1
|
Calì B, Troiani M, Bressan S, Attanasio G, Merler S, Moscarda V, Mosole S, Ricci E, Guo C, Yuan W, Gallagher L, Lundberg A, Bernett I, Figueiredo I, Arzola RA, Abreut EB, D'Ambrosio M, Bancaro N, Brina D, Zumerle S, Pasquini E, Maddalena M, Lai P, Colucci M, Pernigoni N, Rinaldi A, Minardi D, Morlacco A, Moro FD, Sabbadin M, Galuppini F, Fassan M, Rüschoff JH, Moch H, Rescigno P, Francini E, Saieva C, Modesti M, Theurillat JP, Gillessen S, Wilgenbus P, Graf C, Ruf W, de Bono J, Alimonti A. Coagulation factor X promotes resistance to androgen-deprivation therapy in prostate cancer. Cancer Cell 2024; 42:1676-1692.e11. [PMID: 39303726 DOI: 10.1016/j.ccell.2024.08.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 06/13/2024] [Accepted: 08/22/2024] [Indexed: 09/22/2024]
Abstract
Although hypercoagulability is commonly associated with malignancies, whether coagulation factors directly affect tumor cell proliferation remains unclear. Herein, by performing single-cell RNA sequencing (scRNA-seq) of the prostate tumor microenvironment (TME) of mouse models of castration-resistant prostate cancer (CRPC), we report that immunosuppressive neutrophils (PMN-MDSCs) are a key extra-hepatic source of coagulation factor X (FX). FX activation within the TME enhances androgen-independent tumor growth by activating the protease-activated receptor 2 (PAR2) and the phosphorylation of ERK1/2 in tumor cells. Genetic and pharmacological inhibition of factor Xa (FXa) antagonizes the oncogenic activity of PMN-MDSCs, reduces tumor progression, and synergizes with enzalutamide therapy. Intriguingly, F10high PMN-MDSCs express the surface marker CD84 and CD84 ligation enhances F10 expression. Elevated levels of FX, CD84, and PAR2 in prostate tumors associate with worse survival in CRPC patients. This study provides evidence that FXa directly promotes cancer and highlights additional targets for PMN-MDSCs for cancer therapies.
Collapse
Affiliation(s)
- Bianca Calì
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland; Università della Svizzera Italiana, Faculty of Biomedical Sciences, CH6900 Lugano, Switzerland
| | - Martina Troiani
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland; Università della Svizzera Italiana, Faculty of Biomedical Sciences, CH6900 Lugano, Switzerland
| | - Silvia Bressan
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland; Università della Svizzera Italiana, Faculty of Biomedical Sciences, CH6900 Lugano, Switzerland; Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35122 Padova, Italy
| | - Giuseppe Attanasio
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland; Università della Svizzera Italiana, Faculty of Biomedical Sciences, CH6900 Lugano, Switzerland
| | - Sara Merler
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland; Università della Svizzera Italiana, Faculty of Biomedical Sciences, CH6900 Lugano, Switzerland; Section of Oncology, Department of Medicine, University of Verona, 37134 Verona, Italy; Medical Oncology Unit, Oncology Institute of Southern Switzerland, Ente Ospedaliero Cantonale, CH6500 Bellinzona, Switzerland; Veneto Institute of Molecular Medicine, 35129 Padova, Italy
| | - Viola Moscarda
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland; Università della Svizzera Italiana, Faculty of Biomedical Sciences, CH6900 Lugano, Switzerland; Section of Oncology, Department of Medicine, University of Verona, 37134 Verona, Italy
| | - Simone Mosole
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland; Università della Svizzera Italiana, Faculty of Biomedical Sciences, CH6900 Lugano, Switzerland
| | - Elena Ricci
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland; Università della Svizzera Italiana, Faculty of Biomedical Sciences, CH6900 Lugano, Switzerland; Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy
| | - Christina Guo
- The Institute of Cancer Research, The Royal Marsden Hospital, London SW3 6JJ, UK
| | - Wei Yuan
- The Institute of Cancer Research, The Royal Marsden Hospital, London SW3 6JJ, UK
| | - Lewis Gallagher
- The Institute of Cancer Research, The Royal Marsden Hospital, London SW3 6JJ, UK
| | - Arian Lundberg
- The Institute of Cancer Research, The Royal Marsden Hospital, London SW3 6JJ, UK
| | - Ilona Bernett
- The Institute of Cancer Research, The Royal Marsden Hospital, London SW3 6JJ, UK
| | - Ines Figueiredo
- The Institute of Cancer Research, The Royal Marsden Hospital, London SW3 6JJ, UK
| | - Rydell Alvarez Arzola
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland; Department of Immunoregulation, Immunology and Immunotherapy Division, Center of Molecular Immunology, La Habana 3GGH+C9G, Cuba
| | - Ernesto Bermudez Abreut
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland; Department of Immunoregulation, Immunology and Immunotherapy Division, Center of Molecular Immunology, La Habana 3GGH+C9G, Cuba
| | - Mariantonietta D'Ambrosio
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland; Università della Svizzera Italiana, Faculty of Biomedical Sciences, CH6900 Lugano, Switzerland
| | - Nicolò Bancaro
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland; Università della Svizzera Italiana, Faculty of Biomedical Sciences, CH6900 Lugano, Switzerland
| | - Daniela Brina
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland; Università della Svizzera Italiana, Faculty of Biomedical Sciences, CH6900 Lugano, Switzerland
| | - Sara Zumerle
- Veneto Institute of Molecular Medicine, 35129 Padova, Italy; Department of Medicine, University of Padova, 35121 Padova, Italy
| | - Emiliano Pasquini
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland; Università della Svizzera Italiana, Faculty of Biomedical Sciences, CH6900 Lugano, Switzerland
| | - Martino Maddalena
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland; Università della Svizzera Italiana, Faculty of Biomedical Sciences, CH6900 Lugano, Switzerland
| | - Ping Lai
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland; Università della Svizzera Italiana, Faculty of Biomedical Sciences, CH6900 Lugano, Switzerland
| | - Manuel Colucci
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland; Università della Svizzera Italiana, Faculty of Biomedical Sciences, CH6900 Lugano, Switzerland
| | - Nicolò Pernigoni
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland; Università della Svizzera Italiana, Faculty of Biomedical Sciences, CH6900 Lugano, Switzerland
| | - Andrea Rinaldi
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland; Università della Svizzera Italiana, Faculty of Biomedical Sciences, CH6900 Lugano, Switzerland
| | - Davide Minardi
- Veneto Institute of Molecular Medicine, 35129 Padova, Italy; Urology Clinic, Department of Surgery, Oncology and Gastroenterology, University of Padova, 35128 Padova, Italy
| | - Alessandro Morlacco
- Urology Clinic, Department of Surgery, Oncology and Gastroenterology, University of Padova, 35128 Padova, Italy
| | - Fabrizio Dal Moro
- Urology Clinic, Department of Surgery, Oncology and Gastroenterology, University of Padova, 35128 Padova, Italy
| | - Marianna Sabbadin
- Veneto Institute of Molecular Medicine, 35129 Padova, Italy; Department of Medicine, Surgical Pathology Unit, University of Padova, 35121 Padova, Italy
| | - Francesca Galuppini
- Department of Medicine, Surgical Pathology Unit, University of Padova, 35121 Padova, Italy
| | - Matteo Fassan
- Department of Medicine, Surgical Pathology Unit, University of Padova, 35121 Padova, Italy
| | - Jan Hendrik Rüschoff
- Department of Pathology and Molecular Pathology, University Hospital Zurich (USZ), 8091 Zurich, Switzerland
| | - Holger Moch
- Department of Pathology and Molecular Pathology, University Hospital Zurich (USZ), 8091 Zurich, Switzerland
| | | | - Edoardo Francini
- Medical Oncology Unit, Oncology Institute of Southern Switzerland, Ente Ospedaliero Cantonale, CH6500 Bellinzona, Switzerland; Department of Experimental and Clinical Medicine, University of Florence, 50121 Florence, Italy
| | - Calogero Saieva
- Cancer Risk Factors and Lifestyle Epidemiology Unit - ISPRO, 50139 Florence, Italy
| | - Mikol Modesti
- Medical Oncology Unit, Oncology Institute of Southern Switzerland, Ente Ospedaliero Cantonale, CH6500 Bellinzona, Switzerland
| | - Jean-Philippe Theurillat
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland; Università della Svizzera Italiana, Faculty of Biomedical Sciences, CH6900 Lugano, Switzerland
| | - Silke Gillessen
- Università della Svizzera Italiana, Faculty of Biomedical Sciences, CH6900 Lugano, Switzerland; Medical Oncology Unit, Oncology Institute of Southern Switzerland, Ente Ospedaliero Cantonale, CH6500 Bellinzona, Switzerland
| | - Petra Wilgenbus
- Center for Thrombosis and Hemostasis, Johannes Gutenberg University Medical Center, 55131 Mainz, Germany; Department of Immunology and Microbiology, Scripps Research, La Jolla, CA 92037, USA
| | - Claudine Graf
- Center for Thrombosis and Hemostasis, Johannes Gutenberg University Medical Center, 55131 Mainz, Germany; Department of Immunology and Microbiology, Scripps Research, La Jolla, CA 92037, USA
| | - Wolfram Ruf
- Center for Thrombosis and Hemostasis, Johannes Gutenberg University Medical Center, 55131 Mainz, Germany; Department of Immunology and Microbiology, Scripps Research, La Jolla, CA 92037, USA
| | - Johann de Bono
- The Institute of Cancer Research, The Royal Marsden Hospital, London SW3 6JJ, UK
| | - Andrea Alimonti
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland; Università della Svizzera Italiana, Faculty of Biomedical Sciences, CH6900 Lugano, Switzerland; Medical Oncology Unit, Oncology Institute of Southern Switzerland, Ente Ospedaliero Cantonale, CH6500 Bellinzona, Switzerland; Veneto Institute of Molecular Medicine, 35129 Padova, Italy; Department of Medicine, University of Padova, 35121 Padova, Italy; Department of Health Sciences and Technology (D-HEST) ETH Zurich, 8092 Zurich, Switzerland.
| |
Collapse
|
2
|
Garlapati V, Luo Q, Posma J, Aluia M, Nguyen TS, Grunz K, Molitor M, Finger S, Harms G, Bopp T, Ruf W, Wenzel P. Macrophage-Expressed Coagulation Factor VII Promotes Adverse Cardiac Remodeling. Circ Res 2024; 135:841-855. [PMID: 39234697 DOI: 10.1161/circresaha.123.324114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 08/23/2024] [Accepted: 08/25/2024] [Indexed: 09/06/2024]
Abstract
BACKGROUND Excess fibrotic remodeling causes cardiac dysfunction in ischemic heart disease, driven by MAP (mitogen-activated protein) kinase-dependent TGF-ß1 (transforming growth factor-ß1) activation by coagulation signaling of myeloid cells. How coagulation-inflammatory circuits can be specifically targeted to achieve beneficial macrophage reprogramming after myocardial infarction (MI) is not completely understood. METHODS Mice with permanent ligation of the left anterior descending artery were used to model nonreperfused MI and analyzed by single-cell RNA sequencing, protein expression changes, confocal microscopy, and longitudinal monitoring of recovery. We probed the role of the tissue factor (TF)-FVIIa (activated factor VII)-integrin ß1-PAR2 (protease-activated receptor 2) signaling complex by utilizing genetic mouse models and pharmacological intervention. RESULTS Cleavage-insensitive PAR2R38E and myeloid cell integrin ß1-deficient mice had improved cardiac function after MI compared with controls. Proximity ligation assays of monocytic cells demonstrated that colocalization of FVIIa with integrin ß1 was diminished in monocyte/macrophage FVII-deficient mice after MI. Compared with controls, F7fl/fl CX3CR1 (CX3C motif chemokine receptor 1)Cre mice showed reduced TGF-ß1 and MAP kinase activation, as well as cardiac dysfunction after MI, despite unaltered overall recruitment of myeloid cells. Single-cell mRNA sequencing of CD45 (cluster of differentiation 45)+ cells 3 and 7 days after MI uncovered a trajectory from recruited monocytes to inflammatory TF+/TREM (triggered receptor expressed on myeloid cells) 1+ macrophages requiring F7. As early as 7 days after MI, macrophage F7 deletion led to an expansion of reparative Olfml 3 (olfactomedin-like protein 3)+ macrophages and, conversely, to a reduction of TF+/TREM1+ macrophages, which were also reduced in PAR2R38E mice. Short-term treatment from days 1 to 5 after nonreperfused MI with a monoclonal antibody inhibiting the macrophage TF-FVIIa-PAR2 signaling complex without anticoagulant activity improved cardiac dysfunction, decreased excess fibrosis, attenuated vascular endothelial dysfunction, and increased survival 28 days after MI. CONCLUSIONS Extravascular TF-FVIIa-PAR2 complex signaling drives inflammatory macrophage polarization in ischemic heart disease. Targeting this signaling complex for specific therapeutic macrophage reprogramming following MI attenuates cardiac fibrosis and improves cardiovascular function.
