1
|
Chen J, Lynn EG, Sharma H, Byun JH, Kenyon VA, Sahu KK, Tyrrell DL, Houghton M, Gross PL, Trigatti BL, Shayegan B, Austin RC. Small molecules targeting GRP78 mitigate anti-GRP78 autoantibody-mediated tissue factor procoagulant activity in cultured endothelial cells. J Thromb Haemost 2024; 22:3290-3304. [PMID: 39111636 DOI: 10.1016/j.jtha.2024.07.015] [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: 12/02/2023] [Revised: 06/21/2024] [Accepted: 07/08/2024] [Indexed: 09/14/2024]
Abstract
BACKGROUND The 78-kDa glucose-regulated protein (GRP78) expressed on the cell surface (csGRP78) has been reported to regulate tissue factor (TF) procoagulant activity (PCA) in lesion-resident endothelial cells (ECs), which is further enhanced by circulating anti-GRP78 autoantibodies that bind to the Leu98-Leu115 epitope in GRP78. OBJECTIVES Determine the effects of the engagement of the anti-GRP78 autoantibody to csGRP78 on ECs and the underlying mechanisms that impact TF PCA. METHODS Immunofluorescent staining was used to determine the presence of csGRP78 in tumor necrosis factor α-treated ECs. An established TF PCA assay was used to evaluate human ECs following treatment with anti-GRP78 autoantibodies. The Fura 2-AM assay (Abcam) was used to quantify changes in intracellular Ca2+ levels. Small molecules predicted to bind GRP78 were identified using artificial intelligence. Enzyme-linked immunosorbent assays were used to assess the ability of these GRP78 binders to mitigate TF activity and interfere with the autoantibody/csGRP78 complex. RESULTS In tumor necrosis factor α-treated ECs, anti-GRP78 autoantibodies increased TF PCA. This observation was further enhanced by endoplasmic reticulum stress-induced elevation of csGRP78 levels. Anti-GRP78 autoantibody treatment increased intracellular Ca2+ levels. Sequestering the anti-GRP78 autoantibody with a conformational peptide or blocking with heparin attenuated anti-GRP78 autoantibody-induced TF PCA. We identified B07∗, as a GRP78 binder that diminished anti-GRP78 autoantibody-induced TF PCA on ECs. CONCLUSION These findings show how anti-GRP78 autoantibodies enhance TF PCA that contributes to thrombosis and identify novel GRP78 binders that represent a potential novel therapeutic strategy for treating and managing atherothrombotic disease.
Collapse
Affiliation(s)
- Jack Chen
- Department of Medicine, Division of Nephrology, McMaster University, The Research Institute of St. Joe's Hamilton and The Hamilton Center for Kidney Research, Hamilton, Ontario, Canada
| | - Edward G Lynn
- Department of Medicine, Division of Nephrology, McMaster University, The Research Institute of St. Joe's Hamilton and The Hamilton Center for Kidney Research, Hamilton, Ontario, Canada
| | - Hitesh Sharma
- Department of Medicine, Division of Nephrology, McMaster University, The Research Institute of St. Joe's Hamilton and The Hamilton Center for Kidney Research, Hamilton, Ontario, Canada
| | - Jae H Byun
- Department of Medicine, Division of Nephrology, McMaster University, The Research Institute of St. Joe's Hamilton and The Hamilton Center for Kidney Research, Hamilton, Ontario, Canada
| | | | - Kamlesh K Sahu
- Li Ka Shing Applied Virology Institute, Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta, Canada
| | - D Lorne Tyrrell
- Li Ka Shing Applied Virology Institute, Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta, Canada
| | - Michael Houghton
- Li Ka Shing Applied Virology Institute, Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta, Canada
| | - Peter L Gross
- Department of Medicine, Thrombosis and Atherosclerosis Research Institute, McMaster University and Hamilton Health Science, Hamilton, Ontario, Canada
| | - Bernardo L Trigatti
- Department of Biochemistry and Biomedical Sciences, McMaster University and Thrombosis and Atherosclerosis Research Institute, McMaster University and Hamilton Health Sciences, Hamilton, Ontario, Canada
| | - Bobby Shayegan
- Division of Urology, Department of Surgery, McMaster University and The Research Institute of St. Joe's Hamilton, Hamilton, Ontario, Canada
| | - Richard C Austin
- Department of Medicine, Division of Nephrology, McMaster University, The Research Institute of St. Joe's Hamilton and The Hamilton Center for Kidney Research, Hamilton, Ontario, Canada.
| |
Collapse
|
2
|
Bonifay A, Cointe S, Plantureux L, Lacroix R, Dignat-George F. Update on Tissue Factor Detection in Blood in 2024: A Narrative Review. Hamostaseologie 2024; 44:368-376. [PMID: 39442509 DOI: 10.1055/a-2381-6854] [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: 10/25/2024] Open
Abstract
Tissue factor (TF) is a transmembrane protein essential for hemostasis. Different forms of active TF circulate in the blood, either as a component of blood cells and extracellular vesicles (EVs) or as a soluble plasma protein. Accumulating experimental and clinical evidence suggests that TF plays an important role in thrombosis. Many in-house and commercially available assays have been developed to measure TF-dependent procoagulant activity or antigen in blood and have shown promising results for the prediction of disease outcomes or the occurrence of thrombosis events in diseases such as cancer or infectious coagulopathies. This review addresses the different assays that have been published for measuring circulating TF antigen and/or activity in whole blood, cell-free plasma, and EVs and discusses the main preanalytical and analytical parameters that impact results and their interpretation, highlighting their strengths and limitations. In the recent decade, EVTF assays have been significantly developed. Among them, functional assays that use a blocking anti-TF antibody or immunocapture to measure EVTF activity have higher specificity and sensitivity than antigen assays. However, there is still a high variability between assays. Standardization and automatization are prerequisites for the measurement of EVTF in clinical laboratories.
Collapse
Affiliation(s)
- Amandine Bonifay
- Aix-Marseille University, C2VN, INSERM 1263, INRAE 1260, Marseille, France
- Department of Hematology, Biogénopôle, CHU La Timone, APHM, Marseille, France
| | - Sylvie Cointe
- Aix-Marseille University, C2VN, INSERM 1263, INRAE 1260, Marseille, France
- Department of Hematology, Biogénopôle, CHU La Timone, APHM, Marseille, France
| | - Léa Plantureux
- Aix-Marseille University, C2VN, INSERM 1263, INRAE 1260, Marseille, France
| | - Romaric Lacroix
- Aix-Marseille University, C2VN, INSERM 1263, INRAE 1260, Marseille, France
- Department of Hematology, Biogénopôle, CHU La Timone, APHM, Marseille, France
| | - Françoise Dignat-George
- Aix-Marseille University, C2VN, INSERM 1263, INRAE 1260, Marseille, France
- Department of Hematology, Biogénopôle, CHU La Timone, APHM, Marseille, France
| |
Collapse
|
3
|
Noone D, Preston RJS, Rehill AM. The Role of Myeloid Cells in Thromboinflammatory Disease. Semin Thromb Hemost 2024; 50:998-1011. [PMID: 38547918 DOI: 10.1055/s-0044-1782660] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
Inflammation contributes to the development of thrombosis, but the mechanistic basis for this association remains poorly understood. Innate immune responses and coagulation pathways are activated in parallel following infection or injury, and represent an important host defense mechanism to limit pathogen spread in the bloodstream. However, dysregulated proinflammatory activity is implicated in the progression of venous thromboembolism and arterial thrombosis. In this review, we focus on the role of myeloid cells in propagating thromboinflammation in acute inflammatory conditions, such as sepsis and coronavirus disease 2019 (COVID-19), and chronic inflammatory conditions, such as obesity, atherosclerosis, and inflammatory bowel disease. Myeloid cells are considered key drivers of thromboinflammation via upregulated tissue factor activity, formation of neutrophil extracellular traps (NETs), contact pathway activation, and aberrant coagulation factor-mediated protease-activated receptor (PAR) signaling. We discuss how strategies to target the intersection between myeloid cell-mediated inflammation and activation of blood coagulation represent an exciting new approach to combat immunothrombosis. Specifically, repurposed anti-inflammatory drugs, immunometabolic regulators, and NETosis inhibitors present opportunities that have the potential to dampen immunothrombotic activity without interfering with hemostasis. Such therapies could have far-reaching benefits for patient care across many thromboinflammatory conditions.
Collapse
Affiliation(s)
- David Noone
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, Dublin, Ireland
- National Children's Research Centre, Our Lady's Children's Hospital Crumlin, Dublin, Ireland
| | - Roger J S Preston
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, Dublin, Ireland
- National Children's Research Centre, Our Lady's Children's Hospital Crumlin, Dublin, Ireland
| | - Aisling M Rehill
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, Dublin, Ireland
- National Children's Research Centre, Our Lady's Children's Hospital Crumlin, Dublin, Ireland
| |
Collapse
|
4
|
Moruzzi S, Castagna A, Spizzo M, Udali S, Pattini P, Pizzolo F, Friso S, Martinelli N. Activated Factor VII-Antithrombin Complex, a Biomarker of Tissue Factor-Related Pathways in Different Clinical Settings: A Narrative Review from Cardiovascular Diseases to Cancer. Diagnostics (Basel) 2024; 14:1711. [PMID: 39202199 PMCID: PMC11354109 DOI: 10.3390/diagnostics14161711] [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: 07/08/2024] [Revised: 08/02/2024] [Accepted: 08/03/2024] [Indexed: 09/03/2024] Open
Abstract
Tissue factor (TF) is a transmembrane glycoprotein that represents the fundamental physiological initiator of the coagulation cascade through its interaction with factor VII. TF belongs to the cytokine receptor protein superfamily and contributes to the transduction of cellular signaling. Therefore, TF-related pathways are involved in multiple pathophysiological processes, not only in coagulation/thrombosis but in a wider mechanisms' panorama, ranging from infective to neoplastic diseases. Consistently, the measurement of TF activity could have a diagnostic and/or prognostic meaning in different clinical conditions. However, the transmembrane localization, the expression on different cellular types and circulating extracellular vesicles, and the different conformations (encrypted and decrypted) and variants (such as the soluble alternatively spliced TF) hamper TF assessment in clinical practice. The activated factor VII-antithrombin (FVIIa-AT) complex is proposed as an indirect biomarker of the TF-FVIIa interaction and, consequently, of the functionally active TF expression. In this narrative review, we evaluate the clinical studies investigating the role of plasma concentration of FVIIa-AT in health and disease. Although without conclusive data, high FVIIa-AT concentrations predict the worst clinical outcomes in different pathologic conditions, such as cardiovascular disease and cancer, thereby suggesting that overactivation of TF-related pathways may play an unfavorable role in various clinical settings.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Nicola Martinelli
- Department of Medicine, University of Verona, 37134 Verona, Italy; (S.M.); (A.C.); (M.S.); (S.U.); (P.P.); (F.P.); (S.F.)
