1
|
Yogendrakumar V, Vandelanotte S, Mistry EA, Hill MD, Coutts SB, Nogueira RG, Nguyen TN, Medcalf RL, Broderick JP, De Meyer SF, Campbell BCV. Emerging Adjuvant Thrombolytic Therapies for Acute Ischemic Stroke Reperfusion. Stroke 2024; 55:2536-2546. [PMID: 39105286 DOI: 10.1161/strokeaha.124.045755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/07/2024]
Abstract
Thrombolytic therapies for acute ischemic stroke are widely available but only result in recanalization early enough, to be therapeutically useful, in 10% to 30% of cases. This large gap in treatment effectiveness could be filled by novel therapies that can increase the effectiveness of thrombus clearance without significantly increasing the risk of harm. This focused update will describe the current state of emerging adjuvant treatments for acute ischemic stroke reperfusion. We focus on new treatments that are designed to (1) target different components that make up a stroke thrombus, (2) enhance endogenous fibrinolytic systems, (3) reduce stagnant blood flow, and (4) improve recanalization of distal thrombi and postendovascular thrombectomy.
Collapse
Affiliation(s)
- Vignan Yogendrakumar
- Division of Neurology, The Ottawa Hospital and Ottawa Hospital Research Institute, University of Ottawa, Canada (V.Y.)
- Department of Neurology, Melbourne Brain Centre at the Royal Melbourne Hospital, University of Melbourne, Australia (V.Y., B.C.V.C.)
| | - Sarah Vandelanotte
- Laboratory for Thrombosis Research, KU Leuven Campus Kulak, Kortrijk, Belgium (S.V., S.F.D.M.)
| | - Eva A Mistry
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH (E.A.M., J.P.B.)
| | - Michael D Hill
- Department of Clinical Neurosciences, Radiology, and Community Health Sciences, Cumming School of Medicine, Hotchkiss Brain Institute, University of Calgary, Canada (M.D.H., S.B.C.)
| | - Shelagh B Coutts
- Department of Clinical Neurosciences, Radiology, and Community Health Sciences, Cumming School of Medicine, Hotchkiss Brain Institute, University of Calgary, Canada (M.D.H., S.B.C.)
| | - Raul G Nogueira
- Department of Neurology, University of Pittsburgh, PA (R.G.N.)
| | - Thanh N Nguyen
- Department of Neurology, Boston Medical Center, Boston University Chobanian and Avedisian School of Medicine, MA (T.N.N.)
| | - Robert L Medcalf
- Central Clinical School, Australian Centre for Blood Diseases, Monash University, Australia (R.L.M.)
| | - Joseph P Broderick
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH (E.A.M., J.P.B.)
- Gardner Neuroscience Institute, Cincinnati, OH (J.P.B.)
| | - Simon F De Meyer
- Laboratory for Thrombosis Research, KU Leuven Campus Kulak, Kortrijk, Belgium (S.V., S.F.D.M.)
| | - Bruce C V Campbell
- Department of Neurology, Melbourne Brain Centre at the Royal Melbourne Hospital, University of Melbourne, Australia (V.Y., B.C.V.C.)
| |
Collapse
|
2
|
Vandelanotte S, Staessens S, François O, De Wilde M, Desender L, De Sloovere AS, Dewaele T, Tersteeg C, Vanhoorelbeke K, Vanacker P, Andersson T, De Meyer SF. Association between thrombus composition and first-pass recanalization after thrombectomy in acute ischemic stroke. J Thromb Haemost 2024; 22:2555-2561. [PMID: 38897388 DOI: 10.1016/j.jtha.2024.05.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 05/27/2024] [Accepted: 05/30/2024] [Indexed: 06/21/2024]
Abstract
BACKGROUND Achieving first-pass recanalization (FPR) has become the primary procedural objective during thrombectomy in acute ischemic stroke patients as it correlates with the best clinical outcome. Understanding factors contributing to FPR failures is essential to enhance FPR success rates. As the central target of thrombectomy, the thrombus itself may be a significant factor influencing FPR. OBJECTIVES This study aimed to investigate the association between thrombus composition and FPR success rates. METHODS In total, thrombi from 267 ischemic stroke patients were collected in the AZ Groeninge Hospital (Kortrijk, Belgium). Thrombus composition was determined via detailed histologic analysis of red blood cells (RBCs), fibrin, von Willebrand factor, platelets, leukocytes, citrullinated histone 3 (marker for neutrophil extracellular traps), and intracellular and extracellular DNA. FPR was defined as obtaining a modified thrombolysis in cerebral infarction (mTICI) score of 2c/3 after the first pass. RESULTS An mTICI score of 2c/3 was obtained in 180 patients, which was achieved via a successful FPR procedure in 126 cases or after multiple passes in 54 cases. Interestingly, thrombi from FPR cases had a different composition from thrombi that needed multiple passes to obtain an mTICI score of 2c/3. FPR thrombi contained significantly more RBCs (P = .0264), less fibrin (P = .0196), and less extracellular DNA (P = .0457). CONCLUSION Our results indicate that thrombi characterized by lower RBC content, higher fibrin levels, and increased extracellular DNA are less likely to result in an FPR. These results are important to guide future research aiming at improving procedures or technologies to obtain FPR rates in RBC-poor thrombi.
Collapse
Affiliation(s)
| | - Senna Staessens
- Laboratory for Thrombosis Research, KU Leuven Kulak, Kortrijk, Belgium
| | | | - Maaike De Wilde
- Laboratory for Thrombosis Research, KU Leuven Kulak, Kortrijk, Belgium
| | - Linda Desender
- Laboratory for Thrombosis Research, KU Leuven Kulak, Kortrijk, Belgium
| | | | - Tom Dewaele
- Department of Medical Imaging, AZ Groeninge, Kortrijk, Belgium
| | - Claudia Tersteeg
- Laboratory for Thrombosis Research, KU Leuven Kulak, Kortrijk, Belgium
| | | | - Peter Vanacker
- Department of Neurology, AZ Groeninge, Kortrijk, Belgium; Department of Neurology, University Hospitals Antwerp, Antwerp, Belgium; Department of Translational Neuroscience, University of Antwerp, Antwerp, Belgium
| | - Tommy Andersson
- Department of Neuroradiology, Karolinska University Hospital, Stockholm, Sweden; Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Simon F De Meyer
- Laboratory for Thrombosis Research, KU Leuven Kulak, Kortrijk, Belgium.
| |
Collapse
|
3
|
Lou J, Zhang J, Deng Q, Chen X. Neutrophil extracellular traps mediate neuro-immunothrombosis. Neural Regen Res 2024; 19:1734-1740. [PMID: 38103239 PMCID: PMC10960287 DOI: 10.4103/1673-5374.389625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 10/14/2023] [Indexed: 12/18/2023] Open
Abstract
Neutrophil extracellular traps are primarily composed of DNA and histones and are released by neutrophils to promote inflammation and thrombosis when stimulated by various inflammatory reactions. Neutrophil extracellular trap formation occurs through lytic and non-lytic pathways that can be further classified by formation mechanisms. Histones, von Willebrand factor, fibrin, and many other factors participate in the interplay between inflammation and thrombosis. Neuro-immunothrombosis summarizes the intricate interplay between inflammation and thrombosis during neural development and the pathogenesis of neurological diseases, providing cutting-edge insights into post-neurotrauma thrombotic events. The blood-brain barrier defends the brain and spinal cord against external assaults, and neutrophil extracellular trap involvement in blood-brain barrier disruption and immunothrombosis contributes substantially to secondary injuries in neurological diseases. Further research is needed to understand how neutrophil extracellular traps promote blood-brain barrier disruption and immunothrombosis, but recent studies have demonstrated that neutrophil extracellular traps play a crucial role in immunothrombosis, and identified modulators of neuro-immunothrombosis. However, these neurological diseases occur in blood vessels, and the mechanisms are unclear by which neutrophil extracellular traps penetrate the blood-brain barrier to participate in immunothrombosis in traumatic brain injury. This review discusses the role of neutrophil extracellular traps in neuro-immunothrombosis and explores potential therapeutic interventions to modulate neutrophil extracellular traps that may reduce immunothrombosis and improve traumatic brain injury outcomes.
Collapse
Affiliation(s)
- Jianbo Lou
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Jianning Zhang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Neurological Institute, Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, China
| | - Quanjun Deng
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Xin Chen
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Neurological Institute, Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, China
| |
Collapse
|
4
|
Schuermans S, Kestens C, Marques PE. Systemic mechanisms of necrotic cell debris clearance. Cell Death Dis 2024; 15:557. [PMID: 39090111 PMCID: PMC11294570 DOI: 10.1038/s41419-024-06947-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 07/23/2024] [Accepted: 07/24/2024] [Indexed: 08/04/2024]
Abstract
Necrosis is an overarching term that describes cell death modalities caused by (extreme) adverse conditions in which cells lose structural integrity. A guaranteed consequence of necrosis is the production of necrotic cell remnants, or debris. Necrotic cell debris is a strong trigger of inflammation, and although inflammatory responses are required for tissue healing, necrotic debris may lead to uncontrolled immune responses and collateral damage. Besides local phagocytosis by recruited leukocytes, there is accumulating evidence that extracellular mechanisms are also involved in necrotic debris clearance. In this review, we focused on systemic clearance mechanisms present in the bloodstream and vasculature that often cooperate to drive the clearance of cell debris. We reviewed the contribution and cooperation of extracellular DNases, the actin-scavenger system, the fibrinolytic system and reticuloendothelial cells in performing clearance of necrotic debris. Moreover, associations of the (mis)functioning of these clearance systems with a variety of diseases were provided, illustrating the importance of the mechanisms of clearance of dead cells in the organism.
Collapse
Affiliation(s)
- Sara Schuermans
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Caine Kestens
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Pedro Elias Marques
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium.
| |
Collapse
|
5
|
Stanton K, Philippou H, Ariëns RA. Ischaemic Stroke, Thromboembolism and Clot Structure. Neuroscience 2024; 550:3-10. [PMID: 38453129 DOI: 10.1016/j.neuroscience.2024.02.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 02/19/2024] [Accepted: 02/23/2024] [Indexed: 03/09/2024]
Abstract
Ischaemic stroke is a major cause of morbidity and mortality worldwide. Blood clotting and thromboembolism play a central role in the pathogenesis of ischaemic stroke. An increasing number of recent studies indicate changes in blood clot structure and composition in patients with ischaemic stroke. In this review, we aim to summarise and discuss clot structure, function and composition in ischaemic stroke, including its relationships with clinical diagnosis and treatment options such as thrombolysis and thrombectomy. Studies are summarised in which clot structure and composition is analysed both in vitro from patients' plasma samples and ex vivo in thrombi obtained through interventional catheter-mediated thrombectomy. Mechanisms that drive clot composition and architecture such as neutrophil extracellular traps and clot contraction are also discussed. We find that, while in vitro clot structure in plasma samples from ischaemic stroke patients are consistently altered, showing denser clots that are more resistant to fibrinolysis, current data on the composition and architecture of ex vivo clots obtained by thrombectomy are more variable. With the potential of advances in technologies underpinning both the imaging and retrieving of clots, we expect that future studies in this area will generate new data that is of interest for the diagnosis, optimal treatment strategies and clinical management of patients with ischaemic stroke.
Collapse
Affiliation(s)
- Katherine Stanton
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
| | - Helen Philippou
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
| | - Robert As Ariëns
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK.
| |
Collapse
|
6
|
Vandelanotte S, De Meyer SF. Acute Ischemic Stroke Thrombus Composition. Neuroscience 2024; 550:11-20. [PMID: 38185279 DOI: 10.1016/j.neuroscience.2023.12.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 12/18/2023] [Accepted: 12/20/2023] [Indexed: 01/09/2024]
Abstract
Ischemic stroke is caused by a thrombus blocking one or multiple arteries in the brain, resulting in irreversible damage in the associated brain tissue. The aim of therapy is to restore the blood flow as fast as possible. Two recanalization strategies are currently available: pharmacological thrombolysis using recombinant tissue plasminogen activator (rt-PA) and mechanical removal of the thrombus. Despite recent advancements, achieving efficient recanalization remains a challenge. The precise causes of therapy failure are not fully understood but thrombus composition is likely a key factor in successful recanalization. This review explores acute ischemic stroke thrombus composition, its recently identified components, and how it affects stroke treatment. It also discusses how new insights could enhance current recanalization strategies for ischemic stroke patients.
Collapse
Affiliation(s)
| | - Simon F De Meyer
- Laboratory for Thrombosis Research, KU Leuven Kulak, Kortrijk, Belgium.
| |
Collapse
|
7
|
Vandelanotte S, François O, Desender L, Staessens S, Vanhoorne A, Van Gool F, Tersteeg C, Vanhoorelbeke K, Vanacker P, Andersson T, De Meyer SF. R-tPA Resistance Is Specific for Platelet-Rich Stroke Thrombi and Can Be Overcome by Targeting Nonfibrin Components. Stroke 2024; 55:1181-1190. [PMID: 38525646 DOI: 10.1161/strokeaha.123.045880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 02/15/2024] [Indexed: 03/26/2024]
Abstract
BACKGROUND Resistance to r-tPA (recombinant tissue-type plasminogen activator) is a well-known but poorly understood phenomenon that hampers successful recanalization in patients with acute ischemic stroke. Using clinically relevant thrombi from patients with acute ischemic stroke, we investigated if and how thrombus composition impacts r-tPA-mediated lysis. In addition, we explored strategies to overcome r-tPA resistance. METHODS Thrombi were split into 2 parts, 1 of which was used for thrombolysis and the other for detailed histological analysis. Thrombolysis was performed in normal human plasma using r-tPA alone, using r-tPA in combination with DNase-1 or using r-tPA in combination with N,N'-diacetyl-l-cystine. Thrombus lysis was calculated as the percentage of residual thrombus weight compared with its initial weight and the degree of lysis was linked to thrombus composition determined via histology. RESULTS Interestingly, we found that the efficacy of r-tPA-mediated thrombolysis was strongly correlated with the composition of the thrombi. Thrombi containing high amounts of red blood cells and low amounts of DNA and von Willebrand Factor were efficiently degraded by r-tPA, whereas thrombi containing low amounts of red blood cells and higher amounts of DNA and von Willebrand Factor were resistant to r-tPA. Importantly, combination of r-tPA with DNase-1 or N,N'-diacetyl-l-cystine significantly and specifically improved the lysis of these r-tPA-resistant thrombi. CONCLUSIONS Using patient thrombus material, our results for the first time show that the composition of stroke thrombi largely determines their susceptibility to r-tPA-mediated thrombolysis. Red blood cell-poor thrombi have a specific resistance to r-tPA, which can be overcome by targeting nonfibrin components using DNase-1 or N,N'-diacetyl-l-cystine.