Collapse
Affiliation(s)
- Venkata Garlapati
- Center for Thrombosis and Hemostasis (V.G., Q.L., J.P., M.A., T.S.N., K.G., M.M., S.F., W.R., P.W.), University Medical Center Mainz, Germany
- Department of Cardiology (V.G., Q.L., M.A., M.M., S.F., P.W.), University Medical Center Mainz, Germany
- German Center for Cardiovascular Research-Partner site Rhine-Main (V.G., Q.L., M.A., M.M., W.R., P.W.), University Medical Center Mainz, Germany
| | - Qi Luo
- Center for Thrombosis and Hemostasis (V.G., Q.L., J.P., M.A., T.S.N., K.G., M.M., S.F., W.R., P.W.), University Medical Center Mainz, Germany
- Department of Cardiology (V.G., Q.L., M.A., M.M., S.F., P.W.), University Medical Center Mainz, Germany
- German Center for Cardiovascular Research-Partner site Rhine-Main (V.G., Q.L., M.A., M.M., W.R., P.W.), University Medical Center Mainz, Germany
- Department of Biochemistry, Cardiovascular Research Maastricht University, the Netherlands (Q.L.)
| | - Jens Posma
- Center for Thrombosis and Hemostasis (V.G., Q.L., J.P., M.A., T.S.N., K.G., M.M., S.F., W.R., P.W.), University Medical Center Mainz, Germany
| | - Melania Aluia
- Center for Thrombosis and Hemostasis (V.G., Q.L., J.P., M.A., T.S.N., K.G., M.M., S.F., W.R., P.W.), University Medical Center Mainz, Germany
- Department of Cardiology (V.G., Q.L., M.A., M.M., S.F., P.W.), University Medical Center Mainz, Germany
- German Center for Cardiovascular Research-Partner site Rhine-Main (V.G., Q.L., M.A., M.M., W.R., P.W.), University Medical Center Mainz, Germany
| | - Than Son Nguyen
- Center for Thrombosis and Hemostasis (V.G., Q.L., J.P., M.A., T.S.N., K.G., M.M., S.F., W.R., P.W.), University Medical Center Mainz, Germany
| | - Kristin Grunz
- Center for Thrombosis and Hemostasis (V.G., Q.L., J.P., M.A., T.S.N., K.G., M.M., S.F., W.R., P.W.), University Medical Center Mainz, Germany
| | - Michael Molitor
- Center for Thrombosis and Hemostasis (V.G., Q.L., J.P., M.A., T.S.N., K.G., M.M., S.F., W.R., P.W.), University Medical Center Mainz, Germany
- Department of Cardiology (V.G., Q.L., M.A., M.M., S.F., P.W.), University Medical Center Mainz, Germany
- German Center for Cardiovascular Research-Partner site Rhine-Main (V.G., Q.L., M.A., M.M., W.R., P.W.), University Medical Center Mainz, Germany
| | - Stefanie Finger
- Center for Thrombosis and Hemostasis (V.G., Q.L., J.P., M.A., T.S.N., K.G., M.M., S.F., W.R., P.W.), University Medical Center Mainz, Germany
- Department of Cardiology (V.G., Q.L., M.A., M.M., S.F., P.W.), University Medical Center Mainz, Germany
| | - Gregory Harms
- Institute of Immunology and Research Center for Immunotherapy (G.H., T.B.), University Medical Center Mainz, Germany
- Cell Biology Unit (G.H.), University Medical Center Mainz, Germany
- Department of Biology, Wilkes University, Wilkes-Barre, PA (G.H.)
| | - Tobias Bopp
- Institute of Immunology and Research Center for Immunotherapy (G.H., T.B.), University Medical Center Mainz, Germany
| | - Wolfram Ruf
- Center for Thrombosis and Hemostasis (V.G., Q.L., J.P., M.A., T.S.N., K.G., M.M., S.F., W.R., P.W.), University Medical Center Mainz, Germany
- German Center for Cardiovascular Research-Partner site Rhine-Main (V.G., Q.L., M.A., M.M., W.R., P.W.), University Medical Center Mainz, Germany
- Department of Immunology and Microbiology, Scripps Research, La Jolla, CA (W.R.)
| | - Philip Wenzel
- Center for Thrombosis and Hemostasis (V.G., Q.L., J.P., M.A., T.S.N., K.G., M.M., S.F., W.R., P.W.), University Medical Center Mainz, Germany
- Department of Cardiology (V.G., Q.L., M.A., M.M., S.F., P.W.), University Medical Center Mainz, Germany
- German Center for Cardiovascular Research-Partner site Rhine-Main (V.G., Q.L., M.A., M.M., W.R., P.W.), University Medical Center Mainz, Germany
| |
Collapse
|
3
|
Kespohl M, Goetzke CC, Althof N, Bredow C, Kelm N, Pinkert S, Bukur T, Bukur V, Grunz K, Kaur D, Heuser A, Mülleder M, Sauter M, Klingel K, Weiler H, Berndt N, Gaida MM, Ruf W, Beling A. TF-FVIIa PAR2-β-Arrestin Signaling Sustains Organ Dysfunction in Coxsackievirus B3 Infection of Mice. Arterioscler Thromb Vasc Biol 2024; 44:843-865. [PMID: 38385286 DOI: 10.1161/atvbaha.123.320157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 02/05/2024] [Indexed: 02/23/2024]
Abstract
BACKGROUND Accumulating evidence implicates the activation of G-protein-coupled PARs (protease-activated receptors) by coagulation proteases in the regulation of innate immune responses. METHODS Using mouse models with genetic alterations of the PAR2 signaling platform, we have explored contributions of PAR2 signaling to infection with coxsackievirus B3, a single-stranded RNA virus provoking multiorgan tissue damage, including the heart. RESULTS We show that PAR2 activation sustains correlates of severe morbidity-hemodynamic compromise, aggravated hypothermia, and hypoglycemia-despite intact control of the virus. Following acute viral liver injury, canonical PAR2 signaling impairs the restoration process associated with exaggerated type I IFN (interferon) signatures in response to viral RNA recognition. Metabolic profiling in combination with proteomics of liver tissue shows PAR2-dependent reprogramming of liver metabolism, increased lipid droplet storage, and gluconeogenesis. PAR2-sustained hypodynamic compromise, reprograming of liver metabolism, as well as imbalanced IFN responses are prevented in β-arrestin coupling-deficient PAR2 C-terminal phosphorylation mutant mice. Thus, wiring between upstream proteases and immune-metabolic responses results from biased PAR2 signaling mediated by intracellular recruitment of β-arrestin. Importantly, blockade of the TF (tissue factor)-FVIIa (coagulation factor VIIa) complex capable of PAR2 proteolysis with the NAPc2 (nematode anticoagulant protein c2) mitigated virus-triggered pathology, recapitulating effects seen in protease cleavage-resistant PAR2 mice. CONCLUSIONS These data provide insights into a TF-FVIIa signaling axis through PAR2-β-arrestin coupling that is a regulator of inflammation-triggered tissue repair and hemodynamic compromise in coxsackievirus B3 infection and can potentially be targeted with selective coagulation inhibitors.
Collapse
Affiliation(s)
- Meike Kespohl
- Institute of Biochemistry (M.K., C.B., N.K., S.P., A.B.), Charité-Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany
- Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK), partner site Berlin, Germany (M.K., A.B.)
| | - Carl Christoph Goetzke
- Department of Pediatrics, Division of Pulmonology, Immunology and Critical Care Medicine (C.C.G.), Charité-Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany
- Clinician Scientist Program, BIH (Berlin Institute of Health) Academy, BIH, Charité-Universitätsmedizin Berlin, Germany (C.C.G.)
- German Rheumatism Research Center, Leibniz Association, Berlin, Germany (C.C.G.)
| | - Nadine Althof
- German Federal Institute for Risk Assessment, Berlin, Germany (N.A.)
| | - Clara Bredow
- Institute of Biochemistry (M.K., C.B., N.K., S.P., A.B.), Charité-Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany
| | - Nicolas Kelm
- Institute of Biochemistry (M.K., C.B., N.K., S.P., A.B.), Charité-Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany
| | - Sandra Pinkert
- Institute of Biochemistry (M.K., C.B., N.K., S.P., A.B.), Charité-Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany
| | - Thomas Bukur
- Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz (TRON), Germany (T.B., V.B.)
| | - Valesca Bukur
- Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz (TRON), Germany (T.B., V.B.)
| | - Kristin Grunz
- Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK), partner site Rhein-Main, Germany (K.G., D.K., W.R.)
- University Medical Center Mainz, Center for Thrombosis and Hemostasis, Germany (K.G., D.K., W.R.)
| | - Dilraj Kaur
- Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK), partner site Rhein-Main, Germany (K.G., D.K., W.R.)
- University Medical Center Mainz, Center for Thrombosis and Hemostasis, Germany (K.G., D.K., W.R.)
| | - Arnd Heuser
- Max-Delbrueck-Center for Molecular Medicine, Animal Phenotyping Platform, Berlin, Germany (A.H.)
| | - Michael Mülleder
- Core Facility High-Throughput Mass Spectrometry (M.M.), Charité-Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany
| | - Martina Sauter
- University Hospital Tuebingen, Institute for Pathology and Neuropathology, Cardiopathology, Germany (M.S., K.K.)
| | - Karin Klingel
- University Hospital Tuebingen, Institute for Pathology and Neuropathology, Cardiopathology, Germany (M.S., K.K.)
| | | | - Nikolaus Berndt
- Deutsches Herzzentrum der Charité, Institute of Computer-Assisted Cardiovascular Medicine, Berlin, Germany (N.B.)
- Charité-Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany (N.B.)
- German Institute of Human Nutrition Potsdam-Rehbruecke, Department of Molecular Toxicology, Nuthetal, Germany (N.B.)
| | - Matthias M Gaida
- University Medical Center Mainz, Institute for Pathology, Johannes-Gutenberg-Universität Mainz, Germany (M.M.G.)
- University Medical Center Mainz, Research Center for Immunotherapy, Johannes-Gutenberg-Universität Mainz, Germany (M.M.G.)
- Joint Unit Immunopathology, Institute of Pathology, University Medical Center, Johannes Gutenberg University of Mainz, Germany (M.M.G.)
- TRON, Mainz, Germany (M.M.G.)
| | - Wolfram Ruf
- Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK), partner site Rhein-Main, Germany (K.G., D.K., W.R.)
- University Medical Center Mainz, Center for Thrombosis and Hemostasis, Germany (K.G., D.K., W.R.)
| | - Antje Beling
- Institute of Biochemistry (M.K., C.B., N.K., S.P., A.B.), Charité-Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany
- Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK), partner site Berlin, Germany (M.K., A.B.)
| |
Collapse
|
4
|
Müller-Calleja N, Grunz K, Nguyen TS, Posma J, Pedrosa D, Meineck M, Hollerbach A, Braun J, Muth S, Schild H, Saar K, Hübner N, Krishnaswamy S, Royce J, Teyton L, Lemmermann N, Weinmann-Menke J, Lackner KJ, Ruf W. Targeting the tissue factor coagulation initiation complex prevents antiphospholipid antibody development. Blood 2024; 143:1167-1180. [PMID: 38142429 PMCID: PMC10972716 DOI: 10.1182/blood.2023022276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 12/08/2023] [Accepted: 12/13/2023] [Indexed: 12/26/2023] Open
Abstract
ABSTRACT Antiphospholipid antibodies (aPL) in primary or secondary antiphospholipid syndrome (APS) are a major cause for acquired thrombophilia, but specific interventions preventing autoimmune aPL development are an unmet clinical need. Although autoimmune aPL cross react with various coagulation regulatory proteins, lipid-reactive aPL, including those derived from patients with COVID-19, recognize the endolysosomal phospholipid lysobisphosphatidic acid presented by the cell surface-expressed endothelial protein C receptor. This specific recognition leads to complement-mediated activation of tissue factor (TF)-dependent proinflammatory signaling and thrombosis. Here, we show that specific inhibition of the TF coagulation initiation complex with nematode anticoagulant protein c2 (NAPc2) prevents the prothrombotic effects of aPL derived from patients with COVID-19 in mice and the aPL-induced proinflammatory and prothrombotic activation of monocytes. The induction of experimental APS is dependent on the nicotinamide adenine dinucleotide phosphate (NADPH) oxidase complex, and NAPc2 suppresses monocyte endosomal reactive oxygen species production requiring the TF cytoplasmic domain and interferon-α secretion from dendritic cells. Latent infection with murine cytomegalovirus causes TF cytoplasmic domain-dependent development of persistent aPL and circulating phospholipid-reactive B1 cells, which is prevented by short-term intervention with NAPc2 during acute viral infection. In addition, treatment of lupus prone MRL-lpr mice with NAPc2, but not with heparin, suppresses dendritic-cell activation in the spleen, aPL production and circulating phospholipid-reactive B1 cells, and attenuates lupus pathology. These data demonstrate a convergent TF-dependent mechanism of aPL development in latent viral infection and autoimmune disease and provide initial evidence that specific targeting of the TF initiation complex has therapeutic benefits beyond currently used clinical anticoagulant strategies.