| |
Collapse
|
5
|
Yu M, Li X, Xu L, Zheng C, Pan W, Chen H, Liu X, Zhang X, Zhang J. Neutrophil extracellular traps induce intrahepatic thrombotic tendency and liver damage in cholestatic liver disease. Hepatol Commun 2024; 8:e0513. [PMID: 39101776 PMCID: PMC11299992 DOI: 10.1097/hc9.0000000000000513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 06/28/2024] [Indexed: 08/06/2024] Open
Abstract
BACKGROUND Cholestatic liver diseases induce local and systemic hypercoagulation, with neutrophil extracellular traps (NETs) serving as major drivers. These NETs have been linked to decreased liver function in patients with obstructive jaundice. However, the impact of NETs on liver hypercoagulation in cholestatic liver disease remains unknown. METHODS We utilized bile duct ligation to create experimental mice and analyzed NETs formation in the liver. Fibrin deposition, tissue factor expression, and inflammation in the liver were visualized through western blot and immunohistochemical techniques. LSECs were incubated with isolated NETs, and we detected endothelial procoagulant activity using coagulation protein production assays and measuring endothelial permeability. In both in vivo and in vitro settings, DNase I was applied to clarify the effect of NETs on intrahepatic hypercoagulability, hepatotoxicity, LSEC, and macrophage activation or injury. RESULTS Bile duct ligation mice exhibited significantly increased levels of NETs in liver tissue, accompanied by neutrophil infiltration, tissue necrosis, fibrin deposition, and thrombophilia compared to sham mice. Notably, NETs resulted in phosphatidylserine and tissue factor exposure on LSEC, enhancing coagulation Factor Xa and thrombin production. The enhanced procoagulant activity could be reversed by degrading NETs with DNase I. Additionally, NETs-induced permeability changes in LSECs, characterized by increased VE-cadherin expression and F-actin retraction, which could be rescued by DNase I. Meanwhile, NET formation is associated with KC activation and the formation of inflammatory factors. CONCLUSIONS NETs promote intrahepatic activation of coagulation and inflammation, leading to liver tissue injury. Strategies targeting NET formation may offer a potential therapeutic approach for treating cholestatic liver disease.
Collapse
Affiliation(s)
- Muxin Yu
- Department of Clinical Medical Sciences, College of Medicine, Jiaxing University, Jiaxing, China
| | - Xiaowen Li
- Department of Pathology, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Long Xu
- Department of Clinical Medical Sciences, College of Medicine, Jiaxing University, Jiaxing, China
| | - Chuwei Zheng
- Department of Gastroenterology, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Weiwei Pan
- Department of Clinical Medical Sciences, College of Medicine, Jiaxing University, Jiaxing, China
| | - Hui Chen
- Department of Clinical Medical Sciences, College of Medicine, Jiaxing University, Jiaxing, China
| | - Xiaoyu Liu
- Department of Clinical Medical Sciences, College of Medicine, Jiaxing University, Jiaxing, China
| | - Xianshan Zhang
- Department of Clinical Medical Sciences, College of Medicine, Jiaxing University, Jiaxing, China
| | - Jinming Zhang
- Department of Gastroenterology, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| |
Collapse
|
6
|
Mohammad MA, Featherby S, Ettelaie C. Regulation of tissue factor activity by interaction with the first PDZ domain of MAGI1. Thromb J 2024; 22:12. [PMID: 38233821 PMCID: PMC10792917 DOI: 10.1186/s12959-023-00580-6] [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: 12/14/2023] [Accepted: 12/29/2023] [Indexed: 01/19/2024] Open
Abstract
BACKGROUND Tissue factor (TF) activity is stringently regulated through processes termed encryption. Post-translational modification of TF and its interactions with various protein and lipid moieties allows for a multi-step de-encryption of TF and procoagulant activation. Membrane-associated guanylate kinase-with inverted configuration (MAGI) proteins are known to regulate the localisation and activity of a number of proteins including cell-surface receptors. METHODS The interaction of TF with MAGI1 protein was examined as a means of regulating TF activity. MDA-MB-231 cell line was used which express TF and MAGI1, and respond well to protease activated receptor (PAR)2 activation. Proximity ligation assay (PLA), co-immunoprecipitation and pull-down experiments were used to examine the interaction of TF with MAGI1-3 proteins and to investigate the influence of PAR2 activation. Furthermore, by cloning and expressing the PDZ domains from MAGI1, the TF-binding domain was identified. The ability of the recombinant PDZ domains to act as competitors for MAGI1, allowing the induction of TF procoagulant and signalling activity was then examined. RESULTS PLA and fluorescence microscopic analysis indicated that TF predominantly associates with MAGI1 and less with MAGI2 and MAGI3 proteins. The interaction of TF with MAGI1 was also demonstrated by both co-immunoprecipitation of TF with MAGI1, and co-immunoprecipitation of MAGI1 with TF. Moreover, activation of PAR2 resulted in reduction in the association of these two proteins. Pull-down assays using TF-cytoplasmic domain peptides indicated that the phosphorylation of Ser253 within TF prevents its association with MAGI1. Additionally, the five HA-tagged PDZ domains of MAGI1 were overexpressed separately, and the putative TF-binding domain was identified as PDZ1 domain. Expression of this PDZ domain in cells significantly augmented the TF activity measured both as thrombin-generation and also TF-mediated proliferative signalling. CONCLUSIONS Our data indicate a stabilising interaction between TF and the PDZ-1 domain of MAGI1 and demonstrate that the activation of PAR2 disrupts this interaction. The release of TF from MAGI1 appears to be an initial step in TF de-encryption, associated with increased TF-mediated procoagulant and signalling activities. This mechanism is also likely to lead to further interactions and modifications leading to further enhancement of procoagulant activity, or the release of TF.
Collapse
Affiliation(s)
- Mohammad A Mohammad
- Biomedical Sciences/Hull York Medial School, University of Hull, Cottingham Road, Hull, HU6 7RX, UK
- Present address: The Department of Interdisciplinary Oncology, LSUHSC, New Orleans, LA, 70112m, USA
| | - Sophie Featherby
- Biomedical Sciences/Hull York Medial School, University of Hull, Cottingham Road, Hull, HU6 7RX, UK
| | - Camille Ettelaie
- Biomedical Sciences/Hull York Medial School, University of Hull, Cottingham Road, Hull, HU6 7RX, UK.
| |
Collapse
|
7
|
Ranjbar J, Yang Y, Harper AGS. Developing human tissue engineered arterial constructs to simulate human in vivo thrombus formation. Platelets 2023; 34:2153823. [PMID: 36550074 DOI: 10.1080/09537104.2022.2153823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Thrombus formation is highly dependent upon the physico-chemical environment in which it is triggered. Our ability to understand how thrombus formation is initiated, regulated, and resolved in the human body is dependent upon our ability to replicate the mechanical and biological properties of the arterial wall. Current in vitro thrombosis models principally use reductionist approaches to model the complex biochemical and cellular milieu present in the arterial wall, and so researcher have favored the use of in vivo models. The field of vascular tissue engineering has developed a range of techniques for culturing artificial human arteries for use as vascular grafts. These techniques therefore provide a basis for developing more sophisticated 3D replicas of the arterial wall that can be used in in vitro thrombosis models. In this review, we consider how tissue engineering approaches can be used to generate 3D models of the arterial wall that improve upon current in vivo and in vitro approaches. We consider the current benefits and limitations of reported 3D tissue engineered models and consider what additional evidence is required to validate them as alternatives to current in vivo models.
Collapse
Affiliation(s)
| | - Ying Yang
- School of Pharmacy & Bioengineering, Keele University, Keele, UK
| | | |
Collapse
|
8
|
Thaler J, Prager G, Pabinger I, Ay C. Plasma Clot Properties in Patients with Pancreatic Cancer. Cancers (Basel) 2023; 15:4030. [PMID: 37627058 PMCID: PMC10452192 DOI: 10.3390/cancers15164030] [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: 05/04/2023] [Revised: 07/26/2023] [Accepted: 08/03/2023] [Indexed: 08/27/2023] Open
Abstract
Pancreatic cancer is one of the most prothrombotic malignancies. Plasma clot properties may be altered in patients with pancreatic cancer, and circulating tissue factor (TF) may play an important role. We applied a modified plasma clot formation assay (only CaCl2 and phospholipids were added to initiate clotting) and a standard clotting assay (lipidated TF was also added) to investigate whether plasma clot properties are altered in pancreatic cancer patients (n = 40, 23 female) compared to sex-matched healthy controls. The modified assay was also performed in the presence of a TF blocking antibody. With this modified assay, we detected an increased plasma clot formation rate (Vmax) and an increased delta absorbance (ΔAbs, indicating fibrin fiber thickness) in patients compared to controls. These differences were not detected with the standard clotting assay. Following addition of a TF blocking antibody in in our modified assay, Vmax decreased significantly in patients only, ΔAbs significantly decreased in patients and in healthy controls, the lag phase did not change, and the time to peak fibrin generation increased in patients only. Taken together, these findings indicate the presence of a prothrombotic state in pancreatic cancer patients, which depends on TF and is detectable with our modified assay but not with a standard clotting assay.
Collapse
Affiliation(s)
- Johannes Thaler
- Clinical Division of Haematology and Haemostaseology, Department of Medicine I, Medical University of Vienna, 1090 Vienna, Austria; (I.P.); (C.A.)
| | - Gerald Prager
- Clinical Division of Oncology, Department of Medicine I, Medical University of Vienna, 1090 Vienna, Austria;
| | - Ingrid Pabinger
- Clinical Division of Haematology and Haemostaseology, Department of Medicine I, Medical University of Vienna, 1090 Vienna, Austria; (I.P.); (C.A.)
| | - Cihan Ay
- Clinical Division of Haematology and Haemostaseology, Department of Medicine I, Medical University of Vienna, 1090 Vienna, Austria; (I.P.); (C.A.)
| |
Collapse
|
9
|
Marcos-Jubilar M, Lecumberri R, Páramo JA. Immunothrombosis: Molecular Aspects and New Therapeutic Perspectives. J Clin Med 2023; 12:1399. [PMID: 36835934 PMCID: PMC9958829 DOI: 10.3390/jcm12041399] [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: 12/20/2022] [Revised: 02/07/2023] [Accepted: 02/08/2023] [Indexed: 02/12/2023] Open
Abstract
Thromboinflammation or immunothrombosis is a concept that explains the existing link between coagulation and inflammatory response present in many situations, such as sepsis, venous thromboembolism, or COVID-19 associated coagulopathy. The purpose of this review is to provide an overview of the current data regarding the mechanisms involved in immunothrombosis in order to understand the new therapeutic strategies focused in reducing thrombotic risk by controlling the inflammation.
