (from 10-15 nM in control animals to 20-40 nM in hPAI-1-overexpressing animals) resulted in the increased formation of PAI-1/plasminogen activator complexes in vivo. The decrease in intrapleural plasminogen-activating activity observed at 10 to 40 minutes after IPFT correlates linearly with the initial concentration of active PAI-1. Therefore, active PAI-1 in PFs affects the outcome of IPFT, and may be both a biomarker of pleural injury and a molecular target for its treatment.


Experimental Lung Research | 2007

INTRAPLEURAL LOW-MOLECULAR-WEIGHT UROKINASE OR TISSUE PLASMINOGEN ACTIVATOR VERSUS SINGLE-CHAIN UROKINASE IN TETRACYCLINE-INDUCED PLEURAL LOCULATION IN RABBITS

Steven Idell; Ali O. Azghani; Shande Chen; Kathy Koenig; Andrew P. Mazar; Lalitha Kodandapani; Khalil Bdeir; Douglas B. Cines; Irina Kulikovskaya; Timothy Craig Allen

The authors compared the ability of a single dose of the proenzyme single-chain urokinase (scuPA), low-molecular-weight urokinase, tissue plasminogen activator (tPA), or a mutant site-inactive scuPA to resolve intrapleural loculations at 72 to 96 hours after tetracycline-induced pleural injury in rabbits. Both scuPA and tPA reversed loculations at 96 hours after injury P ≤ .001, whereas low-molecular-weight urokinase and the scuPA mutant were ineffective. scuPA and tPA generated inhibitor complexes, induced fibrinolytic activity, and quenched plasminogen activator-1 activity in pleural fluids. The authors conclude that scuPA reverses loculations as effectively as tPA at clinically applied intrapleural doses, whereas low-molecular-weight urokinase was ineffective.


Shock | 2011

Wood Bark Smoke Induces Lung and Pleural Plasminogen Activator Inhibitor 1 and Stabilizes its mRNA in Porcine Lung Cells

Krishna K. Midde; Leopoldo C. Cancio; Sreerama Shetty; Andrey A. Komissarov; Galina Florova; Kerfoot Walker; Kathy Koenig; Zissis C. Chroneos; Timothy Craig Allen; Kevin K. Chung; Michael A. Dubick; Steven Idell

Although aberrant fibrinolysis and plasminogen activator inhibitor 1 (PAI-1) are implicated in acute lung injury, the role of this serpin in the pathogenesis of wood bark smoke (WBS)-induced acute lung injury (SIALI) and its regulation in resident lung cells after exposure to smoke are unclear. A total of 22 mechanically ventilated pigs were included in this study. Immunohistochemical analyses were used to assess fibrin and PAI-1 in the lungs of pigs with SIALI in situ. Plasminogen activator inhibitor 1 was measured in bronchoalveolar lavage fluids by Western blotting. Induction of PAI-1 was determined at the protein and mRNA levels by Western and polymerase chain reaction analyses in primary porcine alveolar type II cells, fibroblasts, and pleural mesothelial cells. Plasminogen activator inhibitor 1 mRNA stability was determined by transcription chase studies. Gel shift analyses were used to characterize the mechanism regulating PAI-1 mRNA stability. Smoke-induced ALI induced PAI-1, with prominent extravascular fibrin deposition in large and small airways as well as alveolar and subpleural compartments. In pleural mesothelial cells, lung fibroblasts, and alveolar type II cells, PAI-1 mRNA was stabilized by WBS extract and contributed to induction of PAI-1. The mechanism involves dissociation of a novel 6-phospho-d-gluconate-NADP oxidoreductase-like PAI-1 mRNA binding protein from PAI-1 mRNA. Exposure to WBS induces prominent airway and mesothelial expression of PAI-1, associated with florid distribution of fibrin in SIALI in vivo Wood bark smoke components induce PAI-1 in vitro in part by stabilization of PAI-1 mRNA, a newly recognized pathway that may promote extravascular fibrin deposition and lung dysfunction in SIALI.ABBREVIATIONS-3&vprime; UTR-3&vprime; Untranslated region; 6-PGD-6-phospho-d-gluconate-NADP oxidoreductase; ARDS-Acute respiratory distress syndrome; APRV-Airway-pressure-release ventilation; ATII-Alveolar type II cells; AU-Arbitrary fluorescence emission units; BAL-Bronchoalveolar lavage; BALF-BAL fluid; CMV-Conventional mechanical ventilation; Fio2-Fraction of inspired oxygen; HBSS-Hanks balanced salt solution; ICU-Intensive care unit; PBS-Phosphate-buffered saline; PL-Plasmin; PGN-Plasminogen; PA-PGN activator; PAI-1-Plasminogen activator inhibitor 1; PMCs-Pleural mesothelial cells; PEEP-Positive end-expiratory pressure; Paco2-Partial pressure of carbon dioxide in arterial blood; Pao2-Partial pressure of oxygen in arterial blood; PIP-Peak inspiratory pressure; PFs-Pleural fluids; PFR-Pao2-to-Fio2 ratio; scuPA-Single-chain urokinase; SIALI-Smoke-induced acute lung injury; TV-tidal volume; uPA-Urokinase-type plasminogen activator; WBSE-Wood bark smoke extract