Collapse
Affiliation(s)
- Nadine Müller-Calleja
- Center for Thrombosis and Hemostasis, Johannes Gutenberg University Medical Center, Mainz, Germany
- Institute of Clinical Chemistry and Laboratory Medicine, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Kristin Grunz
- Center for Thrombosis and Hemostasis, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - T. Son Nguyen
- Center for Thrombosis and Hemostasis, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Jens Posma
- Center for Thrombosis and Hemostasis, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Denise Pedrosa
- Center for Thrombosis and Hemostasis, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Myriam Meineck
- Department of Medicine I, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Anne Hollerbach
- Institute of Clinical Chemistry and Laboratory Medicine, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Johannes Braun
- Center for Thrombosis and Hemostasis, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Sabine Muth
- Institute for Immunology, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Hansjörg Schild
- Institute for Immunology, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Kathrin Saar
- Max Delbrück Center for Molecular Medicine, Berlin, Germany
- Charite-Universitätsmedizin Berlin, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Partner site Berlin, Berlin, Germany
| | - Norbert Hübner
- Max Delbrück Center for Molecular Medicine, Berlin, Germany
- Charite-Universitätsmedizin Berlin, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Partner site Berlin, Berlin, Germany
| | - Sriram Krishnaswamy
- Department of Pediatrics, Children’s Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA
| | - Jennifer Royce
- Department of Immunology and Microbiology, Scripps Research, La Jolla, CA
| | - Luc Teyton
- Department of Immunology and Microbiology, Scripps Research, La Jolla, CA
| | - Niels Lemmermann
- Institute for Virology, Johannes Gutenberg University Medical Center, Mainz, Germany
- Institute of Virology, University Hospital Bonn, Bonn, Germany
| | - Julia Weinmann-Menke
- Department of Medicine I, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Karl J. Lackner
- Institute of Clinical Chemistry and Laboratory Medicine, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Wolfram Ruf
- Center for Thrombosis and Hemostasis, Johannes Gutenberg University Medical Center, Mainz, Germany
- Department of Immunology and Microbiology, Scripps Research, La Jolla, CA
- German Center for Cardiovascular Research (DZHK), Partner site Rhein-Main, Mainz, Germany
| |
Collapse
|
5
|
Garlapati V, Molitor M, Michna T, Harms GS, Finger S, Jung R, Lagrange J, Efentakis P, Wild J, Knorr M, Karbach S, Wild S, Vujacic-Mirski K, Münzel T, Daiber A, Brandt M, Gori T, Milting H, Tenzer S, Ruf W, Wenzel P. Targeting myeloid cell coagulation signaling blocks MAP kinase/TGF-β1-driven fibrotic remodeling in ischemic heart failure. J Clin Invest 2023; 133:156436. [PMID: 36548062 PMCID: PMC9927945 DOI: 10.1172/jci156436] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 12/16/2022] [Indexed: 12/24/2022] Open
Abstract
Despite major advances in acute interventions for myocardial infarction (MI), adverse cardiac remodeling and excess fibrosis after MI causing ischemic heart failure (IHF) remain a leading cause of death worldwide. Here we identify a profibrotic coagulation signaling pathway that can be targeted for improved cardiac function following MI with persistent ischemia. Quantitative phosphoproteomics of cardiac tissue revealed an upregulated mitogen-activated protein kinase (MAPK) pathway in human IHF. Intervention in this pathway with trametinib improves myocardial function and prevents fibrotic remodeling in a murine model of non-reperfused MI. MAPK activation in MI requires myeloid cell signaling of protease-activated receptor 2 linked to the cytoplasmic domain of the coagulation initiator tissue factor (TF). They act upstream of pro-oxidant NOX2 NADPH oxidase, ERK1/2 phosphorylation, and activation of profibrotic TGF-β1. Specific targeting with the TF inhibitor nematode anticoagulant protein c2 (NAPc2) starting 1 day after established experimental MI averts IHF. Increased TF cytoplasmic domain phosphorylation in circulating monocytes from patients with subacute MI identifies a potential thromboinflammatory biomarker reflective of increased risk for IHF and suitable for patient selection to receive targeted TF inhibition therapy.
Collapse
Affiliation(s)
- Venkata Garlapati
- Center for Thrombosis and Hemostasis and.,Department of Cardiology, University Medical Center Mainz, Mainz, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany
| | - Michael Molitor
- Center for Thrombosis and Hemostasis and.,Department of Cardiology, University Medical Center Mainz, Mainz, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany
| | - Thomas Michna
- Institute of Immunology, University Medical Center Mainz, Mainz, Germany
| | - Gregory S Harms
- Cell Biology Unit, University Medical Center Mainz, Mainz, Germany and.,Departments of Biology and Physics, Wilkes University, Wilkes-Barre, Pennsylvania, USA
| | - Stefanie Finger
- Center for Thrombosis and Hemostasis and.,Department of Cardiology, University Medical Center Mainz, Mainz, Germany
| | - Rebecca Jung
- Center for Thrombosis and Hemostasis and.,Department of Cardiology, University Medical Center Mainz, Mainz, Germany.,Institute for Molecular Medicine, University Medical Center Mainz, Mainz, Germany
| | | | | | - Johannes Wild
- Center for Thrombosis and Hemostasis and.,Department of Cardiology, University Medical Center Mainz, Mainz, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany
| | - Maike Knorr
- Center for Thrombosis and Hemostasis and.,Department of Cardiology, University Medical Center Mainz, Mainz, Germany
| | - Susanne Karbach
- Center for Thrombosis and Hemostasis and.,Department of Cardiology, University Medical Center Mainz, Mainz, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany
| | - Sabine Wild
- Center for Thrombosis and Hemostasis and.,Department of Cardiology, University Medical Center Mainz, Mainz, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany
| | | | - Thomas Münzel
- Center for Thrombosis and Hemostasis and.,Department of Cardiology, University Medical Center Mainz, Mainz, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany
| | - Andreas Daiber
- Department of Cardiology, University Medical Center Mainz, Mainz, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany
| | - Moritz Brandt
- Center for Thrombosis and Hemostasis and.,Department of Cardiology, University Medical Center Mainz, Mainz, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany
| | - Tommaso Gori
- Department of Cardiology, University Medical Center Mainz, Mainz, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany
| | - Hendrik Milting
- Erich und Hanna Klessmann-Institut für Kardiovaskuläre Forschung und Entwicklung, Herz- und Diabeteszentrum NRW, Bad Oeynhausen, Germany
| | - Stefan Tenzer
- Institute of Immunology, University Medical Center Mainz, Mainz, Germany.,Helmholtz Institute for Translational Oncology (HI-TRON) Mainz, Germany and.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Wolfram Ruf
- Center for Thrombosis and Hemostasis and.,German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany.,Department of Immunology and Microbiology, Scripps Research, La Jolla, California, USA
| | - Philip Wenzel
- Center for Thrombosis and Hemostasis and.,Department of Cardiology, University Medical Center Mainz, Mainz, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany
| |
Collapse
|
6
|
Bochenek ML, Gogiraju R, Großmann S, Krug J, Orth J, Reyda S, Georgiadis GS, Spronk H, Konstantinides S, Münzel T, Griffin JH, Wild PS, Espinola-Klein C, Ruf W, Schäfer K. EPCR-PAR1 biased signaling regulates perfusion recovery and neovascularization in peripheral ischemia. JCI Insight 2022; 7:157701. [PMID: 35700057 PMCID: PMC9431695 DOI: 10.1172/jci.insight.157701] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 06/08/2022] [Indexed: 11/17/2022] Open
Abstract
Blood clot formation initiates ischemic events, but coagulation roles during postischemic tissue repair are poorly understood. The endothelial protein C receptor (EPCR) regulates coagulation, as well as immune and vascular signaling, by protease activated receptors (PARs). Here, we show that endothelial EPCR-PAR1 signaling supports reperfusion and neovascularization in hindlimb ischemia in mice. Whereas deletion of PAR2 or PAR4 did not impair angiogenesis, EPCR and PAR1 deficiency or PAR1 resistance to cleavage by activated protein C caused markedly reduced postischemic reperfusion in vivo and angiogenesis in vitro. These findings were corroborated by biased PAR1 agonism in isolated primary endothelial cells. Loss of EPCR-PAR1 signaling upregulated hemoglobin expression and reduced endothelial nitric oxide (NO) bioavailability. Defective angiogenic sprouting was rescued by the NO donor DETA-NO, whereas NO scavenging increased hemoglobin and mesenchymal marker expression in human and mouse endothelial cells. Vascular specimens from patients with ischemic peripheral artery disease exhibited increased hemoglobin expression, and soluble EPCR and NO levels were reduced in plasma. Our data implicate endothelial EPCR-PAR1 signaling in the hypoxic response of endothelial cells and identify suppression of hemoglobin expression as an unexpected link between coagulation signaling, preservation of endothelial cell NO bioavailability, support of neovascularization, and prevention of fibrosis.
Collapse
Affiliation(s)
- Magdalena L Bochenek
- Center for Thrombosis and Hemostasis, University Medical Center Mainz, Mainz, Germany
| | | | - Stefanie Großmann
- Department of Cardiology, University Medical Center Mainz, Mainz, Germany
| | - Janina Krug
- Department of Cardiology, University Medical Center Mainz, Mainz, Germany
| | - Jennifer Orth
- Department of Cardiology, University Medical Center Mainz, Mainz, Germany
| | - Sabine Reyda
- Center for Thrombosis and Hemostasis, University Medical Center Mainz, Mainz, Germany
| | - George S Georgiadis
- Department of Vascular Surgery, University Hospital of Alexandroupolis, Alexandroupolis, Greece
| | - Henri Spronk
- CARIM School for Cardiovascular Disease, Maastricht University, Maastricht, Netherlands
| | | | - Thomas Münzel
- Center for Thrombosis and Hemostasis, University Medical Center Mainz, Mainz, Germany
| | - John H Griffin
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, United States of America
| | - Philipp S Wild
- Center for Thrombosis and Hemostasis, University Medical Center Mainz, Mainz, Germany
| | | | - Wolfram Ruf
- Center for Thrombosis and Hemostasis, University Medical Center Mainz, Mainz, Germany
| | - Katrin Schäfer
- Center for Thrombosis and Hemostasis, University Medical Center Mainz, Mainz, Germany
| |
Collapse
|
7
|
Functional Characteristics and Regulated Expression of Alternatively Spliced Tissue Factor: An Update. Cancers (Basel) 2021; 13:cancers13184652. [PMID: 34572880 PMCID: PMC8471299 DOI: 10.3390/cancers13184652] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 09/10/2021] [Accepted: 09/13/2021] [Indexed: 12/11/2022] Open
Abstract
In human and mouse, alternative splicing of tissue factor's primary transcript yields two mRNA species: one features all six TF exons and encodes full-length tissue factor (flTF), and the other lacks exon 5 and encodes alternatively spliced tissue factor (asTF). flTF, which is oftentimes referred to as "TF", is an integral membrane glycoprotein due to the presence of an alpha-helical domain in its C-terminus, while asTF is soluble due to the frameshift resulting from the joining of exon 4 directly to exon 6. In this review, we focus on asTF-the more recently discovered isoform of TF that appears to significantly contribute to the pathobiology of several solid malignancies. There is currently a consensus in the field that asTF, while dispensable to normal hemostasis, can activate a subset of integrins on benign and malignant cells and promote outside-in signaling eliciting angiogenesis; cancer cell proliferation, migration, and invasion; and monocyte recruitment. We provide a general overview of the pioneering, as well as more recent, asTF research; discuss the current concepts of how asTF contributes to cancer progression; and open a conversation about the emerging utility of asTF as a biomarker and a therapeutic target.
Collapse
|
8
|
Abuzeid AMI, Zhou X, Huang Y, Li G. Twenty-five-year research progress in hookworm excretory/secretory products. Parasit Vectors 2020; 13:136. [PMID: 32171305 PMCID: PMC7071665 DOI: 10.1186/s13071-020-04010-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 03/06/2020] [Indexed: 11/16/2022] Open
Abstract
Hookworm infection is a major public health problem that threatens about 500 million people throughout tropical areas of the world. Adult hookworms survive for many years in the host intestine, where they suck blood, causing iron deficiency anemia and malnutrition. Numerous molecules, named excretory/secretory (ES) products, are secreted by hookworm adults and/or larvae to aid in parasite survival and pathobiology. Although the molecular cloning and characterization of hookworm ES products began 25 years ago, the biological role and molecular nature of many of them are still unclear. Hookworm ES products, with distinct structures and functions, have been linked to many essential events in the disease pathogenesis. These events include host invasion and tissue migration, parasite nourishment and reproduction, and immune modulation. Several of these products represent promising vaccine targets for controlling hookworm disease and therapeutic targets for many inflammatory diseases. This review aims to summarize our present knowledge about hookworm ES products, including their role in parasite biology, host-parasite interactions, and as vaccine and pharmaceutical targets and to identify research gaps and future research directions in this field.![]()
Collapse
Affiliation(s)
- Asmaa M I Abuzeid
- Guangdong Provincial Zoonosis Prevention and Control Key Laboratory, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Xue Zhou
- Guangdong Provincial Zoonosis Prevention and Control Key Laboratory, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Yue Huang
- Guangdong Provincial Zoonosis Prevention and Control Key Laboratory, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Guoqing Li
- Guangdong Provincial Zoonosis Prevention and Control Key Laboratory, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China.
| |
Collapse
|
9
|
Duplantier AJ, Shurtleff AC, Miller C, Chiang CY, Panchal RG, Sunay M. Combating biothreat pathogens: ongoing efforts for countermeasure development and unique challenges. DRUG DISCOVERY TARGETING DRUG-RESISTANT BACTERIA 2020. [PMCID: PMC7258707 DOI: 10.1016/b978-0-12-818480-6.00007-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Research to discover and develop antibacterial and antiviral drugs with potent activity against pathogens of biothreat concern presents unique methodological and process-driven challenges. Herein, we review laboratory approaches for finding new antibodies, antibiotics, and antiviral molecules for pathogens of biothreat concern. Using high-throughput screening techniques, molecules that directly inhibit a pathogen’s entry, replication, or growth can be identified. Alternatively, molecules that target host proteins can be interesting targets for development when countering biothreat pathogens, due to the modulation of the host immune response or targeting proteins that interfere with the pathways required by the pathogen for replication. Monoclonal and cocktail antibody therapies approved by the Food and Drug Administration for countering anthrax and under development for treatment of Ebola virus infection are discussed. A comprehensive tabular review of current in vitro, in vivo, pharmacokinetic and efficacy datasets has been presented for biothreat pathogens of greatest concern. Finally, clinical trials and animal rule or traditional drug approval pathways are also reviewed. Opinions; interpretations; conclusions; and recommendations are those of the authors and are not necessarily endorsed by the US Army.