Collapse
Affiliation(s)
- María Marcos-Jubilar
- Hematology and Hemotherapy Service, Clínica Universidad de Navarra, 31008 Pamplona, Spain
| | - Ramón Lecumberri
- Hematology and Hemotherapy Service, Clínica Universidad de Navarra, 31008 Pamplona, Spain
- CIBER-CV, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - José A. Páramo
- Hematology and Hemotherapy Service, Clínica Universidad de Navarra, 31008 Pamplona, Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain
| |
Collapse
|
10
|
Wang J, Keshava S, Das K, Kolesnick R, Jiang XC, Pendurthi UR, Rao LVM. Alterations to Sphingomyelin Metabolism Affect Hemostasis and Thrombosis. Arterioscler Thromb Vasc Biol 2023; 43:64-78. [PMID: 36412194 PMCID: PMC9762718 DOI: 10.1161/atvbaha.122.318443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
BACKGROUND Our recent studies suggest that sphingomyelin levels in the plasma membrane influence TF (tissue factor) procoagulant activity. The current study was performed to investigate how alterations to sphingomyelin metabolic pathway would affect TF procoagulant activity and thereby affect hemostatic and thrombotic processes. METHODS Macrophages and endothelial cells were transfected with specific siRNAs or infected with adenoviral vectors to alter sphingomyelin levels in the membrane. TF activity was measured in factor X activation assay. Saphenous vein incision-induced bleeding and the inferior vena cava ligation-induced flow restriction mouse models were used to evaluate hemostasis and thrombosis, respectively. RESULTS Overexpression of SMS (sphingomyelin synthase) 1 or SMS2 in human monocyte-derived macrophages suppresses ATP-stimulated TF procoagulant activity, whereas silencing SMS1 or SMS2 increases the basal cell surface TF activity to the same level as of ATP-decrypted TF activity. Consistent with the concept that sphingomyelin metabolism influences TF procoagulant activity, silencing of acid sphingomyelinase or neutral sphingomyelinase 2 or 3 attenuates ATP-induced enhanced TF procoagulant activity in macrophages and endothelial cells. Niemann-Pick disease fibroblasts with a higher concentration of sphingomyelin exhibited lower TF activity compared with wild-type fibroblasts. In vivo studies revealed that LPS+ATP-induced TF activity and thrombin generation were attenuated in ASMase-/- mice, while their levels were increased in SMS2-/- mice. Further studies revealed that acid sphingomyelinase deficiency leads to impaired hemostasis, whereas SMS2 deficiency increases thrombotic risk. CONCLUSIONS Overall, our data indicate that alterations in sphingomyelin metabolism would influence TF procoagulant activity and affect hemostatic and thrombotic processes.
Collapse
Affiliation(s)
- Jue Wang
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler (J.W., S.K., K.D., U.R.P., L.V.M.R.)
| | - Shiva Keshava
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler (J.W., S.K., K.D., U.R.P., L.V.M.R.)
| | - Kaushik Das
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler (J.W., S.K., K.D., U.R.P., L.V.M.R.)
| | | | | | - Usha R Pendurthi
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler (J.W., S.K., K.D., U.R.P., L.V.M.R.)
| | - L Vijaya Mohan Rao
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler (J.W., S.K., K.D., U.R.P., L.V.M.R.)
| |
Collapse
|
11
|
Sachetto ATA, Mackman N. Tissue Factor and COVID-19: An Update. Curr Drug Targets 2022; 23:1573-1577. [PMID: 36165519 DOI: 10.2174/1389450123666220926144432] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 08/21/2022] [Accepted: 08/29/2022] [Indexed: 01/25/2023]
Abstract
The coronavirus 2019 (COVID-19) pandemic is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virus. Infection with SARS-CoV-2 is associated with acute respiratory distress syndrome, thrombosis and a high rate of mortality. Thrombotic events increase with severity. Tissue factor (TF) expression is increased during viral and bacterial infections. This review summarizes studies that have examined TF expression in response to SARS-CoV-2 infection. SARS-CoV-2 virus and its proteins upregulate TF mRNA, protein and activity in a variety of cells, including bronchial epithelial cells, neutrophils, monocytes, macrophages, endothelial cells and adventitial fibroblasts. COVID-19 patients have increased TF expression in lungs, bronchoalveolar lavage fluid and circulating extracellular vesicles. The increase in TF was associated with coagulation activation markers, thrombosis, inflammatory markers, severity of disease and mortality. Taken together, the studies suggest that TF plays a central role in thrombosis in COVID- 19. TF may be a useful prognostic marker and therapeutic target to reduce thrombosis and inflammation.
Collapse
Affiliation(s)
- Ana Teresa Azevedo Sachetto
- Laboratory of Pathophysiology, Butantan Institute, São Paulo, São Paulo, Brazil.,University of São Paulo Medical School, São Paulo, São Paulo, Brazil.,Department of Medicine, UNC Blood Research Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Nigel Mackman
- Department of Medicine, UNC Blood Research Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
12
|
Liu L, Deng QJ. Role of platelet-derived extracellular vesicles in traumatic brain injury-induced coagulopathy and inflammation. Neural Regen Res 2022; 17:2102-2107. [PMID: 35259815 PMCID: PMC9083154 DOI: 10.4103/1673-5374.335825] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Extracellular vesicles are composed of fragments of exfoliated plasma membrane, organelles or nuclei and are released after cell activation, apoptosis or destruction. Platelet-derived extracellular vesicles are the most abundant type of extracellular vesicle in the blood of patients with traumatic brain injury. Accumulated laboratory and clinical evidence shows that platelet-derived extracellular vesicles play an important role in coagulopathy and inflammation after traumatic brain injury. This review discusses the recent progress of research on platelet-derived extracellular vesicles in coagulopathy and inflammation and the potential of platelet-derived extracellular vesicles as therapeutic targets for traumatic brain injury.
Collapse
Affiliation(s)
- Liang Liu
- Department of Neurosurgery, Tianjin Institute of Neurology, Tianjin Medical University General Hospital, Tianjin, China
| | - Quan-Jun Deng
- Department of Neurosurgery, Tianjin Institute of Neurology, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
13
|
Wilkinson H, Leonard H, Robson MG, Smith R, Tam E, McVey JH, Kirckhofer D, Chen D, Dorling A. Manipulation of tissue factor-mediated basal PAR-2 signalling on macrophages determines sensitivity for IFNγ responsiveness and significantly modifies the phenotype of murine DTH. Front Immunol 2022; 13:999871. [PMID: 36172348 PMCID: PMC9510775 DOI: 10.3389/fimmu.2022.999871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 08/24/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundTissue factor (TF) generates proteases that can signal through PAR-1 and PAR-2. We have previously demonstrated PAR-1 signalling primes innate myeloid cells to be exquisitely sensitive to interferon-gamma (IFNγ). In this work we explored how TF mediated PAR-2 signalling modulated responsiveness to IFNγ and investigated the interplay between PAR-1/-2 signalling on macrophages.MethodologyWe characterised how TF through PAR-2 influenced IFNγ sensitivity in vitro using PCR and flow cytometry. and how it influenced oxazolone-induced delayed type hypersensitivity (DTH) responses in vivo. We investigated how basal signalling through PAR-2 influenced PAR-1 signalling using a combination of TF-inhibitors and PAR-1 &-2 agonists and antagonists. Finally, we investigated whether this system could be targeted therapeutically using 3-mercaptopropionyl-F-Cha-Cha-RKPNDK (3-MP), which has actions on both PAR-1 and -2.ResultsTF delivered a basal signal through PAR-2 that upregulated SOCS3 expression and blunted M1 polarisation after IFNγ stimulation, opposing the priming achieved by signalling through PAR-1. PAR-1 and -2 agonists or antagonists could be used in combination to modify this basal signal in vitro and in vivo. 3-MP, by virtue of its PAR-2 agonist properties was superior to agents with only PAR-1 antagonist properties at reducing M1 polarisation induced by IFNγ and suppressing DTH. Tethering a myristoyl electrostatic switch almost completely abolished the DTH response.ConclusionsTF-mediated signalling through PARs-1 and -2 act in a homeostatic way to determine how myeloid cells respond to IFNγ. 3-MP, an agent that simultaneously inhibits PAR-1 whilst delivering a PAR-2 signal, can almost completely abolish immune responses dependent on M1 polarisation, particularly if potency is enhanced by targeting to cell membranes; this has potential therapeutic potential in multiple diseases.
Collapse
Affiliation(s)
- Hannah Wilkinson
- Department of Inflammation Biology, School of Immunology & Microbial Sciences, King’s College London, Guy’s Hospital, London, United Kingdom
- *Correspondence: Anthony Dorling, ; Hannah Wilkinson,
| | - Hugh Leonard
- Department of Inflammation Biology, School of Immunology & Microbial Sciences, King’s College London, Guy’s Hospital, London, United Kingdom
| | - Michael G. Robson
- Department of Inflammation Biology, School of Immunology & Microbial Sciences, King’s College London, Guy’s Hospital, London, United Kingdom
| | - Richard Smith
- Department of Inflammation Biology, School of Immunology & Microbial Sciences, King’s College London, Guy’s Hospital, London, United Kingdom
| | - ElLi Tam
- Department of Inflammation Biology, School of Immunology & Microbial Sciences, King’s College London, Guy’s Hospital, London, United Kingdom
| | - John H. McVey
- School of Bioscience & Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom
| | - Daniel Kirckhofer
- Department of Early Discovery Biochemistry, Genentech Inc., South San Francisco, CA, United States
| | - Daxin Chen
- Department of Inflammation Biology, School of Immunology & Microbial Sciences, King’s College London, Guy’s Hospital, London, United Kingdom
| | - Anthony Dorling
- Department of Inflammation Biology, School of Immunology & Microbial Sciences, King’s College London, Guy’s Hospital, London, United Kingdom
- *Correspondence: Anthony Dorling, ; Hannah Wilkinson,
| |
Collapse
|
14
|
Immunothrombosis and the molecular control of tissue factor by pyroptosis: prospects for new anticoagulants. Biochem J 2022; 479:731-750. [PMID: 35344028 DOI: 10.1042/bcj20210522] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 03/09/2022] [Accepted: 03/10/2022] [Indexed: 02/06/2023]
Abstract
The interplay between innate immunity and coagulation after infection or injury, termed immunothrombosis, is the primary cause of disseminated intravascular coagulation (DIC), a condition that occurs in sepsis. Thrombosis associated with DIC is the leading cause of death worldwide. Interest in immunothrombosis has grown because of COVID-19, the respiratory disease caused by SARS-CoV-2, which has been termed a syndrome of dysregulated immunothrombosis. As the relatively new field of immunothrombosis expands at a rapid pace, the focus of academic and pharmacological research has shifted from generating treatments targeted at the traditional 'waterfall' model of coagulation to therapies better directed towards immune components that drive coagulopathies. Immunothrombosis can be initiated in macrophages by cleavage of the non-canonical inflammasome which contains caspase-11. This leads to release of tissue factor (TF), a membrane glycoprotein receptor that forms a high-affinity complex with coagulation factor VII/VIIa to proteolytically activate factors IX to IXa and X to Xa, generating thrombin and leading to fibrin formation and platelet activation. The mechanism involves the post-translational activation of TF, termed decryption, and release of decrypted TF via caspase-11-mediated pyroptosis. During aberrant immunothrombosis, decryption of TF leads to thromboinflammation, sepsis, and DIC. Therefore, developing therapies to target pyroptosis have emerged as an attractive concept to counteract dysregulated immunothrombosis. In this review, we detail the three mechanisms of TF control: concurrent induction of TF, caspase-11, and NLRP3 (signal 1); TF decryption, which increases its procoagulant activity (signal 2); and accelerated release of TF into the intravascular space via pyroptosis (signal 3). In this way, decryption of TF is analogous to the two signals of NLRP3 inflammasome activation, whereby induction of pro-IL-1β and NLRP3 (signal 1) is followed by activation of NLRP3 (signal 2). We describe in detail TF decryption, which involves pathogen-induced alterations in the composition of the plasma membrane and modification of key cysteines on TF, particularly at the location of the critical, allosterically regulated disulfide bond of TF in its 219-residue extracellular domain. In addition, we speculate towards the importance of identifying new therapeutics to block immunothrombotic triggering of TF, which can involve inhibition of pyroptosis to limit TF release, or the direct targeting of TF decryption using cysteine-modifying therapeutics.