Collapse
Affiliation(s)
- Sarah Vandelanotte
- Laboratory for Thrombosis Research, KU Leuven Kulak, Kortrijk, Belgium (S.V., L.D., S.S., C.T., K.V., S.F.D.M.)
| | - Olivier François
- Departments of Medical Imaging (O.F., F.V.G., T.A.), AZ Groeninge, Kortrijk, Belgium
| | - Linda Desender
- Laboratory for Thrombosis Research, KU Leuven Kulak, Kortrijk, Belgium (S.V., L.D., S.S., C.T., K.V., S.F.D.M.)
| | - Senna Staessens
- Laboratory for Thrombosis Research, KU Leuven Kulak, Kortrijk, Belgium (S.V., L.D., S.S., C.T., K.V., S.F.D.M.)
| | | | - Fréderick Van Gool
- Departments of Medical Imaging (O.F., F.V.G., T.A.), AZ Groeninge, Kortrijk, Belgium
| | - Claudia Tersteeg
- Laboratory for Thrombosis Research, KU Leuven Kulak, Kortrijk, Belgium (S.V., L.D., S.S., C.T., K.V., S.F.D.M.)
| | - Karen Vanhoorelbeke
- Laboratory for Thrombosis Research, KU Leuven Kulak, Kortrijk, Belgium (S.V., L.D., S.S., C.T., K.V., S.F.D.M.)
| | - Peter Vanacker
- Neurology (A.V., P.V.), AZ Groeninge, Kortrijk, Belgium
- Department of Translational Neuroscience, University of Antwerp, Belgium (P.V.)
| | - Tommy Andersson
- Departments of Medical Imaging (O.F., F.V.G., T.A.), AZ Groeninge, Kortrijk, Belgium
- Department of Neuroradiology, Karolinska University Hospital, and Clinical Neuroscience Karolinska Institutet, Stockholm, Sweden (T.A.)
| | - Simon F De Meyer
- Laboratory for Thrombosis Research, KU Leuven Kulak, Kortrijk, Belgium (S.V., L.D., S.S., C.T., K.V., S.F.D.M.)
| |
Collapse
|
8
|
Hajdu KS, Baker CE, Moore-Lotridge SN, Schoenecker JG. Sequestration and Involucrum: Understanding Bone Necrosis and Revascularization in Pediatric Orthopedics. Orthop Clin North Am 2024; 55:233-246. [PMID: 38403369 DOI: 10.1016/j.ocl.2023.09.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
Sequestration, a condition where a section of bone becomes necrotic due to a loss of vascularity or thrombosis, can be a challenging complication of osteomyelitis. This review explores the pathophysiology of sequestration, highlighting the role of the periosteum in forming involucrum and creeping substitution which facilitate revascularization and bone formation. The authors also discuss the induced membrane technique, a two-stage surgical procedure for cases of failed healing of sequestration. Future directions include the potential use of prophylactic anticoagulation and novel drugs targeting immunocoagulopathy, as well as the development of advanced imaging techniques and single-stage surgical procedures.
Collapse
Affiliation(s)
- Katherine S Hajdu
- School of Medicine, Vanderbilt University, Nashville, Tennessee, USA
| | - Courtney E Baker
- Department of Orthopedics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Stephanie N Moore-Lotridge
- Department of Orthopedics, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Vanderbilt Center for Bone Biology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Jonathan G Schoenecker
- Department of Orthopedics, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Vanderbilt Center for Bone Biology, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Department of Pharmacology, Vanderbilt University, Nashville, Tennessee, USA; Department of Pediatrics, Monroe Carell Jr. Children's Hospital at Vanderbilt, Nashville, Tennessee, USA.
| |
Collapse
|
9
|
Hetland G, Fagerhol MK, Mirlashari MR, Nissen-Meyer LSH, Croci S, Lonati PA, Bonacini M, Salvarani C, Marvisi C, Bodio C, Muratore F, Borghi MO, Meroni PL. Elevated NET, Calprotectin, and Neopterin Levels Discriminate between Disease Activity in COVID-19, as Evidenced by Need for Hospitalization among Patients in Northern Italy. Biomedicines 2024; 12:766. [PMID: 38672123 PMCID: PMC11048478 DOI: 10.3390/biomedicines12040766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 03/25/2024] [Accepted: 03/28/2024] [Indexed: 04/28/2024] Open
Abstract
Coronavirus disease 2019 (COVID-19) displays clinical heterogeneity, but little information is available for patients with mild or very early disease. We aimed to characterize biomarkers that are useful for discriminating the hospitalization risk in a COVID-19 cohort from Northern Italy during the first pandemic wave. We enrolled and followed for four weeks 76 symptomatic SARS-CoV-2 positive patients and age/sex-matched healthy controls. Patients with mild disease were discharged (n.42), and the remaining patients were hospitalized (n.34). Blood was collected before any anti-inflammatory/immunosuppressive therapy and assessed for soluble C5b-9/C5a, H3-neutrophil extracellular traps (NETs), calprotectin, and DNase plasma levels via ELISA and a panel of proinflammatory cytokines via ELLA. Calprotectin and NET levels discriminate between hospitalized and non-hospitalized patients, while DNase negatively correlates with NET levels; there are positive correlations between calprotectin and both NET and neopterin levels. Neopterin levels increase in patients at the beginning of the disease and do so more in hospitalized than non-hospitalized patients. C5a and sC5b-9, and other acute phase proteins, correlate with neopterin, calprotectin, and DNase. Both NET and neopterin levels negatively correlate with platelet count. We show that calprotectin, NETs, and neopterin are important proinflammatory parameters potentially useful for discriminating between COVID-19 patients at risk of hospitalization.
Collapse
Affiliation(s)
- Geir Hetland
- Department of Immunology and Transfusion Medicine, Oslo University Hospital Ullevål, 0450 Oslo, Norway; (G.H.); (M.K.F.); (M.R.M.); (L.S.H.N.-M.)
- Department of Immunology, Institute of Clinical Medicine, University of Oslo, 0451 Oslo, Norway
| | - Magne Kristoffer Fagerhol
- Department of Immunology and Transfusion Medicine, Oslo University Hospital Ullevål, 0450 Oslo, Norway; (G.H.); (M.K.F.); (M.R.M.); (L.S.H.N.-M.)
- Department of Immunology, Institute of Clinical Medicine, University of Oslo, 0451 Oslo, Norway
| | - Mohammad Reza Mirlashari
- Department of Immunology and Transfusion Medicine, Oslo University Hospital Ullevål, 0450 Oslo, Norway; (G.H.); (M.K.F.); (M.R.M.); (L.S.H.N.-M.)
| | - Lise Sofie Haug Nissen-Meyer
- Department of Immunology and Transfusion Medicine, Oslo University Hospital Ullevål, 0450 Oslo, Norway; (G.H.); (M.K.F.); (M.R.M.); (L.S.H.N.-M.)
| | - Stefania Croci
- Clinical Immunology, Allergy and Advanced Biotechnologies Unit, Azienda USL-IRCCS di Reggio Emilia, 42123 Reggio Emilia, Italy; (S.C.); (M.B.)
| | - Paola Adele Lonati
- Research Laboratory of Immunorheumatology, IRCCS Istituto Auxologico Italiano, 20095 Cusano Milanino, Italy; (P.A.L.); (C.B.); or (M.O.B.)
| | - Martina Bonacini
- Clinical Immunology, Allergy and Advanced Biotechnologies Unit, Azienda USL-IRCCS di Reggio Emilia, 42123 Reggio Emilia, Italy; (S.C.); (M.B.)
| | - Carlo Salvarani
- Azienda USL-IRCCS di Reggio Emilia e Università di Modena e Reggio Emilia, 42123 Reggio Emilia, Italy; (C.S.); (C.M.); (F.M.)
| | - Chiara Marvisi
- Azienda USL-IRCCS di Reggio Emilia e Università di Modena e Reggio Emilia, 42123 Reggio Emilia, Italy; (C.S.); (C.M.); (F.M.)
| | - Caterina Bodio
- Research Laboratory of Immunorheumatology, IRCCS Istituto Auxologico Italiano, 20095 Cusano Milanino, Italy; (P.A.L.); (C.B.); or (M.O.B.)
| | - Francesco Muratore
- Azienda USL-IRCCS di Reggio Emilia e Università di Modena e Reggio Emilia, 42123 Reggio Emilia, Italy; (C.S.); (C.M.); (F.M.)
| | - Maria Orietta Borghi
- Research Laboratory of Immunorheumatology, IRCCS Istituto Auxologico Italiano, 20095 Cusano Milanino, Italy; (P.A.L.); (C.B.); or (M.O.B.)
- Department of Clinical Sciences and Community Health, University of Milan, 20122 Milan, Italy
| | - Pier Luigi Meroni
- Research Laboratory of Immunorheumatology, IRCCS Istituto Auxologico Italiano, 20095 Cusano Milanino, Italy; (P.A.L.); (C.B.); or (M.O.B.)
| |
Collapse
|
10
|
Takeuchi H. The Standard-Dose Heparin-Warfarin Remedy Partially Resolves Thrombi in the Right Superior Pulmonary Vein and Left Atrium and Ameliorates Type 2 Diabetes Mellitus. Cureus 2024; 16:e57323. [PMID: 38559512 PMCID: PMC10981902 DOI: 10.7759/cureus.57323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/31/2024] [Indexed: 04/04/2024] Open
Abstract
Pulmonary vein thrombosis is common and underdiagnosed. Previously, we reported several cases of pulmonary vein thrombi (PVTs) using cardiac computed tomography (CT) and transesophageal echocardiography (TEE). We reported that warfarin and direct oral anticoagulants (DOACs) partially resolved PVTs; however, it is difficult to resolve all PVTs completely. Therefore, we evaluated the effects of standard-dose heparin-warfarin remedy on PVTs and left atrium (LA) thrombi using TEE and cardiac CT. A 64-year-old male with type 2 diabetes mellitus (T2DM) and hypertension was assessed for thrombi in the LA and pulmonary veins using TEE and 80-slice multidetector computed tomography (80-MDCT). After one month of standard-dose heparin-warfarin remedy, the patient's right superior pulmonary vein (RSPV) thrombi and expanded LA thrombi from the RSPV thrombi had partially resolved. The RSPV thrombi and the expanded LA thrombi from the RSPV thrombi were detected using cardiac CT and TEE; however, they were depicted as black areas on TEE. They periodically moved inward with the patient's heartbeats. Additionally, the standard-dose heparin-warfarin remedy ameliorated the patient's T2DM, and the remedy effect could be maintained for five months to some extent by administering a standard dose of warfarin. The standard-dose heparin-warfarin remedy can ameliorate not only T2DM but also diabetic complications such as diabetic nephropathy and gestational diabetes mellitus.
Collapse
Affiliation(s)
- Hidekazu Takeuchi
- Internal Medicine and Cardiology, Takeuchi Naika Clinic, Ogachi-Gun, JPN
| |
Collapse
|
11
|
Ullah M, Mirshahi S, Valinattaj Omran A, Aldybiat I, Crepaux S, Soria J, Contant G, Pocard M, Mirshahi M. Blood Clot Dynamics and Fibrinolysis Impairment in Cancer: The Role of Plasma Histones and DNA. Cancers (Basel) 2024; 16:928. [PMID: 38473289 DOI: 10.3390/cancers16050928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 02/05/2024] [Accepted: 02/14/2024] [Indexed: 03/14/2024] Open
Abstract
BACKGROUND Blood viscoelasticity and plasma protein levels can play an important role in the diagnosis and prognosis of cancer. However, the role of histones and DNA in modulating blood clot properties remains to be investigated. This study investigates the differences in blood viscoelasticity and plasma protein levels among cancer patients, individuals with other diseases, and healthy individuals. METHODS Blood samples were collected from 101 participants, including 45 cancer patients, 22 healthy individuals, and 34 individuals with other diseases. Rheological properties of clots formed in vitro by reconstituted elements of fibrinogen or plasma were analyzed with an Anton Paar Rheometer, USA. Plasma protein levels of D-dimer, TPA, EPCR, fibrinogen, and histone H3 were measured through ELISA. Blood clots were formed with or without DNA and histones (H3) by adding thrombin and calcium to plasma samples, and were evaluated for viscoelasticity, permeability, and degradation. RESULTS Cancer patients show higher blood viscoelasticity and plasma D-dimer levels compared to healthy individuals and individuals with other diseases. Our in vitro analysis showed that the addition of histone to the plasma results in a significant decrease in viscoelasticity and mean fiber thickness of the clot formed thereafter. In parallel studies, using plasma from patients, DNA and histones were detected in fibrin clots and were associated with less degradation by t-PA. Moreover, our results show that the presence of DNA and histones not only increases clots' permeability, but also makes them more prone to degradation. CONCLUSIONS Plasma histones and DNA affect the structure of the clot formed and induce defective fibrinolysis. Moreover, the increased viscoelastic properties of plasma from cancer patients can be used as potential biomarkers in cancer prognosis.