Collapse
|
10
|
van den Boogaard FE, Brands X, Duitman J, de Stoppelaar SF, Borensztajn KS, Roelofs JJTH, Hollenberg MD, Spek CA, Schultz MJ, van 't Veer C, van der Poll T. Protease-Activated Receptor 2 Facilitates Bacterial Dissemination in Pneumococcal Pneumonia. J Infect Dis 2019; 217:1462-1471. [PMID: 29415278 DOI: 10.1093/infdis/jiy010] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 01/18/2018] [Indexed: 11/13/2022] Open
Abstract
Streptococcus pneumoniae is the most common causative pathogen in community-acquired pneumonia. Protease-activated receptor 2 (PAR2) is expressed by different cell types in the lungs and can mediate inflammatory responses. We sought to determine the role of PAR2 during pneumococcal pneumonia. Pneumococcal pneumonia or sepsis was induced in wild-type and PAR2 knock-out (Par2-/-) mice by infection with viable S. pneumoniae. Par2-/- mice demonstrated improved host defense, a largely preserved lung barrier integrity, and reduced mortality during pneumococcal pneumonia. PAR2 deficiency did not influence bacterial growth after intravenous infection. Inhibition of the endogenous PAR2 activating proteases tissue factor/factor VIIa or tryptase did not impact on bacterial burdens during pneumonia. In a PAR2 reporter cell line it was demonstrated that S. pneumoniae-derived proteases are able to cleave PAR2. These results show that S. pneumoniae is able to cleave and exploit PAR2 to disseminate systemically from the airways.
Collapse
Affiliation(s)
- Florry E van den Boogaard
- Center for Experimental and Molecular Medicine (CEMM), The Netherlands.,Center for Infection and Immunity Amsterdam, The Netherlands
| | - Xanthe Brands
- Center for Experimental and Molecular Medicine (CEMM), The Netherlands.,Center for Infection and Immunity Amsterdam, The Netherlands
| | - JanWillem Duitman
- Center for Experimental and Molecular Medicine (CEMM), The Netherlands.,Center for Infection and Immunity Amsterdam, The Netherlands
| | - Sacha F de Stoppelaar
- Center for Experimental and Molecular Medicine (CEMM), The Netherlands.,Center for Infection and Immunity Amsterdam, The Netherlands
| | - Keren S Borensztajn
- Center for Experimental and Molecular Medicine (CEMM), The Netherlands.,Inserm U700, Université Paris Diderot, France.,LabEx Inflamex, PRES Sorbonne Paris Cité, France.,Assistance Publique Hôpitaux de Paris, DHU FIRE, Service de Pneumologie A, Hôpital Bichat, France
| | | | - Morley D Hollenberg
- Department of Physiology and Pharmacology, University of Calgary, Faculty of Medicine, Canada
| | - C Arnold Spek
- Center for Experimental and Molecular Medicine (CEMM), The Netherlands.,Center for Infection and Immunity Amsterdam, The Netherlands
| | - Marcus J Schultz
- Laboratory of Experimental Intensive Care and Anesthesiology, The Netherlands.,Department of Intensive Care Medicine, The Netherlands
| | - Cornelis van 't Veer
- Center for Experimental and Molecular Medicine (CEMM), The Netherlands.,Center for Infection and Immunity Amsterdam, The Netherlands
| | - Tom van der Poll
- Center for Experimental and Molecular Medicine (CEMM), The Netherlands.,Center for Infection and Immunity Amsterdam, The Netherlands.,Division of Infectious Diseases, Academic Medical Center, University of Amsterdam, The Netherlands
| |
Collapse
|
11
|
Rothmeier AS, Versteeg HH, Ruf W. Factor VIIa-induced interaction with integrin controls the release of tissue factor on extracellular vesicles from endothelial cells. J Thromb Haemost 2019; 17:627-634. [PMID: 30740873 DOI: 10.1111/jth.14406] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 01/22/2019] [Accepted: 01/30/2019] [Indexed: 12/16/2022]
Abstract
Essentials Prothrombotic extracellular vesicles (EV) carry agonist pathway-specific proteomes Agonists for protease activated receptor (PAR) 2 signaling have distinct effects on EV composition PAR2 signaling rapidly generates prothrombotic EV and slowly EV with inactive tissue factor (TF) FVIIa integrin ligation restricts TF incorporation into EV from endothelial cells SUMMARY: Background Cell injury signal-induced activation and release of tissue factor (TF) on extracellular vesicles (EVs) from immune and vessel wall cells propagate local and systemic coagulation initiation. TF trafficking and release on EVs occurs in concert with the release of cell adhesion receptors, including integrin β1 heterodimers, which control trafficking of the TF-activated factor VII (FVIIa) complex. Activation of the TF signaling partner, protease-activated receptor (PAR) 2, also triggers TF release on integrin β1+ EVs from endothelial cells, but the physiological signals for PAR2-dependent EV generation at the vascular interface remain unknown. Objective To define relevant protease ligands of TF contributing to PAR2-dependent release on EVs from endothelial cells. Methods In endothelial cells with balanced expression of TF and PAR2, we evaluated TF release on EVs by using a combination of activity and antigen assays, immunocapture, and confocal imaging. Results and Conclusions PAR2 stimulation generated time-dependent release of distinct TF+ EVs with high coagulant activity (early) and high antigen levels (late). Whereas PAR2 agonist peptide and a stabilized TF-FVIIa-activated FX complex triggered TF+ EV release, stimulation with FVIIa alone promoted cellular retention of TF, despite comparable PAR2 activation. On endothelial cells, FVIIa uniquely induced formation of a complex of TF with integrin α5 β1 . Internalization of TF by FVIIa or anti-TF and activating antibodies against integrin β1 prevented PAR2 agonist-induced release of TF on EVs. These data demonstrate that intracellular trafficking controlled by FVIIa forcing interaction with integrin β1 regulates TF availability for release on procoagulant EVs.
Collapse
Affiliation(s)
- Andrea S Rothmeier
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Henri H Versteeg
- Einthoven Laboratory for Experimental Vascular Medicine, Department of Thrombosis and Hemostasis, Leiden, the Netherlands
| | - Wolfram Ruf
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
- Center for Thrombosis and Hemostasis, Johannes Gutenberg University Medical Center, Mainz, Germany
| |
Collapse
|
12
|
Sutherland MR, Simon AY, Shanina I, Horwitz MS, Ruf W, Pryzdial ELG. Virus envelope tissue factor promotes infection in mice. J Thromb Haemost 2019; 17:482-491. [PMID: 30659719 PMCID: PMC6397068 DOI: 10.1111/jth.14389] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Indexed: 01/04/2023]
Abstract
Essentials The coagulation initiator, tissue factor (TF), is on the herpes simplex virus 1 (HSV1) surface. HSV1 surface TF was examined in mice as an antiviral target since it enhances infection in vitro. HSV1 surface TF facilitated infection of all organs evaluated and anticoagulants were antiviral. Protease activated receptor 2 inhibited infection in vivo and its pre-activation was antiviral. SUMMARY: Background Tissue factor (TF) is the essential cell surface initiator of coagulation, and mediates cell signaling through protease-activated receptor (PAR) 2. Having a diverse cellular distribution, TF is involved in many biological pathways and pathologies. Our earlier work identified host cell-derived TF on the envelope covering several viruses, and showed its involvement in enhanced cell infection in vitro. Objective In the current study, we evaluated the in vivo effects of virus surface TF on infection and on the related modulator of infection PAR2. Methods With the use of herpes simplex virus type 1 (HSV1) as a model enveloped virus, purified HSV1 was generated with or without envelope TF through propagation in a TF-inducible cell line. Infection was studied after intravenous inoculation of BALB/c, C57BL/6J or C57BL/6J PAR2 knockout mice with 5 × 105 plaque-forming units of HSV1, mimicking viremia. Three days after inoculation, organs were processed, and virus was quantified with plaque-forming assays and quantitative real-time PCR. Results Infection of brain, lung, heart, spinal cord and liver by HSV1 required viral TF. Demonstrating promise as a therapeutic target, virus-specific anti-TF mAbs or small-molecule inhibitors of coagulation inhibited infection. PAR2 modulates HSV1 in vivo as demonstrated with PAR2 knockout mice and PAR2 agonist peptide. Conclusion TF is a constituent of many permissive host cell types. Therefore, the results presented here may explain why many viruses are correlated with hemostatic abnormalities, and indicate that TF is a novel pan-specific envelope antiviral target.
Collapse
MESH Headings
- Animals
- Anticoagulants/pharmacology
- Antiviral Agents/pharmacology
- Disease Models, Animal
- Female
- Herpes Simplex/blood
- Herpes Simplex/drug therapy
- Herpes Simplex/immunology
- Herpes Simplex/virology
- Herpesvirus 1, Human/drug effects
- Herpesvirus 1, Human/immunology
- Herpesvirus 1, Human/metabolism
- Host-Pathogen Interactions
- Injections, Intravenous
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Knockout
- Receptor, PAR-2/genetics
- Receptor, PAR-2/metabolism
- Th1 Cells/immunology
- Th1 Cells/virology
- Thromboplastin/administration & dosage
- Thromboplastin/metabolism
- Viral Envelope Proteins/administration & dosage
- Viral Envelope Proteins/metabolism
Collapse
Affiliation(s)
- Michael R Sutherland
- Canadian Blood Services, Center for Innovation, Vancouver, Canada
- Centre for Blood Research and Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada
| | - Ayo Y Simon
- Canadian Blood Services, Center for Innovation, Vancouver, Canada
- Centre for Blood Research and Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada
- African Centre of Excellence on Neglected Tropical Diseases and Forensic Biotechnology and Veterinary Teaching Hospital, Ahmadu Bello University, Zaria, Nigeria
- Preclinical Research and Development, Emergent BioSolutions, Winnipeg, Manitoba, Canada
| | - Iryna Shanina
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, Canada
| | - Marc S Horwitz
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, Canada
| | - Wolfram Ruf
- Immunology and Microbial Sciences, The Scripps Research Institute, La Jolla, CA, USA
- Center for Thrombosis and Hemostasis, University Medical Center, Mainz, Germany
| | - Edward L G Pryzdial
- Canadian Blood Services, Center for Innovation, Vancouver, Canada
- Centre for Blood Research and Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada
| |
Collapse
|
13
|
Zelaya H, Rothmeier AS, Ruf W. Tissue factor at the crossroad of coagulation and cell signaling. J Thromb Haemost 2018; 16:1941-1952. [PMID: 30030891 DOI: 10.1111/jth.14246] [Citation(s) in RCA: 97] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Indexed: 12/16/2022]
Abstract
The tissue factor (TF) pathway plays a central role in hemostasis and thrombo-inflammatory diseases. Although structure-function relationships of the TF initiation complex are elucidated, new facets of the dynamic regulation of TF's activities in cells continue to emerge. Cellular pathways that render TF non-coagulant participate in signaling of distinct TF complexes with associated proteases through the protease-activated receptor (PAR) family of G protein-coupled receptors. Additional co-receptors, including the endothelial protein C receptor (EPCR) and integrins, confer signaling specificity by directing subcellular localization and trafficking. We here review how TF is switched between its role in coagulation and cell signaling through thiol-disulfide exchange reactions in the context of physiologically relevant lipid microdomains. Inflammatory mediators, including reactive oxygen species, activators of the inflammasome, and the complement cascade play pivotal roles in TF procoagulant activation on monocytes, macrophages and endothelial cells. We furthermore discuss how TF, intracellular ligands, co-receptors and associated proteases are integrated in PAR-dependent cell signaling pathways controlling innate immunity, cancer and metabolic inflammation. Knowledge of the precise interactions of TF in coagulation and cell signaling is important for understanding effects of new anticoagulants beyond thrombosis and identification of new applications of these drugs for potential additional therapeutic benefits.
Collapse
Affiliation(s)
- H Zelaya
- Center for Thrombosis and Hemostasis, Johannes Gutenberg University Medical Center, Mainz, Germany
- National Scientific and Technical Research Council (CONICET) and National University of Tucumán, Tucumán, Argentina
| | - A S Rothmeier
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
| | - W Ruf
- Center for Thrombosis and Hemostasis, Johannes Gutenberg University Medical Center, Mainz, Germany
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
- German Center for Cardiovascular Research (DZHK), Partnersite Rhein-Main, Mainz, Germany
| |
Collapse
|
14
|
|
15
|
Selective factor VIII activation by the tissue factor-factor VIIa-factor Xa complex. Blood 2017; 130:1661-1670. [PMID: 28729433 DOI: 10.1182/blood-2017-02-767079] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 07/06/2017] [Indexed: 12/23/2022] Open
Abstract
Safe and effective antithrombotic therapy requires understanding of mechanisms that contribute to pathological thrombosis but have a lesser impact on hemostasis. We found that the extrinsic tissue factor (TF) coagulation initiation complex can selectively activate the antihemophilic cofactor, FVIII, triggering the hemostatic intrinsic coagulation pathway independently of thrombin feedback loops. In a mouse model with a relatively mild thrombogenic lesion, TF-dependent FVIII activation sets the threshold for thrombus formation through contact phase-generated FIXa. In vitro, FXa stably associated with TF-FVIIa activates FVIII, but not FV. Moreover, nascent FXa product of TF-FVIIa can transiently escape the slow kinetics of Kunitz-type inhibition by TF pathway inhibitor and preferentially activates FVIII over FV. Thus, TF synergistically primes FIXa-dependent thrombin generation independently of cofactor activation by thrombin. Accordingly, FVIIa mutants deficient in direct TF-dependent thrombin generation, but preserving FVIIIa generation by nascent FXa, can support intrinsic pathway coagulation. In ex vivo flowing blood, a TF-FVIIa mutant complex with impaired free FXa generation but activating both FVIII and FIX supports efficient FVIII-dependent thrombus formation. Thus, a previously unrecognized TF-initiated pathway directly yielding FVIIIa-FIXa intrinsic tenase complex may be prohemostatic before further coagulation amplification by thrombin-dependent feedback loops enhances the risk of thrombosis.