Collapse
|
15
|
Arsiccio A, Metcalfe C, Pisano R, Raut S, Coxon C. A proximity-based in silico approach to identify redox-labile disulfide bonds: The example of FVIII. PLoS One 2022; 17:e0262409. [PMID: 35130281 PMCID: PMC8820644 DOI: 10.1371/journal.pone.0262409] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 12/27/2021] [Indexed: 01/04/2023] Open
Abstract
Allosteric disulfide bonds permit highly responsive, transient 'switch-like' properties that are ideal for processes like coagulation and inflammation that require rapid and localised responses to damage or injury. Haemophilia A (HA) is a rare bleeding disorder managed with exogenous coagulation factor(F) VIII products. FVIII has eight disulfide bonds and is known to be redox labile, but it is not known how reduction/oxidation affects the structure-function relationship, or its immunogenicity-a serious complication for 30% severe HA patients. Understanding how redox-mediated changes influence FVIII can inform molecular engineering strategies aimed at improving activity and stability, and reducing immunogenicity. FVIII is a challenging molecule to work with owing to its poor expression and instability so, in a proof-of-concept study, we used molecular dynamics (MD) to identify which disulfide bonds were most likely to be reduced and how this would affect structure/function; results were then experimentally verified. MD identified Cys1899-Cys1903 disulfide as the most likely to undergo reduction based on energy and proximity criteria. Further MD suggested this reduction led to a more open conformation. Here we present our findings and highlight the value of MD approaches.
Collapse
Affiliation(s)
- Andrea Arsiccio
- Department of Applied Science and Technology, Politecnico di Torino, Torino, Italy
| | - Clive Metcalfe
- National Institute for Biological Standards and Control, Hertfordshire, United Kingdom
| | - Roberto Pisano
- Department of Applied Science and Technology, Politecnico di Torino, Torino, Italy
| | - Sanj Raut
- National Institute for Biological Standards and Control, Hertfordshire, United Kingdom
| | - Carmen Coxon
- National Institute for Biological Standards and Control, Hertfordshire, United Kingdom
| |
Collapse
|
16
|
Vascular thiol isomerases: Structures, regulatory mechanisms, and inhibitor development. Drug Discov Today 2021; 27:626-635. [PMID: 34757205 DOI: 10.1016/j.drudis.2021.10.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 09/15/2021] [Accepted: 10/25/2021] [Indexed: 12/15/2022]
Abstract
Vascular thiol isomerases (VTIs), including PDI, ERp5, ERp57, ERp72, and thioredoxin-related transmembrane protein 1 (TMX1), have important roles in platelet aggregation and thrombosis. Research on VTIs, their substrates in thrombosis, their regulatory mechanisms, and inhibitor development is an emerging and rapidly evolving area in vascular biology. Here, we describe the structures and functions of VTIs, summarize the relationship between the vascular TIs and thrombosis, and focus on the development of VTI inhibitors for antithrombotic applications.
Collapse
|
17
|
Greimel T, Jahnel J, Pohl S, Strini T, Tischitz M, Meier-Allard N, Holasek S, Meinel K, Aguiriano-Moser V, Zobel J, Haidl H, Gallistl S, Panzitt K, Wagner M, Schlagenhauf A. Bile acid-induced tissue factor activity in hepatocytes correlates with activation of farnesoid X receptor. J Transl Med 2021; 101:1394-1402. [PMID: 34145381 PMCID: PMC8440176 DOI: 10.1038/s41374-021-00628-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 05/20/2021] [Accepted: 06/04/2021] [Indexed: 12/13/2022] Open
Abstract
Bile acids (BA) have been found to promote coagulation by increasing tissue factor (TF) activity. The contribution of elevated BA levels and cholestasis to TF decryption within the liver parenchyma and the role of farnesoid X receptor (FXR) in this process remain unclear. We investigated the effects of BA on TF activity and thrombin generation in hepatocytes and correlated these effects with activation of FXR-dependent signaling and apoptosis. HepG2 cells and primary hepatocytes were incubated with chenodeoxycholic acid (CDCA), glycochenodeoxycholic acid (GCDCA), ursodeoxycholic acid (UCDA), or the synthetic FXR agonist GW4064 for 24 h. MTT tests demonstrated cell viability throughout experiments. TF activity was tested via factor Xa generation and thrombin generation was measured by calibrated automated thrombography. Increased TF activity alongside enhanced thrombin generation was observed with CDCA and GW4064 but not with GCDCA and UDCA. TF activity was substantially reduced when FXR activation was blocked with the antagonist DY 268. Quantitative polymerase chain reaction revealed upregulation of FXR target genes only by CDCA and GW4064. Western blot analysis and fluorescence microscopy showed no TF overexpression arguing for TF decryption. Caspase 3 activity measurements and flow cytometric analysis of Annexin V binding showed no signs of apoptosis. Long-term exposure of hepatocytes to nontoxic BA may cause intracellular FXR overstimulation, triggering TF decryption irrespective of the amphiphilic properties of BA. The effect of BA on TF activation correlates with the molecule's ability to enter the cells and activate FXR. TF decryption occurs independently of apoptotic mechanisms.
Collapse
Affiliation(s)
- Theresa Greimel
- Division of General Paediatrics, Department of Paediatrics and Adolescent Medicine, Medical University Graz, Graz, Austria
| | - Jörg Jahnel
- Division of General Paediatrics, Department of Paediatrics and Adolescent Medicine, Medical University Graz, Graz, Austria
| | - Sina Pohl
- Division of General Paediatrics, Department of Paediatrics and Adolescent Medicine, Medical University Graz, Graz, Austria
| | - Tanja Strini
- Division of General Paediatrics, Department of Paediatrics and Adolescent Medicine, Medical University Graz, Graz, Austria
| | - Martin Tischitz
- Division of General Paediatrics, Department of Paediatrics and Adolescent Medicine, Medical University Graz, Graz, Austria
| | - Nathalie Meier-Allard
- Division of Immunology and Pathophysiology, Otto Loewi Research Center, Medical University Graz, Graz, Austria
| | - Sandra Holasek
- Division of Immunology and Pathophysiology, Otto Loewi Research Center, Medical University Graz, Graz, Austria
| | - Katharina Meinel
- Division of General Paediatrics, Department of Paediatrics and Adolescent Medicine, Medical University Graz, Graz, Austria
| | - Victor Aguiriano-Moser
- Division of General Paediatrics, Department of Paediatrics and Adolescent Medicine, Medical University Graz, Graz, Austria
| | - Joachim Zobel
- Division of General Paediatrics, Department of Paediatrics and Adolescent Medicine, Medical University Graz, Graz, Austria
| | - Harald Haidl
- Division of General Paediatrics, Department of Paediatrics and Adolescent Medicine, Medical University Graz, Graz, Austria
| | - Siegfried Gallistl
- Division of General Paediatrics, Department of Paediatrics and Adolescent Medicine, Medical University Graz, Graz, Austria
| | - Katrin Panzitt
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Medical University Graz, Graz, Austria
| | - Martin Wagner
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Medical University Graz, Graz, Austria
| | - Axel Schlagenhauf
- Division of General Paediatrics, Department of Paediatrics and Adolescent Medicine, Medical University Graz, Graz, Austria.
| |
Collapse
|
18
|
Ettelaie C, Featherby S, Rondon AMR, Greenman J, Versteeg HH, Maraveyas A. De-Palmitoylation of Tissue Factor Regulates Its Activity, Phosphorylation and Cellular Functions. Cancers (Basel) 2021; 13:cancers13153837. [PMID: 34359738 PMCID: PMC8345185 DOI: 10.3390/cancers13153837] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 07/21/2021] [Accepted: 07/26/2021] [Indexed: 11/16/2022] Open
Abstract
In this study, the role of de-palmitoylation of tissue factor (TF) in the decryption of its activity was explored. TF-tGFP constructs were prepared by mutagenesis-substitution at Cys245 to prevent or mimic palmitolyation. Additionally, to reduce TF de-palmitoylation, the expression of palmitoyl-protein thioesterases (PPT) was suppressed. Other TF mutants were prepared with altered flexibility, hydrophobicity or length of the transmembrane domain. The outcome of these alterations on fXa-generation, fVIIa binding, Ser253 phosphorylation and TF-microvesicle release were assessed in endothelial cells, and the influence on endothelial and MCF-7 cell proliferation and apoptosis was analysed. Preventing TF palmitoylation (TFSer245-tGFP), increasing the hydrophobicity (TFPhe241-tGFP) or lengthening (TFLongTM-tGFP) of the transmembrane domain enhanced fXa-generation in resting cells compared to cells expressing TFWt-tGFP, but fXa-generation was not further increased following PAR2 activation. Extending the available length of the transmembrane domain enhanced the TF-tGFP release within microvesicles and Ser253 phosphorylation and increased cell proliferation. Moreover, prevention of PKCα-mediated Ser253 phosphorylation with Gö6976 did not preclude fXa-generation. Conversely, reducing the hydrophobicity (TFSer242-tGFP), shortening (TFShortTM-tGFP) or reducing the flexibility (TFVal225-tGFP) of the transmembrane domain suppressed fXa-generation, fVIIa-HRP binding and Ser253 phosphorylation following PAR2 activation. PPT knock-down or mimicking palmitoylation (TFPhe245-tGFP) reduced fXa-generation without affecting fVIIa binding. This study has for the first time shown that TF procoagulant activity is regulated through de-palmitoylation, which alters the orientation of its transmembrane domain and is independent of TF phosphorylation. However, Ser253 phosphorylation is facilitated by changes in the orientation of the transmembrane domain and can induce TF-cellular signalling that influences cellular proliferation/apoptosis.
Collapse
Affiliation(s)
- Camille Ettelaie
- Biomedical Section, University of Hull, Cottingham Road, Hull HU6 7RX, UK; (S.F.); (J.G.)
