451
|
Affiliation(s)
| | - Peter Libby
- Cardiovascular Medicine, Brigham and Women's Hospital, USA
| |
Collapse
|
452
|
Meegan JE, Yang X, Coleman DC, Jannaway M, Yuan SY. Neutrophil-mediated vascular barrier injury: Role of neutrophil extracellular traps. Microcirculation 2018; 24. [PMID: 28120468 DOI: 10.1111/micc.12352] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Accepted: 01/12/2017] [Indexed: 12/19/2022]
Abstract
Neutrophils play an essential role in host defense against infection or injury. While neutrophil activation is necessary for pathogen clearance and tissue repair, a hyperactive response can lead to tissue damage and microcirculatory disorders, a process involving complex neutrophil-endothelium cross talk. This review highlights recent research findings about neutrophil-mediated signaling and structural changes, including those induced by neutrophil extracellular traps, which ultimately lead to vascular barrier injury.
Collapse
Affiliation(s)
- Jamie E Meegan
- Department of Molecular Pharmacology and Physiology, University of South Florida College of Medicine, Tampa, FL, USA
| | - Xiaoyuan Yang
- Department of Molecular Pharmacology and Physiology, University of South Florida College of Medicine, Tampa, FL, USA
| | - Danielle C Coleman
- Department of Molecular Pharmacology and Physiology, University of South Florida College of Medicine, Tampa, FL, USA
| | - Melanie Jannaway
- Department of Molecular Pharmacology and Physiology, University of South Florida College of Medicine, Tampa, FL, USA
| | - Sarah Y Yuan
- Department of Molecular Pharmacology and Physiology, University of South Florida College of Medicine, Tampa, FL, USA
| |
Collapse
|
453
|
Mauracher LM, Posch F, Martinod K, Grilz E, Däullary T, Hell L, Brostjan C, Zielinski C, Ay C, Wagner DD, Pabinger I, Thaler J. Citrullinated histone H3, a biomarker of neutrophil extracellular trap formation, predicts the risk of venous thromboembolism in cancer patients. J Thromb Haemost 2018; 16:508-518. [PMID: 29325226 PMCID: PMC6294121 DOI: 10.1111/jth.13951] [Citation(s) in RCA: 191] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Indexed: 12/12/2022]
Abstract
Essentials Neutrophil extracellular traps (NETs) might play a role in cancer-related coagulopathy. We determined NET biomarkers and followed cancer patients for venous thromboembolism (VTE). We found a constant association with VTE for citrullinated histone H3. Biomarkers of NET formation could reflect a novel pathomechanism of cancer-related VTE. SUMMARY Background Neutrophil extracellular traps (NETs) are decondensed chromatin fibers that might play a role in the prothrombotic state of cancer patients. Objectives To investigate whether the levels of citrullinated histone H3 (H3Cit), a biomarker for NET formation, cell-free DNA (cfDNA) and nucleosomes predict venous thromboembolism (VTE) in cancer patients. Patients/Methods Nine-hundred and forty-six patients with newly diagnosed cancer or progression after remission were enrolled in this prospective observational cohort study. H3Cit, cfDNA and nucleosome levels were determined at study inclusion, and patients were followed for 2 years. VTE occurred in 89 patients; the cumulative 3-month, 6-month, 12-month and 24-month incidence rates of VTE were 3.7%, 6.0%, 8.1%, and 10.0%, respectively. Results Patients with elevated H3Cit levels (> 75th percentile of its distribution, n = 236) experienced a higher cumulative incidence of VTE (2-year risk of 14.5%) than patients with levels below this cut-off (2-year risk of 8.5%, n = 710). In a competing-risk regression analysis, a 100 ng mL-1 increase in H3Cit level was associated with a 13% relative increase in VTE risk (subdistribution hazard ratio [SHR] 1.13, 95% confidence interval [CI] 1.04-1.22). This association remained after adjustment for high VTE risk and very high VTE risk tumor sites, D-dimer level, and soluble P-selectin level (SHR 1.13, 95% CI 1.04-1.22). The association of elevated nucleosome and cfDNA levels with VTE risk was time-dependent, with associations with a higher risk of VTE only during the first 3-6 months. Conclusion These data suggest that biomarkers of NET formation are associated with the occurrence of VTE in cancer patients, indicating a role of NETs in the pathogenesis of cancer-associated thrombosis.
Collapse
Affiliation(s)
- L-M Mauracher
- Clinical Division of Hematology and Hemostaseology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - F Posch
- Clinical Division of Hematology and Hemostaseology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - K Martinod
- Program in Cellular and Molecular Medicine, Boston Children's Hospital and Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - E Grilz
- Clinical Division of Hematology and Hemostaseology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - T Däullary
- Clinical Division of Hematology and Hemostaseology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - L Hell
- Clinical Division of Hematology and Hemostaseology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - C Brostjan
- Department of Surgery, Medical University of Vienna, Austria
| | - C Zielinski
- Clinical Division of Oncology, Department of Medicine I, Comprehensive Cancer Center Vienna, Medical University of Vienna, Vienna, Austria
| | - C Ay
- Clinical Division of Hematology and Hemostaseology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - D D Wagner
- Program in Cellular and Molecular Medicine, Boston Children's Hospital and Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - I Pabinger
- Clinical Division of Hematology and Hemostaseology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - J Thaler
- Clinical Division of Hematology and Hemostaseology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
454
|
Ducroux C, Di Meglio L, Loyau S, Delbosc S, Boisseau W, Deschildre C, Ben Maacha M, Blanc R, Redjem H, Ciccio G, Smajda S, Fahed R, Michel JB, Piotin M, Salomon L, Mazighi M, Ho-Tin-Noe B, Desilles JP. Thrombus Neutrophil Extracellular Traps Content Impair tPA-Induced Thrombolysis in Acute Ischemic Stroke. Stroke 2018; 49:754-757. [PMID: 29438080 DOI: 10.1161/strokeaha.117.019896] [Citation(s) in RCA: 237] [Impact Index Per Article: 33.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 12/22/2017] [Accepted: 01/09/2018] [Indexed: 11/16/2022]
Abstract
BACKGROUND AND PURPOSE Neutrophil Extracellular Traps (NETs) are DNA extracellular networks decorated with histones and granular proteins produced by activated neutrophils. NETs have been identified as major triggers and structural factors of thrombosis. A recent study designated extracellular DNA threads from NETs as a potential therapeutic target for improving tissue-type plasminogen activator (tPA)-induced thrombolysis in acute coronary syndrome. The aim of this study was to assess the presence of NETs in thrombi retrieved during endovascular therapy in patients with acute ischemic stroke (AIS) and their impact on tPA-induced thrombolysis. METHODS We analyzed thrombi from 108 AIS patients treated with endovascular therapy. Thrombi were characterized by hematoxylin/eosin staining, immunostaining, and ex vivo enzymatic assay. Additionally, we assessed ex vivo the impact of deoxyribonuclease 1 (DNAse 1) on thrombolysis of AIS thrombi. RESULTS Histological analysis revealed that NETs contributed to the composition of all AIS thrombi especially in their outer layers. Quantitative measurement of thrombus NETs content was not associated with clinical outcome or AIS pathogenesis but correlated significantly with endovascular therapy procedure length and device number of passes. Ex vivo, recombinant DNAse 1 accelerated tPA-induced thrombolysis, whereas DNAse 1 alone was ineffective. CONCLUSIONS This study suggests that thrombus NETs content may be responsible for reperfusion resistance, including mechanical or pharmacological approaches with intravenous tPA, irrespectively of their etiology. The efficacy of a strategy involving an administration of DNAse 1 in addition to tPA should be explored in the setting of AIS. CLINICAL TRIAL REGISTRATION URL: http://www.clinicaltrials.gov. Unique identifier: NCT02907736.
Collapse
Affiliation(s)
- Celina Ducroux
- From the Université Paris Diderot, Sorbonne Paris Cite, Laboratory of Vascular Translational Science, U1148 Institut National de la Santé et de la Recherche Médicale (INSERM), France (C. Ducroux, L.D.M., S.L., S.D., W.B., C. Deschildre, R.B., J.-B.M., M.P., M.M., B.H.-T.-N., J.-P.D.); Department of Interventional Neuroradiology (W.B., R.B., H.R., G.C., S.S., R.F., M.P., M.M., J.-P.D.) and Department of Clinical Research (M.B.M., L.S.), Rothschild Foundation Hospital, Paris, France; and DHU NeuroVasc, Paris, France (M.M.)
| | - Lucas Di Meglio
- From the Université Paris Diderot, Sorbonne Paris Cite, Laboratory of Vascular Translational Science, U1148 Institut National de la Santé et de la Recherche Médicale (INSERM), France (C. Ducroux, L.D.M., S.L., S.D., W.B., C. Deschildre, R.B., J.-B.M., M.P., M.M., B.H.-T.-N., J.-P.D.); Department of Interventional Neuroradiology (W.B., R.B., H.R., G.C., S.S., R.F., M.P., M.M., J.-P.D.) and Department of Clinical Research (M.B.M., L.S.), Rothschild Foundation Hospital, Paris, France; and DHU NeuroVasc, Paris, France (M.M.)
| | - Stephane Loyau
- From the Université Paris Diderot, Sorbonne Paris Cite, Laboratory of Vascular Translational Science, U1148 Institut National de la Santé et de la Recherche Médicale (INSERM), France (C. Ducroux, L.D.M., S.L., S.D., W.B., C. Deschildre, R.B., J.-B.M., M.P., M.M., B.H.-T.-N., J.-P.D.); Department of Interventional Neuroradiology (W.B., R.B., H.R., G.C., S.S., R.F., M.P., M.M., J.-P.D.) and Department of Clinical Research (M.B.M., L.S.), Rothschild Foundation Hospital, Paris, France; and DHU NeuroVasc, Paris, France (M.M.)
| | - Sandrine Delbosc
- From the Université Paris Diderot, Sorbonne Paris Cite, Laboratory of Vascular Translational Science, U1148 Institut National de la Santé et de la Recherche Médicale (INSERM), France (C. Ducroux, L.D.M., S.L., S.D., W.B., C. Deschildre, R.B., J.-B.M., M.P., M.M., B.H.-T.-N., J.-P.D.); Department of Interventional Neuroradiology (W.B., R.B., H.R., G.C., S.S., R.F., M.P., M.M., J.-P.D.) and Department of Clinical Research (M.B.M., L.S.), Rothschild Foundation Hospital, Paris, France; and DHU NeuroVasc, Paris, France (M.M.)
| | - William Boisseau
- From the Université Paris Diderot, Sorbonne Paris Cite, Laboratory of Vascular Translational Science, U1148 Institut National de la Santé et de la Recherche Médicale (INSERM), France (C. Ducroux, L.D.M., S.L., S.D., W.B., C. Deschildre, R.B., J.-B.M., M.P., M.M., B.H.-T.-N., J.-P.D.); Department of Interventional Neuroradiology (W.B., R.B., H.R., G.C., S.S., R.F., M.P., M.M., J.-P.D.) and Department of Clinical Research (M.B.M., L.S.), Rothschild Foundation Hospital, Paris, France; and DHU NeuroVasc, Paris, France (M.M.)
| | - Catherine Deschildre
- From the Université Paris Diderot, Sorbonne Paris Cite, Laboratory of Vascular Translational Science, U1148 Institut National de la Santé et de la Recherche Médicale (INSERM), France (C. Ducroux, L.D.M., S.L., S.D., W.B., C. Deschildre, R.B., J.-B.M., M.P., M.M., B.H.-T.-N., J.-P.D.); Department of Interventional Neuroradiology (W.B., R.B., H.R., G.C., S.S., R.F., M.P., M.M., J.-P.D.) and Department of Clinical Research (M.B.M., L.S.), Rothschild Foundation Hospital, Paris, France; and DHU NeuroVasc, Paris, France (M.M.)
| | - Malek Ben Maacha
- From the Université Paris Diderot, Sorbonne Paris Cite, Laboratory of Vascular Translational Science, U1148 Institut National de la Santé et de la Recherche Médicale (INSERM), France (C. Ducroux, L.D.M., S.L., S.D., W.B., C. Deschildre, R.B., J.-B.M., M.P., M.M., B.H.-T.-N., J.-P.D.); Department of Interventional Neuroradiology (W.B., R.B., H.R., G.C., S.S., R.F., M.P., M.M., J.-P.D.) and Department of Clinical Research (M.B.M., L.S.), Rothschild Foundation Hospital, Paris, France; and DHU NeuroVasc, Paris, France (M.M.)
| | - Raphael Blanc
- From the Université Paris Diderot, Sorbonne Paris Cite, Laboratory of Vascular Translational Science, U1148 Institut National de la Santé et de la Recherche Médicale (INSERM), France (C. Ducroux, L.D.M., S.L., S.D., W.B., C. Deschildre, R.B., J.-B.M., M.P., M.M., B.H.-T.-N., J.-P.D.); Department of Interventional Neuroradiology (W.B., R.B., H.R., G.C., S.S., R.F., M.P., M.M., J.-P.D.) and Department of Clinical Research (M.B.M., L.S.), Rothschild Foundation Hospital, Paris, France; and DHU NeuroVasc, Paris, France (M.M.)
| | - Hocine Redjem
- From the Université Paris Diderot, Sorbonne Paris Cite, Laboratory of Vascular Translational Science, U1148 Institut National de la Santé et de la Recherche Médicale (INSERM), France (C. Ducroux, L.D.M., S.L., S.D., W.B., C. Deschildre, R.B., J.-B.M., M.P., M.M., B.H.-T.-N., J.-P.D.); Department of Interventional Neuroradiology (W.B., R.B., H.R., G.C., S.S., R.F., M.P., M.M., J.-P.D.) and Department of Clinical Research (M.B.M., L.S.), Rothschild Foundation Hospital, Paris, France; and DHU NeuroVasc, Paris, France (M.M.)
| | - Gabriele Ciccio
- From the Université Paris Diderot, Sorbonne Paris Cite, Laboratory of Vascular Translational Science, U1148 Institut National de la Santé et de la Recherche Médicale (INSERM), France (C. Ducroux, L.D.M., S.L., S.D., W.B., C. Deschildre, R.B., J.-B.M., M.P., M.M., B.H.-T.-N., J.-P.D.); Department of Interventional Neuroradiology (W.B., R.B., H.R., G.C., S.S., R.F., M.P., M.M., J.-P.D.) and Department of Clinical Research (M.B.M., L.S.), Rothschild Foundation Hospital, Paris, France; and DHU NeuroVasc, Paris, France (M.M.)
| | - Stanislas Smajda
- From the Université Paris Diderot, Sorbonne Paris Cite, Laboratory of Vascular Translational Science, U1148 Institut National de la Santé et de la Recherche Médicale (INSERM), France (C. Ducroux, L.D.M., S.L., S.D., W.B., C. Deschildre, R.B., J.-B.M., M.P., M.M., B.H.-T.-N., J.-P.D.); Department of Interventional Neuroradiology (W.B., R.B., H.R., G.C., S.S., R.F., M.P., M.M., J.-P.D.) and Department of Clinical Research (M.B.M., L.S.), Rothschild Foundation Hospital, Paris, France; and DHU NeuroVasc, Paris, France (M.M.)
| | - Robert Fahed
- From the Université Paris Diderot, Sorbonne Paris Cite, Laboratory of Vascular Translational Science, U1148 Institut National de la Santé et de la Recherche Médicale (INSERM), France (C. Ducroux, L.D.M., S.L., S.D., W.B., C. Deschildre, R.B., J.-B.M., M.P., M.M., B.H.-T.-N., J.-P.D.); Department of Interventional Neuroradiology (W.B., R.B., H.R., G.C., S.S., R.F., M.P., M.M., J.-P.D.) and Department of Clinical Research (M.B.M., L.S.), Rothschild Foundation Hospital, Paris, France; and DHU NeuroVasc, Paris, France (M.M.)
| | - Jean-Baptiste Michel
- From the Université Paris Diderot, Sorbonne Paris Cite, Laboratory of Vascular Translational Science, U1148 Institut National de la Santé et de la Recherche Médicale (INSERM), France (C. Ducroux, L.D.M., S.L., S.D., W.B., C. Deschildre, R.B., J.-B.M., M.P., M.M., B.H.-T.-N., J.-P.D.); Department of Interventional Neuroradiology (W.B., R.B., H.R., G.C., S.S., R.F., M.P., M.M., J.-P.D.) and Department of Clinical Research (M.B.M., L.S.), Rothschild Foundation Hospital, Paris, France; and DHU NeuroVasc, Paris, France (M.M.)