Collapse
|
16
|
Abstract
Cancer-associated thrombosis remains a significant complication in the clinical management of cancer and interactions of the hemostatic system with cancer biology continue to be elucidated. Here, we review recent progress in our understanding of tissue factor (TF) regulation and procoagulant activation, TF signaling in cancer and immune cells, and the expanding roles of the coagulation system in stem cell niches and the tumor microenvironment. The extravascular functions of coagulant and anti-coagulant pathways have significant implications not only for tumor progression, but also for the selection of appropriate target specific anticoagulants in the therapy of cancer patients.
Collapse
Affiliation(s)
- Wolfram Ruf
- Center for Thrombosis and Hemostasis, University Medical Center, Mainz, Germany; Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA, USA.
| | - Andrea S Rothmeier
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA, USA
| | - Claudine Graf
- Center for Thrombosis and Hemostasis, University Medical Center, Mainz, Germany; 3(rd) Medical Department, University Medical Center, Mainz, Germany
| |
Collapse
|
17
|
Bode MF, Mackman N. Protective and pathological roles of tissue factor in the heart. Hamostaseologie 2014; 35:37-46. [PMID: 25434707 DOI: 10.5482/hamo-14-09-0042] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Accepted: 11/19/2014] [Indexed: 02/06/2023] Open
Abstract
UNLABELLED Tissue factor (TF) is expressed in the heart where it is required for haemostasis. High levels of TF are also expressed in atherosclerotic plaques and likely contribute to atherothrombosis after plaque rupture. Indeed, risk factors for atherothrombosis, such as diabetes, hypercholesterolaemia, smoking and hypertension, are associated with increased TF expression in circulating monocytes, microparticles and plasma. Several therapies that reduce atherothrombosis, such as statins, ACE inhibitors, beta-blockers and anti-platelet drugs, are associated with reduced TF expression. In addition to its haemostatic and pro-thrombotic functions, the TF : FVIIa complex and downstream coagulation proteases activate cells by cleavage of protease-activated receptors (PARs). In mice, deficiencies in either PAR-1 or PAR-2 reduce cardiac remodelling and heart failure after ischaemia-reperfusion injury. This suggests that inhibition of coagulation proteases and PARs may be protective in heart attack patients. In contrast, the TF/thrombin/PAR-1 pathway is beneficial in a mouse model of Coxsackievirus B3-induced viral myocarditis. We found that stimulation of PAR-1 increases the innate immune response by enhancing TLR3-dependent IFN-β expression. Therefore, inhibition of the TF/thrombin/PAR-1 pathway in patients with viral myocarditis could have detrimental effects. CONCLUSION The TF : FVIIa complex has both protective and pathological roles in the heart.
Collapse
Affiliation(s)
| | - N Mackman
- Nigel Mackman, Ph.D., FAHA, University of North Carolina at Chapel Hill, Division of Hematology and Oncology, Department of Medicine, McAllister Heart Institute, 111 Mason Farm Road, 2312B Medical Biomolecular Research Bldg., CB #7126, Chapel Hill, NC 27599, USA, E-mail:
| |
Collapse
|
18
|
Eriksson O, Ramström M, Hörnaeus K, Bergquist J, Mokhtari D, Siegbahn A. The Eph tyrosine kinase receptors EphB2 and EphA2 are novel proteolytic substrates of tissue factor/coagulation factor VIIa. J Biol Chem 2014; 289:32379-91. [PMID: 25281742 DOI: 10.1074/jbc.m114.599332] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Tissue factor (TF) binds the serine protease factor VIIa (FVIIa) to form a proteolytically active complex that can trigger coagulation or activate cell signaling. Here we addressed the involvement of tyrosine kinase receptors (RTKs) in TF/FVIIa signaling by antibody array analysis and subsequently found that EphB2 and EphA2 of the Eph RTK family were cleaved in their ectodomains by TF/FVIIa. We used N-terminal Edman sequencing and LC-MS/MS analysis to characterize the cleaved Eph isoforms and identified a key arginine residue at the cleavage site, in agreement with the tryptic serine protease activity of FVIIa. Protease-activated receptor 2 (PAR2) signaling and downstream coagulation activity was non-essential in this context, in further support of a direct cleavage by TF/FVIIa. EphB2 was cleaved by FVIIa concentrations in the subnanomolar range in a number of TF expressing cell types, indicating that the active cellular pool of TF was involved. FVIIa caused potentiation of cell repulsion by the EphB2 ligand ephrin-B1, demonstrating a novel proteolytical event to control Eph-mediated cell segregation. These results define Eph RTKs as novel proteolytical targets of TF/FVIIa and provide new insights into how TF/FVIIa regulates cellular functions independently of PAR2.
Collapse
Affiliation(s)
- Oskar Eriksson
- From the Department of Medical Sciences, Clinical Chemistry, Uppsala University Hospital, SE-751 85 Uppsala, Science for Life Laboratory, Uppsala University, Sweden
| | - Margareta Ramström
- the Department of Chemistry, Analytical Chemistry, BMC, SE-751 24 Uppsala, and Science for Life Laboratory, Uppsala University, Sweden
| | - Katarina Hörnaeus
- the Department of Chemistry, Analytical Chemistry, BMC, SE-751 24 Uppsala, and Science for Life Laboratory, Uppsala University, Sweden
| | - Jonas Bergquist
- the Department of Chemistry, Analytical Chemistry, BMC, SE-751 24 Uppsala, and Science for Life Laboratory, Uppsala University, Sweden
| | - Dariush Mokhtari
- From the Department of Medical Sciences, Clinical Chemistry, Uppsala University Hospital, SE-751 85 Uppsala, Science for Life Laboratory, Uppsala University, Sweden
| | - Agneta Siegbahn
- From the Department of Medical Sciences, Clinical Chemistry, Uppsala University Hospital, SE-751 85 Uppsala, Science for Life Laboratory, Uppsala University, Sweden
| |
Collapse
|
19
|
Lange S, Gonzalez I, Pinto MP, Arce M, Valenzuela R, Aranda E, Elliot M, Alvarez M, Henriquez S, Velasquez EV, Orge F, Oliva B, Gonzalez P, Villalon M, Cautivo KM, Kalergis AM, Pereira K, Mendoza C, Saez C, Kato S, Cuello MA, Parborell F, Irusta G, Palma V, Allende ML, Owen GI. Independent Anti-Angiogenic Capacities of Coagulation Factors X and Xa. J Cell Physiol 2014; 229:1673-80. [DOI: 10.1002/jcp.24612] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Accepted: 03/06/2014] [Indexed: 12/15/2022]
Affiliation(s)
- Soledad Lange
- Departamento de Fisiología, Facultad de Ciencias Biológicas; Pontificia Universidad Católica de Chile; Santiago Chile
| | - Ibeth Gonzalez
- Departamento de Fisiología, Facultad de Ciencias Biológicas; Pontificia Universidad Católica de Chile; Santiago Chile
| | - Mauricio P. Pinto
- Departamento de Fisiología, Facultad de Ciencias Biológicas; Pontificia Universidad Católica de Chile; Santiago Chile
| | - Maximiliano Arce
- Departamento de Fisiología, Facultad de Ciencias Biológicas; Pontificia Universidad Católica de Chile; Santiago Chile
| | - Rodrigo Valenzuela
- Departamento de Fisiología, Facultad de Ciencias Biológicas; Pontificia Universidad Católica de Chile; Santiago Chile
| | - Evelyn Aranda
- Departamento de Fisiología, Facultad de Ciencias Biológicas; Pontificia Universidad Católica de Chile; Santiago Chile
| | - Matias Elliot
- Departamento de Biología, Facultad de Ciencias; Universidad de Chile; Santiago Chile
- FONDAP Center for Genome Regulation, Facultad de Ciencias; Universidad de Chile; Santiago Chile
| | - Marjorie Alvarez
- Departamento de Biología, Facultad de Ciencias; Universidad de Chile; Santiago Chile
- FONDAP Center for Genome Regulation, Facultad de Ciencias; Universidad de Chile; Santiago Chile
| | - Soledad Henriquez
- Departamento de Fisiología, Facultad de Ciencias Biológicas; Pontificia Universidad Católica de Chile; Santiago Chile
| | - Ethel V. Velasquez
- Departamento de Fisiología, Facultad de Ciencias Biológicas; Pontificia Universidad Católica de Chile; Santiago Chile
| | - Felipe Orge
- Departamento de Fisiología, Facultad de Ciencias Biológicas; Pontificia Universidad Católica de Chile; Santiago Chile
| | - Barbara Oliva
- Departamento de Fisiología, Facultad de Ciencias Biológicas; Pontificia Universidad Católica de Chile; Santiago Chile
| | - Pamela Gonzalez
- Departamento de Fisiología, Facultad de Ciencias Biológicas; Pontificia Universidad Católica de Chile; Santiago Chile
| | - Manuel Villalon
- Departamento de Fisiología, Facultad de Ciencias Biológicas; Pontificia Universidad Católica de Chile; Santiago Chile
| | - Kelly M. Cautivo
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología; Pontificia Universidad Católica de Chile; Santiago Chile
| | - Alexis M. Kalergis
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología; Pontificia Universidad Católica de Chile; Santiago Chile
- Departamento de Reumatología, Facultad de Medicina; Pontificia Universidad Católica de Chile; Santiago Chile
- Biomedical Research Consortium Chile (BMRC); Santiago Chile
| | - Karla Pereira
- Departamento de Hematología-Oncología, Facultad de Medicina; Pontificia Universidad Católica de Chile; Santiago Chile
| | - Camila Mendoza
- Departamento de Hematología-Oncología, Facultad de Medicina; Pontificia Universidad Católica de Chile; Santiago Chile
| | - Claudia Saez
- Departamento de Hematología-Oncología, Facultad de Medicina; Pontificia Universidad Católica de Chile; Santiago Chile
| | - Sumie Kato
- Departamento de Obstetricia y Ginecología, Facultad de Medicina; Pontificia Universidad Católica de Chile; Santiago Chile
| | - Mauricio A. Cuello
- Departamento de Obstetricia y Ginecología, Facultad de Medicina; Pontificia Universidad Católica de Chile; Santiago Chile
| | - Fernanda Parborell
- Instituto de Biología y Medicina Experimental (IByME-CONICET); Buenos Aires Argentina
| | - Griselda Irusta
- Instituto de Biología y Medicina Experimental (IByME-CONICET); Buenos Aires Argentina
| | - Veronica Palma
- Departamento de Biología, Facultad de Ciencias; Universidad de Chile; Santiago Chile
- FONDAP Center for Genome Regulation, Facultad de Ciencias; Universidad de Chile; Santiago Chile
| | - Miguel L. Allende
- Departamento de Biología, Facultad de Ciencias; Universidad de Chile; Santiago Chile
- FONDAP Center for Genome Regulation, Facultad de Ciencias; Universidad de Chile; Santiago Chile
| | - Gareth I. Owen
- Departamento de Fisiología, Facultad de Ciencias Biológicas; Pontificia Universidad Católica de Chile; Santiago Chile
- Biomedical Research Consortium Chile (BMRC); Santiago Chile
- Centro UC Investigación en Oncología; Pontificia Universidad Católica de Chile; Santiago Chile
- FONDAP Advanced Center forChronicDiseases (ACCDis); Pontificia Universidad Católica de Chile; Santiago Chile
| |
Collapse
|
20
|
Henry BL, Desai UR. Discovery methodology for the development of direct factor VIIa inhibitors. Expert Opin Drug Discov 2014; 9:859-72. [PMID: 24882057 DOI: 10.1517/17460441.2014.923398] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Heparin and warfarin have historically been the only antithrombotics available. Recently, however, newer anticoagulants have been developed. Factor VIIa (fVIIa) inhibitors represent one of the new and potentially exciting classes of anticoagulants currently under development. Indeed, several methodologies have been used to develop fVIIa inhibitors. AREAS COVERED The authors highlight some of the methologies applied for the discovery of fVIIa inhibitors including phage display, isolation of endogenous peptides from hematophagous animals and the use of the 1,5-benzothiazepine molecular scaffolds and screens of large chemical libraries previously used to identify other serine protease inhibitors. Although these screens were intended to identify thrombin and factor Xa inhibitors, the compounds often had concomitant fVIIa activity. The authors also discuss the utilization of medical chemistry techniques for the discovery of these compounds. EXPERT OPINION FVIIa inhibitors represent a viable option for the development of new anticoagulants. There are theoretical advantages that fVIIa inhibitors may possess over existing anticoagulants and highly specific inhibitors that possess oral bioavailability and low bleeding risk may succeed.
Collapse
Affiliation(s)
- Brian L Henry
- University of Pittsburgh Medical Center, Heart and Vascular Institute Pittsburgh, Department of Cardiology , Scaife Hall, Suite B-571.3, 200 Lothrop Street, Pittsburgh, PA 15213 , USA +1 412 647 3429 ; +1 412 647 0481 ;
| | | |
Collapse
|
21
|
Åberg M, Siegbahn A. Tissue factor non-coagulant signaling - molecular mechanisms and biological consequences with a focus on cell migration and apoptosis. J Thromb Haemost 2013; 11:817-25. [PMID: 23384027 DOI: 10.1111/jth.12156] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Tissue factor (TF), a transmembrane glycoprotein, is the main initiator of the blood coagulation cascade. TF is also recognized as a true signaling receptor. There is accumulating evidence that the downstream signaling effects of the TF complexes are transduced by several mechanisms, including: activation of protease-activated receptor (PAR)-1 and PAR-2, and the PAR-dependent pathways, via the TF cytoplasmic domain and by transactivation of receptor tyrosine kinases. Triggering of signaling cascades such as the mitogen-activated protein kinase and phosphoinositide 3-kinase/AKT pathways couples TF to a multitude of functions within the cell, such as proliferation, cell migration, and survival. Thus, TF has a Janus face; on the one hand, it has vital life-maintaining functions, and on the other it has harmful effects, exemplified by inflammation, the acute coronary syndromes, and cancer. TF mediates a broad spectrum of signaling mechanisms. Learning more about these different mechanisms/pathways will lead to new treatment strategies, which can ultimately be personalized.