Collapse
Affiliation(s)
- Matti Ullah
- CAP-Paris Tech., INSERM U1275, Université Paris Cité, Hôpital Lariboisière, 75010 Paris, France
- Faculty of Pharmacy, Hamdard University, Islamabad Campus, Islamabad 45550, Pakistan
| | | | - Azadeh Valinattaj Omran
- CAP-Paris Tech., INSERM U1275, Université Paris Cité, Hôpital Lariboisière, 75010 Paris, France
- Laboratoire des Sciences des Procédés et des Matériaux, Centre National de la Recherche Scientifique (UPR 3407), Université Sorbonne Paris Nord, 93430 Villetaneuse, France
| | - Iman Aldybiat
- CAP-Paris Tech., INSERM U1275, Université Paris Cité, Hôpital Lariboisière, 75010 Paris, France
| | - Sullyvan Crepaux
- CAP-Paris Tech., INSERM U1275, Université Paris Cité, Hôpital Lariboisière, 75010 Paris, France
- Prospective Research, Diagnostica Stago, 92230 Gennevilliers, France
| | - Jeannette Soria
- CAP-Paris Tech., INSERM U1275, Université Paris Cité, Hôpital Lariboisière, 75010 Paris, France
| | - Geneviève Contant
- Prospective Research, Diagnostica Stago, 92230 Gennevilliers, France
| | - Marc Pocard
- CAP-Paris Tech., INSERM U1275, Université Paris Cité, Hôpital Lariboisière, 75010 Paris, France
| | - Massoud Mirshahi
- CAP-Paris Tech., INSERM U1275, Université Paris Cité, Hôpital Lariboisière, 75010 Paris, France
| |
Collapse
|
12
|
Gando S, Levi M, Toh CH. Trauma-induced innate immune activation and disseminated intravascular coagulation. J Thromb Haemost 2024; 22:337-351. [PMID: 37816463 DOI: 10.1016/j.jtha.2023.09.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 09/25/2023] [Accepted: 09/26/2023] [Indexed: 10/12/2023]
Abstract
Dysregulated innate immunity participates in the pathomechanisms of disseminated intravascular coagulation (DIC) in trauma-induced coagulopathy. Accidental and regulated cell deaths and neutrophil extracellular traps release damage-associated molecular patterns (DAMPs), such as histones, nuclear and mitochondrial DNA, and high-mobility group box 1, into circulation immediately after trauma. DAMP-induced inflammation activation releases tissue factor-bearing procoagulant extracellular vesicles through gasdermin D-mediated pore formation and plasma membrane rupture by regulated cell death. DAMPs also evoke systemic inflammation, platelet, coagulation activation, and impaired fibrinolysis associated with endothelial injury, leading to the dysfunction of anticoagulation systems, which are the main pathophysiological mechanisms of DIC. All these processes induce systemic thrombin generation in vivo, not restricted to the injury sites immediately after trauma. Thrombin generation at the site of injury stops bleeding and maintains homeostasis. However, DIC associated with endothelial injury generates massive thrombin, enhancing protease-activated, receptor-mediated bidirectional interplays between inflammation and coagulation, aggravating the diverse actions of thrombin and disturbing homeostasis. Insufficiently regulated thrombin causes disseminated microvascular thrombosis, resulting in tissue hypoxia due to reduced oxygen delivery, and mitochondrial dysfunction due to DAMPs causes tissue dysoxia. In addition, DAMP-induced calcium influx and overload, as well as neutrophil activation, play a role in endothelial cell injury. Tissue hypoxia and cytotoxicity result in multiple organ dysfunction in DIC after trauma. Controls against dysregulated innate immunity evoking systemic inflammation, thrombin generation, and cytotoxicity are key issues in improving the prognosis of DIC in trauma-induced coagulopathy.
Collapse
Affiliation(s)
- Satoshi Gando
- Department of Acute and Critical Care Medicine, Sapporo Higashi Tokushukai Hospital, Sapporo, Japan; Division of Acute and Critical Care Medicine, Department of Anesthesiology and Critical Care Medicine, Hokkaido University Faculty of Medicine, Sapporo, Japan.
| | - Marcel Levi
- Department of Vascular Medicine, Amsterdam University Medical Centers, Amsterdam, The Netherlands; Department of Medicine, University College London Hospitals NHS Foundation Trust, and Cardio-Metabolic Program - NIHR UCLH/UCL BRC London, London, United Kingdom
| | - Cheng-Hock Toh
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, Liverpool, United Kingdom; Liverpool University Hospitals NHS Foundation Trust, Liverpool, United Kingdom
| |
Collapse
|
13
|
Li Y, Shu Y, Yu K, Ni R, Chu L. Analysis and Regulatory Mechanisms of Platelet-Related Genes in Patients with Ischemic Stroke. Cell Mol Neurobiol 2024; 44:15. [PMID: 38175286 DOI: 10.1007/s10571-023-01433-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 11/26/2023] [Indexed: 01/05/2024]
Abstract
It was found that ischemic stroke (IS) was associated with abnormal platelet activity and thrombosis. However, the potential significance of platelet-related genes (PRGs) in IS still needs to be more thorough. This study extracted IS-related transcriptome datasets from the Gene Expression Omnibus (GEO) database. The target genes were obtained by intersecting the differentially expressed genes (DEGs), the module genes related to IS, and PRGs, where the key genes of IS were screened by two machine learning algorithms. The key genes-based diagnostic model was constructed. Gene set enrichment analysis (GSEA) and the immune microenvironment analyses were analyzed targeting key genes in IS. The co-expression, TF-mRNA, and competitive endogenous RNAs (ceRNA) regulatory networks were constructed to reveal the potential regulation of key genes. Potential drugs targeting key genes were predicted as well. Totals of eight target genes were obtained and were associated with immune-related functions. Four platelet-related key genes were acquired, which were related to immunity and energy metabolism. The abnormal expressions of DOCK8, GIMAP5, ICOS were determined by the quantitative real-time polymerase chain reaction (qRT-PCR), and the significant correlations among these key genes were identified. Notably, hsa-miR-17-3p, hsa-miR-3158-3p, hsa-miR-423-3p, and hsa-miR-193a-8p could regulate all key genes at the same time. In addition, Caffeine, Carboplatin, and Vopratelimab were the targeted drugs of these key genes. This study identified four platelet-related key genes of IS, which might help to deepen the understanding of the role of platelet-related genes in the molecular mechanism of IS.
Collapse
Affiliation(s)
- Yuan Li
- Department of Neurology, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China
| | - Yuanlu Shu
- Department of Neurology, Guizhou Medical University, Guiyang, 550004, China
| | - Kun Yu
- Department of Neurology, Guizhou Medical University, Guiyang, 550004, China
| | - Ruihan Ni
- Department of Neurology, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China
| | - Lan Chu
- Department of Neurology, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China.
- Department of Neurology, Guizhou Medical University, Guiyang, 550004, China.
| |
Collapse
|
14
|
Yao MM, Lin TT, Shi X, Chen MG, Wu JX, Zhao YP, Lin BS. Thrombosis density ratio can predict the occurrence of pulmonary embolism and post-thrombotic syndrome in lower-extremity deep vein thrombosis patients. Clin Hemorheol Microcirc 2024; 86:395-405. [PMID: 38073380 PMCID: PMC11091615 DOI: 10.3233/ch-231778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/12/2024]
Abstract
OBJECTIVE Deep vein thrombosis (DVT) formation of lower extremities can lead to serious complications including pulmonary embolism (PE) and chronic post-thrombotic syndrome (PTS). We aimed to explore the relationship between the ratio of thrombotic density and the occurrence of PE and PTS in patients with DVT of the lower extremities. METHODS A retrospective analysis was conducted in patients who performed computed tomography venography, dividing into DVT with PE group (54 patients) and DVT-alone group (34 patients), The clinical data were recorded. Univariate and multivariate logistic regression analysis were used to analysis variables associated with PE. The ability of thrombosis density ratio and Wells score to diagnose PE was evaluated by using the receiver operating characteristic curve (ROC) area under the curve (AUC). According to the treatment and follow-up results, subgroup analysis was performed, and the Villata score was used to determine the presence or absence of PTS and its severity. RESULTS Compare with the DVT-alone group, more patients had dyspnea and chest pain in the DVT with PE group. DVT with PE group had lower the percentage of neutrophils, white blood cell count and platelet count, while had higher blood cell count, D-dimer, wells score, thrombus and thrombus density ratio. Multivariate logistic analysis showed that percentage of neutrophils (OR(95% CIs)=1.15 (1.01,1.31), P = 0.040), platelets (OR(95% CIs)=0.96 (0.93,0.99), P = 0.011), and thrombus density ratio (OR(95% CIs)=5.99 (1.96,18.35), P = 0.002) are independent predictors of PE. The Wells score and thrombosis density ratio were consistent in the diagnostic efficacy of PE. In the subgroup analysis, there was a relevance between the ratio of thrombosis density and the Villalta score. CONCLUSION Percentage of neutrophils, platelets, and thrombus density ratio are independent predictors of PE. The thrombosis density of DVT patients may be an index to predict the risk of PE and PTS in DVT patients.
Collapse
Affiliation(s)
- Miao-Miao Yao
- Imaging Teaching and Research Office, School of Medicine, Quanzhou Medical College, Quanzhou, Fujian, China
| | - Ting-Ting Lin
- Training Teaching and Research Section, School of Medicine, Quanzhou Medical College, Quanzhou, Fujian, China
| | - Xu Shi
- Imaging Teaching and Research Office, School of Medicine, Quanzhou Medical College, Quanzhou, Fujian, China
| | - Mei-Gui Chen
- Department of Imaging, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Jia-Xiang Wu
- Imaging Teaching and Research Office, School of Medicine, Quanzhou Medical College, Quanzhou, Fujian, China
| | - Yan-Ping Zhao
- Imaging Teaching and Research Office, School of Medicine, Quanzhou Medical College, Quanzhou, Fujian, China
| | - Bao-Shan Lin
- Department of Child Health, Quanzhou Women’s and Children’s Hospital, Quanzhou, Fujian, China
| |
Collapse
|
15
|
Chanchal S, Sharma S, Mohd S, Sultan A, Mishra A, Ashraf MZ. Unraveling Epigenetic Interplay between Inflammation, Thrombosis, and Immune-Related Disorders through a Network Meta-analysis. TH OPEN 2024; 8:e81-e92. [PMID: 38313596 PMCID: PMC10837039 DOI: 10.1055/a-2222-9126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 09/25/2023] [Indexed: 02/06/2024] Open
Abstract
Inflammation and thrombosis are two distinct yet interdependent physiological processes. The inflammation results in the activation of the coagulation system that directs the immune system and its activation, resulting in the initiation of the pathophysiology of thrombosis, a process termed immune-thrombosis. Still, the shared underlying molecular mechanism related to the immune system and coagulation has not yet been explored extensively. Inspired to answer this, we carried out a comprehensive gene expression meta-analysis using publicly available datasets of four diseases, including venous thrombosis, systemic lupus erythematosus, rheumatoid arthritis, and inflammatory bowel disease. A total of 609 differentially expressed genes (DEGs) shared by all four datasets were identified based on the combined effect size approach. The pathway enrichment analysis of the DEGs showed enrichment of various epigenetic pathways such as histone-modifying enzymes, posttranslational protein modification, chromatin organization, chromatin-modifying enzymes, HATs acetylate proteins. Network-based protein-protein interaction analysis showed epigenetic enzyme coding genes dominating among the top hub genes. The miRNA-interacting partner of the top 10 hub genes was determined. The predomination of epitranscriptomics regulation opens a layout for the meta-analysis of miRNA datasets of the same four diseases. We identified 30 DEmiRs shared by these diseases. There were 9 common DEmiRs selected from the list of miRNA-interacting partners of top 10 hub genes and shared significant DEmiRs from microRNAs dataset acquisition. These common DEmiRs were found to regulate genes involved in epigenetic modulation and indicate a promising epigenetic aspect that needs to be explored for future molecular studies in the context of immunothrombosis and inflammatory disease.
Collapse
Affiliation(s)
- Shankar Chanchal
- Department of Biotechnology, Faculty of Natural Sciences, Jamia Millia Islamia, Delhi, India
| | - Swati Sharma
- Department of Biotechnology, Faculty of Natural Sciences, Jamia Millia Islamia, Delhi, India
| | - Syed Mohd
- Department of Biotechnology, Faculty of Natural Sciences, Jamia Millia Islamia, Delhi, India
| | - Armiya Sultan
- Department of Biotechnology, Faculty of Natural Sciences, Jamia Millia Islamia, Delhi, India
| | - Aastha Mishra
- Cardio Respiratory Disease unit, CSIR- Institute of Genomics and Integrative Biology, Delhi, India
| | - Mohammad Zahid Ashraf
- Department of Biotechnology, Faculty of Natural Sciences, Jamia Millia Islamia, Delhi, India
| |
Collapse
|
16
|
Yong J, Toh CH. Rethinking coagulation: from enzymatic cascade and cell-based reactions to a convergent model involving innate immune activation. Blood 2023; 142:2133-2145. [PMID: 37890148 DOI: 10.1182/blood.2023021166] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 10/12/2023] [Accepted: 10/20/2023] [Indexed: 10/29/2023] Open
Abstract
ABSTRACT Advancements in the conceptual thinking of hemostasis and thrombosis have been catalyzed by major developments within health research over several decades. The cascade model of coagulation was first described in the 1960s, when biochemistry gained prominence through innovative experimentation and technical developments. This was followed by the cell-based model, which integrated cellular coordination to the enzymology of clot formation and was conceptualized during the growth period in cell biology at the turn of the millennium. Each step forward has heralded a revolution in clinical therapeutics, both in procoagulant and anticoagulant treatments to improve patient care. In current times, the COVID-19 pandemic may also prove to be a catalyst: thrombotic challenges including the mixed responses to anticoagulant treatment and the vaccine-induced immune thrombotic thrombocytopenia have exposed limitations in our preexisting concepts while simultaneously demanding novel therapeutic approaches. It is increasingly clear that innate immune activation as part of the host response to injury is not separate but integrated into adaptive clot formation. Our review summarizes current understanding of the major molecules facilitating such a cross talk between immunity, inflammation and coagulation. We demonstrate how such effects can be layered upon the cascade and cell-based models to evolve conceptual understanding of the physiology of immunohemostasis and the pathology of immunothrombosis.
Collapse
Affiliation(s)
- Jun Yong
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, Liverpool, United Kingdom
- The Roald Dahl Haemostasis and Thrombosis Centre, Liverpool University Hospitals NHS Foundation Trust, Liverpool, United Kingdom
| | - Cheng-Hock Toh
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, Liverpool, United Kingdom
- The Roald Dahl Haemostasis and Thrombosis Centre, Liverpool University Hospitals NHS Foundation Trust, Liverpool, United Kingdom
| |
Collapse
|
17
|
Chooklin S, Chuklin S. The role of neutrophil extracellular traps in thrombosis. EMERGENCY MEDICINE 2023; 19:448-457. [DOI: 10.22141/2224-0586.19.7.2023.1627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
According to the cellular model of hemostasis, the process of blood coagulation is presented in the form of three phases: initiation, amplification and propagation, each of them includes several consecutive stages. At the same time, thrombus formation is often explained by Virchow’s triad: blood stasis, damage to the blood vessel walls, and hypercoagulation. Classically, the appearance of one of the three mentioned parameters can lead to thrombus formation. Over the past decade, our knowledge of the cross-talk between coagulation, inflammation, and innate immune activation and the involvement of neutrophil extracellular traps in these processes has expanded. This brief review shows their role in thrombosis through the mechanisms of activation of platelets, complement, interaction with blood coagulation factors and damage to the vascular endothelium. We searched the literature in the MEDLINE database on the PubMed platform.