| | - Michel Piotin
- From the Université Paris Diderot, Sorbonne Paris Cite, Laboratory of Vascular Translational Science, U1148 Institut National de la Santé et de la Recherche Médicale (INSERM), France (C. Ducroux, L.D.M., S.L., S.D., W.B., C. Deschildre, R.B., J.-B.M., M.P., M.M., B.H.-T.-N., J.-P.D.); Department of Interventional Neuroradiology (W.B., R.B., H.R., G.C., S.S., R.F., M.P., M.M., J.-P.D.) and Department of Clinical Research (M.B.M., L.S.), Rothschild Foundation Hospital, Paris, France; and DHU NeuroVasc, Paris, France (M.M.)
| | - Laurence Salomon
- From the Université Paris Diderot, Sorbonne Paris Cite, Laboratory of Vascular Translational Science, U1148 Institut National de la Santé et de la Recherche Médicale (INSERM), France (C. Ducroux, L.D.M., S.L., S.D., W.B., C. Deschildre, R.B., J.-B.M., M.P., M.M., B.H.-T.-N., J.-P.D.); Department of Interventional Neuroradiology (W.B., R.B., H.R., G.C., S.S., R.F., M.P., M.M., J.-P.D.) and Department of Clinical Research (M.B.M., L.S.), Rothschild Foundation Hospital, Paris, France; and DHU NeuroVasc, Paris, France (M.M.)
| | - Mikael Mazighi
- From the Université Paris Diderot, Sorbonne Paris Cite, Laboratory of Vascular Translational Science, U1148 Institut National de la Santé et de la Recherche Médicale (INSERM), France (C. Ducroux, L.D.M., S.L., S.D., W.B., C. Deschildre, R.B., J.-B.M., M.P., M.M., B.H.-T.-N., J.-P.D.); Department of Interventional Neuroradiology (W.B., R.B., H.R., G.C., S.S., R.F., M.P., M.M., J.-P.D.) and Department of Clinical Research (M.B.M., L.S.), Rothschild Foundation Hospital, Paris, France; and DHU NeuroVasc, Paris, France (M.M.)
| | - Benoit Ho-Tin-Noe
- From the Université Paris Diderot, Sorbonne Paris Cite, Laboratory of Vascular Translational Science, U1148 Institut National de la Santé et de la Recherche Médicale (INSERM), France (C. Ducroux, L.D.M., S.L., S.D., W.B., C. Deschildre, R.B., J.-B.M., M.P., M.M., B.H.-T.-N., J.-P.D.); Department of Interventional Neuroradiology (W.B., R.B., H.R., G.C., S.S., R.F., M.P., M.M., J.-P.D.) and Department of Clinical Research (M.B.M., L.S.), Rothschild Foundation Hospital, Paris, France; and DHU NeuroVasc, Paris, France (M.M.)
| | - Jean-Philippe Desilles
- From the Université Paris Diderot, Sorbonne Paris Cite, Laboratory of Vascular Translational Science, U1148 Institut National de la Santé et de la Recherche Médicale (INSERM), France (C. Ducroux, L.D.M., S.L., S.D., W.B., C. Deschildre, R.B., J.-B.M., M.P., M.M., B.H.-T.-N., J.-P.D.); Department of Interventional Neuroradiology (W.B., R.B., H.R., G.C., S.S., R.F., M.P., M.M., J.-P.D.) and Department of Clinical Research (M.B.M., L.S.), Rothschild Foundation Hospital, Paris, France; and DHU NeuroVasc, Paris, France (M.M.).
| |
Collapse
|
455
|
Dyer MR, Chen Q, Haldeman S, Yazdani H, Hoffman R, Loughran P, Tsung A, Zuckerbraun BS, Simmons RL, Neal MD. Deep vein thrombosis in mice is regulated by platelet HMGB1 through release of neutrophil-extracellular traps and DNA. Sci Rep 2018; 8:2068. [PMID: 29391442 PMCID: PMC5794752 DOI: 10.1038/s41598-018-20479-x] [Citation(s) in RCA: 126] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Accepted: 12/06/2017] [Indexed: 12/21/2022] Open
Abstract
Venous thromboembolic (VTE) disease, consisting of deep venous thrombosis (DVT) and pulmonary embolism (PE) is a leading cause of morbidity and mortality. Current prophylactic measures are insufficient to prevent all occurrence in part due to an incomplete understanding of the underlying pathophysiology. Mounting evidence describes interplay between activation of the innate immune system and thrombus development. Recent work has demonstrated that platelet release of HMGB1 leads to increased microvascular complications following injury. Additionally, platelet HMGB1 was found to enhance DVT and increase the formation of neutrophil extracellular traps (NETs), although the role of HMGB1 induced NET release in thrombosis remains unexplored. Utilizing a transgenic mouse lacking HMGB1 specifically from platelets and megakaryocytes we now demonstrate the specific role of platelet-derived HMGB1 in acute and subacute/chronic venous thrombosis. Platelets account for the majority of circulating HMGB1 and HMGB1 deposition within the developing clot. The pro-thrombotic effect of platelet-derived HMGB1 is mediated through enhanced neutrophil recruitment, NET formation and specifically release of extracellular DNA during NET formation. Taken together, these data suggest that platelet HMGB1 mediated NET release is a primary regulator of DVT formation in mice.
Collapse
Affiliation(s)
- Mitchell R Dyer
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Qiwei Chen
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Shannon Haldeman
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Hamza Yazdani
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Rosemary Hoffman
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Patricia Loughran
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
- Center for Biological Imaging, University of Pittsburgh, Pittsburgh, PA, USA
| | - Allan Tsung
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Brian S Zuckerbraun
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Richard L Simmons
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Matthew D Neal
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA, USA.
| |
Collapse
|
456
|
Gavillet M, Martinod K, Renella R, Wagner DD, Williams DA. A key role for Rac and Pak signaling in neutrophil extracellular traps (NETs) formation defines a new potential therapeutic target. Am J Hematol 2018; 93:269-276. [PMID: 29124783 DOI: 10.1002/ajh.24970] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 11/06/2017] [Accepted: 11/07/2017] [Indexed: 12/13/2022]
Abstract
NET formation in mice (NETosis) is supported by reactive oxygen species (ROS) production by NADPH oxidase and histone hypercitrullination by peptidylarginine deiminase 4 (PAD4). Rac1 and Rac2, expressed in polymorphonuclear neutrophils (PMNs), regulate the cytoskeleton, cell shape, adhesion, and migration and are also essential components of the NADPH oxidase complex. We aimed to explore the role of the Rac signaling pathway including the upstream guanosine exchange factor (GEF) activator, Vav, and a downstream effector, the p21-activated kinase, Pak, on NETosis in PMNs using a previously described flow-cytometry-based assay. Rac2-/- PMNs showed reduced levels of citrullinated histone H3 (H3Cit)-positive cells and defective NETosis. Rac1Δ/Δ ; Rac2-/- PMNs demonstrated a further reduction in PMA-induced H3Cit levels and a more profound impairment of NETosis than deletion of Rac2 alone, suggesting an overlapping role of these two highly related proteins. Genetic knockouts of Vav1, or Vav2, did not impair H3Cit response to phorbol myristate ester (PMA) or NETosis. Combined, Vav1 and Vav3 deletions decreased H3Cit response and caused a modest but significant impairment of NETosis. Pharmacologic inhibition of Pak by two inhibitors with distinct mechanisms of action, led to reduced H3Cit levels after PMA stimulation, as well as significant inhibition of NETosis. We validated the importance of Pak using Pak2Δ/Δ PMNs, which demonstrated significantly impaired histone H3 citrullination and NETosis. These data confirm and more comprehensively define the key role of the Rac signaling pathway in PMN NETosis. The Rac signaling cascade may represent a valuable target for inhibition of NETosis and related pathological processes.
Collapse
Affiliation(s)
- Mathilde Gavillet
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA; Department of Pediatrics; Harvard Medical School; Boston Massachusetts
- Department of Hematology; Lausanne University Hospital; Switzerland
| | - Kimberly Martinod
- Program in Cellular and Molecular Medicine; Boston Children's Hospital; Boston Massachusetts
- Department of Pediatrics; Harvard Medical School; Boston Massachusetts
| | - Raffaele Renella
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA; Department of Pediatrics; Harvard Medical School; Boston Massachusetts
- Pediatric Hematology-Oncology Unit, Division of Pediatrics; Lausanne University Hospital; Switzerland
| | - Denisa D. Wagner
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA; Department of Pediatrics; Harvard Medical School; Boston Massachusetts
- Program in Cellular and Molecular Medicine; Boston Children's Hospital; Boston Massachusetts
| | - David A. Williams
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA; Department of Pediatrics; Harvard Medical School; Boston Massachusetts
- Harvard Stem Cell Institute; Cambridge Massachusetts
| |
Collapse
|
457
|
Witsch T, Martinod K, Sorvillo N, Portier I, De Meyer SF, Wagner DD. Recombinant Human ADAMTS13 Treatment Improves Myocardial Remodeling and Functionality After Pressure Overload Injury in Mice. J Am Heart Assoc 2018; 7:JAHA.117.007004. [PMID: 29367415 PMCID: PMC5850234 DOI: 10.1161/jaha.117.007004] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Background A disintegrin‐like metalloproteinase with thrombospondin motif type 1 member 13 (ADAMTS13), the von Willebrand factor–cleaving enzyme, decreases leukocyte and platelet recruitment and, thus, reduces thrombosis and inflammation. Recombinant human ADAMTS13 (rhADAMTS13) is a novel drug candidate for ischemia/reperfusion injury and has shown short‐term benefits in mouse models of myocardial injury, but long‐term outcome has not been investigated. Methods and Results We evaluated the impact of rhADAMTS13 on cardiac remodeling, scarring, and contractile function, under chronic left ventricular pressure overload. The role of von Willebrand factor and the effect of rhADAMTS13 treatment were studied. This model of heart failure, based on ascending aortic constriction, produces a coronary inflammatory response and microvascular dysfunction, resulting in fibrotic remodeling and cardiac failure. Mice were treated with either rhADAMTS13 or vehicle and assessed for coronary vascular inflammation and ventricular function at several postsurgical time points, as well as for cardiac fibrosis after 4 weeks. Early upon induction of pressure overload under rhADAMTS13 treatment, we detected less endothelial‐lumen–associated von Willebrand factor, fewer platelet aggregates, and decreased activated transforming growth factor‐β1 levels than in vehicle‐treated mice. We observed significant preservation of cardiac function and decrease in fibrotic remodeling as a result of rhADAMTS13 administration. Conclusions Herein, we show that rhADAMTS13 decreases coronary vascular dysfunction and improves cardiac remodeling after left ventricular pressure overload in mice. We propose that this effect may, at least in part, be the result of decreased von Willebrand factor–mediated recruitment of platelets, a major source of the activated profibrotic cytokine transforming growth factor‐β1. Our study further supports the therapeutic potential of rhADAMTS13 for conditions characterized by inflammatory cardiac damage that results in fibrosis.
Collapse
Affiliation(s)
- Thilo Witsch
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA.,Department of Pediatrics, Harvard Medical School, Boston, MA.,Department of Cardiology and Angiology I, Heart Center, University of Freiburg, Freiburg, Germany
| | - Kimberly Martinod
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA.,Department of Pediatrics, Harvard Medical School, Boston, MA.,Laboratory for Thrombosis Research, KU Leuven Campus Kulak Kortrijk, Kortrijk, Belgium
| | - Nicoletta Sorvillo
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA.,Department of Pediatrics, Harvard Medical School, Boston, MA
| | - Irina Portier
- Laboratory for Thrombosis Research, KU Leuven Campus Kulak Kortrijk, Kortrijk, Belgium
| | - Simon F De Meyer
- Laboratory for Thrombosis Research, KU Leuven Campus Kulak Kortrijk, Kortrijk, Belgium
| | - Denisa D Wagner
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA .,Department of Pediatrics, Harvard Medical School, Boston, MA.,Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA
| |
Collapse
|
458
|
Novotny J, Chandraratne S, Weinberger T, Philippi V, Stark K, Ehrlich A, Pircher J, Konrad I, Oberdieck P, Titova A, Hoti Q, Schubert I, Legate KR, Urtz N, Lorenz M, Pelisek J, Massberg S, von Brühl ML, Schulz C. Histological comparison of arterial thrombi in mice and men and the influence of Cl-amidine on thrombus formation. PLoS One 2018; 13:e0190728. [PMID: 29293656 PMCID: PMC5749862 DOI: 10.1371/journal.pone.0190728] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Accepted: 12/19/2017] [Indexed: 02/08/2023] Open
Abstract
Aims Medical treatment of arterial thrombosis is mainly directed against platelets and coagulation factors, and can lead to bleeding complications. Novel antithrombotic therapies targeting immune cells and neutrophil extracellular traps (NETs) are currently being investigated in animals. We addressed whether immune cell composition of arterial thrombi induced in mouse models of thrombosis resemble those of human patients with acute myocardial infarction (AMI). Methods and results In a prospective cohort study of patients suffering from AMI, 81 human arterial thrombi were harvested during percutaneous coronary intervention and subjected to detailed histological analysis. In mice, arterial thrombi were induced using two distinct experimental models, ferric chloride (FeCl3) and wire injury of the carotid artery. We found that murine arterial thrombi induced by FeCl3 were highly concordant with human coronary thrombi regarding their immune cell composition, with neutrophils being the most abundant cell type, as well as the presence of NETs and coagulation factors. Pharmacological treatment of mice with the protein arginine deiminase (PAD)-inhibitor Cl-amidine abrogated NET formation, reduced arterial thrombosis and limited injury in a model of myocardial infarction. Conclusions Neutrophils are a hallmark of arterial thrombi in patients suffering from acute myocardial infarction and in mouse models of arterial thrombosis. Inhibition of PAD could represent an interesting strategy for the treatment of arterial thrombosis to reduce neutrophil-associated tissue damage and improve functional outcome.