Collapse
Affiliation(s)
- M Åberg
- Department of Medical Sciences, Clinical Chemistry, Uppsala University, Uppsala, Sweden.
| | | |
Collapse
|
22
|
Carneiro-Lobo TC, Schaffner F, Disse J, Ostergaard H, Francischetti IMB, Monteiro RQ, Ruf W. The tick-derived inhibitor Ixolaris prevents tissue factor signaling on tumor cells. J Thromb Haemost 2012; 10:1849-58. [PMID: 22823596 PMCID: PMC3433625 DOI: 10.1111/j.1538-7836.2012.04864.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND Tissue factor (TF) is frequently overexpressed in cancer cells and correlated with more aggressive tumor phenotypes and poor prognosis. In addition to promoting coagulation-dependent metastasis and cancer-associated thrombosis, tumor cell-expressed TF mediates direct cell signaling involving the protease-activated receptor (PAR) 2. Ixolaris is a tick-derived inhibitor of the TF-factor (F)VIIa-Xa coagulation initiation complex which blocks primary tumor growth and angiogenesis in glioblastoma and melanoma models. METHODS In this study we address the anti-tumor effects of Ixolaris in TF-VIIa-PAR2 signaling-dependent breast cancer models, a xenograft model of highly aggressive human MDA-MB-231 mfp cells and a syngeneic model of PAR2-deficient and replete PyMT mouse mammary carcinoma cells. RESULTS Ixolaris potently inhibited the procoagulant activity of human MDA-MB-231mfp or murine PyMT breast cancer cells. Ixolaris blocked signaling by the ternary TF-FVIIa-FXa complex, and, surprisingly, at higher concentrations also the binary TF-FVIIa complex on MDA-MB-231 cells. We show that Ixolaris interacts with certain residues in the human VIIa protease domain that are involved in PAR2 cleavage. In contrast to human VIIa, Ixolaris was a poor inhibitor of murine TF-FVIIa signaling and did not attenuate PAR2-dependent tumor growth in a syngeneic mouse model of breast cancer progression. CONCLUSION These data show that Ixolaris inhibits PAR2 cleavage specifically by human TF signaling complexes and suggest that Ixolaris may block tumor growth of human cell models with ectopic FVIIa expression through inhibition of direct TF-FVIIa-PAR2 signaling as well as its anticoagulant activity.
Collapse
Affiliation(s)
- T C Carneiro-Lobo
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla CA, USA
| | | | | | | | | | | | | |
Collapse
|
23
|
Weitz JI, Eikelboom JW, Samama MM. New antithrombotic drugs: Antithrombotic Therapy and Prevention of Thrombosis, 9th ed: American College of Chest Physicians Evidence-Based Clinical Practice Guidelines. Chest 2012; 141:e120S-e151S. [PMID: 22315258 DOI: 10.1378/chest.11-2294] [Citation(s) in RCA: 161] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
This article focuses on new antithrombotic drugs that are in or are entering phase 3 clinical testing. Development of these new agents was prompted by the limitations of existing antiplatelet, anticoagulant, or fibrinolytic drugs. Addressing these unmet needs, this article (1) outlines the rationale for development of new antithrombotic agents; (2) describes the new antiplatelet, anticoagulant, and fibrinolytic drugs; and (3) provides clinical perspectives on the opportunities and challenges faced by these novel agents.
Collapse
Affiliation(s)
- Jeffrey I Weitz
- Thrombosis and Atherosclerosis Research Institute and Department of Medicine, McMaster University, Hamilton, ON, Canada; Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada.
| | - John W Eikelboom
- Thrombosis and Atherosclerosis Research Institute and Department of Medicine, McMaster University, Hamilton, ON, Canada
| | | |
Collapse
|
24
|
Tissue factor/factor VIIa pathway mediates coagulation activation in induced-heat stroke in the baboon. Crit Care Med 2012; 40:1229-36. [DOI: 10.1097/ccm.0b013e3182387bef] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
25
|
Gowen BB, Bray M. Progress in the experimental therapy of severe arenaviral infections. Future Microbiol 2012; 6:1429-41. [PMID: 22122440 DOI: 10.2217/fmb.11.132] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
A number of viruses in the family Arenaviridae cause severe illness in humans. Lassa virus in West Africa and a number of agents in South America produce hemorrhagic fever in persons exposed to aerosolized excretions of the pathogens' rodent hosts. Because arenaviruses are not transmitted by arthropods, and person-to-person spread is rare, human infections occur singly and sporadically, and are usually not diagnosed until the patient is severely ill. Because the arenaviruses are naturally transmitted by the airborne route, they also pose a potential threat as aerosolized bioterror weapons. The broad-spectrum antiviral drug ribavirin was shown to reduce mortality from Lassa fever, and has been tested against Argentine hemorrhagic fever, but it is not an approved treatment for either disease. Human immune convalescent plasma was proven to be effective for Argentine hemorrhagic fever in a controlled trial. New treatments are needed to block viral replication without causing toxicity and to prevent the increased vascular permeability that is responsible for hypotension and shock. In this paper, we review current developments in the experimental therapy of severe arenaviral infections, focusing on drugs that have been tested in animal models, and provide a perspective on future research.
Collapse
Affiliation(s)
- Brian B Gowen
- Institute for Antiviral Research & Department of Animal, Dairy & Veterinary Sciences, Utah State University, Logan, UT, USA.
| | | |
Collapse
|
26
|
Tissue factor and glycoprotein C on herpes simplex virus type 1 are protease-activated receptor 2 cofactors that enhance infection. Blood 2012; 119:3638-45. [PMID: 22374699 DOI: 10.1182/blood-2011-08-376814] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The coagulation system provides physiologic host defense, but it can also be exploited by pathogens for infection. On the HSV1 surface, host-cell-derived tissue factor (TF) and virus-encoded glycoprotein C (gC) can stimulate protease activated receptor 1 (PAR1)-enhanced infection by triggering thrombin production. Using novel engineered HSV1 variants deficient in either TF and/or gC, in the present study, we show that activated coagulation factors X (FXa) or VII (FVIIa) directly affect HSV1 infection of human umbilical vein endothelial cells in a manner that is dependent on viral TF and gC. The combination of FXa and FVIIa maximally enhanced infection for TF(+)/gC(+) HSV1 and receptor desensitization and Ab inhibition demonstrated that both proteases act on PAR2. Inhibitory TF Abs showed that the required TF source was viral. Individually, TF or gC partly enhanced the effect of FXa, but not FVIIa, revealing gC as a novel PAR2 cofactor for FVIIa. In sharp contrast, thrombin enhanced infection via PAR1 independently of viral TF and gC. Thrombin combined with FXa/FVIIa enhanced infection, suggesting that PAR1 and PAR2 are independently involved in virus propagation. These results show that HSV1 surface cofactors promote cellular PAR2-mediated infection, indicating a novel mode by which pathogens exploit the initiation phase of the host hemostatic system.
Collapse
|
27
|
Generation of bioactive recombinant Ancylostoma caninum anticoagulant peptide c2. Protein Expr Purif 2012; 81:49-54. [DOI: 10.1016/j.pep.2011.08.029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2011] [Revised: 08/23/2011] [Accepted: 08/24/2011] [Indexed: 11/24/2022]
|
28
|
|
29
|
Disse J, Petersen HH, Larsen KS, Persson E, Esmon N, Esmon CT, Teyton L, Petersen LC, Ruf W. The endothelial protein C receptor supports tissue factor ternary coagulation initiation complex signaling through protease-activated receptors. J Biol Chem 2010; 286:5756-67. [PMID: 21149441 DOI: 10.1074/jbc.m110.201228] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Protease-activated receptor (PAR) signaling is closely linked to the cellular activation of the pro- and anticoagulant pathways. The endothelial protein C receptor (EPCR) is crucial for signaling by activated protein C through PAR1, but EPCR may have additional roles by interacting with the 4-carboxyglutamic acid domains of procoagulant coagulation factors VII (FVII) and X (FX). Here we show that soluble EPCR regulates the interaction of FX with human or mouse tissue factor (TF)-FVIIa complexes. Mutagenesis of the FVIIa 4-carboxyglutamic acid domain and dose titrations with FX showed that EPCR interacted primarily with FX to attenuate FX activation in lipid-free assay systems. In human cell models of TF signaling, antibody inhibition of EPCR selectively blocked PAR activation by the ternary TF-FVIIa-FXa complex but not by the non-coagulant TF-FVIIa binary complex. Heterologous expression of EPCR promoted PAR1 and PAR2 cleavage by FXa in the ternary complex but did not alter PAR2 cleavage by TF-FVIIa. In murine smooth muscle cells that constitutively express EPCR and TF, thrombin and FVIIa/FX but not FVIIa alone induced PAR1-dependent signaling. Although thrombin signaling was unchanged, cells with genetically reduced levels of EPCR no longer showed a signaling response to the ternary complex. These results demonstrate that EPCR interacts with the ternary TF coagulation initiation complex to enable PAR signaling and suggest that EPCR may play a role in regulating the biology of TF-expressing extravascular and vessel wall cells that are exposed to limited concentrations of FVIIa and FX provided by ectopic synthesis or vascular leakage.
Collapse
Affiliation(s)
- Jennifer Disse
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, California 92037, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Li D, He Q, Kang T, Yin H, Jin X, Li H, Gan W, Yang C, Hu J, Wu Y, Peng L. Identification of an anticoagulant peptide that inhibits both fXIa and fVIIa/tissue factor from the blood-feeding nematode Ancylostoma caninum. Biochem Biophys Res Commun 2010; 392:155-9. [PMID: 20059979 DOI: 10.1016/j.bbrc.2009.12.177] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2009] [Accepted: 12/25/2009] [Indexed: 10/20/2022]
Abstract
Factor VIIa-tissue factor complex (fVIIa/TF) and factor XIa (fXIa) play important roles in the initiation and amplification of coagulation, respectively. They may be good targets for the development of novel anticoagulants to treat and prevent thromboembolic disease. In this study, we cloned, expressed and identified a novel anticoagulant peptide, AcaNAP10, from the blood-feeding nematode Ancylostoma caninum. AcaNAP10 showed potent anticoagulant activity and doubled the activated partial thromboplastin and prothrombin times at estimated concentrations of 92.9 nM and 28.8 nM, respectively. AcaNAP10 demonstrated distinct mechanisms of action compared with known anticoagulants. It inhibited fXIa and fVIIa/TF with IC(50) values of 25.76+/-1.06 nM and 123.9+/-1.71 nM, respectively. This is the first report on an anticoagulant that can inhibit both fXIa and fVIIa/TF. This anticoagulant peptide may be an alternative molecule for the development of novel anticoagulants.
Collapse
Affiliation(s)
- Deng Li
- Department of Parasitology, Guangdong Medical College, Zhanjiang 524023, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Holy EW, Tanner FC. Tissue factor in cardiovascular disease pathophysiology and pharmacological intervention. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2010; 59:259-92. [PMID: 20933205 DOI: 10.1016/s1054-3589(10)59009-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Tissue factor (TF) is the major trigger of the coagulation cascade and thereby crucially involved in the maintenance of vascular hemostasis. By binding factor VIIa, the resulting TF:VIIa complex activates the coagulation factors IX and X ultimately leading to fibrin and clot formation. In the vessel wall, TF expression and activity is detectable in vascular smooth muscle cells and fibroblasts and, at a much lower level, in endothelial cells and can be induced by various stimuli including cytokines. In addition, TF is found in the bloodstream in circulating cells such as monocytes, in TF containing microparticles, and as a soluble splicing isoform. Besides its well-known extracellular role as a trigger of coagulation, TF also functions as a transmembrane receptor, and TF-dependent intracellular signaling events regulate the expression of genes involved in cellular responses such as proliferation and migration. TF indeed appears to be involved in the pathogenesis of neointima formation and tumor growth, and increased levels of TF have been detected in patients with cardiovascular risk factors or coronary artery disease as well as in those with cancer. Therefore, pharmacological or genetic inhibition of TF may be an attractive target for the treatment of cardiovascular disease and cancer. Different strategies for inhibition of TF have been developed such as inhibition of TF synthesis and blockade of TF action. Clinical applications of such strategies need to be tested in appropriate trials, in particular for evaluating the advantages of targeted versus systemic delivery of the inhibitors.
Collapse
Affiliation(s)
- Erik W Holy
- Cardiovascular Research, Physiology Institute, University of Zurich, Zurich, Switzerland
| | | |
Collapse
|
32
|
Abstract
TF (tissue factor) is the main trigger of the coagulation cascade; by binding Factor VIIa it activates Factor IX and Factor X, thereby resulting in fibrin formation. Various stimuli, such as cytokines, growth factors and biogenic amines, induce TF expression and activity in vascular cells. Downstream targets of these mediators include diverse signalling molecules such as MAPKs (mitogen-activated protein kinases), PI3K (phosphoinositide 3-kinase) and PKC (protein kinase C). In addition, TF can be detected in the bloodstream, known as circulating or blood-borne TF. Many cardiovascular risk factors, such as hypertension, diabetes, dyslipidaemia and smoking, are associated with increased expression of TF. Furthermore, in patients presenting with acute coronary syndromes, elevated levels of circulating TF are found. Apart from its role in thrombosis, TF has pro-atherogenic properties, as it is involved in neointima formation by inducing vascular smooth muscle cell migration. As inhibition of TF action appears to be an attractive target for the treatment of cardiovascular disease, therapeutic strategies are under investigation to specifically interfere with the action of TF or, alternatively, promote the effects of TFPI (TF pathway inhibitor).