- Correspondence: ; Tel.: +44-(0)1482-465528; Fax: +44-(0)1482-465458
| | - Sophie Featherby
- Biomedical Section, University of Hull, Cottingham Road, Hull HU6 7RX, UK; (S.F.); (J.G.)
| | - Araci M. R. Rondon
- Einthoven Laboratory for Vascular and Regenerative Medicine, Division of Thrombosis and Hemostasis, Department of Internal Medicine, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (A.M.R.R.); (H.H.V.)
| | - John Greenman
- Biomedical Section, University of Hull, Cottingham Road, Hull HU6 7RX, UK; (S.F.); (J.G.)
| | - Henri H. Versteeg
- Einthoven Laboratory for Vascular and Regenerative Medicine, Division of Thrombosis and Hemostasis, Department of Internal Medicine, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (A.M.R.R.); (H.H.V.)
| | - Anthony Maraveyas
- Division of Cancer-Hull York Medical School, University of Hull, Cottingham Road, Hull HU6 7RX, UK;
| |
Collapse
|
19
|
Novel mechanism regulating tissue factor activity. Blood 2021; 138:289-291. [PMID: 34323941 PMCID: PMC8320428 DOI: 10.1182/blood.2021012459] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 05/25/2021] [Indexed: 11/30/2022] Open
|
20
|
Chen S, Lv K, Sharda A, Deng J, Zeng W, Zhang C, Hu Q, Jin P, Yao G, Xu X, Ming Z, Fang C. Anti-thrombotic effects mediated by dihydromyricetin involve both platelet inhibition and endothelial protection. Pharmacol Res 2021; 167:105540. [PMID: 33711433 DOI: 10.1016/j.phrs.2021.105540] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 03/05/2021] [Accepted: 03/06/2021] [Indexed: 01/04/2023]
Abstract
Classical antithrombotics and antiplatelets are associated with high frequencies of bleeding complications or treatment failure when used as single agents. The platelet-independent fibrin generation by activated endothelium highlights the importance of vascular protection in addition to platelet inhibition in thrombosis prevention. Dihydromyricetin (DHM), the most abundant flavonoid in Ampelopsis grossedentata, has unique vasoprotective effects. This study aims to characterize the antithrombotic potential of DHM. The effects of DHM on the activation of platelets and endothelial cells were evaluated in vitro. Calcium mobilization and activation of mitogen-activated protein kinases (MAPKs) were examined as the potential targets of DHM based on molecular docking analysis. The in vivo effects of DHM were determined in FeCl3-injured carotid arteries and laser-injured cremasteric arterioles. The results showed that DHM suppressed a range of platelet responses including aggregation, secretion, adhesion, spreading and integrin activation, and inhibited exocytosis, phosphatidylserine exposure and tissue factor expression in activated endothelial cells. Mechanistically, DHM attenuated thrombin-induced calcium mobilization and phosphorylation of ERK1/2 and p38 both in platelets and endothelial cells. Intravenous treatment with DHM delayed FeCl3-induced carotid arterial thrombosis. Furthermore, DHM treatment inhibited both platelet accumulation and fibrin generation in the presence or absence of eptifibatide in the laser injury-induced thrombosis model, without prolonging ex vivo plasma coagulation or tail bleeding time. DHM represents a novel antithrombotic agent whose effects involve both inhibition of platelet activation and reduction of fibrin generation as a result of endothelial protection.
Collapse
Affiliation(s)
- Shuai Chen
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Department of Pharmacology, School of Basic Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou 550025, China
| | - Keyu Lv
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Anish Sharda
- Division of Hemostasis and Thrombosis, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Jun Deng
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Wanjiang Zeng
- Department of Perinatal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Chao Zhang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei 430030, China
| | - Qinghua Hu
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology and Key Laboratory for Respiratory Diseases, Health Ministry of China, Wuhan, Hubei 430030, China
| | - Pengfei Jin
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Guangmin Yao
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Xulin Xu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Zhangyin Ming
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Chao Fang
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Hubei 430030, China.
| |
Collapse
|
21
|
Wilkinson H, Leonard H, Chen D, Lawrence T, Robson M, Goossens P, McVey JH, Dorling A. PAR-1 signaling on macrophages is required for effective in vivo delayed-type hypersensitivity responses. iScience 2021; 24:101981. [PMID: 33458623 PMCID: PMC7797913 DOI: 10.1016/j.isci.2020.101981] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 11/13/2020] [Accepted: 12/17/2020] [Indexed: 12/15/2022] Open
Abstract
Delayed-type hypersensitivity (DTH) responses underpin chronic inflammation. Using a model of oxazolone-induced dermatitis and a combination of transgenic mice, adoptive cell transfer, and selective agonists/antagonists against protease activated receptors, we show that that PAR-1 signaling on macrophages by thrombin is required for effective granuloma formation. Using BM-derived macrophages (BMMs) in vitro, we show that thrombin signaling induced (a) downregulation of cell membrane reverse cholesterol transporter ABCA1 and (b) increased expression of IFNγ receptor and enhanced co-localization within increased areas of cholesterol-rich membrane microdomains. These two key phenotypic changes combined to make thrombin-primed BMMs sensitive to M1 polarization by 1000-fold less IFNγ, compared to resting BMMs. We confirm that changes in ABCA1 expression were directly responsible for the exquisite sensitivity to IFNγ in vitro and for the impact on granuloma formation in vivo. These data indicate that PAR-1 signaling plays a hitherto unrecognized and critical role in DTH responses.
Collapse
Affiliation(s)
- Hannah Wilkinson
- Department of Inflammation Biology, School of Immunology & Microbial Sciences, King's College London, Guy's Hospital, London SE1 9RT, UK
| | - Hugh Leonard
- Department of Inflammation Biology, School of Immunology & Microbial Sciences, King's College London, Guy's Hospital, London SE1 9RT, UK
| | - Daxin Chen
- Department of Inflammation Biology, School of Immunology & Microbial Sciences, King's College London, Guy's Hospital, London SE1 9RT, UK
| | - Toby Lawrence
- Centre for Inflammation Biology and Cancer Immunology, School of Immunology & Microbial Sciences, King's College London, London SE1 9RT, UK
| | - Michael Robson
- Department of Inflammation Biology, School of Immunology & Microbial Sciences, King's College London, Guy's Hospital, London SE1 9RT, UK
| | - Pieter Goossens
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6229HX Maastricht, the Netherlands
| | - John H McVey
- School of Bioscience & Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7XH, UK
| | - Anthony Dorling
- Department of Inflammation Biology, School of Immunology & Microbial Sciences, King's College London, Guy's Hospital, London SE1 9RT, UK
| |
Collapse
|
22
|
Bleeding Disorders in Bothrops atrox Envenomations in the Brazilian Amazon: Participation of Hemostatic Factors and the Impact of Tissue Factor. Toxins (Basel) 2020; 12:toxins12090554. [PMID: 32872404 PMCID: PMC7551922 DOI: 10.3390/toxins12090554] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 08/13/2020] [Accepted: 08/13/2020] [Indexed: 02/06/2023] Open
Abstract
Bleeding is a common hemostatic disorder that occurs in Bothrops envenomations. We evaluated the changes in coagulation, fibrinolysis components, and platelets in Bothrops atrox envenomations with bleeding. This is an observational study with B. atrox snakebite patients (n = 100) treated in Manaus, Brazilian Amazon. Bleeding was recorded on admission and during hospitalization. We found that the platelet count in our patients presented a weak correlation to tissue factor, factor II, and plasminogen. Tissue factor presented weak correlation to factor V, II, D-dimer, plasminogen, alpha 2-antiplasmin, and moderate correlation to fibrinogen and fibrin/fibrinogen degradation product (FDP). Patients with systemic bleeding (n = 20) presented low levels of factor V, II, fibrinogen, plasminogen, and alpha 2-antiplasmin, and high levels of tissue factor and FDP compared to those without bleeding. Patients with only local bleeding (n = 41) and without bleeding showed similar levels of hemostatic factors. Thrombocytopenia was observed mainly in patients with systemic bleeding and increased levels of serum venom. No association was found between venom levels and systemic bleeding, or between venom levels and clinical severity of envenomation. This is the first report that shows the participation of the extrinsic coagulation pathway in the consumption coagulopathy of B. atrox envenomations with systemic bleeding due to tissue factor release.
Collapse
|
23
|
Mazetto BDM, Lazarini M, Tobaldini LQ, Arantes FT, Dos Santos APR, Jacinto BC, Vaz CDO, Mesquita GTV, Saraiva SDS, Annichino-Bizzacchi J, Orsi FA. Expression of tissue factor mRNA in thrombosis associated with antiphospholipid syndrome. J Thromb Thrombolysis 2020; 51:370-378. [PMID: 32627125 DOI: 10.1007/s11239-020-02209-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Tissue factor (TF) is a procoagulant protein associated with increased risk of thrombotic events in antiphospholipid syndrome (t-APS). The mechanisms by which TF levels are increased in APS have not yet been established. The aim of this study was to investigate whether TF mRNA expression is associated with TF levels and thrombosis in APS. We compared levels of circulating TF and high sensitivity C-reactive protein (hs-CRP) between t-APS and controls (individuals without thrombosis). The association between TF mRNA expression, quantified by real time quantitative polymerase chain reaction, and t-APS was accessed using regression analysis. We included 41 controls and 42 t-APS patients, mean age was 41 years old (SD 14) in both groups. Hs-CRP and TF levels were higher in t-APS patients (mean hs-CRP levels 0.81 mg/dL [SD 1.88] and median TF levels 249.0 pg/mL [IQR 138.77-447.61]) as compared to controls (mean hs-CRP levels 0.24 mg/dL [SD 0.26] and median TF levels 113.0 pg/mL [IQR 81.17-161.53]; P = 0.02 and P < 0.0001, respectively). There was no correlation between TF mRNA expression and TF levels in t-APS (r - 0.209, P = 0.19). TF mRNA expression was not associated with t-APS (adjusted OR 1.16; 95%CI 0.72-1.87). Despite circulating TF levels being higher in patients with t-APS than in controls, TF mRNA expression was similar between groups. The results demonstrate that TF mRNA expression is not associated with levels of circulating TF and hypercoagulability in t-APS.