Collapse
Affiliation(s)
- Julia Novotny
- Medizinische Klinik und Poliklinik I, Ludwig-Maximilians-Universität, Munich, Germany
- Walter-Brendel-Centre of Experimental Medicine, Ludwig-Maximilians-Universität, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Sue Chandraratne
- Medizinische Klinik und Poliklinik I, Ludwig-Maximilians-Universität, Munich, Germany
- Walter-Brendel-Centre of Experimental Medicine, Ludwig-Maximilians-Universität, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Tobias Weinberger
- Medizinische Klinik und Poliklinik I, Ludwig-Maximilians-Universität, Munich, Germany
- Walter-Brendel-Centre of Experimental Medicine, Ludwig-Maximilians-Universität, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Vanessa Philippi
- Medizinische Klinik und Poliklinik I, Ludwig-Maximilians-Universität, Munich, Germany
- Walter-Brendel-Centre of Experimental Medicine, Ludwig-Maximilians-Universität, Munich, Germany
| | - Konstantin Stark
- Medizinische Klinik und Poliklinik I, Ludwig-Maximilians-Universität, Munich, Germany
- Walter-Brendel-Centre of Experimental Medicine, Ludwig-Maximilians-Universität, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Andreas Ehrlich
- Medizinische Klinik und Poliklinik I, Ludwig-Maximilians-Universität, Munich, Germany
- Walter-Brendel-Centre of Experimental Medicine, Ludwig-Maximilians-Universität, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Joachim Pircher
- Medizinische Klinik und Poliklinik I, Ludwig-Maximilians-Universität, Munich, Germany
- Walter-Brendel-Centre of Experimental Medicine, Ludwig-Maximilians-Universität, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Ildiko Konrad
- Medizinische Klinik und Poliklinik I, Ludwig-Maximilians-Universität, Munich, Germany
- Walter-Brendel-Centre of Experimental Medicine, Ludwig-Maximilians-Universität, Munich, Germany
| | - Paul Oberdieck
- Medizinische Klinik und Poliklinik I, Ludwig-Maximilians-Universität, Munich, Germany
- Walter-Brendel-Centre of Experimental Medicine, Ludwig-Maximilians-Universität, Munich, Germany
| | - Anna Titova
- Medizinische Klinik und Poliklinik I, Ludwig-Maximilians-Universität, Munich, Germany
- Walter-Brendel-Centre of Experimental Medicine, Ludwig-Maximilians-Universität, Munich, Germany
| | - Qendresa Hoti
- Medizinische Klinik und Poliklinik I, Ludwig-Maximilians-Universität, Munich, Germany
- Walter-Brendel-Centre of Experimental Medicine, Ludwig-Maximilians-Universität, Munich, Germany
| | - Irene Schubert
- Medizinische Klinik und Poliklinik I, Ludwig-Maximilians-Universität, Munich, Germany
- Walter-Brendel-Centre of Experimental Medicine, Ludwig-Maximilians-Universität, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Kyle R. Legate
- Medizinische Klinik und Poliklinik I, Ludwig-Maximilians-Universität, Munich, Germany
- Department of Applied Physics, Center for NanoSciences, Ludwig-Maximilians-Universität, Munich, Germany
| | - Nicole Urtz
- Medizinische Klinik und Poliklinik I, Ludwig-Maximilians-Universität, Munich, Germany
- Walter-Brendel-Centre of Experimental Medicine, Ludwig-Maximilians-Universität, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Michael Lorenz
- Medizinische Klinik und Poliklinik I, Ludwig-Maximilians-Universität, Munich, Germany
- Walter-Brendel-Centre of Experimental Medicine, Ludwig-Maximilians-Universität, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Jaroslav Pelisek
- Department of Vascular and Endovascular Surgery, Klinikum rechts der Isar der Technischen Universität, Munich, Germany
| | - Steffen Massberg
- Medizinische Klinik und Poliklinik I, Ludwig-Maximilians-Universität, Munich, Germany
- Walter-Brendel-Centre of Experimental Medicine, Ludwig-Maximilians-Universität, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Marie-Luise von Brühl
- Medizinische Klinik und Poliklinik I, Ludwig-Maximilians-Universität, Munich, Germany
- Walter-Brendel-Centre of Experimental Medicine, Ludwig-Maximilians-Universität, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Christian Schulz
- Medizinische Klinik und Poliklinik I, Ludwig-Maximilians-Universität, Munich, Germany
- Walter-Brendel-Centre of Experimental Medicine, Ludwig-Maximilians-Universität, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
- * E-mail:
| |
Collapse
|
459
|
Ortmann W, Kolaczkowska E. Age is the work of art? Impact of neutrophil and organism age on neutrophil extracellular trap formation. Cell Tissue Res 2017; 371:473-488. [PMID: 29250748 PMCID: PMC5820386 DOI: 10.1007/s00441-017-2751-4] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Accepted: 11/21/2017] [Indexed: 12/15/2022]
Abstract
Neutrophil extracellular traps or NETs are released by highly activated neutrophils in response to infectious agents, sterile inflammation, autoimmune stimuli and cancer. In the cells, the nuclear envelop disintegrates and decondensation of chromatin occurs that depends on peptidylarginine deiminase 4 (PAD4) and neutrophil elastase (NE). Subsequently, proteins from neutrophil granules (e.g., NE, lactoferrin and myeloperoxidase) and the nucleus (histones) bind to decondensed DNA and the whole structure is ejected from the cell. The DNA decorated with potent antimicrobials and proteases can act to contain dissemination of infection and in sterile inflammation NETs were shown to degrade cytokines and chemokines via serine proteases. On the other hand, overproduction of NETs, or their inadequate removal and prolonged presence in vasculature or tissues, can lead to bystander damage or even initiation of diseases. Considering the pros and cons of NET formation, it is of relevance if the stage of neutrophil maturation (immature, mature and senescent cells) affects the capacity to produce NETs as the cells of different age-related phenotypes dominate in given (pathological) conditions. Moreover, the immune system of neonates and elderly individuals is weaker than in adulthood. Is the same pattern followed when it comes to NETs? The overall importance of individual and neutrophil age on the capacity to release NETs is reviewed in detail and the significance of these facts is discussed.
Collapse
Affiliation(s)
- Weronika Ortmann
- Department of Evolutionary Immunology, Institute of Zoology and Biomedical Research, Jagiellonian University, ul. Gronostajowa 9, 30-387, Krakow, Poland
| | - Elzbieta Kolaczkowska
- Department of Evolutionary Immunology, Institute of Zoology and Biomedical Research, Jagiellonian University, ul. Gronostajowa 9, 30-387, Krakow, Poland.
| |
Collapse
|
460
|
Barliya T, Dardik R, Nisgav Y, Dachbash M, Gaton D, Kenet G, Ehrlich R, Weinberger D, Livnat T. Possible involvement of NETosis in inflammatory processes in the eye: Evidence from a small cohort of patients. Mol Vis 2017; 23:922-932. [PMID: 29296072 PMCID: PMC5741378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Accepted: 12/11/2017] [Indexed: 11/23/2022] Open
Abstract
PURPOSE To evaluate whether NETosis is involved in cytokine-induced ocular inflammation and to track neutrophil extracellular traps (NET) complexes in patients with proliferative diabetic retinopathy (PDR). METHODS For the animal model, the eyes of C57BL/6J mice were intravitreally injected with interleukin-8 (IL-8), tumor necrosis factor alpha (TNF-α), or saline. Histology and immunofluorescence staining for CD11b, neutrophil elastase (NE), myeloperoxidase (MPO), citrullinated histone 3 (H3Cit), and net-like structure were performed. Vitreous samples were collected from patients with PDR; the PDR1 group had no need for repeated surgical intervention, and the PDR2 group had repeated vitreous bleeding or other complication and controls. Levels of MPO, H3Cit-MPO, and NE-MPO complex were measured with enzyme-linked immunosorbent assay (ELISA). RESULTS Massive influx of CD11+ inflammatory cells, involving the anterior and posterior chambers, was observed in the murine eyes 24 h after the IL-8 or TNF-α injections. Cells excreted to their surroundings an extracellular net-like structure positive for NE, MPO, and H3Cit. H3Cit staining was abolished with the DNase I treatment, indicating the presence of extracellular DNA in the net-like structures. The vitreous samples of the patients with PDR2 contained statistically significantly higher levels of MPO (173±230) compared to those of the patients with PDR1 (12.0±33.0, p<0.05) or the controls (0.00, p<0.01). The levels of H3Cit-MPO and NE-MPO complexes were also statistically significantly higher in the patients with PDR2 (776.0±1274, 573.0±911.0, respectively) compared to those in the patients with PDR1 (0, p<0.05) and the controls (0, p<0.05). CONCLUSIONS This study showed the existence of NETosis in cytokine-induced ocular inflammation in a mouse model and human samples. Furthermore, the extent of NET complex formation was higher in a subset of patients who exhibited more complicated PDR.
Collapse
Affiliation(s)
- Tilda Barliya
- Laboratory of Eye research Felsenstein Medical Research Center (FMRC), Rabin Medical Center, Petah Tikva, Israel
| | - Rima Dardik
- The Israeli National Hemophilia Center, Sheba Medical Center, Tel Hashomer, Israel
| | - Yael Nisgav
- Laboratory of Eye research Felsenstein Medical Research Center (FMRC), Rabin Medical Center, Petah Tikva, Israel
| | - Mor Dachbash
- Laboratory of Eye research Felsenstein Medical Research Center (FMRC), Rabin Medical Center, Petah Tikva, Israel
| | - Dan Gaton
- Division of Ophthalmology, Rabin Medical Center- Beilinson campus, Petah Tikva, Israel,Sackler School of Medicine, Tel-Aviv University, Israel
| | - Gili Kenet
- The Israeli National Hemophilia Center, Sheba Medical Center, Tel Hashomer, Israel,Sackler School of Medicine, Tel-Aviv University, Israel
| | - Rita Ehrlich
- Division of Ophthalmology, Rabin Medical Center- Beilinson campus, Petah Tikva, Israel,Sackler School of Medicine, Tel-Aviv University, Israel
| | - Dov Weinberger
- Laboratory of Eye research Felsenstein Medical Research Center (FMRC), Rabin Medical Center, Petah Tikva, Israel,Division of Ophthalmology, Rabin Medical Center- Beilinson campus, Petah Tikva, Israel,Sackler School of Medicine, Tel-Aviv University, Israel
| | - Tami Livnat
- Laboratory of Eye research Felsenstein Medical Research Center (FMRC), Rabin Medical Center, Petah Tikva, Israel,The Israeli National Hemophilia Center, Sheba Medical Center, Tel Hashomer, Israel,Sackler School of Medicine, Tel-Aviv University, Israel
| |
Collapse
|
461
|
Hayashi H, Cherpokova D, Martinod K, Witsch T, Wong SL, Gallant M, Cifuni SM, Guarente LP, Wagner DD. Sirt3 deficiency does not affect venous thrombosis or NETosis despite mild elevation of intracellular ROS in platelets and neutrophils in mice. PLoS One 2017; 12:e0188341. [PMID: 29236713 PMCID: PMC5728566 DOI: 10.1371/journal.pone.0188341] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Accepted: 11/06/2017] [Indexed: 11/23/2022] Open
Abstract
Inflammation is a common denominator in chronic diseases of aging. Yet, how inflammation fuels these diseases remains unknown. Neutrophils are the primary leukocytes involved in the early phase of innate immunity and inflammation. As part of their anti-microbial defense, neutrophils form extracellular traps (NETs) by releasing decondensed chromatin lined with cytotoxic proteins. NETs have been shown to induce tissue injury and thrombosis. Here, we demonstrated that Sirt3, a nicotinamide adenine dinucleotide (NAD+)-dependent protein deacetylase, an enzyme linked to human longevity, was expressed in mouse neutrophils and platelets. Using Sirt3-/- mice as a model of accelerated aging, we investigated the effects of Sirt3 deficiency on NETosis and platelet function, aiming to detect enhancement of thrombosis. More mitochondrial reactive oxygen species (ROS) were generated in neutrophils and platelets of Sirt3-/- mice compared to WT, when stimulated with a low concentration of phorbol 12-myristate 13-acetate (PMA) and a high concentration of thrombin, respectively. There were no differences in in vitro NETosis, with or without stimulation. Platelet aggregation was mildly augmented in Sirt3-/- mice compared to WT mice, when stimulated with a low concentration of collagen. The effect of Sirt3 deficiency on platelet and neutrophil activation in vivo was examined by the venous thrombosis model of inferior vena cava stenosis. Elevation of plasma DNA concentration was observed after stenosis in both genotypes, but no difference was shown between the two genotypes. The systemic response to thrombosis was enhanced in Sirt3-/- mice with significantly elevated neutrophil count and reduced platelet count. However, no differences were observed in incidence of thrombus formation, thrombus weight and thrombin-antithrombin complex generation between WT and Sirt3-/- mice. We conclude that Sirt3 does not considerably impact NET formation, platelet function, or venous thrombosis in healthy young mice.
Collapse
Affiliation(s)
- Hideki Hayashi
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, Massachusetts, United States of America
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Deya Cherpokova
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, Massachusetts, United States of America
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Kimberly Martinod
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, Massachusetts, United States of America
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Thilo Witsch
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, Massachusetts, United States of America
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Siu Ling Wong
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, Massachusetts, United States of America
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Maureen Gallant
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, Massachusetts, United States of America
| | - Stephen M. Cifuni
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, Massachusetts, United States of America
| | - Leonard P. Guarente
- Department of Biology, Paul F. Glenn Center for the Science of Aging, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| | - Denisa D. Wagner
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, Massachusetts, United States of America
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, United States of America
- Division of Hematology/Oncology, Boston Children’s Hospital, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
462
|
Date K, Ettelaie C, Maraveyas A. Tissue factor-bearing microparticles and inflammation: a potential mechanism for the development of venous thromboembolism in cancer. J Thromb Haemost 2017; 15:2289-2299. [PMID: 29028284 DOI: 10.1111/jth.13871] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Indexed: 12/31/2022]
Abstract
Cancer is associated with an increased risk of venous thromboembolism (VTE); the exact mechanisms for the induction of VTE remain to be fully elucidated, but it is widely acknowledged that tissue factor (TF)-bearing microparticles (TF-MPs) may play a significant role. However, TF-MPs have yet to be accepted as a genuine biomarker for cancer-associated VTE, as the presence of elevated TF-MP levels is not always accompanied by thrombosis; interestingly, in certain cases, particularly in pancreatic cancer, VTE seems to be more likely in the context of acute inflammation. Although several potential mechanisms for the development of VTE in cancer have been postulated, this review explores the homeostatic disruption of TF-MPs, as the main reservoir of bloodborne TF, in the context of cancer and inflammation, and considers the abrogated responses of the activated endothelium and mononuclear phagocyte system in mediating this disruption.
Collapse
Affiliation(s)
- K Date
- Hull York Medical School, University of Hull, Hull, UK
| | - C Ettelaie
- School of Life Sciences, University of Hull, Hull, UK
| | - A Maraveyas
- Hull York Medical School, University of Hull, Hull, UK
- Queen's Centre for Oncology and Haematology, Castle Hill Hospital, Cottingham, UK
| |
Collapse
|
463
|
Matan M, King D, Peled E, Ackerman S, Bar-Lavi Y, Brenner B, Nadir Y. Heparanase level and procoagulant activity are reduced in severe sepsis. Eur J Haematol 2017; 100:182-188. [PMID: 29120525 DOI: 10.1111/ejh.12997] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/02/2017] [Indexed: 12/19/2022]
Abstract
BACKGROUND During severe sepsis, levels and activity of all coagulation proteins are reduced. Heparanase is implicated in angiogenesis and tumor progression. We previously demonstrated that heparanase also affected the hemostatic system. It forms a complex and increases the activity of the blood coagulation initiator tissue factor. AIM To evaluate heparanase levels and procoagulant activity as predictors of sepsis severity. MATERIALS AND METHODS Twenty-one patients with non-trauma, non-surgical sepsis admitted to the intensive care unit and 35 controls were recruited. Plasma samples were drawn from the study participants on days 1 and 7 following admission. RESULTS Heparanase levels and procoagulant activity on day 1 were significantly reduced in patients compared to controls (P < .0001, P < .0001, respectively). Day 1 heparanase procoagulant activity ≥350 ng/mL yielded a negative predictive value for severe sepsis of 89%. Additionally, heparanase procoagulant activity on day 7 correlated with the change in the APACHE score between days 1 and 7 (r = .66, P = .007). CONCLUSIONS Heparanase procoagulant activity decreases during sepsis and returns to normal levels as soon as the patient recovers. Hence, it can be potentially used to predict the risk of severe sepsis. These findings need to be further explored in large-scale studies.
Collapse
Affiliation(s)
- Moshe Matan
- Intensive Care Unit, Rambam Health Care Campus, The Bruce Rappaport Faculty of Medicine, The Technion, Haifa, Israel
| | - Daniel King
- Intensive Care Unit, Rambam Health Care Campus, The Bruce Rappaport Faculty of Medicine, The Technion, Haifa, Israel
| | - Eli Peled
- Department of Orthopedic B, Rambam Health Care Campus, The Bruce Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Shanny Ackerman
- Thrombosis and Hemostasis Unit, Rambam Health Care Campus, The Bruce Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Yaron Bar-Lavi
- Intensive Care Unit, Rambam Health Care Campus, The Bruce Rappaport Faculty of Medicine, The Technion, Haifa, Israel
| | - Benjamin Brenner
- Thrombosis and Hemostasis Unit, Rambam Health Care Campus, The Bruce Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Yona Nadir
- Thrombosis and Hemostasis Unit, Rambam Health Care Campus, The Bruce Rappaport Faculty of Medicine, Technion, Haifa, Israel
| |
Collapse
|
464
|
Platelet-neutrophil interactions as drivers of inflammatory and thrombotic disease. Cell Tissue Res 2017; 371:567-576. [PMID: 29178039 PMCID: PMC5820397 DOI: 10.1007/s00441-017-2727-4] [Citation(s) in RCA: 160] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Accepted: 10/27/2017] [Indexed: 02/07/2023]
Abstract
Neutrophils are well known for their role in infection and inflammatory disease and are first responders at sites of infection or injury. Platelets have an established role in hemostasis and thrombosis and are first responders at sites of vascular damage. However, neutrophils are increasingly recognized for their role in thrombosis, while the immunemodulatory properties of platelets are being increasingly studied. Platelets and neutrophils interact during infection, inflammation and thrombosis and modulate each other’s functions. This review will discuss the consequences of platelet–neutrophil interactions in infection, thrombosis, atherosclerosis and tissue injury and repair.