Collapse
|
18
|
Ząbczyk M, Kruk A, Natorska J, Undas A. Low-grade endotoxemia in acute pulmonary embolism: Links with prothrombotic plasma fibrin clot phenotype. Thromb Res 2023; 232:70-76. [PMID: 37949000 DOI: 10.1016/j.thromres.2023.10.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 10/25/2023] [Accepted: 10/30/2023] [Indexed: 11/12/2023]
Abstract
BACKGROUND Lipopolysaccharide (LPS) can traverse the intestinal barrier and enter bloodstream, causing endotoxemia and triggering inflammation. Increased circulating LPS was reported in arterial thromboembolism. We investigated whether increased LPS levels occur in acute pulmonary embolism (PE) and if it is associated with a prothrombotic state. METHODS We studied 120 normotensive PE patients (aged 59 [48-68] years) on admission, after 5-7 days, and after a 3-month anticoagulation. Serum LPS levels, along with zonulin, a marker of gut permeability, endogenous thrombin potential (ETP), fibrin clot permeability (Ks), clot lysis time (CLT), fibrinolysis proteins, and platelet markers were assessed. RESULTS Median LPS concentration on admission was 70.5 (61.5-82) pg/mL (min-max, 34-134 pg/mL), in association with C-reactive protein (r = 0.22, p = 0.018), but not with fibrinogen, D-dimer or platelet markers. Patients with more severe PE had higher LPS levels compared with the remainder. Median zonulin level was 3.26 (2.74-4.08) ng/mL and correlated with LPS (r = 0.66, p < 0.0001). Patients with baseline LPS levels in the top quartile (≥82 pg/mL; n = 29) compared to lower quartiles had 18.6 % increased ETP, 14.5 % reduced Ks, and 25.3 % prolonged CLT, related to higher plasminogen activator inhibitor type 1 (PAI-1) levels. LPS decreased by 23.4 % after 5-7 days and by 40.4 % after 3-month anticoagulation together with reduced zonulin by 18.4 % and 22.3 %, respectively, compared to baseline (all p < 0.001). LPS levels were not related with fibrin characteristics and other variables assessed at 3 months. CONCLUSIONS Low-grade endotoxemia is detectable in patients with acute PE and may contribute to increased thrombin generation and PAI-1-mediated hypofibrinolysis.
Collapse
Affiliation(s)
- Michał Ząbczyk
- St. John Paul II Hospital, Kraków, Poland; Institute of Cardiology, Jagiellonian University Medical College, Kraków, Poland
| | | | - Joanna Natorska
- St. John Paul II Hospital, Kraków, Poland; Institute of Cardiology, Jagiellonian University Medical College, Kraków, Poland
| | - Anetta Undas
- St. John Paul II Hospital, Kraków, Poland; Institute of Cardiology, Jagiellonian University Medical College, Kraków, Poland.
| |
Collapse
|
19
|
Kumar R, Patil G, Dayal S. NLRP3-Induced NETosis: A Potential Therapeutic Target for Ischemic Thrombotic Diseases? Cells 2023; 12:2709. [PMID: 38067137 PMCID: PMC10706381 DOI: 10.3390/cells12232709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 11/22/2023] [Accepted: 11/24/2023] [Indexed: 12/18/2023] Open
Abstract
Ischemic thrombotic disease, characterized by the formation of obstructive blood clots within arteries or veins, is a condition associated with life-threatening events, such as stroke, myocardial infarction, deep vein thrombosis, and pulmonary embolism. The conventional therapeutic strategy relies on treatments with anticoagulants that unfortunately pose an inherent risk of bleeding complications. These anticoagulants primarily target clotting factors, often overlooking upstream events, including the release of neutrophil extracellular traps (NETs). Neutrophils are integral components of the innate immune system, traditionally known for their role in combating pathogens through NET formation. Emerging evidence has now revealed that NETs contribute to a prothrombotic milieu by promoting platelet activation, increasing thrombin generation, and providing a scaffold for clot formation. Additionally, NET components enhance clot stability and resistance to fibrinolysis. Clinical and preclinical studies have underscored the mechanistic involvement of NETs in the pathogenesis of thrombotic complications, since the clots obtained from patients and experimental models consistently exhibit the presence of NETs. Given these insights, the inhibition of NETs or NET formation is emerging as a promising therapeutic approach for ischemic thrombotic diseases. Recent investigations also implicate a role for the nucleotide-binding oligomerization domain (NOD)-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome as a mediator of NETosis and thrombosis, suggesting that NLRP3 inhibition may also hold potential for mitigating thrombotic events. Therefore, future preclinical and clinical studies aimed at identifying and validating NLRP3 inhibition as a novel therapeutic intervention for thrombotic disorders are imperative.
Collapse
Affiliation(s)
- Rahul Kumar
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA; (R.K.); (G.P.)
- Department of Biotechnology, GITAM School of Sciences, GITAM (Deemed to be) University, Visakhapatnam 530045, India
| | - Gokul Patil
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA; (R.K.); (G.P.)
| | - Sanjana Dayal
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA; (R.K.); (G.P.)
- Holden Comprehensive Cancer Center, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
- Iowa City VA Healthcare System, Iowa City, IA 52246, USA
| |
Collapse
|
20
|
Singh R, Gautam P, Sharma C, Osmolovskiy A. Fibrin and Fibrinolytic Enzyme Cascade in Thrombosis: Unravelling the Role. Life (Basel) 2023; 13:2196. [PMID: 38004336 PMCID: PMC10672518 DOI: 10.3390/life13112196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 10/30/2023] [Accepted: 11/02/2023] [Indexed: 11/26/2023] Open
Abstract
Blood clot formation in blood vessels (thrombosis) is a major cause of life-threatening cardiovascular diseases. These clots are formed by αA-, βB-, and ϒ-peptide chains of fibrinogen joined together by isopeptide bonds with the help of blood coagulation factor XIIIa. These clot structures are altered by various factors such as thrombin, platelets, transglutaminase, DNA, histones, and red blood cells. Various factors are used to dissolve the blood clot, such as anticoagulant agents, antiplatelets drugs, fibrinolytic enzymes, and surgical operations. Fibrinolytic enzymes are produced by microorganisms (bacteria, fungi, etc.): streptokinase of Streptococcus hemolyticus, nattokinase of Bacillus subtilis YF 38, bafibrinase of Bacillus sp. AS-S20-I, longolytin of Arthrobotrys longa, versiase of Aspergillus versicolor ZLH-1, etc. They act as a thrombolytic agent by either enhancing the production of plasminogen activators (tissue or urokinase types), which convert inactive plasminogen to active plasmin, or acting as plasmin-like proteins themselves, forming fibrin degradation products which cause normal blood flow again in blood vessels. Fibrinolytic enzymes may be classified in two groups, as serine proteases and metalloproteases, based on their catalytic properties, consisting of a catalytic triad responsible for their fibrinolytic activity having different physiochemical properties (such as molecular weight, pH, and temperature). The analysis of fibrinolysis helps to detect hyperfibrinolysis (menorrhagia, renal failure, etc.) and hypofibrinolysis (diabetes, obesity, etc.) with the help of various fibrinolytic assays such as a fibrin plate assay, fibrin microplate assay, the viscoelastic method, etc. These fibrinolytic activities serve as a key aspect in the recognition of numerous cardiovascular diseases and can be easily produced on a large scale with a short generation time by microbes and are less expensive.
Collapse
Affiliation(s)
- Rajni Singh
- Amity Institute of Microbial Technology, Amity University Uttar Pradesh, Noida 201301, India; (P.G.); (C.S.)
| | - Prerna Gautam
- Amity Institute of Microbial Technology, Amity University Uttar Pradesh, Noida 201301, India; (P.G.); (C.S.)
| | - Chhavi Sharma
- Amity Institute of Microbial Technology, Amity University Uttar Pradesh, Noida 201301, India; (P.G.); (C.S.)
| | | |
Collapse
|
21
|
Nurden AT. Molecular basis of clot retraction and its role in wound healing. Thromb Res 2023; 231:159-169. [PMID: 36008192 DOI: 10.1016/j.thromres.2022.08.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 08/08/2022] [Accepted: 08/10/2022] [Indexed: 11/30/2022]
Abstract
Clot retraction is important for the prevention of bleeding, in the manifestations of thrombosis and for tissue repair. The molecular mechanisms behind clot formation are complex. Platelet involvement begins with adhesion at sites of vessel injury followed by platelet aggregation, thrombin generation and fibrin production. Other blood cells incorporate into a fibrin mesh that is consolidated by FXIIIa-mediated crosslinking and platelet contractile activity. The latter results in the asymmetric redistribution of erythrocytes into a tighter central mass providing the clot with stability and resistance to fibrinolysis. Integrin αIIbβ3 on platelets is the key player in these events, bridging fibrin and the platelet cytoskeleton. Glycoprotein VI participates in thrombus formation but not in the retraction. Rheological and environmental factors influence clot construction with retraction driven by the platelet cytoskeleton with actomyosin acting as the motor. Activated platelets provide procoagulant activity stimulating thrombin generation together with the release of a plethora of biologically active proteins and substances from storage pools; many form chemotactic gradients within the fibrin or the underlying matrix. Also released are newly synthesized metabolites and lipid-rich vesicles that circulate within the vasculature and mimic platelet functions. Platelets and their released elements play key roles in wound healing. This includes promoting stem cell and mesenchymal stromal cell recruitment, fibroblast and endothelial cell migration, angiogenesis and matrix formation. These properties have led to the use of autologous clots in therapies designed to accelerate tissue repair while offering the potential for genetic manipulation in both inherited and acquired diseases.
Collapse
Affiliation(s)
- Alan T Nurden
- Institut Hospitalo-Universitaire LIRYC, Pessac, France.
| |
Collapse
|
22
|
Liu Y, Ma YH, Yang JW, Man JW, Wang HB, Li Y, Liang C, Cao JL, Chen SY, Li KP, Yang L. Rethinking neutrophil extracellular traps. Int Immunopharmacol 2023; 124:110834. [PMID: 37625368 DOI: 10.1016/j.intimp.2023.110834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 08/07/2023] [Accepted: 08/18/2023] [Indexed: 08/27/2023]
Abstract
Neutrophils are a major subset of leukocytes in human circulating blood. In some circumstances, neutrophils release neutrophil extracellular traps (NETs). lnitially, NETs were considered to have a strong antibacterial capacity. However, currently, NETs have been shown to have a pivotal impact on various diseases. Different stimulators induce the production of different types of NETs, and their biological functions and modes of clearance do not appear to be the same. In this review, we will discuss several important issues related to NETs in order to better understand the relationship between NETs and diseases, as well as how to utilize the characteristics of NETs for disease treatment.
Collapse
Affiliation(s)
- Yi Liu
- Department of Urology, The Second Hospital of Lanzhou University, Gansu Province Clinical Research Center for Urology, Second Clinical School Lanzhou University, China
| | - Yu-Hua Ma
- Department of Urology, The Second Hospital of Lanzhou University, Gansu Province Clinical Research Center for Urology, Second Clinical School Lanzhou University, China
| | - Jian-Wei Yang
- Department of Urology, The Second Hospital of Lanzhou University, Gansu Province Clinical Research Center for Urology, Second Clinical School Lanzhou University, China
| | - Jiang-Wei Man
- Department of Urology, The Second Hospital of Lanzhou University, Gansu Province Clinical Research Center for Urology, Second Clinical School Lanzhou University, China
| | - Hua-Bin Wang
- Department of Urology, The Second Hospital of Lanzhou University, Gansu Province Clinical Research Center for Urology, Second Clinical School Lanzhou University, China
| | - Yi Li
- Department of Urology, The Second Hospital of Lanzhou University, Gansu Province Clinical Research Center for Urology, Second Clinical School Lanzhou University, China
| | - Cheng Liang
- Department of Urology, The Second Hospital of Lanzhou University, Gansu Province Clinical Research Center for Urology, Second Clinical School Lanzhou University, China
| | - Jin-Long Cao
- Department of Urology, The Second Hospital of Lanzhou University, Gansu Province Clinical Research Center for Urology, Second Clinical School Lanzhou University, China
| | - Si-Yu Chen
- Department of Urology, The Second Hospital of Lanzhou University, Gansu Province Clinical Research Center for Urology, Second Clinical School Lanzhou University, China
| | - Kun-Peng Li
- Department of Urology, The Second Hospital of Lanzhou University, Gansu Province Clinical Research Center for Urology, Second Clinical School Lanzhou University, China
| | - Li Yang
- Department of Urology, The Second Hospital of Lanzhou University, Gansu Province Clinical Research Center for Urology, Second Clinical School Lanzhou University, China.
| |
Collapse
|
23
|
Islam MM, Takeyama N. Role of Neutrophil Extracellular Traps in Health and Disease Pathophysiology: Recent Insights and Advances. Int J Mol Sci 2023; 24:15805. [PMID: 37958788 PMCID: PMC10649138 DOI: 10.3390/ijms242115805] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 10/27/2023] [Accepted: 10/27/2023] [Indexed: 11/15/2023] Open
Abstract
Neutrophils are the principal trouper of the innate immune system. Activated neutrophils undergo a noble cell death termed NETosis and release a mesh-like structure called neutrophil extracellular traps (NETs) as a part of their defensive strategy against microbial pathogen attack. This web-like architecture includes a DNA backbone embedded with antimicrobial proteins like myeloperoxidase (MPO), neutrophil elastase (NE), histones and deploys in the entrapment and clearance of encountered pathogens. Thus NETs play an inevitable beneficial role in the host's protection. However, recent accumulated evidence shows that dysregulated and enhanced NET formation has various pathological aspects including the promotion of sepsis, pulmonary, cardiovascular, hepatic, nephrological, thrombotic, autoimmune, pregnancy, and cancer diseases, and the list is increasing gradually. In this review, we summarize the NET-mediated pathophysiology of different diseases and focus on some updated potential therapeutic approaches against NETs.