Collapse
|
33
|
Schaffner F, Ruf W. Tissue factor and PAR2 signaling in the tumor microenvironment. Arterioscler Thromb Vasc Biol 2009; 29:1999-2004. [PMID: 19661489 DOI: 10.1161/atvbaha.108.177428] [Citation(s) in RCA: 89] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Diverse oncogenic transformations result in the constitutive expression of tissue factor (TF) in cancer cells. The local and systemic activation of the coagulation cascade has long been a recognized hallmark for aggressive cancer, but genetic mouse models and new experimental therapeutics have only recently demonstrated crucial roles for TF initiated cell signaling in the pathogenesis of cancer. On tumor cells, the TF-VIIa binary complex mediates activation of protease activated receptor (PAR) 2 and thereby shapes the tumor microenvironment by inducing an array of proangiogenic and immune modulating cytokines, chemokines, and growth factors. PAR2 also uniquely triggers tumor cell migration by G protein-independent pathways through beta-arrestin scaffolding. Metastatic tumor cells use additional signaling networks of the coagulation cascade by activating PAR1 through thrombin or the ternary TF-VIIa-Xa signaling complex in the vascular and potentially lymphatic system. Selective antagonists of TF-VIIa-PAR2 signaling may be used as antiangiogenic therapy without increasing the risk of bleeding, whereas coagulation and associated signaling pathways on platelets and other host cells may be targeted for therapeutic benefit in advanced cancer and metastatic disease.
Collapse
Affiliation(s)
- Florence Schaffner
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA 92037, USA
| | | |
Collapse
|
34
|
Zhao J, Aguilar G, Palencia S, Newton E, Abo A. rNAPc2 inhibits colorectal cancer in mice through tissue factor. Clin Cancer Res 2009; 15:208-16. [PMID: 19118048 DOI: 10.1158/1078-0432.ccr-08-0407] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Recombinant nematode anticoagulant protein c2 (rNAPc2) is a specific inhibitor of tissue factor (TF)/factor VIIa complex with novel antithrombotic activity. TF is highly expressed in human colorectal tumors, and levels are positively correlated with disease progression. EXPERIMENTAL DESIGN To explore the therapeutic potential and mechanism of action of rNAPc2 during tumor growth and metastasis, we tested rNAPc2 in several experimental colorectal cancer models in mice. RESULTS Administration of rNAPc2 inhibited pulmonary metastasis in mice systemically disseminated with CT26 murine colon carcinoma cells in a dose-dependent fashion. Combining rNAPc2 with the cytotoxic agent 5-fluorouracil or bevacizumab (humanized anti-vascular endothelial growth factor monoclonal antibody) resulted in additive growth inhibition and simultaneous reduction of microvessel density in HCT116 human colorectal tumor xenografts in nude mice. Furthermore, rNAPc2 potentiated CPT-11 in inhibiting hepatic metastasis in nude mice with portal vein injection of HCT116 tumor cells. Long-term administration of rNAPc2 significantly suppressed spontaneous formation of intestinal tumors in Apc(Min/+) mice. Using a RNA interference approach, we showed that TF expression is necessary for rNAPc2-mediated inhibition of HCT116 human colorectal tumor xenograft growth in nude mice, indicating that the antitumor effect of rNAPc2 may be transduced through TF that is expressed on tumor cells. CONCLUSIONS rNAPc2 is a potent anticancer agent when used in combination with chemotherapy or antiangiogenic therapy in mouse models of colorectal cancer, and TF positivity appears to be required for its activity.
Collapse
Affiliation(s)
- Jingsong Zhao
- Department of Research, Nuvelo, Inc., San Carlos, California 94070, USA.
| | | | | | | | | |
Collapse
|
35
|
Weitz JI, Hirsh J, Samama MM. New antithrombotic drugs: American College of Chest Physicians Evidence-Based Clinical Practice Guidelines (8th Edition). Chest 2008; 133:234S-256S. [PMID: 18574267 DOI: 10.1378/chest.08-0673] [Citation(s) in RCA: 190] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
This chapter focuses on new antithrombotic drugs that are in phase II or III clinical testing. Development of these new agents was prompted by limitations of existing antiplatelet, anticoagulant, or fibrinolytic drugs. Addressing these unmet needs, this chapter (1) outlines the rationale for development of new antithrombotic agents, (2) describes the new antiplatelet, anticoagulant, and fibrinolytic drugs, and (3) provides clinical perspectives on the opportunities and challenges faced by these novel agents.
Collapse
Affiliation(s)
- Jeffrey I Weitz
- From the Henderson Research Center, McMaster University, Hamilton, ON, Canada.
| | - Jack Hirsh
- From the Henderson Research Center, McMaster University, Hamilton, ON, Canada
| | | |
Collapse
|
36
|
Monteiro RQ, Rezaie AR, Bae JS, Calvo E, Andersen JF, Francischetti IMB. Ixolaris binding to factor X reveals a precursor state of factor Xa heparin-binding exosite. Protein Sci 2007; 17:146-53. [PMID: 18042685 DOI: 10.1110/ps.073016308] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Ixolaris is a two-Kunitz tick salivary gland tissue factor pathway inhibitor (TFPI). In contrast to human TFPI, Ixolaris specifically binds to factor Xa (FXa) heparin-binding exosite (HBE). In addition, Ixolaris interacts with zymogen FX. In the present work we characterized the interaction of Ixolaris with human FX quantitatively, and identified a precursor state of the heparin-binding exosite (proexosite, HBPE) as the Ixolaris-binding site on the zymogen. Gel-filtration chromatography demonstrated 1:1 complex formation between fluorescein-labeled Ixolaris and FX. Isothermal titration calorimetry confirmed that the binding of Ixolaris to FX occurs at stoichiometric concentrations in a reaction which is characteristically exothermic, with a favorable enthalpy (DeltaH) of -10.78 kcal/mol. ELISA and plasmon resonance experiments also indicate that Ixolaris binds to plasma FX and FXa, or to recombinant Gla domain-containing FX/FXa with comparable affinities ( approximately 1 nM). Using a series of mutants on the HBPE, we identified the most important amino acids involved in zymogen/Ixolaris interaction-Arg-93 >>> Arg-165 > or = Lys-169 > Lys-236 > Arg-125-which was identical to that observed for FXa/Ixolaris interaction. Remarkably, Ixolaris strongly inhibited FX activation by factor IXa in the presence but not in the absence of factor VIIIa, suggesting a specific interference in the cofactor activity. Further, solid phase assays demonstrated that Ixolaris inhibits FX interaction with immobilized FVIIIa. Altogether, Ixolaris is the first inhibitor characterized to date that specifically binds to FX HBPE. Ixolaris may be a useful tool to study the physiological role of the FX HBPE and to evaluate this domain as a target for anticoagulant drugs.
Collapse
Affiliation(s)
- Robson Q Monteiro
- Instituto de Bioquimica Medica, Centro de Ciencias de Saude, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | | | | | | | | |
Collapse
|
37
|
Fluture A, Giugliano GR, Giugliano RP. Recombinant nematode anticoagulant protein c2 in non-ST segment elevation acute coronary syndrome and beyond. Future Cardiol 2007; 3:365-75. [DOI: 10.2217/14796678.3.4.365] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Patients with acute coronary syndromes (ACS) have high recurrent ischemic event rates despite management with current guideline-based therapies. Recombinant nematode anticoagulant protein (rNAP)c2 provides factor Xa-dependent inhibition of the tissue factor/factor VIIa complex acting proximally on the clotting cascade. It may be administered either intravenously or subcutaneously and has an elimination half-life of approximately 50–60 h. rNAPc2 reduces thrombin formation in patients undergoing elective percutaneous coronary interventions (PCI) and in patients with non-ST segment elevation ACS managed with an early invasive strategy, while bleeding rates are comparable with currently used anticoagulants. Patients receiving rNAPc2 undergoing emergent coronary artery bypass surgery within 96 h of dosing have increased rates of major bleeding. Some heparin coadministration may be necessary to avoid PCI-related thrombotic complications. Large-scale trials are needed to confirm these findings and to evaluate the impact of rNAPc2 on clinical events.
Collapse
Affiliation(s)
- Adrian Fluture
- Baystate Medical Center/Tufts Unicersity School of Medicine, 759 Chestnut Street, Springfield, MA 01199, USA
| | - Gregory R Giugliano
- Associate Director, Baystate Medical Education & Research Foundation, Cardiac Catheterization Laboratory and Cardiology Research, 759 Chestnut Street. BMC # S4659, Springfield, MA 01199, USA
| | - Robert P Giugliano
- TIMI Study Group, 350 Longwood Avenue, 1st Floor Offices, Boston, MA 02115, USA
| |
Collapse
|
38
|
Giugliano RP, Wiviott SD, Stone PH, Simon DI, Schweiger MJ, Bouchard A, Leesar MA, Goulder MA, Deitcher SR, McCabe CH, Braunwald E. Recombinant nematode anticoagulant protein c2 in patients with non-ST-segment elevation acute coronary syndrome: the ANTHEM-TIMI-32 trial. J Am Coll Cardiol 2007; 49:2398-407. [PMID: 17599602 DOI: 10.1016/j.jacc.2007.02.065] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2007] [Revised: 01/31/2007] [Accepted: 02/12/2007] [Indexed: 10/23/2022]
Abstract
OBJECTIVES We sought to evaluate the safety and efficacy of recombinant nematode anticoagulant protein c2 (rNAPc2) in patients with non-ST-segment elevation acute coronary syndrome (nSTE-ACS). BACKGROUND Recombinant NAPc2 is a potent inhibitor of the tissue factor/factor VIIa complex that has the potential to reduce ischemic complications mediated by thrombin generation. METHODS A total of 203 patients were randomized 4:1 to double-blinded intravenous rNAPc2 or placebo every 48 h for a total of 1 to 3 doses in 8 ascending panels (1.5 to 10 microg/kg). All patients received aspirin, unfractionated heparin (UFH), or enoxaparin and early catheterization; clopidogrel and glycoprotein IIb/IIIa blockers were encouraged. Two subsequent open-label panels evaluated 10 mug/kg rNAPc2 with half-dose UFH (n = 26) and no UFH (n = 26). The primary end point was the rate of major plus minor bleeding. Pharmacokinetics, pharmacodynamics, continuous electrocardiography, and clinical events were assessed. RESULTS Recombinant NAPc2 did not significantly increase major plus minor bleeding (3.7% vs. 2.5%; p = NS) despite increasing the international normalized ratio in a dose-related fashion (trend p < or = 0.0001). Higher-dose rNAPc2 (> or =7.5 microg/kg) suppressed prothrombin fragment F1.2 generation compared with placebo and reduced ischemia by >50% compared to placebo and lower-dose rNAPc2. Thrombotic bailout requiring open-label anticoagulant occurred in 5 of 26 patients treated without UFH, but none in the half-dose UFH group (19% vs. 0%; p = 0.051). CONCLUSIONS In patients with nSTE-ACS managed with standard antithrombotics and an early invasive approach, additional proximal inhibition of the coagulation cascade with rNAPc2 was well tolerated. rNAPc2 doses > or =7.5 microg/kg suppressed F1.2 and reduced ischemia, though some heparin may be necessary to avoid procedure-related thrombus formation. (Anticoagulation With rNAPc2 to Eliminate MACE/TIMI 32; http://www.clinicaltrial.gov/ct/show/NCT00116012?order=1; NCT00116012).
Collapse
|
39
|
McRae SJ, Eikelboom JW. Latest medical treatment strategies for venous thromboembolism. Expert Opin Pharmacother 2007; 8:1221-33. [PMID: 17563258 DOI: 10.1517/14656566.8.9.1221] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Anticoagulant therapy with unfractionated heparin (UFH) followed by warfarin prevents thrombus extension, reduces the risk of recurrent thrombosis and prevents death in patients with venous thromboembolism (VTE). Low-molecular-weight heparin (LMWH) has replaced UFH as the preferred initial anticoagulant therapy for VTE because it is as effective and safe as UFH, but does not require laboratory monitoring and is less likely to cause immune thrombocytopenia and osteoporosis. More recently, fondaparinux has been shown to be an effective and safe alternative to LMWH and several new parenteral anticoagulants are being evaluated. The most important unmet need in the anticoagulant management of VTE is a replacement for warfarin. New oral anticoagulants that selectively target individual steps in the coagulation cascade have been shown to be effective for the long-term treatment of VTE in Phase II and III trials and are likely to become available in the near future.