Collapse
|
465
|
Ayoade F, Cotelingam J, Joel Chandranesan AS. Disseminated Mycobacterium avium-Intracellulare Complex Infection Presenting With Disseminated Intravascular Coagulation in an AIDS Patient. J Investig Med High Impact Case Rep 2017; 5:2324709617740904. [PMID: 29164156 PMCID: PMC5692144 DOI: 10.1177/2324709617740904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 09/20/2017] [Accepted: 09/24/2017] [Indexed: 12/01/2022] Open
Abstract
Disseminated Mycobacterium avium-intracellulare complex (MAC) infection is one of the relatively common opportunistic infections seen in severely immunocompromised AIDS patients. A constellation of clinical, laboratory, and pathological features involving multiple organ systems are often present in disseminated MAC infection but disseminated intravascular coagulation (DIC) has not been previously described in association with this condition. To our knowledge, this is the first reported case of DIC complicating disseminated MAC infection in an AIDS patient. In this article, we present the case of a 33-year-old AIDS patient with high viral load, CD4 lymphocyte count of 1/mm3, who presented with nonspecific symptoms, anemia, thrombocytopenia, and increased lactate dehydrogenase, alkaline phosphatase, and ferritin. She also had abnormal coagulation parameters and features compatible with chronic DIC. Bone marrow biopsy assisted in making the correct diagnosis. She also later grew MAC from blood and sputum cultures. There were no other factors identified after a complete workup to explain DIC in this patient. After commencement of appropriate MAC therapy, she initially had a good response with some improvement of her coagulation parameters. Few months later, however, probably attributable to poor medication compliance, her condition deteriorated with development of thromboembolism, full-fledged DIC, sepsis, and an eventual fatal outcome. This case illustrates the importance of including disseminated MAC in the differential diagnosis of DIC in an AIDS patient.
Collapse
Affiliation(s)
- Folusakin Ayoade
- Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - James Cotelingam
- Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | | |
Collapse
|
466
|
Hosseini E, Ghasemzadeh M. Intravascular leukocyte migration through platelet thrombi: directing leukocytes to sites of vascular injury. Thromb Haemost 2017; 113:1224-35. [DOI: 10.1160/th14-08-0662] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2014] [Accepted: 01/13/2015] [Indexed: 12/15/2022]
Abstract
SummaryLeukocytes recruitment to thrombi supports an intimate cellular interaction leading to the enhancement of pro-coagulant functions and pro-inflammatory responses at site of vascular injury. Recent observations of neutrophil extracellular traps (NETs) formation and its mutual reactions with platelet thrombi adds more clinical interest to the growing body of knowledge in the field of platelet-leukocyte crosstalk. However, having considered thrombus as a barrier between leukocytes and injured endothelium, the full inflammatory roles of these cells during thrombosis is still ill defined. The most recent observation of neutrophils migration into the thrombi is a phenomenon that highlights the inflammatory functions of leukocytes at the site of injury. It has been hypothesised that leukocytes migration might be associated with the conveyance of highly reactive pro-inflammatory and/or procoagulant mediators to sites of vascular injury. In addition, the evidence of neutrophils migration into arterial thrombi following traumatic and ischaemia-reperfusion injury highlights the already described role of these cells in atherosclerosis. Regardless of the mechanisms behind leukocyte migration, whether these migrated cells benefit normal homeostasis by their involvement in wound healing and vascular rebuilding or they increase unwilling inflammatory responses, could be of interest for future researches that provide new insight into biological importance of leukocyte recruitment to thrombi.
Collapse
|
467
|
Therapeutic targeting of extracellular DNA improves the outcome of intestinal ischemic reperfusion injury in neonatal rats. Sci Rep 2017; 7:15377. [PMID: 29133856 PMCID: PMC5684414 DOI: 10.1038/s41598-017-15807-6] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 11/02/2017] [Indexed: 12/15/2022] Open
Abstract
Thrombosis and inflammation cooperate in the development of intestinal infarction. Recent studies suggest that extracellular DNA released by damaged cells or neutrophils in form of extracellular traps (NETs) contributes to organ damage in experimental models of ischemia-reperfusion injury. Here we compared the therapeutic effects of targeting fibrin or extracellular DNA in intestinal infarction after midgut volvulus in rats. Following iatrogenic midgut volvulus induction for 3 hours, we treated animals with a combination of tissue plasminogen activator (tPA) and low molecular weight heparin (LMWH) to target fibrin or with DNase1 to degrade extracellular DNA. The therapeutic effects of tPA/LMWH and DNase1 were analyzed after 7 days. We observed that both therapeutic interventions ameliorated tissue injury, apoptosis, and oxidative stress in the intestine. DNase1, but not tPA/LMWH, reduced intestinal neutrophil infiltration and histone-myeloperoxidase-complexes, a surrogate marker of NETs, in circulation. Importantly, tPA/LMWH, but not DNase1, interfered with hemostasis as evidenced by a prolonged tail bleeding time. In conclusion, our data suggest that the therapeutic targeting of fibrin and extracellular DNA improves the outcome of midgut volvulus in rats. DNase1 therapy reduces the inflammatory response including NETs without increasing the risk of bleeding. Thus, targeting of extracellular DNA may provide a safe therapy for patients with intestinal infarction in future.
Collapse
|
468
|
Tripodi A. Detection of procoagulant imbalance. Thromb Haemost 2017; 117:830-836. [DOI: 10.1160/th16-10-0806] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Accepted: 01/25/2017] [Indexed: 01/14/2023]
Abstract
SummaryEach individual possesses his/her own endogenous-thrombin-potential (ETP) (i. e. the ability to generate thrombin) which depends on the relative strength of the pro- and anticoagulant drivers operating in plasma. This ability depends in turn on the clinical conditions in which the balance between the two drivers is variably affected. One of the major determinants of this balance is the factor (F)VIII-protein C(PC) axis and its effect can be conveniently explored by the thrombin generation procedures with results expressed as ETP ratio with/without thrombomodulin (TM) (ETP-TM ratio). Furthermore, owing to the many feedback mechanisms mediated by thrombin (e. g. activation of PC, FXI, FV, FVIII, platelets etc.) it is also possible that any perturbation of the balance between pro- and anticoagulants that may occur in plasma even outside the FVIII-PC axis could result in an increased ETPTM ratio and therefore may suggest a procoagulant imbalance. Indeed, other non-coagulation moieties (e. g. microparticles, neutrophil extracellular traps, pro-inflammatory cytokines and others) circulating in blood of patients with various clinical conditions may also contribute to the procoagulant imbalance even when FVIII and/or PC are apparently normal. It can be postulated that dual ETP measurements performed in the presence and absence of TM with results expressed as their ratio may be the candidate procedure to detect subtle procoagulant imbalance in many clinical conditions characterised by an increased risk of thromboembolism. This article aimed at reviewing the clinical conditions in which evidence for the value of the ETP-TM ratio has been provided.
Collapse
|
469
|
Amulic B, Knackstedt SL, Abu Abed U, Deigendesch N, Harbort CJ, Caffrey BE, Brinkmann V, Heppner FL, Hinds PW, Zychlinsky A. Cell-Cycle Proteins Control Production of Neutrophil Extracellular Traps. Dev Cell 2017; 43:449-462.e5. [DOI: 10.1016/j.devcel.2017.10.013] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Revised: 07/12/2017] [Accepted: 10/09/2017] [Indexed: 01/09/2023]
|
470
|
Gorog DA, Fayad ZA, Fuster V. Arterial Thrombus Stability. J Am Coll Cardiol 2017; 70:2036-2047. [DOI: 10.1016/j.jacc.2017.08.065] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Revised: 08/15/2017] [Accepted: 08/31/2017] [Indexed: 01/27/2023]
|
471
|
Abstract
PURPOSE OF REVIEW The present review explores the mechanisms of superficial intimal erosion, a common cause of thrombotic complications of atherosclerosis. RECENT FINDINGS Human coronary artery atheroma that give rise to thrombosis because of erosion differ diametrically from those associated with fibrous cap rupture. Eroded lesions characteristically contain few inflammatory cells, abundant extracellular matrix, and neutrophil extracellular traps (NETs). Innate immune mechanisms such as engagement of Toll-like receptor 2 (TLR2) on cultured endothelial cells can impair their viability, attachment, and ability to recover a wound. Hyaluronan fragments may serve as endogenous TLR2 ligands. Mouse experiments demonstrate that flow disturbance in arteries with neointimas tailored to resemble features of human eroded plaques disturbs endothelial cell barrier function, impairs endothelial cell viability, recruits neutrophils, and provokes endothelial cells desquamation, NET formation, and thrombosis in a TLR2-dependent manner. SUMMARY Mechanisms of erosion have received much less attention than those that provoke plaque rupture. Intensive statin treatment changes the characteristic of plaques that render them less susceptible to rupture. Thus, erosion may contribute importantly to the current residual burden of risk. Understanding the mechanisms of erosion may inform the development and deployment of novel therapies to combat the remaining atherothrombotic risk in the statin era.
Collapse
Affiliation(s)
- Thibaut Quillard
- Department of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Grégory Franck
- Department of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Thomas Mawson
- Department of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Eduardo Folco
- Department of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Peter Libby
- Department of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| |
Collapse
|
472
|
Knight JS, Meng H, Coit P, Yalavarthi S, Sule G, Gandhi AA, Grenn RC, Mazza LF, Ali RA, Renauer P, Wren JD, Bockenstedt PL, Wang H, Eitzman DT, Sawalha AH. Activated signature of antiphospholipid syndrome neutrophils reveals potential therapeutic target. JCI Insight 2017; 2:93897. [PMID: 28931754 DOI: 10.1172/jci.insight.93897] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Accepted: 08/15/2017] [Indexed: 01/05/2023] Open
Abstract
Antiphospholipid antibodies, present in one-third of lupus patients, increase the risk of thrombosis. We recently reported a key role for neutrophils - neutrophil extracellular traps (NETs), in particular - in the thrombotic events that define antiphospholipid syndrome (APS). To further elucidate the role of neutrophils in APS, we performed a comprehensive transcriptome analysis of neutrophils isolated from patients with primary APS. Moreover, APS-associated venous thrombosis was modeled by treating mice with IgG prepared from APS patients, followed by partial restriction of blood flow through the inferior vena cava. In patients, APS neutrophils demonstrated a proinflammatory signature with overexpression of genes relevant to IFN signaling, cellular defense, and intercellular adhesion. For in vivo studies, we focused on P-selectin glycoprotein ligand-1 (PSGL-1), a key adhesion molecule overexpressed in APS neutrophils. The introduction of APS IgG (as compared with control IgG) markedly potentiated thrombosis in WT mice, but not PSGL-1-KOs. PSGL-1 deficiency was also associated with reduced leukocyte vessel wall adhesion and NET formation. The thrombosis phenotype was restored in PSGL-1-deficient mice by infusion of WT neutrophils, while an anti-PSGL-1 monoclonal antibody inhibited APS IgG-mediated thrombosis in WT mice. PSGL-1 represents a potential therapeutic target in APS.
Collapse
Affiliation(s)
- Jason S Knight
- Division of Rheumatology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - He Meng
- Division of Rheumatology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Patrick Coit
- Division of Rheumatology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Srilakshmi Yalavarthi
- Division of Rheumatology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Gautam Sule
- Division of Rheumatology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Alex A Gandhi
- Division of Rheumatology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Robert C Grenn
- Division of Rheumatology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Levi F Mazza
- Division of Rheumatology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Ramadan A Ali
- Division of Rheumatology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Paul Renauer
- Division of Rheumatology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Jonathan D Wren
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA.,Department of Biochemistry and Molecular Biology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | | | - Hui Wang
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Daniel T Eitzman
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Amr H Sawalha
- Division of Rheumatology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA.,Center for Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
473
|
Hattori Y, Hattori K, Suzuki T, Matsuda N. Recent advances in the pathophysiology and molecular basis of sepsis-associated organ dysfunction: Novel therapeutic implications and challenges. Pharmacol Ther 2017; 177:56-66. [DOI: 10.1016/j.pharmthera.2017.02.040] [Citation(s) in RCA: 99] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
474
|
|
475
|
Kaufman T, Magosevich D, Moreno MC, Guzman MA, D'Atri LP, Carestia A, Fandiño ME, Fondevila C, Schattner M. Nucleosomes and neutrophil extracellular traps in septic and burn patients. Clin Immunol 2017; 183:254-262. [PMID: 28863968 DOI: 10.1016/j.clim.2017.08.014] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Revised: 07/28/2017] [Accepted: 08/28/2017] [Indexed: 11/30/2022]
Abstract
NETosis is a host defense mechanism associated with inflammation and tissue damage. Experimental models show that platelets and von Willebrand factor (VWF) are key elements for intravascular NETosis. We determined NETosis in septic and burn patients at 1 and 4days post-admission (dpa). Nucleosomes were elevated in patients. In septics, they correlated with Human Neutrophil Elastase (HNE)-DNA complexes and SOFA score at 1dpa, and were associated with mortality. Patient's neutrophils had spontaneous NETosis and were unresponsive to stimulation. Although platelet P-selectin and TNF-α were increased in both groups, higher platelet TLR-4 expression, VWF levels and IL-6 were found in septics at 1dpa. Neither platelet activation markers nor cytokines correlated with nucleosomes or HNE-DNA. Nucleosomes could be indicators of organ damage and predictors of mortality in septic but not in burn patients. Platelet activation, VWF and cytokines do not appear to be key mediators of NETosis in these patient groups.
Collapse
Affiliation(s)
- Tomás Kaufman
- Laboratory of Experimental Thrombosis, Institute of Experimental Medicine-CONICET, National Academy of Medicine, José Andrés Pacheco de Melo 3081, Buenos Aires, Argentina
| | - Débora Magosevich
- Sagrado Corazón Clinic, Bartolomé Mitre 1955, Buenos Aires, Argentina
| | | | | | - Lina Paola D'Atri
- Laboratory of Experimental Thrombosis, Institute of Experimental Medicine-CONICET, National Academy of Medicine, José Andrés Pacheco de Melo 3081, Buenos Aires, Argentina
| | - Agostina Carestia
- Laboratory of Experimental Thrombosis, Institute of Experimental Medicine-CONICET, National Academy of Medicine, José Andrés Pacheco de Melo 3081, Buenos Aires, Argentina
| | | | | | - Mirta Schattner
- Laboratory of Experimental Thrombosis, Institute of Experimental Medicine-CONICET, National Academy of Medicine, José Andrés Pacheco de Melo 3081, Buenos Aires, Argentina.
| |
Collapse
|
476
|
Laridan E, Denorme F, Desender L, François O, Andersson T, Deckmyn H, Vanhoorelbeke K, De Meyer SF. Neutrophil extracellular traps in ischemic stroke thrombi. Ann Neurol 2017; 82:223-232. [PMID: 28696508 DOI: 10.1002/ana.24993] [Citation(s) in RCA: 357] [Impact Index Per Article: 44.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Revised: 07/04/2017] [Accepted: 07/06/2017] [Indexed: 12/11/2022]
Abstract
OBJECTIVE Neutrophil extracellular traps (NETs) have been shown to promote thrombus formation. Little is known about the exact composition of thrombi that cause ischemic stroke. In particular, no information is yet available on the presence of NETs in cerebral occlusions. Such information is, however, essential to improve current thrombolytic therapy with tissue plasminogen activator (t-PA). This study aimed at investigating the presence of neutrophils and more specifically NETs in ischemic stroke thrombi. METHODS Sixty-eight thrombi retrieved from ischemic stroke patients undergoing endovascular treatment were characterized by immunostaining using neutrophil markers (CD66b and neutrophil elastase) and NET markers (citrullinated histone H3 [H3Cit] and extracellular DNA). Neutrophils and NETs were quantified. In addition, extracellular DNA was targeted by performing ex vivo lysis of retrieved thrombi with DNase 1 and t-PA. RESULTS Neutrophils were detected extensively throughout all thrombi. H3Cit, a hallmark of NETs, was observed in almost all thrombi. H3Cit-positive area varied up to 13.45% of total thrombus area. Colocalization of H3Cit with extracellular DNA released from neutrophils confirmed the specific presence of NETs. H3Cit was more abundant in thrombi of cardioembolic origin compared to other etiologies. Older thrombi contained significantly more neutrophils and H3Cit compared to fresh thrombi. Interestingly, ex vivo lysis of patient thrombi was more successful when adding DNase 1 to standard t-PA. INTERPRETATION Neutrophils and NETs form important constituents of cerebral thrombi. Targeting of NETs with DNase 1 might have prothrombolytic potential in treatment of acute ischemic stroke. Ann Neurol 2017;82:223-232.