American Journal of Respiratory Cell and Molecular Biology | 2010

The urokinase receptor supports tumorigenesis of human malignant pleural mesothelioma cells.

Torry A. Tucker; Candice Dean; Andrey A. Komissarov; Kathy Koenig; Andrew P. Mazar; Usha R. Pendurthi; Timothy Craig Allen; Steven Idell

Malignant pleural mesothelioma (MPM) is a lethal neoplasm for which current therapy is unsatisfactory. The urokinase plasminogen activator receptor (uPAR) is associated with increased virulence of many solid neoplasms, but its role in the pathogenesis of MPM is currently unclear. We found that REN human pleural MPM cells expressed 4- to 10-fold more uPAR than MS-1 or M9K MPM cells or MeT5A human pleural mesothelial cells. In a new orthotopic murine model of MPM, we found that the kinetics of REN cell tumorigenesis is accelerated versus MS-1 or M9K cells, and that REN instillates generated larger tumors expressing increased uPAR, were more invasive, and caused earlier mortality. While REN, MS-1, and M9K tumors were all associated with prominent extravascular fibrin deposition, excised REN tumor homogenates were characterized by markedly increased uPAR at both the mRNA and protein levels. REN cells exhibited increased thymidine incorporation, which was attenuated in uPAR-silenced cells (P < 0.01). REN cells traversed three-dimensional fibrin gels while MS-1, M9K, and MeT5A cells did not. uPAR siRNA or uPAR blocking antibodies decreased REN cell migration and invasion, while uPA and fetal bovine serum augmented the effects. Transfection of relatively low uPAR expressing MS-1 cells with uPAR cDNA increased proliferation and migration in vitro and tumor formation in vivo. These observations link overexpression of uPAR to the pathogenesis of MPM, demonstrate that this receptor contributes to accelerated tumor growth in part through interactions with uPA, and suggest that uPAR may be a promising target for therapeutic intervention.


Experimental Lung Research | 2009

SINGLE-CHAIN UROKINASE IN EMPYEMA INDUCED BY PASTURELLA MULTOCIDA

Steven Idell; Moon Jun Na; Huai Liao; A. E. Gazar; Wonder P. Drake; Kirk B. Lane; Kathy Koenig; Andrey A. Komissarov; Torry A. Tucker; Richard W. Light

Intrapleural fibrin deposition and subsequent fibrosis characterize evolving empyema and contribute to the morbidity associated with this condition. Single-chain urokinase (scuPA) is proenzyme form of the urokinase plasminogen activator, which has recently been shown to effectively clear intrapleural loculation in tetracycline-induced pleurodesis in rabbits. The authors therefore hypothesized that scuPA could likewise improve intrapleural injury associated with empyema. The authors used a rabbit model of empyema induced by intrapleural administration of Pasturella multocida to test this hypothesis and determined the effects of intrapleural scuPA on pleural fluids indices of inflammation and intrapleural fibrosis. The authors found that intrapleural administration of scuPA was well tolerated, generated readily detectable fibrinolytic activity in the empyema fluids and did not induce intrapleural or systemic bleeding. Pleural fluid volume, intrapleural protein, and D-dimer concentrations were increased at 24 and 48 hours (P < .01, respectively) after induction of empyema. Intrapleural loculation did not occur in the scuPA- or vehicle control–treated animals and there was no significant change in the pleural empyema or thickening scores. These findings confirm that intrapleural scuPA generates fibrinolysis in empyema fluids but does not alter fibrotic repair at the pleural surface or the intensity of intrapleural inflammation in this empyema model.


European Respiratory Journal | 1995

Inhibition of factor Xa-mediated procoagulant activity of human lung fibroblasts and pleural mesothelial cells.