Collapse
Affiliation(s)
- Md Monirul Islam
- Department of Emergency and Critical Care Medicine, Aichi Medical University, Aichi 480-1195, Japan
- Department of Biochemistry and Biotechnology, University of Science and Technology Chittagong (USTC), Chattogram 4202, Bangladesh
| | - Naoshi Takeyama
- Department of Emergency and Critical Care Medicine, Aichi Medical University, Aichi 480-1195, Japan
| |
Collapse
|
24
|
Moore-Lotridge SN, Hajdu KS, Hou BQ, Gibson BHY, Schoenecker JG. Maintaining the balance: the critical role of plasmin activity in orthopedic surgery injury response. J Thromb Haemost 2023; 21:2653-2665. [PMID: 37558131 PMCID: PMC10926148 DOI: 10.1016/j.jtha.2023.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 08/01/2023] [Accepted: 08/01/2023] [Indexed: 08/11/2023]
Abstract
The musculoskeletal system plays vital roles in the body, facilitating movement, protecting vital structures, and regulating hematopoiesis and mineral metabolism. Injuries to this system are common and can cause chronic pain, loss of range of motion, and disability. The acute phase response (APR) is a complex process necessary for surviving and repairing injured musculoskeletal tissue. To conceptualize the APR, it is useful to divide it into 2 distinct phases, survival and repair. During the survival-APR, a "damage matrix" primarily composed of fibrin, via thrombin activity, is produced to contain the zone of injury. Once containment is achieved, the APR transitions to the repair phase, where reparative inflammatory cells use plasmin to systematically remove the damage matrix and replace it with new permanent matrices produced by differentiated mesenchymal stem cells. The timing of thrombin and plasmin activation during their respective APR phases is crucial for appropriate regulation of the damage matrix. This review focuses on evidence indicating that inappropriate exuberant activation of plasmin during the survival-APR can result in an overactive APR, leading to an "immunocoagulopathy" that may cause "immunothrombosis" and death. Conversely, preclinical data suggest that too little plasmin activity during the repair-APR may contribute to failed tissue repair, such as a fracture nonunion, and chronic inflammatory degenerative diseases like osteoporosis. Future clinical studies are required to affirm these findings. Therefore, the temporal-spatial functions of plasmin in response to musculoskeletal injury and its pharmacologic manipulation are intriguing new targets for improving orthopedic care.
Collapse
Affiliation(s)
- Stephanie N Moore-Lotridge
- Department of Orthopaedics, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Center for Bone Biology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Katherine S Hajdu
- School of Medicine, Vanderbilt University, Nashville, Tennessee, USA
| | - Brian Q Hou
- School of Medicine, Vanderbilt University, Nashville, Tennessee, USA
| | - Breanne H Y Gibson
- Department of Orthopaedics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Jonathan G Schoenecker
- Department of Orthopaedics, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Center for Bone Biology, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Department of Pharmacology, Vanderbilt University, Nashville, Tennessee, USA.
| |
Collapse
|
25
|
Chen R, Huang M, Xu P. Polyphosphate as an antithrombotic target and hemostatic agent. J Mater Chem B 2023; 11:7855-7872. [PMID: 37534776 DOI: 10.1039/d3tb01152f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/04/2023]
Abstract
Polyphosphate (PolyP) is a polymer comprised of linear phosphate units connected by phosphate anhydride bonds. PolyP exists in a diverse range of eukaryotes and prokaryotes with varied chain lengths ranging from six to thousands of phosphate units. Upon activation, human platelets and neutrophils release short-chain PolyP, along with other components, to initiate the coagulation pathway. Long-chain PolyP derived from cellular or bacterial organelles exhibits higher proinflammatory and procoagulant effects compared to short-chain PolyP. Notably, PolyP has been identified as a low-hemorrhagic antithrombotic target since neutralizing plasma PolyP suppresses the thrombotic process without impairing the hemostatic functions. As an inorganic polymer without uniform steric configuration, PolyP is typically targeted by cationic polymers or recombinant polyphosphatases rather than conventional antibodies, small-molecule compounds, or peptides. Additionally, because of its procoagulant property, PolyP has been incorporated in wound-dressing materials to facilitate blood hemostasis. This review summarizes current studies on PolyP as a low-hemorrhagic antithrombotic target and the development of hemostatic materials based on PolyP.
Collapse
Affiliation(s)
- Ruoyu Chen
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, Fujian, 350108, P. R. China.
| | - Mingdong Huang
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, Fujian, 350108, P. R. China.
- College of Chemistry, Fuzhou University, Fuzhou, Fujian, 350108, P. R. China
| | - Peng Xu
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, Fujian, 350108, P. R. China.
| |
Collapse
|
26
|
Yao M, Ma J, Wu D, Fang C, Wang Z, Guo T, Mo J. Neutrophil extracellular traps mediate deep vein thrombosis: from mechanism to therapy. Front Immunol 2023; 14:1198952. [PMID: 37680629 PMCID: PMC10482110 DOI: 10.3389/fimmu.2023.1198952] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Accepted: 08/10/2023] [Indexed: 09/09/2023] Open
Abstract
Deep venous thrombosis (DVT) is a part of venous thromboembolism (VTE) that clinically manifests as swelling and pain in the lower limbs. The most serious clinical complication of DVT is pulmonary embolism (PE), which has a high mortality rate. To date, its underlying mechanisms are not fully understood, and patients usually present with clinical symptoms only after the formation of the thrombus. Thus, it is essential to understand the underlying mechanisms of deep vein thrombosis for an early diagnosis and treatment of DVT. In recent years, many studies have concluded that Neutrophil Extracellular Traps (NETs) are closely associated with DVT. These are released by neutrophils and, in addition to trapping pathogens, can mediate the formation of deep vein thrombi, thereby blocking blood vessels and leading to the development of disease. Therefore, this paper describes the occurrence and development of NETs and discusses the mechanism of action of NETs on deep vein thrombosis. It aims to provide a direction for improved diagnosis and treatment of deep vein thrombosis in the near future.
Collapse
Affiliation(s)
- Mengting Yao
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China
- Department of Orthopedic Surgery, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Jiacheng Ma
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China
- Department of Orthopedic Surgery, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Dongwen Wu
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China
- Department of Orthopedic Surgery, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Chucun Fang
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China
- Department of Orthopedic Surgery, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Zilong Wang
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China
- Department of Orthopedic Surgery, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Tianting Guo
- Department of Orthopedics, Guangdong Provincial People’s Hospital Ganzhou Hospital, Ganzhou Municipal Hospital, Ganzhou, Jiangxi, China
| | - Jianwen Mo
- Department of Orthopedic Surgery, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| |
Collapse
|
27
|
Komorowicz E, Farkas VJ, Szabó L, Cherrington S, Thelwell C, Kolev K. DNA and histones impair the mechanical stability and lytic susceptibility of fibrin formed by staphylocoagulase. Front Immunol 2023; 14:1233128. [PMID: 37662916 PMCID: PMC10470048 DOI: 10.3389/fimmu.2023.1233128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 07/31/2023] [Indexed: 09/05/2023] Open
Abstract
Background Staphylocoagulase (SCG) is a virulence factor of Staphylococcus aureus, one of the most lethal pathogens of our times. The complex of SCG with prothrombin (SCG/ProT) can clot fibrinogen, and SCG/ProT-induced fibrin and plasma clots have been described to show decreased mechanical and lytic resistance, which may contribute to septic emboli from infected cardiac vegetations. At infection sites, neutrophils can release DNA and histones, as parts of neutrophil extracellular traps (NETs), which in turn favor thrombosis, inhibit fibrinolysis and strengthen clot structure. Objectives To characterize the combined effects of major NET-components (DNA, histone H1 and H3) on SCG/ProT-induced clot structure, mechanical and lytic stability. Methods Recombinant SCG was used to clot purified fibrinogen and plasma. The kinetics of formation and lysis of fibrin and plasma clots containing H1 or core histones+/-DNA were followed by turbidimetry. Fibrin structure and mechanical stability were characterized with scanning electron microscopy, pressure-driven permeation, and oscillation rheometry. Results Histones and DNA favored the formation of thicker fibrin fibers and a more heterogeneous clot structure including high porosity with H1 histone, whereas low porosity with core histones and DNA. As opposed to previous observations with thrombin-induced clots, SCG/ProT-induced fibrin was not mechanically stabilized by histones. Similarly to thrombin-induced clots, the DNA-histone complexes prolonged fibrinolysis with tissue-type plasminogen activator (up to 2-fold). The anti-fibrinolytic effect of the DNA and DNA-H3 complex was observed in plasma clots too. Heparin (low molecular weight) accelerated the lysis of SCG/ProT-clots from plasma, even if DNA and histones were also present. Conclusions In the interplay of NETs and fibrin formed by SCG, DNA and histones promote structural heterogeneity in the clots, and fail to stabilize them against mechanical stress. The DNA-histone complexes render the SCG-fibrin more resistant to lysis and thereby less prone to embolization.
Collapse
Affiliation(s)
- Erzsébet Komorowicz
- Institute of Biochemistry and Molecular Biology, Department of Biochemistry, Semmelweis University, Budapest, Hungary
| | - Veronika J. Farkas
- Institute of Biochemistry and Molecular Biology, Department of Biochemistry, Semmelweis University, Budapest, Hungary
| | - László Szabó
- Institute of Biochemistry and Molecular Biology, Department of Biochemistry, Semmelweis University, Budapest, Hungary
- Plasma Chemistry Research Group, Institute of Materials and Environmental Chemistry, Research Centre for Natural Sciences, Budapest, Hungary
| | - Sophie Cherrington
- South Mimms Laboratories, Medicines and Healthcare Products Regulatory Agency, Potters Bar, United Kingdom
| | - Craig Thelwell
- South Mimms Laboratories, Medicines and Healthcare Products Regulatory Agency, Potters Bar, United Kingdom
| | - Krasimir Kolev
- Institute of Biochemistry and Molecular Biology, Department of Biochemistry, Semmelweis University, Budapest, Hungary
| |
Collapse
|
28
|
Bhargavan B, Kanmogne GD. SARS-CoV-2 Spike Proteins and Cell-Cell Communication Induce P-Selectin and Markers of Endothelial Injury, NETosis, and Inflammation in Human Lung Microvascular Endothelial Cells and Neutrophils: Implications for the Pathogenesis of COVID-19 Coagulopathy. Int J Mol Sci 2023; 24:12585. [PMID: 37628764 PMCID: PMC10454213 DOI: 10.3390/ijms241612585] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 07/31/2023] [Accepted: 08/03/2023] [Indexed: 08/27/2023] Open
Abstract
COVID-19 progression often involves severe lung injury, inflammation, coagulopathy, and leukocyte infiltration into pulmonary tissues. The pathogenesis of these complications is unknown. Because vascular endothelium and neutrophils express angiotensin-converting enzyme-2 and spike (S)-proteins, which are present in bodily fluids and tissues of SARS-CoV-2-infected patients, we investigated the effect of S-proteins and cell-cell communication on human lung microvascular endothelial cells and neutrophils expression of P-selectin, markers of coagulopathy, NETosis, and inflammation. Exposure of endothelial cells or neutrophils to S-proteins and endothelial-neutrophils co-culture induced P-selectin transcription and expression, significantly increased expression/secretion of IL-6, von Willebrand factor (vWF, pro-coagulant), and citrullinated histone H3 (cit-H3, NETosis marker). Compared to the SARS-CoV-2 Wuhan variant, Delta variant S-proteins induced 1.4-15-fold higher P-selectin and higher IL-6 and vWF. Recombinant tissue factor pathway inhibitor (rTFPI), 5,5'-dithio-bis-(2-nitrobenzoic acid) (thiol blocker), and thrombomodulin (anticoagulant) blocked S-protein-induced vWF, IL-6, and cit-H3. This suggests that following SARS-CoV-2 contact with the pulmonary endothelium or neutrophils and endothelial-neutrophil interactions, S-proteins increase adhesion molecules, induce endothelial injury, inflammation, NETosis and coagulopathy via the tissue factor pathway, mechanisms involving functional thiol groups, and/or the fibrinolysis system. Using rTFPI, effectors of the fibrinolysis system and/or thiol-based drugs could be viable therapeutic strategies against SARS-CoV-2-induced endothelial injury, inflammation, NETosis, and coagulopathy.
Collapse
Affiliation(s)
| | - Georgette D. Kanmogne
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198-5800, USA;
| |
Collapse
|
29
|
Moore HB. Fibrinolysis Shutdown and Hypofibrinolysis Are Not Synonymous Terms: The Clinical Significance of Differentiating Low Fibrinolytic States. Semin Thromb Hemost 2023; 49:433-443. [PMID: 36318960 PMCID: PMC10366941 DOI: 10.1055/s-0042-1758057] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Low fibrinolytic activity has been associated with pathologic thrombosis and multiple-organ failure. Low fibrinolytic activity has two commonly associated terms, hypofibrinolysis and fibrinolysis shutdown. Hypofibrinolysis is a chronic state of lack of ability to generate an appropriate fibrinolytic response when anticipated. Fibrinolysis shutdown is the shutdown of fibrinolysis after systemic activation of the fibrinolytic system. There has been interchanging of these terms to describe critically ill patients in multiple settings. This is problematic in understanding the pathophysiology of disease processes related to these conditions. There is also a lack of research on the cellular mediators of these processes. The purpose of this article is to review the on and off mechanisms of fibrinolysis in the context of low fibrinolytic states to define the importance in differentiating hypofibrinolysis from fibrinolysis shutdown. In many clinical scenarios, the etiology of a low fibrinolytic state cannot be determined due to ambiguity if a preceding fibrinolytic activation event occurred. In this scenario, the term "low fibrinolytic activity" or "fibrinolysis resistance" is a more appropriate descriptor, rather than using assumptive of hypofibrinolysis and fibrinolysis shutdown, particularly in the acute setting of infection, injury, and surgery.
Collapse
Affiliation(s)
- Hunter B. Moore
- Division of Transplant Surgery, Department of Surgery, University of Colorado Denver, Aurora, Colorado Semin Thromb Hemost
| |
Collapse
|
30
|
Yong J, Abrams ST, Wang G, Toh CH. Cell-free histones and the cell-based model of coagulation. J Thromb Haemost 2023; 21:1724-1736. [PMID: 37116754 DOI: 10.1016/j.jtha.2023.04.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 04/06/2023] [Accepted: 04/20/2023] [Indexed: 04/30/2023]
Abstract
The cell-based model of coagulation remains the basis of our current understanding of clinical hemostasis and thrombosis. Its advancement on the coagulation cascade model has enabled new prohemostatic and anticoagulant treatments to be developed. In the past decade, there has been increasing evidence of the procoagulant properties of extracellular, cell-free histones (CFHs). Although high levels of circulating CFHs released following extensive cell death in acute critical illnesses, such as sepsis and trauma, have been associated with adverse coagulation outcomes, including disseminated intravascular coagulation, new information has also emerged on how its local effects contribute to physiological clot formation. CFHs initiate coagulation by tissue factor exposure, either by destruction of the endovascular barrier or induction of endoluminal tissue factor expression on endothelia and monocytes. CFHs can also bind prothrombin directly, generating thrombin via the alternative prothrombinase pathway. In amplifying and augmenting the procoagulant signal, CFHs activate and aggregate platelets, increase procoagulant material bioavailability through platelet degranulation and Weibel-Palade body exocytosis, activate intrinsic coagulation via platelet polyphosphate release, and induce phosphatidylserine exposure. CFHs also inhibit protein C activation and downregulate thrombomodulin expression to reduce anti-inflammatory and anticoagulant effects. In consolidating clot formation, CFHs augment the fibrin polymer to confer fibrinolytic resistance and integrate neutrophil extracellular traps into the clot structure. Such new information holds the promise of new therapeutic developments, including improved targeting of immunothrombotic pathologies in acute critical illnesses.