Collapse
Affiliation(s)
- Elodie Laridan
- Laboratory for Thrombosis Research, KU Leuven, Campus Kulak Kortrijk, Kortrijk, Belgium
| | - Frederik Denorme
- Laboratory for Thrombosis Research, KU Leuven, Campus Kulak Kortrijk, Kortrijk, Belgium
| | - Linda Desender
- Laboratory for Thrombosis Research, KU Leuven, Campus Kulak Kortrijk, Kortrijk, Belgium
| | | | - Tommy Andersson
- Department of Medical Imaging, AZ Groeninge, Kortrijk, Belgium.,Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Hans Deckmyn
- Laboratory for Thrombosis Research, KU Leuven, Campus Kulak Kortrijk, Kortrijk, Belgium
| | - Karen Vanhoorelbeke
- Laboratory for Thrombosis Research, KU Leuven, Campus Kulak Kortrijk, Kortrijk, Belgium
| | - Simon F De Meyer
- Laboratory for Thrombosis Research, KU Leuven, Campus Kulak Kortrijk, Kortrijk, Belgium
| |
Collapse
|
477
|
|
478
|
Korkmaz HI, Ulrich MMW, Vogels S, de Wit T, van Zuijlen PPM, Krijnen PAJ, Niessen HWM. Neutrophil extracellular traps coincide with a pro-coagulant status of microcirculatory endothelium in burn wounds. Wound Repair Regen 2017; 25:609-617. [PMID: 28727215 DOI: 10.1111/wrr.12560] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Accepted: 06/19/2017] [Indexed: 01/31/2023]
Abstract
Burn-induced tissue loss is partly related to secondary expansion of necrosis into vital dermis neighboring the initial burn injury. An important factor herein is the severe loss of perfusion of the burn wound, probably caused by microvascular damage induced by the intense local inflammatory responses as well as burn-induced hypercoagulation. We hypothesize that the formation of neutrophilic extracellular traps (NETs) play an important role in this. The purpose of this study was to investigate postburn intravascular thrombosis, NETs formation and the coagulant state in the microvasculature of burns in both animal models and patients. We used two in vivo burn wound models: rats and pigs. In rats, the entire wound was excised at day 14 postburn and in pigs burn wound biopsies were collected at different time points up to 60 days postburn. To confirm the data in patients, eschar from the burn wound was obtained from burn wound patients at different time points after wounding. The number of intravascular thrombi, the presence of intravascular NETs and the number of tissue factor (TF) positive blood vessels in the burn wound was determined. In rats, a significant increase in intravascular thrombi and TF expression was observed 14 days postburn, that in majority coincided with NETs. In pigs, a significant increase in intravascular thrombi and TF expression was found over time up to 60 days postburn, that in majority coincided with NETs too. Also in eschar of burn wound patients, a significant increase in intravascular thrombi was noted, that in majority coincided with NETs, already 0.5 days postburn and remained elevated up to 46 days postburn. This study shows the presence of NETosis in microcirculatory thrombosis of burn wounds and a switch in the microcirculatory endothelium toward a procoagulant phenotype.
Collapse
Affiliation(s)
- H Ibrahim Korkmaz
- Department of Pathology, MOVE Research Institute, VU University Medical Center, Amsterdam, The Netherlands.,Institute of Cardiovascular Research (ICaR-VU), MOVE Research Institute, VU University Medical Center, Amsterdam, The Netherlands
| | - Magda M W Ulrich
- Department of Molecular Cell Biology and Immunology, MOVE Research Institute, VU University Medical Center, Amsterdam, The Netherlands.,Department of Plastic, Reconstructive and Hand Surgery, MOVE Research Institute, VU University Medical Center, Amsterdam, The Netherlands.,Association of Dutch Burn Centers (ADBC), Beverwijk, The Netherlands
| | - Sanne Vogels
- Department of Pathology, MOVE Research Institute, VU University Medical Center, Amsterdam, The Netherlands
| | - Tim de Wit
- Department of Pathology, MOVE Research Institute, VU University Medical Center, Amsterdam, The Netherlands
| | - Paul P M van Zuijlen
- Department of Plastic, Reconstructive and Hand Surgery, MOVE Research Institute, VU University Medical Center, Amsterdam, The Netherlands.,Association of Dutch Burn Centers (ADBC), Beverwijk, The Netherlands.,Burn Center and Department of Plastic and Reconstructive Surgery, Red Cross Hospital, Beverwijk, The Netherlands
| | - Paul A J Krijnen
- Department of Pathology, MOVE Research Institute, VU University Medical Center, Amsterdam, The Netherlands.,Institute of Cardiovascular Research (ICaR-VU), MOVE Research Institute, VU University Medical Center, Amsterdam, The Netherlands
| | - Hans W M Niessen
- Department of Pathology, MOVE Research Institute, VU University Medical Center, Amsterdam, The Netherlands.,Institute of Cardiovascular Research (ICaR-VU), MOVE Research Institute, VU University Medical Center, Amsterdam, The Netherlands.,Cardiac Surgery, VU University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
479
|
Chabert A, Damien P, Verhoeven PO, Grattard F, Berthelot P, Zeni F, Panicot-Dubois L, Robert S, Dignat-George F, Eyraud MA, Pozzetto B, Payrastre B, Cognasse F, Garraud O, Hamzeh-Cognasse H. Acetylsalicylic acid differentially limits the activation and expression of cell death markers in human platelets exposed to Staphylococcus aureus strains. Sci Rep 2017; 7:5610. [PMID: 28717145 PMCID: PMC5514152 DOI: 10.1038/s41598-017-06024-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Accepted: 06/07/2017] [Indexed: 12/14/2022] Open
Abstract
Beyond their hemostatic functions, platelets alter their inflammatory response according to the bacterial stimulus. Staphylococcus aureus is associated with exacerbated inflammation and thrombocytopenia, which is associated with poor prognosis during sepsis. Acetylsalicylic acid and statins prevent platelet aggregation and decrease the mortality rate during sepsis. Therefore, we assessed whether these two molecules could reduce in vitro platelet activation and the inflammatory response to S. aureus. Platelets were exposed to clinical strains of S. aureus in the presence or absence of acetylsalicylic acid or fluvastatin. Platelet activation, aggregation, and release of soluble sCD62P, sCD40 Ligand, RANTES and GROα were assessed. Platelet cell death was evaluated by analyzing the mitochondrial membrane potential, phosphatidylserine exposure, platelet microparticle release and caspase-3 activation. All S. aureus strains induced platelet activation but not aggregation and decreased the platelet count, the expression of cell death markers and the release of RANTES and GROα. Acetylsalicylic acid but not fluvastatin limited platelet activation and inflammatory factor release and restored the platelet count by protecting platelets from Staphylococcus-induced expression of cell death markers. This study demonstrates that acetylsalicylic acid limits S. aureus-induced effects on platelets by reducing cell death, revealing new strategies to reduce the platelet contribution to bacteremia-associated inflammation.
Collapse
Affiliation(s)
- Adrien Chabert
- EA3064-GIMAP, Université de Lyon, 42023, Saint-Etienne, France
| | - Pauline Damien
- EA3064-GIMAP, Université de Lyon, 42023, Saint-Etienne, France
| | - Paul O Verhoeven
- EA3064-GIMAP, Université de Lyon, 42023, Saint-Etienne, France
- Laboratoire des Agents Infectieux et d'Hygiène, CHU de Saint-Etienne, 42055, Saint-Etienne, France
| | - Florence Grattard
- EA3064-GIMAP, Université de Lyon, 42023, Saint-Etienne, France
- Laboratoire des Agents Infectieux et d'Hygiène, CHU de Saint-Etienne, 42055, Saint-Etienne, France
| | - Philippe Berthelot
- EA3064-GIMAP, Université de Lyon, 42023, Saint-Etienne, France
- Laboratoire des Agents Infectieux et d'Hygiène, CHU de Saint-Etienne, 42055, Saint-Etienne, France
| | - Fabrice Zeni
- EA3064-GIMAP, Université de Lyon, 42023, Saint-Etienne, France
- Service de Réanimation polyvalente, CHU de Saint-Etienne, 42055, Saint-Etienne, France
| | | | - Stéphane Robert
- Université Aix Marseille, INSERM UMR-S1076, VRCM, Marseille, France
| | | | | | - Bruno Pozzetto
- EA3064-GIMAP, Université de Lyon, 42023, Saint-Etienne, France
- Laboratoire des Agents Infectieux et d'Hygiène, CHU de Saint-Etienne, 42055, Saint-Etienne, France
| | | | - Fabrice Cognasse
- EA3064-GIMAP, Université de Lyon, 42023, Saint-Etienne, France
- EFS Auvergne-Rhone-Alpes, 42023, Saint-Etienne, France
| | - Olivier Garraud
- EA3064-GIMAP, Université de Lyon, 42023, Saint-Etienne, France
- Institut National de la Transfusion Sanguine, 75015, Paris, France
| | | |
Collapse
|
480
|
Circulating dsDNA, endothelial injury, and complement activation in thrombotic microangiopathy and GVHD. Blood 2017; 130:1259-1266. [PMID: 28705839 DOI: 10.1182/blood-2017-05-782870] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 07/04/2017] [Indexed: 12/25/2022] Open
Abstract
Transplant-associated thrombotic microangiopathy (TA-TMA) is a common and poorly recognized complication of hematopoietic stem cell transplantation (HSCT) associated with excessive complement activation, likely triggered by endothelial injury. An important missing piece is the link between endothelial injury and complement activation. We hypothesized that neutrophil extracellular traps (NETs) mechanistically link endothelial damage with complement activation and subsequent TA-TMA. Neutrophil activation releases granule proteins together with double-stranded DNA (dsDNA) to form extracellular fibers known as NETs. NETs have been shown to activate complement and can be assessed in humans by quantification of dsDNA in serum. We measured levels of dsDNA, as a surrogate for NETs in 103 consecutive pediatric allogeneic transplant recipients at day 0, +14, +30, +60, and +100. A spike in dsDNA production around day +14 during engraftment was associated with subsequent TA-TMA development. Peak dsDNA production around day +14 was associated with interleukin-8-driven neutrophil recovery. Increased dsDNA levels at days +30, +60, and +100 were also associated with increased mortality and gastrointestinal graft-versus-host disease (GVHD). NETs may serve as a mechanistic link between endothelial injury and complement activation. NET formation may be one mechanism contributing to the clinical overlap between GVHD and TA-TMA.
Collapse
|
481
|
Meng H, Yalavarthi S, Kanthi Y, Mazza LF, Elfline MA, Luke CE, Pinsky DJ, Henke PK, Knight JS. In Vivo Role of Neutrophil Extracellular Traps in Antiphospholipid Antibody-Mediated Venous Thrombosis. Arthritis Rheumatol 2017; 69:655-667. [PMID: 27696751 DOI: 10.1002/art.39938] [Citation(s) in RCA: 156] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Accepted: 09/15/2016] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Antiphospholipid syndrome (APS) is a leading acquired cause of thrombotic events. Although antiphospholipid antibodies have been shown to promote thrombosis in mice, the role of neutrophils has not been explicitly studied. The aim of this study was to characterize neutrophils in the context of a new model of antiphospholipid antibody-mediated venous thrombosis. METHODS Mice were administered fractions of IgG obtained from patients with APS. At the same time, blood flow through the inferior vena cava was reduced by induction of stenosis. Resulting thrombi were characterized for size and neutrophil content. Circulating factors and the vessel wall were also assessed. RESULTS As measured by both thrombus weight and thrombosis frequency, mice treated with IgG from patients with APS (APS IgG) demonstrated exaggerated thrombosis as compared with control IgG-treated mice. Thrombi in mice treated with APS IgG were enriched for citrullinated histone H3 (a marker of neutrophil extracellular traps [NETs]). APS IgG-treated mice also demonstrated elevated levels of circulating cell-free DNA and human IgG bound to the neutrophil surface. In contrast, circulating neutrophil numbers and markers of vessel wall activation were not appreciably different between APS IgG-treated mice and control mice. Treatment with either DNase (which dissolves NETs) or a neutrophil-depleting antibody reduced thrombosis in APS IgG-treated mice to the level in control mice. CONCLUSION These data support a mechanism whereby circulating neutrophils are primed by antiphospholipid antibodies to accelerate thrombosis. This line of investigation suggests new, immunomodulatory approaches for the treatment of APS.
Collapse
Affiliation(s)
- He Meng
- University of Michigan Medical School, Ann Arbor
| | | | - Yogendra Kanthi
- University of Michigan Medical School and Ann Arbor Veterans Administration Healthcare System, Ann Arbor
| | - Levi F Mazza
- University of Michigan Medical School, Ann Arbor
| | | | | | | | | | | |
Collapse
|
482
|
Lood C, Hughes GC. Neutrophil extracellular traps as a potential source of autoantigen in cocaine-associated autoimmunity. Rheumatology (Oxford) 2017; 56:638-643. [PMID: 27354687 DOI: 10.1093/rheumatology/kew256] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Indexed: 11/14/2022] Open
Abstract
Objective Exposure to illicit cocaine and its frequent adulterant, levamisole, is associated with ANCAs targeting neutrophil elastase (NE), neutropenia and vasculitic/thrombotic skin purpura. The mechanisms of cocaine/levamisole-associated autoimmunity (CLAA) are unknown. The aim of this study was to assess the ability of cocaine and levamisole to induce the release of neutrophil extracellular traps (NETs), a potential source of autoantigen and tissue injury. Methods We performed quantitative and qualitative assessment of NET formation in neutrophils from healthy donors exposed to either drug in vitro . In addition, IgG from sera of individuals with CLAA (CLAA-IgG) was assessed for its ability to enhance formation of, and to bind to, drug-induced NETs. Results Both cocaine and levamisole could induce formation of NETs enriched in NE and, potentially, inflammatory mitochondrial DNA. Both drugs could also augment simultaneous release of B cell-activating factor belonging to the TNF family (BAFF). CLAA-IgG, but not IgG from healthy individuals, could potentiate drug-induced NETosis. Furthermore, CLAA-IgG, but not ANCA + control IgG, bound to drug-induced NETs in a pattern consistent with NE targeting. Conclusion Both cocaine and levamisole may contribute to the development of ANCAs by inducing release of potentially inflammatory NETs in association with NE autoantigen and BAFF. Enhancement of drug-induced NET release by CLAA-IgG provides a potential mechanism linking vasculitis/pupuric skin disease to acute drug exposure in patients with CLAA. Further study of this under-recognized form of autoimmunity will be likely to provide mechanistic insight into ANCA-associated vasculitis and other diseases associated with NETosis.