Collapse
Affiliation(s)
- Simon J McRae
- The Queen Elizabeth Hospital, Institute of Medical and Veterinary Sciences, Department of Haematology-Oncology, 28 Woodville Rd, Woodville 5011, Adelaide, South Australia, Australia.
| | | |
Collapse
|
40
|
Uusitalo-Jarvinen H, Kurokawa T, Mueller BM, Andrade-Gordon P, Friedlander M, Ruf W. Role of protease activated receptor 1 and 2 signaling in hypoxia-induced angiogenesis. Arterioscler Thromb Vasc Biol 2007; 27:1456-62. [PMID: 17363687 DOI: 10.1161/atvbaha.107.142539] [Citation(s) in RCA: 97] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
OBJECTIVE Tissue factor (TF) initiates coagulation and indirectly triggers thrombin-dependent protease activated receptor (PAR) signaling. The TF-VIIa complex also directly cleaves PAR2 and promotes angiogenesis in vitro in TF cytoplasmic domain-deleted (TF(deltaCT)) mice. Here we address the effect of PAR1 and PAR2 deficiency on angiogenesis in vivo. METHODS AND RESULTS In hypoxia-driven angiogenesis of oxygen induced retinopathy (OIR), wild-type, PAR1-/-, PAR2-/-, and TF(deltaCT) mice showed a comparable regression of the superficial vascular plexus during the initial exposure of mice to hyperoxia. However, TF(deltaCT) mice revascularized areas of central vaso-obliteration significantly faster than wild-type animals. Pharmacological inhibition of the TF-VIIa complex, but not of Xa, and blockade of tyrosine kinase receptor pathways with Gleevec reversed accelerated angiogenesis of TF(deltaCT) mice to revascularization rates observed in wild-type mice. Genetic deletion of PAR2, but not of PAR1, abolished enhanced revascularization of TF(deltaCT) mice. PAR1 knock-out animals were indistinguishable from wild-type mice in the model of retinal neoangiogenesis and angiogenesis-dependent subcutaneous tumor growth was unaltered in PAR1- and PAR2-deficient animals. CONCLUSION Loss of the TF cytoplasmic domain results in accelerated hypoxia-induced angiogenesis mediated by TF-VIIa signaling. PAR2 signaling is sufficient for this proangiogenic effect without apparent contributions of mouse host cell PAR1.
Collapse
MESH Headings
- Animals
- Benzamides
- Blood Coagulation Factor Inhibitors/pharmacology
- Cell Line, Tumor
- Disease Models, Animal
- Factor VIIa/metabolism
- Hyperoxia/chemically induced
- Hyperoxia/metabolism
- Hyperoxia/pathology
- Hypoxia/complications
- Hypoxia/metabolism
- Hypoxia/pathology
- Imatinib Mesylate
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Neoplasms, Experimental/blood supply
- Neoplasms, Experimental/metabolism
- Neoplasms, Experimental/pathology
- Neovascularization, Pathologic/metabolism
- Neovascularization, Pathologic/pathology
- Oxygen
- Piperazines/pharmacology
- Protein Kinase Inhibitors/pharmacology
- Pyrimidines/pharmacology
- Receptor, PAR-1/deficiency
- Receptor, PAR-1/genetics
- Receptor, PAR-1/metabolism
- Receptor, PAR-2/deficiency
- Receptor, PAR-2/genetics
- Receptor, PAR-2/metabolism
- Retinal Neovascularization/etiology
- Retinal Neovascularization/metabolism
- Retinal Neovascularization/pathology
- Retinal Vessels/metabolism
- Retinal Vessels/pathology
- Signal Transduction/drug effects
- Thromboplastin/genetics
- Thromboplastin/metabolism
- Time Factors
Collapse
Affiliation(s)
- Hannele Uusitalo-Jarvinen
- Department of Cell Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | | | | | | | | | | |
Collapse
|
41
|
NG HENGJOO, CROWTHER MARK. New anticoagulants and the management of their bleeding complications. ACTA ACUST UNITED AC 2006. [DOI: 10.1111/j.1778-428x.2006.00026.x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
42
|
Mahajan AL, Tenorio X, Pepper MS, Baetens D, Montandon D, Schlaudraff KU, Pittet B. Progressive tissue injury in burns is reduced by rNAPc2. Burns 2006; 32:957-63. [PMID: 16905262 DOI: 10.1016/j.burns.2006.02.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2005] [Accepted: 02/16/2006] [Indexed: 11/16/2022]
Abstract
INTRODUCTION Burn wounds are characterised by central necrosis surrounded by an area of stasis with compromised perfusion. Secondary aggravation of the burn wound due to ischaemia in the zone of stasis can also result in necrosis. This study aims to improve circulation in the zone of stasis by reducing microthrombus formation and thereby to reduce secondary aggravation. MATERIAL AND METHODS Recombinant nematode anticoagulant protein (rNAPc2) was administered to Wistar rats at 3 or 30 microg/kg as a single or daily dose. A comb pattern burn was induced on the dorsum of these rats and its evolution monitored by serial photography, planimetry, laser doppler flowmetry and immunohistochemistry. RESULTS In the 30 microg/kg daily group, extension of the burn wound was curbed, limiting the burn area to 1.99+/-0.67 cm(2) on day 28, compared to 3.51+/-0.37 cm(2) in the control group (p=0.015). Laser doppler evaluation showed a significant (p<0.001) increase in circulation in the first day post-burn. Significantly less (p<0.001) microvascular fibrin formation was observed by immunohistochemistry. CONCLUSION Anticoagulation with rNAPc2 improved perfusion of the burn wound. The resultant reduction in the area of the burn led to earlier healing and less scar contracture.
Collapse
Affiliation(s)
- Ajay L Mahajan
- Department of Plastic and Reconstructive Surgery, University Hospitals of Geneva, Geneva, Switzerland
| | | | | | | | | | | | | |
Collapse
|
43
|
Mayr FB, Jilma B. Coagulation interventions in experimental human endotoxemia. Transl Res 2006; 148:263-71. [PMID: 17145572 DOI: 10.1016/j.trsl.2006.08.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2006] [Accepted: 08/04/2006] [Indexed: 11/20/2022]
Abstract
Recognition of the link between coagulation activation and inflammation has led to the hypothesis that anticoagulants may be effective in the treatment of septic patients by altering the inflammatory response. However, only limited methodologies exist that can be used in human volunteers to mimic the physiologic alterations observed in critically ill patients. The human endotoxemia model represents a model of inflammation-induced tissue factor triggered coagulation activation. As it permits elucidation of a key player in this proinflammatory and procoagulant response, it serves as a useful tool to investigate novel therapeutics in a standardized setting. The aim of this review is to focus on coagulation interventions in the human endotoxemia model.
Collapse
Affiliation(s)
- Florian B Mayr
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | | |
Collapse
|
44
|
Rijneveld AW, Weijer S, Bresser P, Florquin S, Vlasuk GP, Rote WE, Spek CA, Reitsma PH, van der Zee JS, Levi M, van der Poll T. Local activation of the tissue factor-factor VIIa pathway in patients with pneumonia and the effect of inhibition of this pathway in murine pneumococcal pneumonia. Crit Care Med 2006; 34:1725-30. [PMID: 16625114 DOI: 10.1097/01.ccm.0000218807.20570.c2] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
OBJECTIVE The tissue factor (TF)-factor VIIa (FVIIa) complex not only is essential for activation of blood coagulation but also affect the inflammatory response during sepsis. The objective of this study was to determine the role of TF-FVIIa in pneumonia caused by Streptococcus pneumoniae, the most important causative organism in community-acquired pneumonia and a major cause of sepsis. DESIGN A controlled, in vivo laboratory study. SETTING Research laboratory of a health sciences university. PATIENTS AND SUBJECTS Patients with unilateral community-acquired pneumonia and female BALB/c mice. INTERVENTIONS Bilateral bronchoalveolar lavage was performed in patients with community-acquired pneumonia. In mice, pneumonia was induced by intranasal inoculation with S. pneumoniae with or without concurrent inhibition of TF-FVIIa by subcutaneous injections of recombinant nematode anticoagulant protein (rNAPc2). MEASUREMENTS AND MAIN RESULTS Patients with unilateral community-acquired pneumonia demonstrated elevated concentrations of FVIIa, soluble TF, and thrombin-antithrombin complexes in bronchoalveolar lavage fluid obtained from the infected site compared with the uninfected site. Mice with S. pneumoniae pneumonia displayed increased TF expression and fibrin deposits in lungs together with elevated thrombin-antithrombin complex levels in bronchoalveolar lavage fluid; inhibition of TF-FVIIa by rNAPc2 attenuated the procoagulant response in the lung but did not affect host defense, as reflected by an unaltered outgrowth of pneumococci and an unchanged survival. CONCLUSIONS These data suggest that TF-FVIIa activity contributes to activation of coagulation in the lung during pneumococcal pneumonia but does not play an important role in the antibacterial host defense in this murine model.
Collapse
Affiliation(s)
- Anita W Rijneveld
- Center of Infection and Immunity Amsterdam, Laboratory of Experimental Internal Medicine, Department of Pulmonology, University of Amsterdam, the Netherlands
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Ng HJ, Crowther MA. New Anti-thrombotic Agents: Emphasis on Hemorrhagic Complications and Their Management. Semin Hematol 2006; 43:S77-83. [PMID: 16427390 DOI: 10.1053/j.seminhematol.2005.11.023] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Our advanced knowledge of coagulation has led to the synthesis of novel procoagulant substances, such as recombinant activated factor VII (rFVIIa; NovoSeven, Novo Nordisk, Bagsvaerd, Denmark). Similarly, in-depth understanding of the interaction between anticoagulant proteins and their natural inhibitors has led to the synthesis of various novel anticoagulants. Novel anticoagulants are characterized by highly specific coagulation-inhibiting activities and, frequently, a complete lack of effective antidotes. This lack of antidotes is particularly important in the case of novel inhibitors with extended half-lives; for example, idraparinux may produce effective anticoagulation for as long as one week after subcutaneous administration. As novel anticoagulants complete licensing evaluations and are used in clinical practice, the likelihood of anticoagulant-associated hemorrhage will increase. This will require physicians to have an understanding of the mechanism of action of these anticoagulants, and to have an advanced degree of knowledge of the potential specific and nonspecific inhibitors of these anticoagulant agents. This paper will briefly review the biochemistry of coagulation, focusing on the complexes inhibited by currently available and novel anticoagulants. Specific and nonspecific prohemostatic agents will be reviewed and discussed. The ability of nonspecific procoagulant agents (particularly rFVIIa) to reverse the effects of novel anticoagulants will also be reviewed.
Collapse
Affiliation(s)
- Heng Joo Ng
- Department of Haematology, Singapore General Hospital, Singapore
| | | |
Collapse
|
46
|
Abstract
Unfractionated heparin and vitamin K antagonists such as warfarin have been used as the anticoagulants of choice for over five decades. Subsequently, low molecular weight heparins (LMWHs) became widely available and have provided several advantages, especially in infants and children. The field of anticoagulation, however, has undergone a major revolution with better understanding of the structure of coagulation proteins and the development of a host of new drugs with highly specific actions. Many of these drugs have undergone extensive clinical testing in adults and have been approved for specific indications in adults. Unfortunately, clinical data and the reported use of these drugs in children are extremely limited. A lack of familiarity with the actions and pharmacokinetic properties of these drugs could be a major contributing factor. This review focuses on several of the new anticoagulants, with a special emphasis on those that could be potentially beneficial in pediatric patients with thromboembolic disorders. The need for well-designed trials with large-scale participation by pediatric hematologists in order to improve the antithrombotic care of young infants and children is also emphasized.
Collapse
Affiliation(s)
- Vinod V Balasa
- Hemophilia and Thrombosis Center, Division of Hematology/Oncology, Cincinnati Children's Hospital Medical Center, University of Cincinnati School of Medicine, Cincinnati, OH 45229, USA.
| |
Collapse
|
47
|
Whitlock R, Crowther MA, Ng HJ. Bleeding in Cardiac Surgery: Its Prevention and Treatment—an Evidence-Based Review. Crit Care Clin 2005; 21:589-610. [PMID: 15992674 DOI: 10.1016/j.ccc.2005.04.003] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Expected and unexpected bleeding occur frequently in patients undergoing cardiac surgery. Bleeding after cardiac surgery can be broadly divided into two groups: surgical (unrecognized bleeding vessel, anastomosis, or other suture line) or nonsurgical bleeding (caused by coagulopathy). Factors influencing both surgical and nonsurgical bleeding can be further broken down into those occurring preoperatively and those that occur intraoperatively and postoperatively. A thorough understanding of these factors is necessary to reduce bleeding. This is a desirable clinical goal, because excessive bleeding is associated with adverse outcomes.
Collapse
Affiliation(s)
- Richard Whitlock
- Department of Medicine, McMaster University, Room L208, St. Joseph's Hospital, 50 Charlton Avenue East, Hamilton, Ontario L8N 4A6, Canada
| | | | | |
Collapse
|
48
|
Abstract
Macromolecular substrate recognition and serine proteinase specificity lie at the heart of the tightly regulated hemostatic response. Mechanisms established for the less specific serine proteinases of digestion have played a dominant role in guiding investigations of the basis for the narrow specificities exhibited by the coagulation enzymes. These concepts have also dominated the development of specific inhibitors of coagulation for therapeutic purposes. Studies of the enzymology and physical biochemistry of prothrombinase challenge these prevailing ideas by establishing a principal role for exosites within the enzyme in determining substrate recognition and directing the action of the enzyme on its biological substrate. Mechanisms by which narrow protein substrate specificity is achieved by prothrombinase also apply to several other reactions of coagulation. These strategies are increasingly evident in the action of other families of enzymes that act with high specificity on protein substrates. Exosite-driven enzymic function probably represents a widely employed biological strategy for the achievement of high macromolecular substrate specificity.
Collapse
Affiliation(s)
- S Krishnaswamy
- Joseph Stokes Research Institute, Children's Hospital of Philadelphia & Department of Pediatrics, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
49
|
Affiliation(s)
- Kyung W Park
- Department of Anesthesia and Critical Care, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
| |
Collapse
|
50
|
Abstract
Activation of coagulation by tissue factor (TF) is frequently observed in sepsis syndrome and is documented in certain viral hemorrhagic fevers. Coagulation protease complexes signal by activating the G-protein coupled, protease-activated receptors that regulate inflammation. Blockade of TF attenuates lethality in experimental models of Ebola virus infection but - similar to findings in bacterial sepsis - reduction of inflammation, rather than attenuation of coagulation, predicts survival of treated animals. Thus, targeting TF appears to aid the antiviral immune response in hemorrhagic fevers, and further studies are encouraged to define how TF-dependent signaling regulates immunity.
Collapse
Affiliation(s)
- Wolfram Ruf
- Department of Immunology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA.
| |
Collapse
|