Collapse
Affiliation(s)
- Jun Yong
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, Liverpool, UK
| | - Simon T Abrams
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, Liverpool, UK; Liverpool Clinical Laboratories, Liverpool, UK
| | - Guozheng Wang
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, Liverpool, UK; Liverpool Clinical Laboratories, Liverpool, UK
| | - Cheng-Hock Toh
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, Liverpool, UK; The Roald Dahl Haemostasis and Thrombosis Centre, Liverpool University Hospitals NHS Foundation Trust, Liverpool, UK.
| |
Collapse
|
31
|
Li W, Wang Z, Su C, Liao Z, Pei Y, Wang J, Li Z, Fu S, Liu J. The effect of neutrophil extracellular traps in venous thrombosis. Thromb J 2023; 21:67. [PMID: 37328882 DOI: 10.1186/s12959-023-00512-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Accepted: 06/11/2023] [Indexed: 06/18/2023] Open
Abstract
Neutrophil extracellular traps (NETs) as special release products of neutrophils have received extensive attention. They are composed of decondensed chromatin and coated with nucleoproteins, including histones and some granulosa proteins. NETs can form a network structure to effectively capture and eliminate pathogens and prevent their spread. Not only that, recent studies have shown that NETs also play an important role in venous thrombosis. This review provides the most important updated evidence regarding the mechanism of NETs formation and the role of NETs in the process of venous thrombosis. The potential prophylactic and therapeutic value of NETs in venous thrombotic disease will also be discussed.
Collapse
Affiliation(s)
- Weiwei Li
- Department of Hepatobiliary Surgery, Affiliated Hospital of Chengde Medical University, Chengde, Hebei Province, 067000, China
| | - Zixiang Wang
- Department of Hepatobiliary Surgery, Affiliated Hospital of Chengde Medical University, Chengde, Hebei Province, 067000, China
| | - Chen'guang Su
- Department of Hepatobiliary Surgery, Affiliated Hospital of Chengde Medical University, Chengde, Hebei Province, 067000, China
| | - Zheng Liao
- Department of Hepatobiliary Surgery, Affiliated Hospital of Chengde Medical University, Chengde, Hebei Province, 067000, China
| | - Yinxuan Pei
- Department of Hepatobiliary Surgery, Affiliated Hospital of Chengde Medical University, Chengde, Hebei Province, 067000, China
| | - Jianli Wang
- Department of Hepatobiliary Surgery, Affiliated Hospital of Chengde Medical University, Chengde, Hebei Province, 067000, China
| | - Zixin Li
- Department of Pathology, Affiliated Hospital of Chengde Medical University, Chengde, Hebei Province, 067000, China
| | - Shijie Fu
- Department of Orthopedic, Affiliated Hospital of Chengde Medical University, Chengde, Hebei Province, 067000, China
| | - Jinlong Liu
- Department of Hepatobiliary Surgery, Affiliated Hospital of Chengde Medical University, Chengde, Hebei Province, 067000, China.
| |
Collapse
|
32
|
Whyte CS. All tangled up: interactions of the fibrinolytic and innate immune systems. Front Med (Lausanne) 2023; 10:1212201. [PMID: 37332750 PMCID: PMC10272372 DOI: 10.3389/fmed.2023.1212201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 05/17/2023] [Indexed: 06/20/2023] Open
Abstract
The hemostatic and innate immune system are intertwined processes. Inflammation within the vasculature promotes thrombus development, whilst fibrin forms part of the innate immune response to trap invading pathogens. The awareness of these interlinked process has resulted in the coining of the terms "thromboinflammation" and "immunothrombosis." Once a thrombus is formed it is up to the fibrinolytic system to resolve these clots and remove them from the vasculature. Immune cells contain an arsenal of fibrinolytic regulators and plasmin, the central fibrinolytic enzyme. The fibrinolytic proteins in turn have diverse roles in immunoregulation. Here, the intricate relationship between the fibrinolytic and innate immune system will be discussed.
Collapse
|
33
|
Liu Y, Dai D, Abbasi M, Mereuta OM, Gamb SI, Kadirvel R, Kallmes DF, Brinjikji W. An in vitro model for Extracellular DNA Traps (ETs)-rich Human Thrombus Analogs. J Neurointerv Surg 2023; 15:589-593. [PMID: 35483911 PMCID: PMC11094997 DOI: 10.1136/neurintsurg-2022-018790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 04/16/2022] [Indexed: 11/03/2022]
Abstract
BACKGROUND Extracellular DNA traps (ETs) have important implications in both thrombosis and thrombolysis. Thus, developing benchtop thrombus analogs that recapitulate clinical ETs is potentially of great value for preclinical development and testing of thrombolytic agents and thrombectomy devices. In this study, we aimed to develop ETs-rich thrombus analogs for preclinical testing. METHODS Red blood cell (RBC)-rich, fibrin-rich, and platelet-rich thrombus analogs were created using human whole blood, platelet-poor plasma, and platelet-rich plasma obtained from the blood bank following institutional approval. Peripheral blood mononuclear cells (9.9×106 cells/mL) isolated from human whole blood and lipopolysaccharide (1 µg/mL) were added to induce ETs. Histochemical, immunohistochemistry and immunofluorescence were used to identify thrombus components and ETs. Scanning electronic microscopy was used to investigate the ultrastructure of the thrombus analogs. The thrombus compositions, morphologic features of ETs and citrullinated histone H3 (H3Cit) expression were compared with those of thrombi retrieved from patients by thrombectomy. RESULTS ETs-rich thrombus analogs were more compacted th-an the ETs-poor thrombus analogs. ETs were identified in both ETs-rich thrombus analogs and patient thrombi showing morphologic features including nuclear lobulation, nuclear swelling, diffused chromatin within cytoplasm, DNA/chromatin extending intracellularly and extracellularly, and extracellular chromatin patches and bundles. In the ETs-poor thrombus analogs, ETs were not observed and H3Cit expression was absent to minimal. The compositions and H3Cit expression in the ETs-rich thrombus analogs fell in the range of patient thrombi. CONCLUSIONS ETs-rich thrombus analogs can be consistently created in vitro and may benefit the preclinical development and testing of new thrombolytic agents and thrombectomy devices.
Collapse
Affiliation(s)
- Yang Liu
- Radiology, Mayo Clinic, Rochester, Minnesota, USA
- Global Institute of Future Technology, Shanghai Jiao Tong University, Shanghai, China
| | - Daying Dai
- Radiology, Mayo Clinic, Rochester, Minnesota, USA
| | - Mehdi Abbasi
- Radiology, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Scott I Gamb
- Electron Microscopy Core Facility, Mayo Clinic, Rochester, Minnesota, USA
| | | | | | | |
Collapse
|
34
|
Thakur M, Junho CVC, Bernhard SM, Schindewolf M, Noels H, Döring Y. NETs-Induced Thrombosis Impacts on Cardiovascular and Chronic Kidney Disease. Circ Res 2023; 132:933-949. [PMID: 37053273 PMCID: PMC10377271 DOI: 10.1161/circresaha.123.321750] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/15/2023]
Abstract
Arterial and venous thrombosis constitute a major source of morbidity and mortality worldwide. Association between thrombotic complications and cardiovascular and other chronic inflammatory diseases are well described. Inflammation and subsequent initiation of thrombotic events, termed immunothrombosis, also receive growing attention but are still incompletely understood. Nevertheless, the clinical relevance of aberrant immunothrombosis, referred to as thromboinflammation, is evident by an increased risk of thrombosis and cardiovascular events in patients with inflammatory or infectious diseases. Proinflammatory mediators released from platelets, complement activation, and the formation of NETs (neutrophil extracellular traps) initiate and foster immunothrombosis. In this review, we highlight and discuss prominent and emerging interrelationships and functions between NETs and other mediators in immunothrombosis in cardiovascular disease. Also, with patients with chronic kidney disease suffering from increased cardiovascular and thrombotic risk, we summarize current knowledge on neutrophil phenotype, function, and NET formation in chronic kidney disease. In addition, we elaborate on therapeutic targeting of NETs-induced immunothrombosis. A better understanding of the functional relevance of antithrombotic mediators which do not increase bleeding risk may provide opportunities for successful therapeutic interventions to reduce thrombotic risk beyond current treatment options.
Collapse
Affiliation(s)
- Manovriti Thakur
- Division of Angiology, Swiss Cardiovascular Center, Inselspital (M.T., S.M.B., M.S., Y.D.), Bern University Hospital, University of Bern, Switzerland
- Department for BioMedical Research (DBMR) (M.T., S.M.B., M.S., Y.D.), Bern University Hospital, University of Bern, Switzerland
| | - Carolina Victoria Cruz Junho
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital RWTH Aachen, Germany (C.V.C.J., H.N.)
| | - Sarah Maike Bernhard
- Division of Angiology, Swiss Cardiovascular Center, Inselspital (M.T., S.M.B., M.S., Y.D.), Bern University Hospital, University of Bern, Switzerland
- Department for BioMedical Research (DBMR) (M.T., S.M.B., M.S., Y.D.), Bern University Hospital, University of Bern, Switzerland
| | - Marc Schindewolf
- Division of Angiology, Swiss Cardiovascular Center, Inselspital (M.T., S.M.B., M.S., Y.D.), Bern University Hospital, University of Bern, Switzerland
- Department for BioMedical Research (DBMR) (M.T., S.M.B., M.S., Y.D.), Bern University Hospital, University of Bern, Switzerland
| | - Heidi Noels
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital RWTH Aachen, Germany (C.V.C.J., H.N.)
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, the Netherlands (H.N.)
| | - Yvonne Döring
- Division of Angiology, Swiss Cardiovascular Center, Inselspital (M.T., S.M.B., M.S., Y.D.), Bern University Hospital, University of Bern, Switzerland
- Department for BioMedical Research (DBMR) (M.T., S.M.B., M.S., Y.D.), Bern University Hospital, University of Bern, Switzerland
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany (Y.D.)
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University Munich (LMU), Munich, Germany (Y.D.)
| |
Collapse
|
35
|
Medeiros SK, Sharma N, Dwivedi D, Cani E, Zhou J, Dwivedi N, Sohrabipour S, Liaw PC. THE EFFECTS OF DNASE I AND LOW-MOLECULAR-WEIGHT HEPARIN IN A MURINE MODEL OF POLYMICROBIAL ABDOMINAL SEPSIS. Shock 2023; 59:666-672. [PMID: 36852972 DOI: 10.1097/shk.0000000000002095] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2023]
Abstract
ABSTRACT Introduction: Cell-free DNA (CFDNA) has emerged as a prognostic biomarker in patients with sepsis. Circulating CFDNA is hypothesized to be associated with histones in the form of nucleosomes. In vitro, DNA activates coagulation and inhibits fibrinolysis, whereas histones activate platelets and are cytotoxic to endothelial cells. Previous studies have targeted CFDNA or histones in animal models of sepsis using DNase I or heparins, respectively, which has reduced inflammatory and thrombosis markers, thereby improving survival. In this study, we explored the possibility that the combination of DNase I and a low-molecular weight heparin (LMWH) may be a better therapeutic approach than monotherapy in a murine model of abdominal sepsis. Methods: C57Bl/6 mice (8-12 weeks old, both sexes) were subjected to either cecal ligation and puncture or sham surgery. Mice were given antibiotics, fluids, and either saline, DNase I (intraperitoneally, 20 mg/kg/8 h), LMWH (dalteparin, subcutaneously 500 IU/kg/12 h), or a combination of both (n = 12-31). Mice were monitored over 72 h for survival. Organs and blood were harvested for analysis. Levels of LMWH, CFDNA, IL-6, citrullinated histone-H3, thrombin-antithrombin complexes, and protein C were measured in plasma. Results: Administration of either DNase I (81.8%) or LMWH (83.3%, prophylactic range of 0.12 ± 0.07 IU/mL achieved) improved the survival of septic mice compared with saline- (38.7%) and combination-treated mice (48.8%, P < 0.05). Combination-treated mice also showed a small but insignificant improvement in survival compared with saline-treated cecal ligation and puncture mice. Monotherapies may be improving survival by reducing blood bacterial loads, citrullinated histone-H3, and thrombin-antithrombin complexes, and improving protein C levels. Conclusions: Compared with saline- and combination-treated mice, administration of monotherapies to septic mice improved survival. These findings suggest that there may be a negative drug-drug interaction between DNase I and LMWH when DNase I is administered intraperitoneally in a murine model of polymicrobial abdominal sepsis.
Collapse
Affiliation(s)
| | | | | | | | | | - Naviya Dwivedi
- Thrombosis and Atherosclerosis Research Institute, Hamilton, Canada
| | | | | |
Collapse
|
36
|
Ząbczyk M, Ariëns RAS, Undas A. Fibrin clot properties in cardiovascular disease: from basic mechanisms to clinical practice. Cardiovasc Res 2023; 119:94-111. [PMID: 36662542 PMCID: PMC10377755 DOI: 10.1093/cvr/cvad017] [Citation(s) in RCA: 36] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 10/28/2022] [Accepted: 11/14/2022] [Indexed: 01/21/2023] Open
Abstract
Fibrinogen conversion into insoluble fibrin and the formation of a stable clot is the final step of the coagulation cascade. Fibrin clot porosity and its susceptibility to plasmin-mediated lysis are the key fibrin measures, describing the properties of clots prepared ex vivo from citrated plasma. Cardiovascular disease (CVD), referring to coronary heart disease, heart failure, stroke, and hypertension, has been shown to be associated with the formation of dense fibrin networks that are relatively resistant to lysis. Denser fibrin mesh characterized acute patients at the onset of myocardial infarction or ischaemic stroke, while hypofibrinolysis has been identified as a persistent fibrin feature in patients following thrombotic events or in those with stable coronary artery disease. Traditional cardiovascular risk factors, such as smoking, diabetes mellitus, hyperlipidaemia, obesity, and hypertension, have also been linked with unfavourably altered fibrin clot properties, while some lifestyle modifications and pharmacological treatment, in particular statins and anticoagulants, may improve fibrin structure and function. Prospective studies have suggested that prothrombotic fibrin clot phenotype can predict cardiovascular events in short- and long-term follow-ups. Mutations and splice variants of the fibrinogen molecule that have been proved to be associated with thrombophilia or increased cardiovascular risk, along with fibrinogen post-translational modifications, prothrombotic state, inflammation, platelet activation, and neutrophil extracellular traps formation, contribute also to prothrombotic fibrin clot phenotype. Moreover, about 500 clot-bound proteins have been identified within plasma fibrin clots, including fibronectin, α2-antiplasmin, factor XIII, complement component C3, and histidine-rich glycoprotein. This review summarizes the current knowledge on the mechanisms underlying unfavourable fibrin clot properties and their implications in CVD and its thrombo-embolic manifestations.