Collapse
Affiliation(s)
- Christian Lood
- Department of Medicine, Division of Rheumatology, University of Washington, Seattle, WA, USA
| | - Grant C Hughes
- Department of Medicine, Division of Rheumatology, University of Washington, Seattle, WA, USA
| |
Collapse
|
483
|
O’Connell GC, Petrone AB, Tennant CS, Lucke-Wold N, Kabbani Y, Tarabishy AR, Chantler PD, Barr TL. Circulating extracellular DNA levels are acutely elevated in ischaemic stroke and associated with innate immune system activation. Brain Inj 2017; 31:1369-1375. [PMID: 28585898 DOI: 10.1080/02699052.2017.1312018] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Grant C. O’Connell
- Center for Basic and Translational Stroke Research, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, West Virginia, USA
- Department of Pharmaceutical Sciences, School of Pharmacy, West Virginia University, Morgantown, West Virginia, USA
| | - Ashley B. Petrone
- Center for Basic and Translational Stroke Research, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, West Virginia, USA
| | - Connie S. Tennant
- Center for Basic and Translational Stroke Research, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, West Virginia, USA
| | - Noelle Lucke-Wold
- Center for Basic and Translational Stroke Research, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, West Virginia, USA
| | - Yasser Kabbani
- Department of Neuroradiology, Ruby Memorial Hospital, Morgantown, West Virginia, USA
| | - Abdul R. Tarabishy
- Department of Neuroradiology, Ruby Memorial Hospital, Morgantown, West Virginia, USA
| | - Paul D. Chantler
- Center for Cardiovascular and Respiratory Sciences, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, West Virginia, USA
- Division of Exercise Physiology, School of Medicine, West Virginia University, Morgantown, West Virginia, USA
| | - Taura L. Barr
- Valtari Bio Incorporated, Morgantown, West Virginia, USA
| |
Collapse
|
484
|
Aldrich MB, Velasquez FC, Kwon S, Azhdarinia A, Pinkston K, Harvey BR, Chan W, Rasmussen JC, Ross RF, Fife CE, Sevick-Muraca EM. Lymphatic delivery of etanercept via nanotopography improves response to collagen-induced arthritis. Arthritis Res Ther 2017; 19:116. [PMID: 28566090 PMCID: PMC5452411 DOI: 10.1186/s13075-017-1323-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Accepted: 05/09/2017] [Indexed: 12/21/2022] Open
Abstract
Background Evidence suggests lymphatic function mediates local rheumatoid arthritis (RA) flares. Yet biologics that target the immune system are dosed systemically via the subcutaneous (SC) administration route, thereby inefficiently reaching local lymphatic compartments. Nanotopography has previously been shown to disrupt tight cellular junctions, potentially enhancing local lymphatic delivery and potentially improving overall therapeutic efficacy. Method We first characterized nanotopography (SOFUSA™) delivery of an anti-TNF drug, etanercept, by comparing pharmacokinetic profiles to those obtained by conventional SC, intravenous (IV), and intradermal (ID) routes of administration, and assessed uptake of radiolabeled etanercept in draining lymph nodes (LNs) in single dosing studies. We then compared etanercept efficacy in a progressive rat model of collagen-induced arthritis (CIA), administered systemically via SC route of administration; via the regional lymphatics through ID delivery; or through a nanotopography (SOFUSA™) device at 10, 12, and 14 days post CIA induction. Measurements of hind limb swelling and near-infrared fluorescence (NIRF) imaging of afferent lymph pumping function and reflux were conducted on days 11, 13, and 18 post CIA induction and compared to untreated CIA animals. Univariate and multivariate analysis of variance were used to compare the group differences for percentage swelling and lymphatic contractile activity. Results Even though all three modes of administration delivered an equal amount of etanercept, SOFUSA™ delivery resulted in increased lymphatic pumping and significantly reduced swelling as compared to untreated, ID, and SC groups. Pharmacokinetic profiles in serum and LN uptake studies showed that using the nanotopography device resulted in the greatest uptake and retention in draining LNs. Conclusions Locoregional lymphatic delivery of biologics that target the immune system may have more favorable pharmacodynamics than SC or IV administration. Nanotopography may provide a more efficient method for delivery of anti-TNF drugs to reverse impairment of lymphatic function and reduce swelling associated with RA flares. Electronic supplementary material The online version of this article (doi:10.1186/s13075-017-1323-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Melissa B Aldrich
- The Center for Molecular Imaging, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center, Houston, TX, 77030, USA
| | - Fred C Velasquez
- The Center for Molecular Imaging, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center, Houston, TX, 77030, USA
| | - Sunkuk Kwon
- The Center for Molecular Imaging, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center, Houston, TX, 77030, USA
| | - Ali Azhdarinia
- The Center for Molecular Imaging, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center, Houston, TX, 77030, USA
| | - Kenneth Pinkston
- The Center for Molecular Imaging, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center, Houston, TX, 77030, USA
| | - Barrett R Harvey
- The Center for Molecular Imaging, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center, Houston, TX, 77030, USA
| | - Wenyaw Chan
- Department of Biostatistics, The School of Public Health, The University of Texas Health Science Center, Houston, TX, 77030, USA
| | - John C Rasmussen
- The Center for Molecular Imaging, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center, Houston, TX, 77030, USA
| | | | - Caroline E Fife
- The Wound Care Clinic, CHI St. Luke's Health, The Woodlands Hospital, The Woodlands, TX, 77382, USA
| | - E M Sevick-Muraca
- The Center for Molecular Imaging, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center, Houston, TX, 77030, USA.
| |
Collapse
|
485
|
Abstract
Beyond its role in hemostasis, von Willebrand factor (VWF) is an emerging mediator of vascular inflammation. Recent studies highlight the involvement of VWF and its regulator, ADAMTS13, in mechanisms that underlie vascular inflammation and immunothrombosis, like leukocyte rolling, adhesion, and extravasation; vascular permeability; ischemia/reperfusion injury; complements activation; and NETosis. The VWF/ADAMTS13 axis is implicated in the pathogenesis of atherosclerosis, promoting plaque formation and inflammation through macrophage and neutrophil recruitment in inflamed lesions. Moreover, VWF and ADAMTS13 have been recently proposed as prognostic biomarkers in cardiovascular, metabolic, and inflammatory diseases, such as diabetes, stroke, myocardial infarction, and sepsis. All these features make VWF an attractive therapeutic target in thromboinflammation. Several lines of research have recently investigated “tailor-made” inhibitors of VWF. Results from animal models and clinical studies support the potent anti-inflammatory and antithrombotic effect of VWF antagonism, providing reassuring data on its safety profile. This review describes the role of VWF in vascular inflammation “from bench to bedside” and provides an updated overview of the drugs that can directly interfere with the VWF/ADAMTS13 axis.
Collapse
|
486
|
Silica particles contribute to the procoagulant activity of DNA and polyphosphate isolated using commercial kits. Blood 2017; 130:88-91. [PMID: 28533308 DOI: 10.1182/blood-2017-03-772848] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
|
487
|
Liu Z, Yago T, Zhang N, Panicker SR, Wang Y, Yao L, Mehta-D'souza P, Xia L, Zhu C, McEver RP. L-selectin mechanochemistry restricts neutrophil priming in vivo. Nat Commun 2017; 8:15196. [PMID: 28497779 PMCID: PMC5437312 DOI: 10.1038/ncomms15196] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Accepted: 03/09/2017] [Indexed: 01/02/2023] Open
Abstract
Circulating neutrophils must avoid premature activation to prevent tissue injury. The leukocyte adhesion receptor L-selectin forms bonds with P-selectin glycoprotein ligand-1 (PSGL-1) on other leukocytes and with peripheral node addressin (PNAd) on high endothelial venules. Mechanical forces can strengthen (catch) or weaken (slip) bonds between biological molecules. How these mechanochemical processes influence function in vivo is unexplored. Here we show that mice expressing an L-selectin mutant (N138G) have altered catch bonds and prolonged bond lifetimes at low forces. Basal lymphocyte homing and neutrophil recruitment to inflamed sites are normal. However, circulating neutrophils form unstable aggregates and are unexpectedly primed to respond robustly to inflammatory mediators. Priming requires signals transduced through L-selectin N138G after it engages PSGL-1 or PNAd. Priming enhances bacterial clearance but increases inflammatory injury and enlarges venous thrombi. Thus, L-selectin mechanochemistry limits premature activation of neutrophils. Our results highlight the importance of probing how mechanochemistry functions in vivo.
Collapse
Affiliation(s)
- Zhenghui Liu
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma 73104, USA
| | - Tadayuki Yago
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma 73104, USA
| | - Nan Zhang
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, USA
| | - Sumith R. Panicker
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma 73104, USA
| | - Ying Wang
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, USA
| | - Longbiao Yao
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma 73104, USA
| | - Padmaja Mehta-D'souza
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma 73104, USA
| | - Lijun Xia
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma 73104, USA
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, USA
| | - Cheng Zhu
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, USA
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, USA
- Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia 30332, USA
| | - Rodger P. McEver
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma 73104, USA
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, USA
| |
Collapse
|
488
|
Pieterse E, Rother N, Garsen M, Hofstra JM, Satchell SC, Hoffmann M, Loeven MA, Knaapen HK, van der Heijden OWH, Berden JHM, Hilbrands LB, van der Vlag J. Neutrophil Extracellular Traps Drive Endothelial-to-Mesenchymal Transition. Arterioscler Thromb Vasc Biol 2017; 37:1371-1379. [PMID: 28495931 DOI: 10.1161/atvbaha.117.309002] [Citation(s) in RCA: 189] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 05/01/2017] [Indexed: 12/12/2022]
Abstract
OBJECTIVE An excessive release and impaired degradation of neutrophil extracellular traps (NETs) leads to the continuous exposure of NETs to the endothelium in a variety of hematologic and autoimmune disorders, including lupus nephritis. This study aims to unravel the mechanisms through which NETs jeopardize vascular integrity. APPROACH AND RESULTS Microvascular and macrovascular endothelial cells were exposed to NETs, and subsequent effects on endothelial integrity and function were determined in vitro and in vivo. We found that endothelial cells have a limited capacity to internalize NETs via the receptor for advanced glycation endproducts. An overflow of the phagocytic capacity of endothelial cells for NETs resulted in the persistent extracellular presence of NETs, which rapidly altered endothelial cell-cell contacts and induced vascular leakage and transendothelial albumin passage through elastase-mediated proteolysis of the intercellular junction protein VE-cadherin. Furthermore, NET-associated elastase promoted the nuclear translocation of junctional β-catenin and induced endothelial-to-mesenchymal transition in cultured endothelial cells. In vivo, NETs could be identified in kidney samples of diseased MRL/lpr mice and patients with lupus nephritis, in whom the glomerular presence of NETs correlated with the severity of proteinuria and with glomerular endothelial-to-mesenchymal transition. CONCLUSIONS These results indicate that an excess of NETs exceeds the phagocytic capacity of endothelial cells for NETs and promotes vascular leakage and endothelial-to-mesenchymal transition through the degradation of VE-cadherin and the subsequent activation of β-catenin signaling. Our data designate NET-associated elastase as a potential therapeutic target in the prevention of endothelial alterations in diseases characterized by aberrant NET release.
Collapse
Affiliation(s)
- Elmar Pieterse
- From the Nephrology Research Laboratory, Department of Nephrology (E.P., N.R., M.G., J.M.H., M.A.L., J.H.M.B., L.B.H., J.v.d.V.), Department of Rheumatology (H.K.K.), and Department of Obstetrics & Gynecology (O.W.H.v.d.H.), Radboud University Medical Center, Nijmegen, The Netherlands; Academic Renal Unit, School of Clinical Science, University of Bristol, United Kingdom (S.C.S.); and Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich Alexander University of Erlangen-Nuremberg, Germany (M.H.)
| | - Nils Rother
- From the Nephrology Research Laboratory, Department of Nephrology (E.P., N.R., M.G., J.M.H., M.A.L., J.H.M.B., L.B.H., J.v.d.V.), Department of Rheumatology (H.K.K.), and Department of Obstetrics & Gynecology (O.W.H.v.d.H.), Radboud University Medical Center, Nijmegen, The Netherlands; Academic Renal Unit, School of Clinical Science, University of Bristol, United Kingdom (S.C.S.); and Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich Alexander University of Erlangen-Nuremberg, Germany (M.H.)
| | - Marjolein Garsen
- From the Nephrology Research Laboratory, Department of Nephrology (E.P., N.R., M.G., J.M.H., M.A.L., J.H.M.B., L.B.H., J.v.d.V.), Department of Rheumatology (H.K.K.), and Department of Obstetrics & Gynecology (O.W.H.v.d.H.), Radboud University Medical Center, Nijmegen, The Netherlands; Academic Renal Unit, School of Clinical Science, University of Bristol, United Kingdom (S.C.S.); and Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich Alexander University of Erlangen-Nuremberg, Germany (M.H.)
| | - Julia M Hofstra
- From the Nephrology Research Laboratory, Department of Nephrology (E.P., N.R., M.G., J.M.H., M.A.L., J.H.M.B., L.B.H., J.v.d.V.), Department of Rheumatology (H.K.K.), and Department of Obstetrics & Gynecology (O.W.H.v.d.H.), Radboud University Medical Center, Nijmegen, The Netherlands; Academic Renal Unit, School of Clinical Science, University of Bristol, United Kingdom (S.C.S.); and Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich Alexander University of Erlangen-Nuremberg, Germany (M.H.)
| | - Simon C Satchell
- From the Nephrology Research Laboratory, Department of Nephrology (E.P., N.R., M.G., J.M.H., M.A.L., J.H.M.B., L.B.H., J.v.d.V.), Department of Rheumatology (H.K.K.), and Department of Obstetrics & Gynecology (O.W.H.v.d.H.), Radboud University Medical Center, Nijmegen, The Netherlands; Academic Renal Unit, School of Clinical Science, University of Bristol, United Kingdom (S.C.S.); and Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich Alexander University of Erlangen-Nuremberg, Germany (M.H.)
| | - Markus Hoffmann
- From the Nephrology Research Laboratory, Department of Nephrology (E.P., N.R., M.G., J.M.H., M.A.L., J.H.M.B., L.B.H., J.v.d.V.), Department of Rheumatology (H.K.K.), and Department of Obstetrics & Gynecology (O.W.H.v.d.H.), Radboud University Medical Center, Nijmegen, The Netherlands; Academic Renal Unit, School of Clinical Science, University of Bristol, United Kingdom (S.C.S.); and Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich Alexander University of Erlangen-Nuremberg, Germany (M.H.)
| | - Markus A Loeven
- From the Nephrology Research Laboratory, Department of Nephrology (E.P., N.R., M.G., J.M.H., M.A.L., J.H.M.B., L.B.H., J.v.d.V.), Department of Rheumatology (H.K.K.), and Department of Obstetrics & Gynecology (O.W.H.v.d.H.), Radboud University Medical Center, Nijmegen, The Netherlands; Academic Renal Unit, School of Clinical Science, University of Bristol, United Kingdom (S.C.S.); and Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich Alexander University of Erlangen-Nuremberg, Germany (M.H.)
| | - Hanneke K Knaapen
- From the Nephrology Research Laboratory, Department of Nephrology (E.P., N.R., M.G., J.M.H., M.A.L., J.H.M.B., L.B.H., J.v.d.V.), Department of Rheumatology (H.K.K.), and Department of Obstetrics & Gynecology (O.W.H.v.d.H.), Radboud University Medical Center, Nijmegen, The Netherlands; Academic Renal Unit, School of Clinical Science, University of Bristol, United Kingdom (S.C.S.); and Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich Alexander University of Erlangen-Nuremberg, Germany (M.H.)
| | - Olivier W H van der Heijden
- From the Nephrology Research Laboratory, Department of Nephrology (E.P., N.R., M.G., J.M.H., M.A.L., J.H.M.B., L.B.H., J.v.d.V.), Department of Rheumatology (H.K.K.), and Department of Obstetrics & Gynecology (O.W.H.v.d.H.), Radboud University Medical Center, Nijmegen, The Netherlands; Academic Renal Unit, School of Clinical Science, University of Bristol, United Kingdom (S.C.S.); and Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich Alexander University of Erlangen-Nuremberg, Germany (M.H.)
| | - Jo H M Berden
- From the Nephrology Research Laboratory, Department of Nephrology (E.P., N.R., M.G., J.M.H., M.A.L., J.H.M.B., L.B.H., J.v.d.V.), Department of Rheumatology (H.K.K.), and Department of Obstetrics & Gynecology (O.W.H.v.d.H.), Radboud University Medical Center, Nijmegen, The Netherlands; Academic Renal Unit, School of Clinical Science, University of Bristol, United Kingdom (S.C.S.); and Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich Alexander University of Erlangen-Nuremberg, Germany (M.H.)
| | - Luuk B Hilbrands
- From the Nephrology Research Laboratory, Department of Nephrology (E.P., N.R., M.G., J.M.H., M.A.L., J.H.M.B., L.B.H., J.v.d.V.), Department of Rheumatology (H.K.K.), and Department of Obstetrics & Gynecology (O.W.H.v.d.H.), Radboud University Medical Center, Nijmegen, The Netherlands; Academic Renal Unit, School of Clinical Science, University of Bristol, United Kingdom (S.C.S.); and Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich Alexander University of Erlangen-Nuremberg, Germany (M.H.)
| | - Johan van der Vlag
- From the Nephrology Research Laboratory, Department of Nephrology (E.P., N.R., M.G., J.M.H., M.A.L., J.H.M.B., L.B.H., J.v.d.V.), Department of Rheumatology (H.K.K.), and Department of Obstetrics & Gynecology (O.W.H.v.d.H.), Radboud University Medical Center, Nijmegen, The Netherlands; Academic Renal Unit, School of Clinical Science, University of Bristol, United Kingdom (S.C.S.); and Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich Alexander University of Erlangen-Nuremberg, Germany (M.H.).
| |
Collapse
|
489
|
Granger V, Faille D, Marani V, Noël B, Gallais Y, Szely N, Flament H, Pallardy M, Chollet-Martin S, de Chaisemartin L. Human blood monocytes are able to form extracellular traps. J Leukoc Biol 2017; 102:775-781. [PMID: 28465447 DOI: 10.1189/jlb.3ma0916-411r] [Citation(s) in RCA: 111] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 03/13/2017] [Accepted: 03/31/2017] [Indexed: 12/17/2022] Open
Abstract
Neutrophil extracellular traps (NETs) are extracellular DNA filaments formed during neutrophil activation. This process, called netosis, was originally associated with neutrophil antibacterial properties. However, several lines of evidence now suggest a major role for netosis in thrombosis, autoimmune diseases, and cancer. We demonstrate here that highly purified human blood monocytes are also capable of extracellular trap (ET) release in response to several stimuli. Monocyte ETs display a morphology analogous to NETs and are associated with myeloperoxidase (MPO), lactoferrin (LF), citrullinated histones, and elastase. Monocyte ET release depends on oxidative burst but not on MPO activity, in contrast to neutrophils. Moreover, we demonstrate procoagulant activity for monocyte ETs, a feature that could be relevant to monocyte thrombogenic properties. This new cellular mechanism is likely to have implications in the multiple pathologic contexts where monocytes are implicated, such as inflammatory disorders, infection, or thrombosis.