Collapse
Affiliation(s)
| | - Mariana Lazarini
- Hematology and Hemotherapy Center, University of Campinas, Campinas, Brazil
| | - Lais Quinteiro Tobaldini
- Faculty of Medical Sciences, University of Campinas, Campinas, Brazil.,Hematology and Hemotherapy Center, University of Campinas, Campinas, Brazil
| | - Fernanda Talge Arantes
- Faculty of Medical Sciences, University of Campinas, Campinas, Brazil.,Hematology and Hemotherapy Center, University of Campinas, Campinas, Brazil
| | - Ana Paula Rosa Dos Santos
- Faculty of Medical Sciences, University of Campinas, Campinas, Brazil.,Hematology and Hemotherapy Center, University of Campinas, Campinas, Brazil
| | - Bruna Cardoso Jacinto
- Faculty of Medical Sciences, University of Campinas, Campinas, Brazil.,Hematology and Hemotherapy Center, University of Campinas, Campinas, Brazil
| | - Camila de Oliveira Vaz
- Faculty of Medical Sciences, University of Campinas, Campinas, Brazil.,Hematology and Hemotherapy Center, University of Campinas, Campinas, Brazil
| | | | - Sabrina da Silva Saraiva
- Faculty of Medical Sciences, University of Campinas, Campinas, Brazil.,Hematology and Hemotherapy Center, University of Campinas, Campinas, Brazil
| | - Joyce Annichino-Bizzacchi
- Hematology and Hemotherapy Center, University of Campinas, Campinas, Brazil.,Department of Clinical Medicine, University of Campinas, Campinas, Brazil
| | - Fernanda Andrade Orsi
- Hematology and Hemotherapy Center, University of Campinas, Campinas, Brazil. .,Department of Clinical Pathology, Faculty of Medical Sciences, University of Campinas, Campinas R. Tessália Vieira de Camargo, 126, Cidade Universitária Zeferino Vaz, Campinas, 13083-887, Brazil.
| |
Collapse
|
24
|
Tripisciano C, Weiss R, Karuthedom George S, Fischer MB, Weber V. Extracellular Vesicles Derived From Platelets, Red Blood Cells, and Monocyte-Like Cells Differ Regarding Their Ability to Induce Factor XII-Dependent Thrombin Generation. Front Cell Dev Biol 2020; 8:298. [PMID: 32478066 PMCID: PMC7232549 DOI: 10.3389/fcell.2020.00298] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 04/06/2020] [Indexed: 12/18/2022] Open
Abstract
As transmitters of biological information, extracellular vesicles (EVs) are crucial for the maintenance of physiological homeostasis, but also contribute to pathological conditions, such as thrombotic disorders. The ability of EVs to support thrombin generation has been linked to their exposure of phosphatidylserine, an anionic phospholipid that is normally restricted to the inner leaflet of the plasma membrane but exposed on the outer leaflet during EV biogenesis. Here, we investigated whether EVs of different cellular origin and from different settings, namely platelets and red blood cells from blood bank units and a monocyte-like cell line (THP-1), differ regarding their potential to support factor XII-dependent thrombin generation. EVs were isolated from blood products or THP-1 cell culture supernatants using differential centrifugation and characterized by a combination of flow cytometry, nanoparticle tracking analysis, and Western blotting. Soluble factors co-enriched during the isolation of EVs were depleted from blood-cell derived EV fractions using size exclusion chromatography, while proteins bound to the surface of EVs were degraded by mild protease treatment. We found that platelet-derived and red blood cell-derived EVs supported factor XII-dependent thrombin generation to comparable extents, while monocytic EVs failed to support thrombin generation when added to EV-depleted human plasma. We excluded a major contribution of co-enriched soluble proteins or of proteins bound to the EV surface to the thrombogenicity of blood cell-derived EVs. Our data suggest that the enhanced potential of blood cell-derived EVs to support thrombin generation is rather due to enhanced exposure of phosphatidylserine on the surface of blood cell-derived EVs. Extending these investigations to EVs from other cell types, such as mesenchymal stromal cells, will be crucial for their future therapeutic applications.
Collapse
Affiliation(s)
- Carla Tripisciano
- Christian Doppler Laboratory for Innovative Therapy Approaches in Sepsis, Department for Biomedical Research, Danube University Krems, Krems, Austria
| | - René Weiss
- Christian Doppler Laboratory for Innovative Therapy Approaches in Sepsis, Department for Biomedical Research, Danube University Krems, Krems, Austria
| | - Sobha Karuthedom George
- Center for Biomedical Technology, Department for Biomedical Research, Danube University Krems, Krems, Austria
| | - Michael B Fischer
- Center for Experimental Medicine, Department for Biomedical Research, Danube University Krems, Krems, Austria
| | - Viktoria Weber
- Christian Doppler Laboratory for Innovative Therapy Approaches in Sepsis, Department for Biomedical Research, Danube University Krems, Krems, Austria.,Center for Biomedical Technology, Department for Biomedical Research, Danube University Krems, Krems, Austria.,Center for Experimental Medicine, Department for Biomedical Research, Danube University Krems, Krems, Austria
| |
Collapse
|
25
|
Zhou P, Li T, Jin J, Liu Y, Li B, Sun Q, Tian J, Zhao H, Liu Z, Ma S, Zhang S, Novakovic VA, Shi J, Hu S. Interactions between neutrophil extracellular traps and activated platelets enhance procoagulant activity in acute stroke patients with ICA occlusion. EBioMedicine 2020; 53:102671. [PMID: 32114386 PMCID: PMC7047181 DOI: 10.1016/j.ebiom.2020.102671] [Citation(s) in RCA: 88] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Revised: 01/14/2020] [Accepted: 01/27/2020] [Indexed: 02/06/2023] Open
Abstract
Background The role of neutrophil extracellular traps (NETs) in procoagulant activity (PCA) in stroke patients caused by thromboembolic occlusion of the internal carotid artery (ICA) remains unclear. Our objectives were to evaluate the critical role of NETs in the induction of hypercoagulability in stroke and to identify the functional significance of NETs during atherothrombosis. Methods The levels of NETs, activated platelets (PLTs), and PLT-derived microparticles (PMPs) were detected in the plasma of 55 stroke patients and 35 healthy controls. NET formation and thrombi were analysed using immunofluorescence. Exposed phosphatidylserine (PS) was evaluated with flow cytometry and confocal microscopy. PCA was analysed using purified coagulation complex, thrombin, and fibrin formation assays. Findings The plasma levels of NETs, activated PLTs, and PMP markers in the carotid lesion site (CLS) were significantly higher than those in the aortic blood. NETs were decorated with PS in thrombi and the CLS plasma of ICA occlusion patients. Notably, the complementary roles of CLS plasma and thrombin-activated PLTs were required for NET formation and subsequent PS exposure. PS-bearing NETs provided functional platforms for PMPs and coagulation factor deposition and thus increased thrombin and fibrin formation. DNase I and lactadherin markedly inhibited these effects. In addition, NETs were cytotoxic to endothelial cells, converting these cells to a procoagulant phenotype. Sivelestat, anti-MMP9 antibody, and activated protein C (APC) blocked this cytotoxicity by 25%, 39%, or 52%, respectively. Interpretation NETs played a pivotal role in the hypercoagulability of stroke patients. Strategies that prevent NET formation may offer a potential therapeutic strategy for thromboembolism interventions. Funding This study was supported by grants from the National Natural Science Foundation of China (61575058, 81873433 and 81670128) and Graduate Innovation Fund of Harbin Medical University (YJSKYCX2018-58HYD).
Collapse
Affiliation(s)
- Peng Zhou
- Department of Neurosurgery, The Second Affiliated Hospital, Harbin Medical University, Harbin, PR China; The Key Laboratory of Myocardial Ischemia, Ministry of Education, Heilongjiang Province, Harbin, PR China
| | - Tao Li
- Department of Hematology, The First Affiliated Hospital, Harbin Medical University, Harbin, PR China
| | - Jiaqi Jin
- Department of Neurosurgery, The Second Affiliated Hospital, Harbin Medical University, Harbin, PR China
| | - Yingmiao Liu
- Department of Stomatology, The First Affiliated Hospital, Harbin Medical University, Harbin, PR China
| | - Baorong Li
- Department of Stomatology, The First Affiliated Hospital, Harbin Medical University, Harbin, PR China
| | - Quanye Sun
- Department of Clinical Laboratory, Qingdao Municipal Hospital Group, Qingdao, PR China
| | - Jiawei Tian
- Department of Neurosurgery, The Second Affiliated Hospital, Harbin Medical University, Harbin, PR China
| | - Hongtao Zhao
- Department of Neurosurgery, The Second Affiliated Hospital, Harbin Medical University, Harbin, PR China
| | - Zhihui Liu
- Department of Neurosurgery, The Second Affiliated Hospital, Harbin Medical University, Harbin, PR China
| | - Shuai Ma
- Department of Neurosurgery, The Second Affiliated Hospital, Harbin Medical University, Harbin, PR China
| | - Shuoqi Zhang
- Department of Neurology, The Second Affiliated Hospital, Harbin Medical University, Harbin, PR China
| | - Valerie A Novakovic
- Department of Research, Brigham and Women's Hospital, VA Boston Healthcare System, Harvard Medical School, Boston, MA, USA
| | - Jialan Shi
- Department of Hematology, The First Affiliated Hospital, Harbin Medical University, Harbin, PR China; Department of Surgery, Brigham and Women's Hospital, VA Boston Healthcare System, Harvard Medical School, Boston, MA, USA.
| | - Shaoshan Hu
- Department of Neurosurgery, The Second Affiliated Hospital, Harbin Medical University, Harbin, PR China.
| |
Collapse
|
26
|
Diaz-Cañestro C, Bonetti NR, Wüst P, Nageswaran V, Liberale L, Beer JH, Montecucco F, Lüscher TF, Bohacek J, Camici GG. Apold1 deficiency associates with increased arterial thrombosis in vivo. Eur J Clin Invest 2020; 50:e13191. [PMID: 31797367 DOI: 10.1111/eci.13191] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 11/20/2019] [Accepted: 12/02/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND Endothelial cells regulate the formation of blood clots; thus, genes selectively expressed in these cells could primarily determine thrombus formation. Apold1 (apolipoprotein L domain containing 1) is a gene expressed by endothelial cells; whether Apold1 directly contributes to arterial thrombosis has not yet been investigated. Here, we assessed the effect of Apold1 deletion on arterial thrombus formation using an in vivo model of carotid thrombosis induced by photochemical injury. MATERIAL AND METHODS Apold1 knockout (Apold1-/- ) mice and wild-type (WT) littermates underwent carotid thrombosis induced by photochemical injury, and time to occlusion was recorded. Tissue factor (TF) activity and activation of mitogen-activated protein kinases (MAPKs) and phosphatidyl-inositol-3 kinase (PI3K)/Akt pathways were analysed by colorimetric assay and Western blotting in both Apold1-/- and WT mice. Finally, platelet reactivity was assessed using light transmission aggregometry. RESULTS After photochemical injury, Apold1-/- mice exhibited shorter time to occlusion as compared to WT mice. Moreover, TF activity was increased in carotid arteries of Apold1-/- when compared to WT mice. Underlying mechanistic markers such as TF mRNA and MAPKs activation were unaffected in Apold1-/- mice. In contrast, phosphorylation of Akt was reduced in Apold1-/- as compared to WT mice. Additionally, Apold1-/- mice displayed increased platelet reactivity to stimulation with collagen compared with WT animals. CONCLUSIONS Deficiency of Apold1 results in a prothrombotic phenotype, accompanied by increased vascular TF activity, decreased PI3K/Akt activation and increased platelet reactivity. These findings suggest Apold1 as an interesting new therapeutic target in the context of arterial thrombosis.