Anuradha Kumar; Kathy Koenig; Alice R. Johnson; Daryl S. Fair; Steven Idell

Extravascular fibrin deposition characterizes diverse forms of lung and pleural injury. Fibrin formation in these compartments is locally potentiated by the assembly and expression of the prothrombinase procoagulant complex (factors Xa, Va and II) at the surface of human lung fibroblasts and pleural mesothelial cells. We sought to identify structural domains on factor Xa that mediate expression of prothrombinase activity by these cells. In order to accomplish this objective, we used panels of monoclonal antibodies (MoAbs) to factor X to block prothrombinase assembly and function on the surface of cultured human lung fibroblasts and pleural mesothelial cells. Of 30 factor X MoAbs that recognized native factors X and Xa, 10 completely inhibited factor Xa function (prothrombin activation), and five others neutralized Xa function without affecting cell-binding, presumably by blocking the prothrombin binding site. Western blots showed that these inhibitory MoAbs reacted with the Xa heavy-chain. One MoAb that recognized the factor Xa light-chain blocked prothrombin activation at the factor Va binding site. Our results indicate that prothrombinase activity at the surface of lung parachymal or pleural cells can be blocked by MoAbs that interact with either the heavy- or light-chain of factors X. Antibodies that neutralize cell surface-expressed prothrombin activation offer a potential means to arrest pericellular fibrin formation in the lung and pleural space.


Clinical Pulmonary Medicine | 2017

Precision-guided, Personalized Intrapleural Fibrinolytic Therapy for Empyema and Complicated Parapneumonic Pleural Effusions: The Case for The Fibrinolytic Potential

Steven Idell; Galina Florova; Sreerama Shetty; Torry A. Tucker; Richard Idell; Kathy Koenig; Ali O. Azghani; Najib M. Rahman; Andrey A. Komissarov

Complicated pleural effusions and empyema with loculation and failed drainage are common clinical problems. In adults, intrapleural fibrinolytic therapy is commonly used with variable results and therapy remains empiric. Despite the intrapleural use of various plasminogen activators; fibrinolysins, for about sixty years, there is no clear consensus about which agent is most effective. Emerging evidence demonstrates that intrapleural administration of plasminogen activators is subject to rapid inhibition by plasminogen activator inhibitor-1 and that processing of fibrinolysins is importantly influenced by other factors including the levels and quality of pleural fluid DNA. Current therapy for loculation that accompanies pleural infections also includes surgery, which is invasive and for which patient selection can be problematic. Most of the clinical literature published to date has used flat dosing of intrapleural fibrinolytic therapy in all subjects but little is known about how that strategy influences the processing of the administered fibrinolysin or how this influences outcomes. We developed a new test of pleural fluids ex vivo, which is called the Fibrinolytic Potential or FP, in which a dose of a fibrinolysin is added to pleural fluids ex vivo after which the fibrinolytic activity is measured and normalized to baseline levels. Testing in preclinical and clinical empyema fluids reveals a wide range of responses, indicating that individual patients will likely respond differently to flat dosing of fibrinolysins. The test remains under development but is envisioned as a guide for dosing of these agents, representing a novel candidate approach to personalization of intrapleural fibrinolytic therapy.


American Journal of Respiratory and Critical Care Medicine | 2002

Single-chain urokinase alone or complexed to its receptor in tetracycline-induced pleuritis in rabbits

Steven Idell; Andrew P. Mazar; Douglas B. Cines; Alice Kuo; Graham Parry; Susan Gawlak; Jose Juarez; Kathy Koenig; Ali Azghani; Will Hadden; Jerry W. McLarty; Edmund J. Miller


American Journal of Pathology | 1995

Regulation of fibrin deposition by malignant mesothelioma.

Steven Idell; Siegfried Pueblitz; S. Emri; Y. Gungen; L. Gray; Anuradha Kumar; D. Holiday; Kathy Koenig; Alice R. Johnson

Collaboration


Dive into the Kathy Koenig's collaboration.

Top Co-Authors

Avatar

Steven Idell

University of Texas Health Science Center at San Antonio

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Torry A. Tucker

University of Texas Health Science Center at Tyler

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Timothy Craig Allen

University of Texas Health Science Center at Tyler

View shared research outputs
Top Co-Authors

Avatar

Galina Florova

University of Texas Health Science Center at Tyler

View shared research outputs
Top Co-Authors

Avatar

Ali O. Azghani

University of Texas at Tyler

View shared research outputs
Top Co-Authors

Avatar

Alice R. Johnson

University of Texas Health Science Center at Tyler

View shared research outputs
Top Co-Authors

Avatar

Anuradha Kumar

University of Texas Health Science Center at Tyler

View shared research outputs
Top Co-Authors

Avatar

Sreerama Shetty

University of Texas Health Science Center at Tyler

View shared research outputs
Researchain Logo
Decentralizing Knowledge