Collapse
Affiliation(s)
- Vanessa Granger
- Unité mixte de Recherche 996-Inflammation, Chemokines and Immunopathology, Institut National de la Santé et de la Recherche Médicale, Université Paris Sud, Université Paris-Saclay, Châtenay-Malabry, France.,Assistance Publique Hopitaux de Paris, Bichat Hospital, Immunology Department, Paris, France; and
| | - Dorothée Faille
- Assistance Publique Hopitaux de Paris, Bichat Hospital, Hematology Department, Paris, France
| | - Vanessa Marani
- Unité mixte de Recherche 996-Inflammation, Chemokines and Immunopathology, Institut National de la Santé et de la Recherche Médicale, Université Paris Sud, Université Paris-Saclay, Châtenay-Malabry, France
| | - Benoît Noël
- Unité mixte de Recherche 996-Inflammation, Chemokines and Immunopathology, Institut National de la Santé et de la Recherche Médicale, Université Paris Sud, Université Paris-Saclay, Châtenay-Malabry, France
| | - Yann Gallais
- Unité mixte de Recherche 996-Inflammation, Chemokines and Immunopathology, Institut National de la Santé et de la Recherche Médicale, Université Paris Sud, Université Paris-Saclay, Châtenay-Malabry, France
| | - Natacha Szely
- Unité mixte de Recherche 996-Inflammation, Chemokines and Immunopathology, Institut National de la Santé et de la Recherche Médicale, Université Paris Sud, Université Paris-Saclay, Châtenay-Malabry, France
| | - Héloïse Flament
- Assistance Publique Hopitaux de Paris, Bichat Hospital, Immunology Department, Paris, France; and
| | - Marc Pallardy
- Unité mixte de Recherche 996-Inflammation, Chemokines and Immunopathology, Institut National de la Santé et de la Recherche Médicale, Université Paris Sud, Université Paris-Saclay, Châtenay-Malabry, France
| | - Sylvie Chollet-Martin
- Unité mixte de Recherche 996-Inflammation, Chemokines and Immunopathology, Institut National de la Santé et de la Recherche Médicale, Université Paris Sud, Université Paris-Saclay, Châtenay-Malabry, France.,Assistance Publique Hopitaux de Paris, Bichat Hospital, Immunology Department, Paris, France; and
| | - Luc de Chaisemartin
- Unité mixte de Recherche 996-Inflammation, Chemokines and Immunopathology, Institut National de la Santé et de la Recherche Médicale, Université Paris Sud, Université Paris-Saclay, Châtenay-Malabry, France; .,Assistance Publique Hopitaux de Paris, Bichat Hospital, Immunology Department, Paris, France; and
| |
Collapse
|
490
|
Tripodi A, Ammollo CT, Semeraro F, Colucci M, Malchiodi E, Verrua E, Ferrante E, Arnaldi G, Trementino L, Padovan L, Chantarangkul V, Peyvandi F, Mantovani G. Hypercoagulability in patients with Cushing disease detected by thrombin generation assay is associated with increased levels of neutrophil extracellular trap-related factors. Endocrine 2017; 56:298-307. [PMID: 27448294 DOI: 10.1007/s12020-016-1027-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 06/16/2016] [Indexed: 10/21/2022]
Abstract
Patients with Cushing disease (CD) are at increased risk of venous thromboembolism (VTE). It was surmised, but not conclusively shown that the risk is related to plasma hypercoagulability secondary to the glucocorticoids effect. This study is aimed at detecting hypercoagulability in patients with CD. Case-control study of 48 CD patients and controls enrolled at two Italian clinics for whom we assessed the thrombin-forming-potential in the presence of optimal activation of protein C obtained by adding into the assay system its main endothelial activator, thrombomodulin. These experimental conditions mimic more closely than any other test the in vivo situation. We observed enhanced thrombin-generation in CD patients, as shown by the modification of thrombin-generation parameters [i.e., shortened lag-time and time-to-peak, increased thrombin peak and endogenous thrombin potential (ETP)]. Moreover, the ETP ratio (with/without thrombomodulin), recognized as an index of hypercoagulability, was increased in patients as compared to controls. We attempted to explain such hypercoagulability by measuring both procoagulant and anticoagulant factors, and some other non-coagulation parameters (i.e., neutrophil extracellular traps (NET), recently associated with the VTE risk and/or increased hypercoagulability. We showed that the hypercoagulability in patients with CD is associated with increased levels of factor VIII and NET-related variables. We detected plasma hypercoagulability in patients with CD and found experimental explanation for its occurrence. Whether this hypercoagulability can entirely explain the occurrence of VTE in patients with CD should be investigated by ad-hoc clinical trials. However, until these studies will be available the evidence supports the concept that patients with CD are candidates for antithrombotic prophylaxis.
Collapse
Affiliation(s)
- Armando Tripodi
- Angelo Bianchi Bonomi Hemophilia and Thrombosis Center, Milan, Italy.
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milano, Italy.
- IRCCS Cà Granda Maggiore Hospital Foundation, Milano, Italy.
| | - Concetta T Ammollo
- Department of Biomedical Sciences and Human Oncology, Aldo Moro University, Bari, Italy
| | - Fabrizio Semeraro
- Department of Biomedical Sciences and Human Oncology, Aldo Moro University, Bari, Italy
| | - Mario Colucci
- Department of Biomedical Sciences and Human Oncology, Aldo Moro University, Bari, Italy
| | - Elena Malchiodi
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milano, Italy
- Unit of Endocrinology and Diabetology, Milano, Italy
- IRCCS Cà Granda Maggiore Hospital Foundation, Milano, Italy
| | - Elisa Verrua
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milano, Italy
- Unit of Endocrinology and Diabetology, Milano, Italy
- IRCCS Cà Granda Maggiore Hospital Foundation, Milano, Italy
| | - Emanuele Ferrante
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milano, Italy
- Unit of Endocrinology and Diabetology, Milano, Italy
- IRCCS Cà Granda Maggiore Hospital Foundation, Milano, Italy
| | - Giorgio Arnaldi
- Clinica di Endocrinologia e Malattie del Metabolismo, Ospedali Riuniti di Ancona, Ancona, Italy
| | - Laura Trementino
- Clinica di Endocrinologia e Malattie del Metabolismo, Ospedali Riuniti di Ancona, Ancona, Italy
| | - Lidia Padovan
- Angelo Bianchi Bonomi Hemophilia and Thrombosis Center, Milan, Italy
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milano, Italy
- IRCCS Cà Granda Maggiore Hospital Foundation, Milano, Italy
| | - Veena Chantarangkul
- Angelo Bianchi Bonomi Hemophilia and Thrombosis Center, Milan, Italy
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milano, Italy
- IRCCS Cà Granda Maggiore Hospital Foundation, Milano, Italy
| | - Flora Peyvandi
- Angelo Bianchi Bonomi Hemophilia and Thrombosis Center, Milan, Italy
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milano, Italy
- IRCCS Cà Granda Maggiore Hospital Foundation, Milano, Italy
| | - Giovanna Mantovani
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milano, Italy
- Unit of Endocrinology and Diabetology, Milano, Italy
- IRCCS Cà Granda Maggiore Hospital Foundation, Milano, Italy
| |
Collapse
|
491
|
Foley JH, Conway EM. Cross Talk Pathways Between Coagulation and Inflammation. Circ Res 2017; 118:1392-408. [PMID: 27126649 DOI: 10.1161/circresaha.116.306853] [Citation(s) in RCA: 409] [Impact Index Per Article: 51.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2016] [Accepted: 03/21/2016] [Indexed: 02/06/2023]
Abstract
Anatomic pathology studies performed over 150 years ago revealed that excessive activation of coagulation occurs in the setting of inflammation. However, it has taken over a century since these seminal observations were made to delineate the molecular mechanisms by which these systems interact and the extent to which they participate in the pathogenesis of multiple diseases. There is, in fact, extensive cross talk between coagulation and inflammation, whereby activation of one system may amplify activation of the other, a situation that, if unopposed, may result in tissue damage or even multiorgan failure. Characterizing the common triggers and pathways are key for the strategic design of effective therapeutic interventions. In this review, we highlight some of the key molecular interactions, some of which are already showing promise as therapeutic targets for inflammatory and thrombotic disorders.
Collapse
Affiliation(s)
- Jonathan H Foley
- From the Department of Haematology, UCL Cancer Institute, University College London, London, United Kingdom (J.H.F.); Katharine Dormandy Haemophilia Centre and Thrombosis Unit, Royal Free NHS Trust, London, United Kingdom (J.H.F.); and Centre for Blood Research, Department of Medicine, University of British Columbia, Vancouver, Canada (E.M.C.)
| | - Edward M Conway
- From the Department of Haematology, UCL Cancer Institute, University College London, London, United Kingdom (J.H.F.); Katharine Dormandy Haemophilia Centre and Thrombosis Unit, Royal Free NHS Trust, London, United Kingdom (J.H.F.); and Centre for Blood Research, Department of Medicine, University of British Columbia, Vancouver, Canada (E.M.C.).
| |
Collapse
|
492
|
Laurance S, Bertin FR, Ebrahimian T, Kassim Y, Rys RN, Lehoux S, Lemarié CA, Blostein MD. Gas6 Promotes Inflammatory (CCR2 hiCX3CR1 lo) Monocyte Recruitment in Venous Thrombosis. Arterioscler Thromb Vasc Biol 2017; 37:1315-1322. [PMID: 28450294 DOI: 10.1161/atvbaha.116.308925] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 04/17/2017] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Coagulation and inflammation are inter-related. Gas6 (growth arrest-specific 6) promotes venous thrombosis and participates to inflammation through endothelial-innate immune cell interactions. Innate immune cells can provide the initiating stimulus for venous thrombus development. We hypothesize that Gas6 promotes monocyte recruitment during venous thrombosis. APPROACH AND RESULTS Deep venous thrombosis was induced in wild-type and Gas6-deficient (-/-) mice using 5% FeCl3 and flow reduction in the inferior vena cava. Total monocyte depletion was achieved by injection of clodronate before deep venous thrombosis. Inflammatory monocytes were depleted using an anti-C-C chemokine receptor type 2 (CCR2) antibody. Similarly, injection of an anti-chemokine ligand 2 (CCL2) antibody induced CCL2 depletion. Flow cytometry and immunofluorescence were used to characterize the monocytes recruited to the thrombus. In vivo, absence of Gas6 was associated with a reduction of monocyte recruitment in both deep venous thrombosis models. Global monocyte depletion by clodronate leads to smaller thrombi in wild-type mice. Compared with wild type, the thrombi from Gas6-/- mice contain less inflammatory (CCR2hiCX3CR1lo) monocytes, consistent with a Gas6-dependent recruitment of this monocyte subset. Correspondingly, selective depletion of CCR2hiCX3CR1lo monocytes reduced the formation of venous thrombi in wild-type mice demonstrating a predominant role of the inflammatory monocytes in thrombosis. In vitro, the expression of both CCR2 and CCL2 were Gas6 dependent in monocytes and endothelial cells, respectively, impacting monocyte migration. Moreover, Gas6-dependent CCL2 expression and monocyte migration were mediated via JNK (c-Jun N-terminal kinase). CONCLUSIONS This study demonstrates that Gas6 specifically promotes the recruitment of inflammatory CCR2hiCX3CR1lo monocytes through the regulation of both CCR2 and CCL2 during deep venous thrombosis.
Collapse
MESH Headings
- Animals
- CX3C Chemokine Receptor 1
- Cells, Cultured
- Chemokine CCL2/genetics
- Chemokine CCL2/metabolism
- Chemotaxis, Leukocyte/drug effects
- Clodronic Acid/pharmacology
- Disease Models, Animal
- Endothelial Cells/metabolism
- Genetic Predisposition to Disease
- Inflammation/genetics
- Inflammation/metabolism
- Inflammation/pathology
- Inflammation/prevention & control
- Intercellular Signaling Peptides and Proteins/deficiency
- Intercellular Signaling Peptides and Proteins/genetics
- Intercellular Signaling Peptides and Proteins/metabolism
- JNK Mitogen-Activated Protein Kinases/metabolism
- Male
- Mice, Inbred C57BL
- Mice, Knockout
- Monocytes/drug effects
- Monocytes/metabolism
- Paracrine Communication
- Phenotype
- Receptors, CCR2/genetics
- Receptors, CCR2/metabolism
- Receptors, Chemokine/metabolism
- Signal Transduction
- Vena Cava, Inferior/drug effects
- Vena Cava, Inferior/metabolism
- Vena Cava, Inferior/pathology
- Venous Thrombosis/genetics
- Venous Thrombosis/metabolism
- Venous Thrombosis/pathology
- Venous Thrombosis/prevention & control
Collapse
Affiliation(s)
- Sandrine Laurance
- From the Lady Davis Institute for Medical Research (S.L., F.-R.B., T.E., Y.K., R.N.R., S.L., C.A.L., M.D.B.); and Department of Medicine (S.L., C.A.L., M.D.B.), Jewish General Hospital, McGill University, Montreal, Québec, Canada
| | - François-René Bertin
- From the Lady Davis Institute for Medical Research (S.L., F.-R.B., T.E., Y.K., R.N.R., S.L., C.A.L., M.D.B.); and Department of Medicine (S.L., C.A.L., M.D.B.), Jewish General Hospital, McGill University, Montreal, Québec, Canada
| | - Talin Ebrahimian
- From the Lady Davis Institute for Medical Research (S.L., F.-R.B., T.E., Y.K., R.N.R., S.L., C.A.L., M.D.B.); and Department of Medicine (S.L., C.A.L., M.D.B.), Jewish General Hospital, McGill University, Montreal, Québec, Canada
| | - Yusra Kassim
- From the Lady Davis Institute for Medical Research (S.L., F.-R.B., T.E., Y.K., R.N.R., S.L., C.A.L., M.D.B.); and Department of Medicine (S.L., C.A.L., M.D.B.), Jewish General Hospital, McGill University, Montreal, Québec, Canada
| | - Ryan N Rys
- From the Lady Davis Institute for Medical Research (S.L., F.-R.B., T.E., Y.K., R.N.R., S.L., C.A.L., M.D.B.); and Department of Medicine (S.L., C.A.L., M.D.B.), Jewish General Hospital, McGill University, Montreal, Québec, Canada
| | - Stéphanie Lehoux
- From the Lady Davis Institute for Medical Research (S.L., F.-R.B., T.E., Y.K., R.N.R., S.L., C.A.L., M.D.B.); and Department of Medicine (S.L., C.A.L., M.D.B.), Jewish General Hospital, McGill University, Montreal, Québec, Canada
| | - Catherine A Lemarié
- From the Lady Davis Institute for Medical Research (S.L., F.-R.B., T.E., Y.K., R.N.R., S.L., C.A.L., M.D.B.); and Department of Medicine (S.L., C.A.L., M.D.B.), Jewish General Hospital, McGill University, Montreal, Québec, Canada.