Collapse
Affiliation(s)
- Michał Ząbczyk
- Thromboembolic Disorders Department, Institute of Cardiology, Jagiellonian University Medical College, 80 Pradnicka St, 31-202 Krakow, Poland
- Krakow Center for Medical Research and Technologies, John Paul II Hospital, Krakow, Poland
| | - Robert A S Ariëns
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
| | - Anetta Undas
- Thromboembolic Disorders Department, Institute of Cardiology, Jagiellonian University Medical College, 80 Pradnicka St, 31-202 Krakow, Poland
- Krakow Center for Medical Research and Technologies, John Paul II Hospital, Krakow, Poland
| |
Collapse
|
37
|
Coagulation Disorders in Sepsis and COVID-19-Two Sides of the Same Coin? A Review of Inflammation-Coagulation Crosstalk in Bacterial Sepsis and COVID-19. J Clin Med 2023; 12:jcm12020601. [PMID: 36675530 PMCID: PMC9866352 DOI: 10.3390/jcm12020601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/27/2022] [Accepted: 01/10/2023] [Indexed: 01/15/2023] Open
Abstract
Sepsis is a major cause of morbidity and mortality worldwide. Sepsis-associated coagulation disorders are involved in the pathogenesis of multiorgan failure and lead to a subsequently worsening prognosis. Alongside the global impact of the COVID-19 pandemic, a great number of research papers have focused on SARS-CoV-2 pathogenesis and treatment. Significant progress has been made in this regard and coagulation disturbances were once again found to underlie some of the most serious adverse outcomes of SARS-CoV-2 infection, such as acute lung injury and multiorgan dysfunction. In the attempt of untangling the mechanisms behind COVID-19-associated coagulopathy (CAC), a series of similarities with sepsis-induced coagulopathy (SIC) became apparent. Whether they are, in fact, the same disease has not been established yet. The clinical picture of CAC shows the unique feature of an initial phase of intravascular coagulation confined to the respiratory system. Only later on, patients can develop a clinically significant form of systemic coagulopathy, possibly with a consumptive pattern, but, unlike SIC, it is not a key feature. Deepening our understanding of CAC pathogenesis has to remain a major goal for the research community, in order to design and validate accurate definitions and classification criteria.
Collapse
|
38
|
Platelet-Neutrophil Crosstalk in Thrombosis. Int J Mol Sci 2023; 24:ijms24021266. [PMID: 36674781 PMCID: PMC9861587 DOI: 10.3390/ijms24021266] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/02/2023] [Accepted: 01/04/2023] [Indexed: 01/11/2023] Open
Abstract
Platelets are essential for the formation of a haemostatic plug to prevent bleeding, while neutrophils are the guardians of our immune defences against invading pathogens. The interplay between platelets and innate immunity, and subsequent triggering of the activation of coagulation is part of the host system to prevent systemic spread of pathogen in the blood stream. Aberrant immunothrombosis and excessive inflammation can however, contribute to the thrombotic burden observed in many cardiovascular diseases. In this review, we highlight how platelets and neutrophils interact with each other and how their crosstalk is central to both arterial and venous thrombosis and in COVID-19. While targeting platelets and coagulation enables efficient antithrombotic treatments, they are often accompanied with a bleeding risk. We also discuss how novel approaches to reduce platelet-mediated recruitment of neutrophils could represent promising therapies to treat thrombosis without affecting haemostasis.
Collapse
|
39
|
Whyte CS, Mutch NJ. "Going with the flow" in modeling fibrinolysis. Front Cardiovasc Med 2022; 9:1054541. [PMID: 36531720 PMCID: PMC9755328 DOI: 10.3389/fcvm.2022.1054541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 11/18/2022] [Indexed: 09/10/2024] Open
Abstract
The formation of thrombi is shaped by intravascular shear stress, influencing both fibrin architecture and the cellular composition which has downstream implications in terms of stability against mechanical and fibrinolytic forces. There have been many advancements in the development of models that incorporate flow rates akin to those found in vivo. Both thrombus formation and breakdown are simultaneous processes, the balance of which dictates the size, persistence and resolution of thrombi. Therefore, there is a requirement to have models which mimic the physiological shear experienced within the vasculature which in turn influences the fibrinolytic degradation of the thrombus. Here, we discuss various assays for fibrinolysis and importantly the development of novel models that incorporate physiological shear rates. These models are essential tools to untangle the molecular and cellular processes which govern fibrinolysis and can recreate the conditions within normal and diseased vessels to determine how these processes become perturbed in a pathophysiological setting. They also have utility to assess novel drug targets and antithrombotic drugs that influence thrombus stability.
Collapse
Affiliation(s)
- Claire S. Whyte
- Aberdeen Cardiovascular and Diabetes Centre, Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, United Kingdom
| | | |
Collapse
|
40
|
Varjú I, Tóth E, Farkas ÁZ, Farkas VJ, Komorowicz E, Feller T, Kiss B, Kellermayer MZ, Szabó L, Wacha A, Bóta A, Longstaff C, Kolev K. Citrullinated fibrinogen forms densely packed clots with decreased permeability. J Thromb Haemost 2022; 20:2862-2872. [PMID: 36083779 PMCID: PMC9828116 DOI: 10.1111/jth.15875] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 08/17/2022] [Accepted: 09/07/2022] [Indexed: 01/13/2023]
Abstract
BACKGROUND Fibrin, the main scaffold of thrombi, is susceptible to citrullination by PAD (peptidyl arginine deiminase) 4, secreted from neutrophils during the formation of neutrophil extracellular traps. Citrullinated fibrinogen (citFg) has been detected in human plasma as well as in murine venous thrombi, and it decreases the lysability and mechanical resistance of fibrin clots. OBJECTIVE To investigate the effect of fibrinogen citrullination on the structure of fibrin clots. METHODS Fibrinogen was citrullinated with PAD4 and clotted with thrombin. Scanning electron microscopy (SEM) and atomic force microscopy (AFM) were used to measure fiber thickness, fiber height/width ratio, and fiber persistence length in clots containing citFg. Fiber density was measured with laser scanning microscopy (LSM) and permeability measurements were carried out to estimate the porosity of the clots. The intra-fiber structure of fibrin was analyzed with small-angle X-ray scattering (SAXS). RESULTS SEM images revealed a decrease in the median fiber diameter that correlated with the fraction of citFg in the clot, while the fiber width/length ratio remained unchanged according to AFM. With SAXS we observed that citrullination resulted in the formation of denser clots in line with increased fiber density shown by LSM. The permeability constant of citrullinated fibrin decreased more than 3-fold indicating significantly decreased porosity. SAXS also showed largely preserved periodicity in the longitudinal assembly of fibrin monomers. CONCLUSION The current observations of thin fibers combined with dense packing and low porosity in the presence of citFg can provide a structural framework for the mechanical fragility and lytic resistance of citrullinated fibrin.
Collapse
Affiliation(s)
- Imre Varjú
- Program in Cellular and Molecular MedicineBoston Children's HospitalBostonMassachusettsUSA
- Department of PediatricsHarvard Medical SchoolBostonMassachusettsUSA
- Department of Biochemistry, Institute of Biochemistry and Molecular BiologySemmelweis UniversityBudapestHungary
- Department of Sociomedical Sciences, Mailman School of Public HealthColumbia UniversityNew YorkNew YorkUSA
| | - Erzsébet Tóth
- Department of Biochemistry, Institute of Biochemistry and Molecular BiologySemmelweis UniversityBudapestHungary
| | - Ádám Z. Farkas
- Department of Biochemistry, Institute of Biochemistry and Molecular BiologySemmelweis UniversityBudapestHungary
| | - Veronika J. Farkas
- Department of Biochemistry, Institute of Biochemistry and Molecular BiologySemmelweis UniversityBudapestHungary
| | - Erzsébet Komorowicz
- Department of Biochemistry, Institute of Biochemistry and Molecular BiologySemmelweis UniversityBudapestHungary
| | - Tímea Feller
- Department of Biophysics and Radiation BiologySemmelweis UniversityBudapestHungary
| | - Balázs Kiss
- Department of Biophysics and Radiation BiologySemmelweis UniversityBudapestHungary
| | | | - László Szabó
- Department of Biochemistry, Institute of Biochemistry and Molecular BiologySemmelweis UniversityBudapestHungary
- Department of Functional and Structural Materials, Institute of Materials and Environmental Chemistry, Research Centre for Natural SciencesHungarian Academy of SciencesBudapestHungary
| | - András Wacha
- Biological Nanochemistry Research Group, Institute of Materials and Environmental Chemistry, Centre for Natural SciencesHungarian Academy of SciencesBudapestHungary
| | - Attila Bóta
- Biological Nanochemistry Research Group, Institute of Materials and Environmental Chemistry, Centre for Natural SciencesHungarian Academy of SciencesBudapestHungary
| | - Colin Longstaff
- National Institute for Biological Standards and ControlSouth MimmsUK
| | - Krasimir Kolev
- Department of Biochemistry, Institute of Biochemistry and Molecular BiologySemmelweis UniversityBudapestHungary
| |
Collapse
|
41
|
Platelet-Neutrophil Association in NETs-Rich Areas in the Retrieved AIS Patient Thrombi. Int J Mol Sci 2022; 23:ijms232214477. [PMID: 36430952 PMCID: PMC9694992 DOI: 10.3390/ijms232214477] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 11/08/2022] [Accepted: 11/10/2022] [Indexed: 11/23/2022] Open
Abstract
Histological structure of thrombi is a strong determinant of the outcome of vascular recanalization therapy, the only treatment option for acute ischemic stroke (AIS) patients. A total of 21 AIS patients from this study after undergoing non-enhanced CT scan and multimodal MRI were treated with mechanical stent-based and manual aspiration thrombectomy, and thromboembolic retrieved from a cerebral artery. Complementary histopathological and imaging analyses were performed to understand their composition with a specific focus on fibrin, von Willebrand factor, and neutrophil extracellular traps (NETs). Though distinct RBC-rich and platelet-rich areas were found, AIS patient thrombi were overwhelmingly platelet-rich, with 90% of thrombi containing <40% total RBC-rich contents (1.5 to 37%). Structurally, RBC-rich areas were simple, consisting of tightly packed RBCs in thin fibrin meshwork with sparsely populated nucleated cells and lacked any substantial von Willebrand factor (VWF). Platelet-rich areas were structurally more complex with thick fibrin meshwork associated with VWF. Plenty of leukocytes populated the platelet-rich areas, particularly in the periphery and border areas between platelet-rich and RBC-rich areas. Platelet-rich areas showed abundant activated neutrophils (myeloperoxidase+ and neutrophil-elastase+) containing citrullinated histone-decorated DNA. Citrullinated histone-decorated DNA also accumulated extracellularly, pointing to NETosis by the activated neutrophils. Notably, NETs-containing areas showed strong reactivity to VWF, platelets, and high-mobility group box 1 (HMGB1), signifying a close interplay between these components.
Collapse
|
42
|
Li J, Tong D, Song B, Xie F, Zhang G, Hao X, Li W, Chi H, Wang W, Shao Y. Inflammatory cytokines induce neutrophil extracellular traps interaction with activated platelets and endothelial cells exacerbate coagulation in moderate and severe essential hypertension. J Hypertens 2022; 40:2219-2229. [PMID: 35950987 DOI: 10.1097/hjh.0000000000003250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Essential hypertension (EH) patients suffer from paradoxically thrombotic rather than haemorrhagic, although the exact mechanism remains elusive. Our aim is to explore whether and how neutrophil extracellular traps (NETs) play the procoagulant role in EH patients, as well as evaluated whether the NET releasing were triggered by inflammatory cytokines. METHODS The concentration of plasma NETs components were detected by ELISA. The morphology of cells and NETs formation were analysed using immunofluorescence. Procoagulant activity was analysed by clotting time, purified coagulation complex and fibrin generation assays. Phosphatidylserine (PS) exposure on endothelial cells (ECs) was analysed with flow cytometry. RESULTS Moderate to severe EH patients plasma NETs levels were significantly higher compared to mild EH patients or controls. Furthermore, inflammatory cytokines can induce NETs generation, depleting these patients plasma inflammatory cytokines led to a reduction in NET releasing. NETs from moderate to severe EH patients neutrophils led to significantly decreased clotting time (CT), increased potency to generate thrombin and fibrin (all P < 0.05). These procoagulant effects were markedly attenuated by approximately 70% using DNase I. Additionally, high concentrations NETs exerted a strong cytotoxic effect on ECs, conferring them a procoagulant phenotype. CONCLUSION Our study reveals that EH drives a systemic inflammatory environment, which, in turn, drives neutrophils to prime and NET releasing, and found a link between hypercoagulability and NETs levels in moderate to severe EH patients. Therefore, anti-inflammatory combined with block the generation of NETs may represent a new therapeutic target for preventing thrombosis in EH patients.
Collapse
Affiliation(s)
- Jihe Li
- Department of Cardiology
- Heilongjiang Academy of Chinese Medicine Sciences, Harbin, Heilongjiang Province, China
| | - Dongxia Tong
- Department of Oncology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao
| | - Bin Song
- Department of Geriatrics, Second Hospital of Traditional Chinese Medicine, Tai'an
| | | | - Guixin Zhang
- Department of General Surgery, Qingdao FUWAI Cardiovascular Hospital, Qingdao
| | - Xin Hao
- Department of Ophthalmology, Second Hospital of Traditional Chinese Medicine, Tai'an, Shandong Province
| | | | | | - Weiming Wang
- Heilongjiang Academy of Chinese Medicine Sciences, Harbin, Heilongjiang Province, China
| | | |
Collapse
|
43
|
Komorowicz E, Kolev K. Fibrin structure, viscoelasticity and lysis face the interplay of biorelevant polyions. Curr Opin Hematol 2022; 29:244-250. [PMID: 35916559 DOI: 10.1097/moh.0000000000000725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE OF REVIEW In the past 5 decades, heparins have been widely used as anticoagulants in the prevention and treatment of thrombosis. Subsequent development of heparin variants of various size and charge facilitated the discovery of their multiple biological actions and nonanticoagulant benefits. Platelet-derived or microbial polyphosphates, as well as DNA released in the course of neutrophil extracellular trap-formation are additional polyanions, which can modulate the development and stability of thrombi associated with cancer or inflammation. In this review, we focus on the size-dependent and electric charge-dependent modulatory effects of the three polyanions of different chemical structure. RECENT FINDINGS The polycationic histones have been recognized as potential biomarkers and therapeutic targets in several diseases related to inflammation and thrombosis. Since combating histones with activated protein C or heparin could cause unwanted bleeding, the quest for nonanticoagulant histone-neutralizing agents is ongoing. Polyanions may neutralize or exaggerate certain histone-mediated effects depending on their electric charge, size and histone effects under investigation. Several prothrombotic effects of polyphosphates and DNA are also size-dependent. SUMMARY The efficiency of future therapeutics targeting prothrombotic polyanions or histones is not a simple matter of electric charge, but may rely on a delicate combination of size, charge and chemical composition.