Collapse
Affiliation(s)
| | - Nicole R Bonetti
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | - Patricia Wüst
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | - Vanasa Nageswaran
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | - Luca Liberale
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland.,First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - Jürg H Beer
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland.,Department of Internal Medicine, Cantonal Hospital Baden, Baden, Switzerland
| | - Fabrizio Montecucco
- IRCCS Ospedale Policlinico San Martino Genoa - Italian Cardiovascular Network, Genoa, Italy.,Department of Internal Medicine, First Clinic of Internal Medicine, Centre of Excellence for Biomedical Research (CEBR), University of Genoa, Genoa, Italy
| | - Thomas F Lüscher
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | - Johannes Bohacek
- Lab of Molecular and Behavioral Neuroscience, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland.,Neuroscience Center Zurich, ETH Zurich and University of Zurich, Zurich, Switzerland
| | - Giovanni G Camici
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland.,Department of Cardiology, University Heart Center, University Hospital Zurich, Zurich, Switzerland.,Department of Research and Education, University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
27
|
Di Pillo E, Carrasco K, Brustolin B, Boufenzer A, Jolly L, Derive M, Lacolley P, Regnault V, Gibot S. Inhibition of triggering receptor expressed on myeloid cells-1 impairs thrombin generation. J Thromb Haemost 2020; 18:454-462. [PMID: 31680426 DOI: 10.1111/jth.14677] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Accepted: 10/30/2019] [Indexed: 01/24/2023]
Abstract
BACKGROUND New evidence has shown the link between inflammation and thrombosis. Triggering receptor expressed on myeloid cells-1 (TREM-1) is an immunoreceptor expressed mostly on neutrophils and monocytes/macrophages. TREM-1 acts as an amplifier of the inflammatory response, and its pharmacological inhibition displays protective effects in various models of inflammatory disorders, in particular by dampening coagulation abnormalities and thrombocytopenia observed during acute inflammation. OBJECTIVES We aimed to decipher the role of TREM-1 in fostering thrombin generation. METHODS We measured thrombin generation (TG) by the use of calibrated automated thrombography with whole blood, and isolated primary human neutrophils and monocytes upon stimulation with lipopolysaccharide (LPS). Tissue factor (TF) expression was measured by flow cytometry and its activity by ELISA. Phosphatidylserine (PtdSer) exposure was determined by flow cytometry. A dodecapeptide (LR12) was used as a specific inhibitor of TREM-1. RESULTS LPS increased TG, TF expression, and activity, as well as the exposure of PtdSer on the surface of monocytes. LR12 dampened TF activity through the decrease of PtdSer exposure, leading to a reduction of thrombin generation. CONCLUSIONS TREM-1 inhibition decreases thrombin generation and could be an interesting target for the development of new inhibitors of leukocyte-associated thrombotic activity.
Collapse
Affiliation(s)
- Elisa Di Pillo
- Inserm UMR_S1116, Faculté de Médecine de Nancy, Université de Lorraine, Vandœuvre-lès-Nancy, France
| | | | - Benjamin Brustolin
- Inserm UMR_S1116, Faculté de Médecine de Nancy, Université de Lorraine, Vandœuvre-lès-Nancy, France
| | | | | | | | - Patrick Lacolley
- Inserm UMR_S1116, Faculté de Médecine de Nancy, Université de Lorraine, Vandœuvre-lès-Nancy, France
| | - Véronique Regnault
- Inserm UMR_S1116, Faculté de Médecine de Nancy, Université de Lorraine, Vandœuvre-lès-Nancy, France
| | - Sébastien Gibot
- Inserm UMR_S1116, Faculté de Médecine de Nancy, Université de Lorraine, Vandœuvre-lès-Nancy, France
- Service de Médecine Intensive Réanimation, Hôpital Central, Nancy, France
| |
Collapse
|
28
|
Phosphatidylserine-exposing blood cells, microparticles and neutrophil extracellular traps increase procoagulant activity in patients with pancreatic cancer. Thromb Res 2020; 188:5-16. [PMID: 32032826 DOI: 10.1016/j.thromres.2020.01.025] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 01/23/2020] [Accepted: 01/27/2020] [Indexed: 01/05/2023]
Abstract
Patients with pancreatic cancer (PC) are at increased risk of venous thrombosis, but the precise mechanisms of hypercoagulable state in PC remain unclear. We aimed to identify how phosphatidylserine positive (PS+) blood cells (BCs), PS+ microparticles (MPs) and neutrophil extracellular traps (NETs) regulate procoagulant activity (PCA) in PC, and to assess the relationship between PCA and PC staging. A total of 83 PC patients with different stages of disease were compared to 30 healthy controls, with confocal microscopy and flow cytometry used to assess MP and cellular PS exposure. MP and cell PCA was determined using both fibrin production assays and procoagulant enzyme complex analyses, and coagulation time was further measured. Patients with stage I PC and healthy controls exhibited significantly lower frequencies of PS+ MPs and BCs relative to those with more advanced disease, which may partly due to the increased levels of inflammation cytokines in advanced disease. Functional coagulation assays indicated that PS+ MPs and BCs derived from patients with stage II/III/IV PC directly contribute to elevated FXa, thrombin, and fibrin formation, and to more rapid coagulation relative to healthy control samples. In inhibition assays, lactadherin, which antagonizes PS, led to a roughly 80% inhibition of PCA. We further used isolated NETs to stimulate endothelial cells, revealing that this led to morphological changes including retraction from cell-cell junctions and a more pro-coagulative phenotype, with DNase I and activated protein C treatment reversing these changes. In patients with stage III PC, curative resection surgery significantly reduced PCA, whereas non-curative surgery did not have a marked impact based on studies of pre- and post-operative samples. These results highlight the pathogenic activity of PS+ cells, MPs, and NETs in promoting a prothrombotic environment within individuals suffering from advanced PC. Targeting PS and NETs in these patients may thus be a viable means of preventing pathological thrombosis.
Collapse
|
29
|
Yu M, Xie R, Zhang Y, Liang H, Hou L, Yu C, Zhang J, Dong Z, Tian Y, Bi Y, Kou J, Novakovic VA, Shi J. Phosphatidylserine on microparticles and associated cells contributes to the hypercoagulable state in diabetic kidney disease. Nephrol Dial Transplant 2019. [PMID: 29529237 DOI: 10.1093/ndt/gfy027] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Background Relatively little is known about the role of phosphatidylserine (PS) in procoagulant activity (PCA) in patients with diabetic kidney disease (DKD). This study was designed to evaluate whether exposed PS on microparticles (MPs) and MP-origin cells were involved in the hypercoagulability in DKD patients. Methods DKD patients (n = 90) were divided into three groups based on urinary albumin excretion rate, defined as normoalbuminuria (No-A) (<30 mg/24 h), microalbuminuria (Mi-A) (30-299 mg/24 h) or macroalbuminuria (Ma-A) (>300 mg/24 h), and compared with healthy controls (n = 30). Lactadherin was used to quantify PS exposure on MPs and their original cells. Healthy blood cells (BCs) and human umbilical vein endothelial cells (HUVECs) were treated with 25, 5 or 2.5 mmol/L glucose as well as 3-12 mg/dL uric acid and cells were evaluated by clotting time and purified coagulation complex assays. Fibrin production was determined by turbidity. PS exposure and fibrin strands were observed using confocal microscopy. Results Using flow cytometry, we found that PS+ MPs (derived from platelets, erythrocytes, HUVECs, neutrophils, monocytes and lymphocytes) and BCs were significantly higher in patients than in controls. Furthermore, the number of PS+ MPs and BCs in patients with Ma-A was significantly higher than in patients with No-A. Similarly, we observed markedly elevated PS exposure on HUVECs cultured with serum from patients with Ma-A versus serum from patients with Mi-A or normoalbuminuria. In addition, circulating PS+ MPs cooperated with PS+ cells, contributing to markedly shortened coagulation time and dramatically increased FXa/thrombin generation and fibrin formation in each DKD group. Confocal microscopy images demonstrated colocalization of fibrin with PS on HUVECs. Moreover, blockade of exposed PS on MPs and cells with lactadherin inhibited PCA by ∼80%. In vitro, BCs and endothelial cells exposed more PS in hypoglycemia or hyperglycemia. Interestingly, reconstitution experiments showed that hypoglycemia-treated cells could be further activated or injured when recovery is obtained reaching hyperglycemia. Moreover, uric acid induced PS exposure on cells (excluding platelets) at concentrations >6 mg/dL. Linear regression analysis showed that levels of PS+ BCs and microparticles were positively correlated with uric acid and proteinuria, but negatively correlated with glomerular filtration rate. Conclusions Our results suggest that PS+ MPs and MP-origin cells play procoagulant roles in patients with DKD. Blockade of PS could become a novel therapeutic modality for the prevention of thrombosis in these patients.
Collapse
Affiliation(s)
- Muxin Yu
- Department of Nephrology, the First Hospital, Harbin, China
| | - Rujuan Xie
- Department of Nephrology, the First Hospital, Harbin, China
| | - Yan Zhang
- Department of Hematology, the First Hospital, Harbin, China
| | - Hui Liang
- Department of Nephrology, the First Hospital, Harbin, China
| | - Li Hou
- Department of Nephrology, the First Hospital, Harbin, China
| | - Chengyuan Yu
- Department of Nephrology, the First Hospital, Harbin, China
| | - Jinming Zhang
- Department of Gastroenterology, the Fourth Hospital, Harbin, China
| | - Zengxiang Dong
- Department of Cardiology, the First Hospital, Harbin, China
| | - Ye Tian
- Department of Cardiology, the First Hospital, Harbin, China
| | - Yayan Bi
- Department of Cardiology, the First Hospital, Harbin, China
| | - Junjie Kou
- Department of Cardiology, the Second Hospital, Harbin Medical University, Harbin, China
| | - Valerie A Novakovic
- Department of Research, VA Boston Healthcare System, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Jialan Shi
- Department of Hematology, the First Hospital, Harbin, China.,Department of Research, VA Boston Healthcare System, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.,Department of Surgery, VA Boston Healthcare System, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
30
|
Ivanov II, Apta BHR, Bonna AM, Harper MT. Platelet P-selectin triggers rapid surface exposure of tissue factor in monocytes. Sci Rep 2019; 9:13397. [PMID: 31527604 PMCID: PMC6746844 DOI: 10.1038/s41598-019-49635-7] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 08/23/2019] [Indexed: 12/13/2022] Open
Abstract
Tissue factor (TF) plays a central role in haemostasis and thrombosis. Following vascular damage, vessel wall TF initiates the extrinsic coagulation cascade. TF can also be exposed by monocytes. Inflammatory or infectious stimuli trigger synthesis of new TF protein by monocytes over the course of hours. It has also been suggested that monocytes can expose TF within minutes when stimulated by activated platelets. Here, we have confirmed that monocytes rapidly expose TF in whole blood and further demonstrate that platelet P-selectin exposure is necessary and sufficient. Monocyte TF exposure increased within five minutes in response to platelet activation by PAR1-AP, PAR4-AP or CRP-XL. PAR1-AP did not trigger TF exposure on isolated monocytes unless platelets were also present. In whole blood, PAR1-AP-triggered TF exposure required P-selectin and PGSL-1. In isolated monocytes, although soluble recombinant P-selectin had no effect, P-selectin coupled to 2 µm beads triggered TF exposure. Cycloheximide did not affect rapid TF exposure, indicating that de novo protein synthesis was not required. These data show that P-selectin on activated platelets rapidly triggers TF exposure on monocytes. This may represent a mechanism by which platelets and monocytes rapidly contribute to intravascular coagulation.