| | - Mark D Blostein
- From the Lady Davis Institute for Medical Research (S.L., F.-R.B., T.E., Y.K., R.N.R., S.L., C.A.L., M.D.B.); and Department of Medicine (S.L., C.A.L., M.D.B.), Jewish General Hospital, McGill University, Montreal, Québec, Canada
| |
Collapse
|
493
|
Recent Progress in Research on the Pathogenesis of Pulmonary Thromboembolism: An Old Story with New Perspectives. BIOMED RESEARCH INTERNATIONAL 2017; 2017:6516791. [PMID: 28484717 PMCID: PMC5397627 DOI: 10.1155/2017/6516791] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Revised: 02/26/2017] [Accepted: 03/27/2017] [Indexed: 12/25/2022]
Abstract
Pulmonary thromboembolism (PTE) is part of a larger clinicopathological entity, venous thromboembolism. It is also a complex, multifactorial disorder divided into four major disease processes including venous thrombosis, thrombus in transit, acute pulmonary embolism, and pulmonary circulation reconstruction. Even when treated, some patients develop chronic thromboembolic pulmonary hypertension. PTE is also a common fatal type of pulmonary vascular disease worldwide, but earlier studies primarily focused on the pathological changes in the blood component of the disease. With contemporary advances in molecular and cellular biology, people are becoming increasingly aware of coagulation pathways, the function of vascular smooth muscle cells, microparticles, and the inflammatory pathways that play key roles in PTE. Combined hypoxia and immune research has revealed that PTE should be regarded as a class of complex diseases caused by multiple factors involving the vascular microenvironment and vascular cell dysfunction.
Collapse
|
494
|
Nurden AT. Should studies on Glanzmann thrombasthenia not be telling us more about cardiovascular disease and other major illnesses? Blood Rev 2017; 31:287-299. [PMID: 28395882 DOI: 10.1016/j.blre.2017.03.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Accepted: 03/23/2017] [Indexed: 12/17/2022]
Abstract
Glanzmann thrombasthenia (GT) is a rare inherited bleeding disorder caused by loss of αIIbβ3 integrin function in platelets. Most genetic variants of β3 also affect the widely expressed αvβ3 integrin. With brief mention of mouse models, I now look at the consequences of disease-causing ITGA2B and ITGB3 mutations on the non-hemostatic functions of platelets and other cells. Reports of arterial thrombosis in GT patients are rare, but other aspects of cardiovascular disease do occur including deep vein thrombosis and congenital heart defects. Thrombophilic and other risk factors for thrombosis and lessons from heterozygotes and variant forms of GT are discussed. Assessed for GT patients are reports of leukemia and cancer, loss of fertility, bone pathology, inflammation and wound repair, infections, kidney disease, autism and respiratory disease. This survey shows an urgent need for a concerted international effort to better determine how loss of αIIbβ3 and αvβ3 influences health and disease.
Collapse
Affiliation(s)
- Alan T Nurden
- Institut de Rhythmologie et de Modélisation Cardiaque, Plateforme Technologique d'Innovation Biomédicale, Hôpital Xavier Arnozan, Pessac, France.
| |
Collapse
|
495
|
Kushnir M, Cohen HW, Billett HH. Persistent neutrophilia is a marker for an increased risk of venous thrombosis. J Thromb Thrombolysis 2017; 42:545-51. [PMID: 27383828 DOI: 10.1007/s11239-016-1398-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
In patients with cancer and myeloproliferative disorders, leukocytosis has been associated with an increased venous thromboembolic (VTE) risk. Our goal was to determine whether persistent neutrophilia (PN), not associated with known causes such as malignancies, infections or steroids, is independently associated with VTE. All adult patients with >3 outpatient complete blood counts (CBCs) within 3 years were included. PN was defined as having an absolute neutrophil count >95 % (>2SD) of the population (≥7.8 × 10(9)/L) on at least three CBCs, at least 2 months apart. Separate analyses for neutrophil counts ≥9 × 10(9)/L and ≥10 × 10(9)/L were also performed. Blood counts from inpatients were excluded. Primary outcome was diagnosis of VTE, as determined by ICD-9 codes. Odds ratios were adjusted for diabetes, smoking, obesity, gender, and age. Charlson score was utilized as a morbidity measure. Data on 43,538 outpatients were collected. Although there was no association of VTE with neutrophil counts ≥7.8 × 10(9)/L, patients with ≥9.0 × 10(9)/L neutrophils were twice as likely to be diagnosed with VTE compared to those with normal neutrophil counts (OR 2.0, 95 % CI 1.3, 3.1; p = 0.003). Patients with neutrophil counts ≥10.0 × 10(9)/L were at an even higher risk (OR 2.3, 95 % CI 1.2, 4.8; p = 0.019). Charlson scores significantly modified this risk when incorporated into analysis. Elevated neutrophil counts are associated with an increased risk of venous thrombosis even when they are not due to cancer, infection or steroids. In patients with significant comorbidities, neutrophilia may be a marker of VTE risk.
Collapse
Affiliation(s)
- Margarita Kushnir
- Division of Hematology, Department of Oncology, Montefiore Medical Center and The Albert Einstein College of Medicine, 3411 Wayne Avenue, Bronx, NY, 10467, USA.
| | - Hillel W Cohen
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY, 10461, USA
| | - Henny H Billett
- Division of Hematology, Department of Oncology, Montefiore Medical Center and The Albert Einstein College of Medicine, 3411 Wayne Avenue, Bronx, NY, 10467, USA
| |
Collapse
|
496
|
Wang Y, Attar BM, Fuentes HE, Yu J, Zhang H, Tafur AJ. Performance of Khorana Risk Score for Prediction of Venous Thromboembolism in Patients With Hepatocellular Carcinoma. Clin Appl Thromb Hemost 2017; 24:471-476. [PMID: 28288526 DOI: 10.1177/1076029617699088] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Cancer-associated venous thromboembolism (VTE) is one of the leading causes of mortality and morbidity among patients with malignancy. The Khorana risk score (KRS) is currently the best validated risk assessment model to stratify risks of VTE development in ambulatory patients with cancer. In the current study, we assessed the performance of KRS in patients with hepatocellular carcinoma (HCC). We retrospectively analyzed patients with diagnosis of HCC (screened by International Classification of Diseases [ ICD-9] and ICD-10 code, confirmed with radiographic examination and/or histopathology) at a large public hospital over 15 years (January 2000 through July 2015). Cases with VTE were identified through radiographic examination and blindly adjudicated. Khorana risk score was calculated for each patient, and its association with VTE development and mortality was assessed. Among 270 patients with HCC, 16 (5.9%) cases of VTE were identified, including 7 (43.8%) pulmonary embolism, 4 (25%) peripheral deep vein thrombosis, and 6 (37.5%) intra-abdominal thrombosis. One hundred eighty-four (68.1%) patients had a KRS of 0 and 86 (31.9%) patients had a KRS >0. Most of the thrombotic (n = 9, 56%) events occurred in the low-risk group. In univariate analysis, only prechemotherapy leukocyte count equal to or greater than 11 000/μL was statistically significant in the prediction of VTE incidence. After adjusting for confounding factors in multivariate analysis, KRS >0 was not predictive of VTE (hazard ratio [HR] = 1.83, 95% confidence interval [CI] = 0.81-4.15, P = .15) or mortality (HR = 1.61, 95% CI = 0.92-2.81, P = .09). Khorana risk score did not predict VTE development or mortality in patients with HCC. Design of HCC-specific risk assessment model for VTE development is necessary.
Collapse
Affiliation(s)
- Y Wang
- 1 Department of Internal Medicine, John Stroger Jr. Hospital, Chicago, IL, USA
| | - B M Attar
- 2 Division of Gastroenterology and Hepatology, John Stroger Jr. Hospital, Chicago, IL, USA
| | - H E Fuentes
- 1 Department of Internal Medicine, John Stroger Jr. Hospital, Chicago, IL, USA
| | - J Yu
- 1 Department of Internal Medicine, John Stroger Jr. Hospital, Chicago, IL, USA
| | - Huiyuan Zhang
- 3 Collaborative Research Unit, John Stroger Jr. Hospital, Chicago, IL, USA
| | - A J Tafur
- 4 Cardiology-Vascular Medicine, Northshore University Healthsystem, Evanston, IL, USA
| |
Collapse
|
497
|
Koupenova M, Kehrel BE, Corkrey HA, Freedman JE. Thrombosis and platelets: an update. Eur Heart J 2017; 38:785-791. [PMID: 28039338 PMCID: PMC11110018 DOI: 10.1093/eurheartj/ehw550] [Citation(s) in RCA: 173] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Revised: 10/03/2016] [Accepted: 10/25/2016] [Indexed: 12/11/2022] Open
Abstract
Haemostasis and thrombosis are complex, multifactorial processes. There is an evolving understanding of the mechanisms influencing vascular occlusion and the role of inflammation and immunity. Despite major advances in elucidating the mechanistic pathways mediating platelet function and thrombosis, challenges in the treatment of vascular occlusive diseases persist. Pharmacological advances have greatly affected thrombotic outcomes, but this has led to the unwanted side effect of bleeding. Detailed assessment of the impact of non-thrombotic diseases on haemostasis and thrombosis is necessary to better evaluate thrombotic risk and establish optimal treatment. This review will focus on recent advances in understanding the contribution of evolving risk factors to thrombosis.
Collapse
Affiliation(s)
- Milka Koupenova
- University of Massachusetts Medical School, Albert Sherman Center, 368
Plantation St, Worcester, MA 01605, USA
| | - Beate E. Kehrel
- Westfälische Wilhelms University Muenster, Münster, University Hospital,
Department of Anaesthesiology, Intensive Care and Pain Medicine, Experimental and Clinical
Haemostasis Research, D-48149 Muenster, Germany Mendelstrasse 11, UK
| | - Heather A. Corkrey
- University of Massachusetts Medical School, Albert Sherman Center, 368
Plantation St, Worcester, MA 01605, USA
| | - Jane E. Freedman
- University of Massachusetts Medical School, Albert Sherman Center, 368
Plantation St, Worcester, MA 01605, USA
| |
Collapse
|
498
|
Toh CH, Alhamdi Y, Abrams ST. Current Pathological and Laboratory Considerations in the Diagnosis of Disseminated Intravascular Coagulation. Ann Lab Med 2017; 36:505-12. [PMID: 27578502 PMCID: PMC5011102 DOI: 10.3343/alm.2016.36.6.505] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Revised: 06/21/2016] [Accepted: 07/22/2016] [Indexed: 01/06/2023] Open
Abstract
Systemically sustained thrombin generation in vivo is the hallmark of disseminated intravascular coagulation (DIC). Typically, this is in response to a progressing disease state that is associated with significant cellular injury. The etiology could be infectious or noninfectious, with the main pathophysiological mechanisms involving cross-activation among coagulation, innate immunity, and inflammatory responses. This leads to consumption of both pro- and anticoagulant factors as well as endothelial dysfunction and disrupted homeostasis at the blood vessel wall interface. In addition to the release of tissue plasminogen activator (tPA) and soluble thrombomodulin (sTM) following cellular activation and damage, respectively, there is the release of damage-associated molecular patterns (DAMPs) such as extracellular histones and cell-free DNA. Extracellular histones are increasingly recognized as significantly pathogenic in critical illnesses through direct cell toxicity, the promotion of thrombin generation, and the induction of neutrophil extracellular trap (NET) formation. Clinically, high circulating levels of histones and histone–DNA complexes are associated with multiorgan failure, DIC, and adverse patient outcomes. Their measurements as well as that of other DAMPs and molecular markers of thrombin generation are not yet applicable in the routine diagnostic laboratory. To provide a practical diagnostic tool for acute DIC, a composite scoring system using rapidly available coagulation tests is recommended by the International Society on Thrombosis and Haemostasis. Its usefulness and limitations are discussed alongside the advances and unanswered questions in DIC pathogenesis.
Collapse
Affiliation(s)
- Cheng Hock Toh
- Institute of Infection and Global Health, University of Liverpool, Liverpool, United Kingdom.,Roald Dahl Haemostasis & Thrombosis Centre, Royal Liverpool University Hospital, Liverpool, United Kingdom.
| | - Yasir Alhamdi
- Institute of Infection and Global Health, University of Liverpool, Liverpool, United Kingdom
| | - Simon T Abrams
- Institute of Infection and Global Health, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
499
|
Fetz AE, Neeli I, Rodriguez IA, Radic MZ, Bowlin GL. Electrospun Template Architecture and Composition Regulate Neutrophil NETosis In Vitro and In Vivo<sup/>. Tissue Eng Part A 2017; 23:1054-1063. [PMID: 28068879 DOI: 10.1089/ten.tea.2016.0452] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Mounting evidence indicates that neutrophils, first responders to an implanted biomaterial, prime the microenvironment for recruited immune cells by secreting factors and releasing neutrophil extracellular traps (NETs) through NETosis. In this study, we investigated the role of electrospun template architecture and composition in regulating NETosis. Electrospun polydioxanone (PDO), collagen type I (COL), and blended PDO-COL templates (PC) were fabricated with small-diameter (0.25-0.35 μm) and large-diameter (1.0-2.00 μm) fibers. Neutrophil-template interactions were evaluated in vitro for 3 and 24 h with human neutrophils, and the PDO templates were studied in vivo (rat subcutaneous model) for 1 and 7 days. Template-bound NETs were quantified by fluorescent microscopy and an On-cell Western assay. The in vitro results indicate that larger fiber diameters reduced NETosis on PDO templates, whereas the incorporation of COL attenuated NETosis independent of fiber diameter. The in vivo results similarly revealed a lower degree of NETs on large-diameter PDO templates at 1 day, resulting in marginal tissue integration of the templates at 7 days. In contrast, the small-diameter PDO templates, which were coated in a large amount of NETs at 24 h in vivo, were surrounded by capsule-like tissue at 7 days. These preliminary in vivo results validate the in vitro model and signify NETosis as a potentially significant physiological response and a critical preconditioning event for the innate immune response to templates. In conclusion, these results demonstrate the importance of characterizing the neutrophil's acute confrontation with biomaterials to engineer templates capable of promoting in situ regeneration.
Collapse
Affiliation(s)
- Allison E Fetz
- 1 Department of Biomedical Engineering, University of Memphis , Memphis, Tennessee
| | - Indira Neeli
- 2 Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center , Memphis, Tennessee
| | - Isaac A Rodriguez
- 1 Department of Biomedical Engineering, University of Memphis , Memphis, Tennessee
| | - Marko Z Radic
- 2 Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center , Memphis, Tennessee
| | - Gary L Bowlin
- 1 Department of Biomedical Engineering, University of Memphis , Memphis, Tennessee
| |
Collapse
|
500
|
Shen YH, LeMaire SA. Molecular pathogenesis of genetic and sporadic aortic aneurysms and dissections. Curr Probl Surg 2017; 54:95-155. [PMID: 28521856 DOI: 10.1067/j.cpsurg.2017.01.001] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Accepted: 01/16/2017] [Indexed: 12/20/2022]
Affiliation(s)
- Ying H Shen
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX; Department of Cardiovascular Surgery, Texas Heart Institute, Houston, TX; Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX.
| | - Scott A LeMaire
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX; Department of Cardiovascular Surgery, Texas Heart Institute, Houston, TX; Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX; Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX.
| |
Collapse
|