Collapse
Affiliation(s)
- Erzsébet Komorowicz
- Department of Biochemistry, Institute of Biochemistry and Molecular Biology, Semmelweis University, Budapest, Hungary
| | | |
Collapse
|
44
|
Pryzdial ELG, Leatherdale A, Conway EM. Coagulation and complement: Key innate defense participants in a seamless web. Front Immunol 2022; 13:918775. [PMID: 36016942 PMCID: PMC9398469 DOI: 10.3389/fimmu.2022.918775] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 07/06/2022] [Indexed: 12/30/2022] Open
Abstract
In 1969, Dr. Oscar Ratnoff, a pioneer in delineating the mechanisms by which coagulation is activated and complement is regulated, wrote, “In the study of biological processes, the accumulation of information is often accelerated by a narrow point of view. The fastest way to investigate the body’s defenses against injury is to look individually at such isolated questions as how the blood clots or how complement works. We must constantly remind ourselves that such distinctions are man-made. In life, as in the legal cliché, the devices through which the body protects itself form a seamless web, unwrinkled by our artificialities.” Our aim in this review, is to highlight the critical molecular and cellular interactions between coagulation and complement, and how these two major component proteolytic pathways contribute to the seamless web of innate mechanisms that the body uses to protect itself from injury, invading pathogens and foreign surfaces.
Collapse
Affiliation(s)
- Edward L. G. Pryzdial
- Centre for Blood Research, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
- Canadian Blood Services, Medical Affairs and Innovation, Vancouver, BC, Canada
- *Correspondence: Edward L. G. Pryzdial, ; Edward M. Conway,
| | - Alexander Leatherdale
- Centre for Blood Research, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada
- Division of Hematology, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Edward M. Conway
- Centre for Blood Research, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
- Canadian Blood Services, Medical Affairs and Innovation, Vancouver, BC, Canada
- Division of Hematology, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
- *Correspondence: Edward L. G. Pryzdial, ; Edward M. Conway,
| |
Collapse
|
45
|
Wienkamp AK, Erpenbeck L, Rossaint J. Platelets in the NETworks interweaving inflammation and thrombosis. Front Immunol 2022; 13:953129. [PMID: 35979369 PMCID: PMC9376363 DOI: 10.3389/fimmu.2022.953129] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 07/07/2022] [Indexed: 12/18/2022] Open
Abstract
Platelets are well characterized for their indispensable role in primary hemostasis to control hemorrhage. Research over the past years has provided a substantial body of evidence demonstrating that platelets also participate in host innate immunity. The surface expression of pattern recognition receptors, such as TLR2 and TLR4, provides platelets with the ability to sense bacterial products in their environment. Platelet α-granules contain microbicidal proteins, chemokines and growth factors, which upon release may directly engage pathogens and/or contribute to inflammatory signaling. Additionally, platelet interactions with neutrophils enhance neutrophil activation and are often crucial to induce a sufficient immune response. In particular, platelets can activate neutrophils to form neutrophil extracellular traps (NETs). This specific neutrophil effector function is characterized by neutrophils expelling chromatin fibres decorated with histones and antimicrobial proteins into the extracellular space where they serve to trap and kill pathogens. Until now, the mechanisms and signaling pathways between platelets and neutrophils inducing NET formation are still not fully characterized. NETs were also detected in thrombotic lesions in several disease backgrounds, pointing towards a role as an interface between neutrophils, platelets and thrombosis, also known as immunothrombosis. The negatively charged DNA within NETs provides a procoagulant surface, and in particular NET-derived proteins may directly activate platelets. In light of the current COVID-19 pandemic, the topic of immunothrombosis has become more relevant than ever, as a majority of COVID-19 patients display thrombi in the lung capillaries and other vascular beds. Furthermore, NETs can be found in the lung and other tissues and are associated with an increased mortality. Here, virus infiltration may lead to a cytokine storm that potently activates neutrophils and leads to massive neutrophil infiltration into the lung and NET formation. The resulting NETs presumably activate platelets and coagulation factors, further contributing to the subsequent emergence of microthrombi in pulmonary capillaries. In this review, we will discuss the interplay between platelets and NETs and the potential of this alliance to influence the course of inflammatory diseases. A better understanding of the underlying molecular mechanisms and the identification of treatment targets is of utmost importance to increase patients’ survival and improve the clinical outcome.
Collapse
Affiliation(s)
- Ann-Katrin Wienkamp
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Münster, Münster, Germany
| | - Luise Erpenbeck
- Department of Dermatology, University Hospital Münster, Münster, Germany
| | - Jan Rossaint
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Münster, Münster, Germany
- *Correspondence: Jan Rossaint,
| |
Collapse
|
46
|
Colciaghi F, Costanza M. Unveiling Leukocyte Extracellular Traps in Inflammatory Responses of the Central Nervous System. Front Immunol 2022; 13:915392. [PMID: 35844591 PMCID: PMC9283689 DOI: 10.3389/fimmu.2022.915392] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 06/07/2022] [Indexed: 11/13/2022] Open
Abstract
Over the past nearly two decades, increasing evidence has uncovered how immune cells can actively extrude genetic material to entrap invading pathogens or convey sterile inflammatory signals that contribute to shaping immune responses. Originally identified in neutrophils, the release of decondensed chromatin fibers decorated with antimicrobial proteins, called extracellular traps (ETs), has been recognized as a specific form of programmed inflammatory cell death, which is now known to occur in several other leukocytes. Subsequent reports have shown that self-DNA can be extruded from immune cells even in the absence of cell death phenomena. More recent data suggest that ETs formation could exacerbate neuroinflammation in several disorders of the central nervous system (CNS). This review article provides an overview of the varied types, sources, and potential functions of extracellular DNA released by immune cells. Key evidence suggesting the involvement of ETs in neurodegenerative, traumatic, autoimmune, and oncological disorders of the CNS will be discussed, outlining ongoing challenges and drawing potentially novel lines of investigation.
Collapse
Affiliation(s)
- Francesca Colciaghi
- Epilepsy Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Massimo Costanza
- Molecular Neuro-Oncology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
- *Correspondence: Massimo Costanza,
| |
Collapse
|
47
|
Bryk-Wiązania AH, Cysewski D, Ocłoń E, Undas A. Mass-spectrometric identification of oxidative modifications in plasma-purified plasminogen: Association with hypofibrinolysis in patients with acute pulmonary embolism. Biochem Biophys Res Commun 2022; 621:53-58. [PMID: 35810591 DOI: 10.1016/j.bbrc.2022.06.063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 06/21/2022] [Indexed: 11/02/2022]
Abstract
OBJECTIVES Mechanisms behind disturbed fibrinolysis in pulmonary embolism (PE) are poorly understood. We hypothesized that oxidative stress-induced changes in plasminogen contribute to impaired fibrinolysis in patients with acute PE. METHODS Oxidative and other modifications were investigated using mass-spectrometry in plasminogen purified from pooled plasma of 5 acute PE patients on admission and after 3 months of anticoagulant treatment, along with plasma clot lysis time, a measure of global efficiency of fibrinolysis, and a stable oxidative stress marker, plasma 8-isoprostane. RESULTS Twenty sites of oxidation, 3 sites of carbonylation and 4 sites of S-nitrosylation were identified in plasminogen. The intensity of peptides oxidized at cysteine residues with respect to unmodified peptides decreased after 3 months of anticoagulation (p = 0.018). This was not observed for oxidized methionine residues (p = 0.9). Oxidized tryptophan (n = 4) and proline (n = 2), as well as carbonylation at 3 threonine residues were selectively identified in acute PE episode, not after 3 months. This was accompanied by 12.8% decrease in clot lysis time (p = 0.043). Deamidation occurred at the arginine, previously identified to undergo the cleavage by plasminogen activator. Methylated were two lysine-binding sites important for an interaction of plasminogen with fibrin. Other identified modifications involved: glycation, acetylation, phosphorylation, homocysteinylation, carbamylation and dichlorination (88 modifications at 162 sites). CONCLUSIONS Data suggest that oxidative stress-induced changes in plasminogen molecules may contribute to less effective global fibrinolysis in patients with acute PE. The comprehensive library of posttranslational modifications in plasminogen molecules was provided, including modifications of sites reported to be involved in important biological functions.
Collapse
Affiliation(s)
- Agata Hanna Bryk-Wiązania
- Department of Endocrinology, Jagiellonian University Medical College, Krakow, Poland; University Hospital, Krakow, Poland.
| | - Dominik Cysewski
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | - Ewa Ocłoń
- Centre for Experimental and Innovative Medicine, Laboratory of Recombinant Proteins Production, University of Agriculture in Krakow, Krakow, Poland
| | - Anetta Undas
- Institute of Cardiology, Jagiellonian University Medical College, Krakow, Poland; John Paul II Hospital, Krakow, Poland
| |
Collapse
|
48
|
Kostyunin A, Glushkova T, Stasev A, Mukhamadiyarov R, Velikanova E, Bogdanov L, Sinitskaya A, Asanov M, Ovcharenko E, Barbarash L, Kutikhin A. Early Postoperative Immunothrombosis of Bioprosthetic Mitral Valve and Left Atrium: A Case Report. Int J Mol Sci 2022; 23:ijms23126736. [PMID: 35743174 PMCID: PMC9224391 DOI: 10.3390/ijms23126736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 06/13/2022] [Accepted: 06/15/2022] [Indexed: 11/16/2022] Open
Abstract
A 72-year-old female patient with mixed rheumatic mitral valve disease and persistent atrial fibrillation underwent mitral valve replacement and suffered from a combined thrombosis of the bioprosthetic valve and the left atrium as soon as 2 days post operation. The patient immediately underwent repeated valve replacement and left atrial thrombectomy. Yet, four days later the patient died due to the recurrent prosthetic valve and left atrial thrombosis which both resulted in an extremely low cardiac output. In this patient's case, the thrombosis was notable for the resistance to anticoagulant therapy as well as for aggressive neutrophil infiltration and release of neutrophil extracellular traps (NETs) within the clot, as demonstrated by immunostaining. The reasons behind these phenomena remained unclear, as no signs of sepsis or contamination of the BHV were documented, although the patient was diagnosed with inherited thrombophilia that could impede the fibrinolysis. The described case highlights the hazard of immunothrombosis upon valve replacement and elucidates its mechanisms in this surgical setting.
Collapse
|
49
|
Block H, Rossaint J, Zarbock A. The Fatal Circle of NETs and NET-Associated DAMPs Contributing to Organ Dysfunction. Cells 2022; 11:1919. [PMID: 35741047 PMCID: PMC9222025 DOI: 10.3390/cells11121919] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 06/02/2022] [Accepted: 06/10/2022] [Indexed: 02/07/2023] Open
Abstract
The innate immune system is the first line of defense against invading pathogens or sterile injuries. Pattern recognition receptors (PRR) sense molecules released from inflamed or damaged cells, or foreign molecules resulting from invading pathogens. PRRs can in turn induce inflammatory responses, comprising the generation of cytokines or chemokines, which further induce immune cell recruitment. Neutrophils represent an essential factor in the early immune response and fulfill numerous tasks to fight infection or heal injuries. The release of neutrophil extracellular traps (NETs) is part of it and was originally attributed to the capture and elimination of pathogens. In the last decade studies revealed a detrimental role of NETs during several diseases, often correlated with an exaggerated immune response. Overwhelming inflammation in single organs can induce remote organ damage, thereby further perpetuating release of inflammatory molecules. Here, we review recent findings regarding damage-associated molecular patterns (DAMPs) which are able to induce NET formation, as well as NET components known to act as DAMPs, generating a putative fatal circle of inflammation contributing to organ damage and sequentially occurring remote organ injury.
Collapse
Affiliation(s)
| | | | - Alexander Zarbock
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Muenster, 48149 Muenster, Germany; (H.B.); (J.R.)
| |
Collapse
|
50
|
Polyphosphate nanoparticles enhance the fibrin stabilization by histones more efficiently than linear polyphosphates. PLoS One 2022; 17:e0266782. [PMID: 35468161 PMCID: PMC9037942 DOI: 10.1371/journal.pone.0266782] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 03/26/2022] [Indexed: 01/02/2023] Open
Abstract
Introduction Beyond the three-dimensional fibrin network, the mechanical and lytic stability of thrombi is supported by the matrix of neutrophil extracellular traps (NETs) composed of polyanionic DNA meshwork with attached proteins including polycationic histones. Polyphosphates represent another type of polyanions, which in their linear form are known to enhance the fibrin stabilizing effects of DNA and histones. However, in vivo polyphosphates are also present in the form of nanoparticles (PolyP-NP), the interference of which with the fibrin/NET matrix is poorly characterized. Aims To compare the effects of linear and nanoparticulate polyphosphates, and their combinations with relevant NET components (DNA, histone H3) on fibrin formation, structure, and lysis in in vitro assays focusing on histone-polyphosphate interactions. Methods Transmission electron microscopy and dynamic light scattering for stability of the PolyP-NP preparations. Turbidimetry for kinetics of fibrinogen clotting by thrombin and fibrin dissolution by tissue-type plasminogen activator/plasminogen. Scanning electron microscopy for fibrin structure. Surface plasmon resonance for strength of histone-PolyP interactions. Results Both linear PolyP and PolyP-NP accelerated the fibrin formation and slowed down its dissolution and these effects were strongly dependent on the number of individual PolyP particles and not on their size. Addition of DNA did not modify significantly the PolyP-NP effects on fibrin formation and lysis. Both linear and nanoparticulate PolyP counteracted the effect of histone in the acceleration of fibrinogen clotting by thrombin. PolyP-NP, but not linear PolyP enhanced the prolongation of lysis time in fibrin containing histone and caused more pronounced thickening of the fibrin fibers than the linear form. Finally, PolyP-NP bound weaker to histone than the linear form. Conclusions The interaction of PolyP with histone was a stronger modulator of fibrin formation and lysis than its interaction with DNA. In addition, the PolyP nanoparticles enhanced the thrombus stabilizing effects of histone more effectively than linear PolyP.
Collapse
|