Collapse
Affiliation(s)
- Ivelin I Ivanov
- Department of Pharmacology, University of Cambridge, Cambridge, United Kingdom
| | - Bonita H R Apta
- Department of Pharmacology, University of Cambridge, Cambridge, United Kingdom
| | - Arkadiusz M Bonna
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| | - Matthew T Harper
- Department of Pharmacology, University of Cambridge, Cambridge, United Kingdom.
| |
Collapse
|
31
|
Scatena C, Franceschi S, Franzini M, Sanguinetti C, Romiti N, Caponi L, Mandalà M, Mazzanti CM, Naccarato AG. Dabrafenib and Trametinib prolong coagulation through the inhibition of tissue factor in BRAF v600e mutated melanoma cells in vitro. Cancer Cell Int 2019; 19:223. [PMID: 31467489 PMCID: PMC6712666 DOI: 10.1186/s12935-019-0938-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Accepted: 08/18/2019] [Indexed: 01/01/2023] Open
Abstract
Background Neoplastic cells promote a hypercoagulable state by the expression of cell surface proteins, such as tissue factor. In BRAFv600 mutated melanoma patients upon BRAF inhibitors, a hypercoagulable state correlates with prognosis, while a down-regulation of the hemostatic parameters is observed in patients responders as compared to non responders. The present study was intended to better clarify the strict relationship between coagulation mediators and target therapy in melanoma. Methods The expression of tissue factor was investigated after the treatment with the BRAF inhibitor Dabrafenib and the MEK inhibitor Trametinib in the BRAFv600e mutated melanoma cell lines A-375 and SK-MEL-28, together with its ability to activate the coagulation cascade. Results Dabrafenib and Trametinib caused the down-regulation of TF in both cell lines A-375 and SK-MEL-28. For the cell line A-375 the effect was evident both at RNA and procoagulant activity; for the cell line SK-MEL-28 only at RNA level without any variation of the protein. Interestingly, when in contact with plasma deficient of factor VII, both cell lines were not able to activate the coagulation cascade. Conclusions The present study provides the first in vitro observation that tissue factor expressed in melanoma cells may contribute to the modulation of the coagulation state of patients in the treatment with BRAF inhibitors.
Collapse
Affiliation(s)
- Cristian Scatena
- 1Division of Surgical Pathology, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma 57, 56126 Pisa, Italy
| | | | - Maria Franzini
- 3Division of Clinical Pathology, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Chiara Sanguinetti
- 3Division of Clinical Pathology, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Nadia Romiti
- 3Division of Clinical Pathology, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Laura Caponi
- 3Division of Clinical Pathology, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Mario Mandalà
- Unit of Medical Oncology, Department of Oncology and Haematology, Papa Giovanni XXIII Cancer Center Hospital, Bergamo, Italy
| | | | - Antonio Giuseppe Naccarato
- 1Division of Surgical Pathology, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma 57, 56126 Pisa, Italy
| |
Collapse
|
32
|
Liu Y, Li B, Hu TL, Li T, Zhang Y, Zhang C, Yu M, Wang C, Hou L, Dong Z, Hu TS, Novakovic VA, Shi J. Increased Phosphatidylserine on Blood Cells in Oral Squamous Cell Carcinoma. J Dent Res 2019; 98:763-771. [PMID: 30979345 DOI: 10.1177/0022034519843106] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The specific function of phosphatidylserine (PS) in the context of the development of a hypercoagulable state among individuals with oral squamous cell carcinoma (OSCC) is uncertain. The goal of this study was therefore to assess the exposure of PS on microparticles (MPs) as well as on endothelial and blood cells and to assess procoagulant activity (PCA) as a function of the stage of OSCC progression. We recruited patients with OSCC (n = 63) as well as healthy controls (n = 26) to participate in this study. PS exposure was then assessed via confocal microscopy and flow cytometry, revealing that patients with stage III/IV OSCC exhibited higher frequencies of PS-exposing blood cells, MPs, and serum-cultured endothelial cells (ECs) than did patients with stage I/II OSCC or healthy controls. When we conducted functional coagulation assays, we discovered that PS+ blood cells, MPs, and serum-cultured ECs from patients with stage III/IV OSCC mediated more rapid coagulation and more substantial production of FXa, thrombin, and fibrin as compared with controls. When samples were treated with the PS antagonist lactadherin, this resulted in an 80% disruption of PCA. Strikingly, when pre- and postoperative samples were compared from patients with stage III/IV OSCC undergoing resective surgery, PCA was significantly reduced in the postoperative samples. After stimulating ECs with inflammatory cytokines, we found by confocal microscopy that they expose PS on their cell membranes, thus generating FVa and FXa binding sites and mediating the formation of fibrin. Together our findings provide evidence that PS+ blood cells and MPs are important mediators of the development of a hypercoagulable and prothrombotic state among individuals afflicted by advanced-stage OSCC. As such, a PS blockade may be a viable therapeutic strategy for treating such patients.
Collapse
Affiliation(s)
- Y Liu
- 1 Department of Stomatology, First Hospital, Harbin Medical University, Harbin, China
| | - B Li
- 1 Department of Stomatology, First Hospital, Harbin Medical University, Harbin, China
| | - T L Hu
- 1 Department of Stomatology, First Hospital, Harbin Medical University, Harbin, China.,2 Department of Oral Anatomy and Physiology, Stomatology School, Harbin Medical University, Harbin, China
| | - T Li
- 3 Department of Hematology, First Hospital, Harbin Medical University, Harbin, China
| | - Y Zhang
- 3 Department of Hematology, First Hospital, Harbin Medical University, Harbin, China
| | - C Zhang
- 3 Department of Hematology, First Hospital, Harbin Medical University, Harbin, China
| | - M Yu
- 3 Department of Hematology, First Hospital, Harbin Medical University, Harbin, China
| | - C Wang
- 3 Department of Hematology, First Hospital, Harbin Medical University, Harbin, China
| | - L Hou
- 3 Department of Hematology, First Hospital, Harbin Medical University, Harbin, China
| | - Z Dong
- 4 Department of Cardiology, First Hospital, Harbin Medical University, Harbin, China
| | - T S Hu
- 3 Department of Hematology, First Hospital, Harbin Medical University, Harbin, China
| | - V A Novakovic
- 5 Departments of Research and Surgery, VA Boston Healthcare System, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - J Shi
- 3 Department of Hematology, First Hospital, Harbin Medical University, Harbin, China.,5 Departments of Research and Surgery, VA Boston Healthcare System, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
33
|
O'Donnell JS, O'Sullivan JM, Preston RJS. Advances in understanding the molecular mechanisms that maintain normal haemostasis. Br J Haematol 2019; 186:24-36. [DOI: 10.1111/bjh.15872] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- James S. O'Donnell
- Haemostasis Research Group Department of Molecular and Cellular Therapeutics Irish Centre for Vascular Biology Royal College of Surgeons in Ireland Dublin Ireland
| | - Jamie M. O'Sullivan
- Haemostasis Research Group Department of Molecular and Cellular Therapeutics Irish Centre for Vascular Biology Royal College of Surgeons in Ireland Dublin Ireland
| | - Roger J. S. Preston
- Haemostasis Research Group Department of Molecular and Cellular Therapeutics Irish Centre for Vascular Biology Royal College of Surgeons in Ireland Dublin Ireland
| |
Collapse
|
34
|
Yamaguchi R, Sakamoto A, Yamaguchi R, Haraguchi M, Narahara S, Sugiuchi H, Katoh T, Yamaguchi Y. Di-(2-Ethylhexyl) Phthalate Promotes Release of Tissue Factor-Bearing Microparticles From Macrophages via the TGFβ1/Smad/PAI-1 Signaling Pathway. Am J Med Sci 2019; 357:492-506. [PMID: 30910165 DOI: 10.1016/j.amjms.2019.02.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 01/26/2019] [Accepted: 02/07/2019] [Indexed: 01/03/2023]
Abstract
BACKGROUND Plasminogen activator inhibitor type 1 promotes formation of endothelial microparticles with procoagulant activity. However, it remains unclear whether di-(2-ethylhexyl) phthalate, a peroxisome proliferator-activated receptor α agonist, influences microparticle formation. MATERIALS AND METHODS The effect of di-(2-ethylhexyl) phthalate on release of tissue factor-bearing microparticles was investigated using human M1 macrophages. RESULTS Exposure of M1 macrophages to di-(2-ethylhexyl) phthalate significantly upregulated expression of plasminogen activator inhibitor type 1, whereas incubation of macrophages with small interfering RNA for peroxisome proliferator-activated receptor α attenuated it. Di-(2-ethylhexyl) phthalate significantly increased the tissue factor protein level in culture supernatants of M1 macrophages, but not M2 macrophages. After purification of proteins by centrifugal filtration, western blotting detected 2 high molecular weight bands of tissue factor-bearing microparticles in culture supernatants of M1 macrophages. The upper band showed binding to factor VIIa and tissue factor pathway inhibitor, unlike the lower band. This suggested heterogeneity of the procoagulant activity of tissue factor-bearing microparticles, presumably dependent upon encryption/decryption of tissue factor. Phosphatidylserine contributes to tissue factor decryption, and western blotting revealed that the density of phosphatidylserine was reduced in the upper tissue factor band compared with the lower band. Di-(2-ethylhexyl) phthalate also upregulated transforming growth factor-β1 protein production by M1 macrophages. Moreover, silencing of Smad2, Smad3 or Smad4 attenuated plasminogen activator inhibitor type 1 expression and tissue factor-release from macrophages after di-(2-ethylhexyl) phthalate stimulation. CONCLUSIONS Di-(2-ethylhexyl) phthalate promotes formation of tissue factor-bearing microparticles in human M1 macrophages via the transforming growth factor-β1/Smad/ plasminogen activator inhibitor type 1 signaling pathway.
Collapse
Affiliation(s)
- Rui Yamaguchi
- Department of Public Health, Faculty of Life Sciences, Kumamoto University School of Medicine, Kumamoto, Japan; Graduate School of Medical Science, Kumamoto Health Science University, Kumamoto, Japan
| | - Arisa Sakamoto
- Graduate School of Medical Science, Kumamoto Health Science University, Kumamoto, Japan
| | - Reona Yamaguchi
- Department of Neuroscience, Graduate School of Medicine and Faculty of Medicine, Kyoto University, Kyoto, Japan
| | - Misa Haraguchi
- Graduate School of Medical Science, Kumamoto Health Science University, Kumamoto, Japan
| | - Shinji Narahara
- Graduate School of Medical Science, Kumamoto Health Science University, Kumamoto, Japan
| | - Hiroyuki Sugiuchi
- Graduate School of Medical Science, Kumamoto Health Science University, Kumamoto, Japan
| | - Takahiko Katoh
- Department of Public Health, Faculty of Life Sciences, Kumamoto University School of Medicine, Kumamoto, Japan
| | - Yasuo Yamaguchi
- Graduate School of Medical Science, Kumamoto Health Science University, Kumamoto, Japan.
| |
Collapse
|
35
|
Ansari SA, Pendurthi UR, Rao LVM. Role of Cell Surface Lipids and Thiol-Disulphide Exchange Pathways in Regulating the Encryption and Decryption of Tissue Factor. Thromb Haemost 2019; 119:860-870. [PMID: 30861549 DOI: 10.1055/s-0039-1681102] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Tissue factor (TF), a transmembrane glycoprotein, is the cellular receptor of the coagulation factors VII (FVII) and VIIa (FVIIa). The formation of TF-FVIIa complex triggers the initiation of the blood coagulation pathway. TF plays an essential role in haemostasis, but an aberrant expression of TF activity contributes to thrombotic disorders. In health, TF pro-coagulant activity on cells is controlled tightly to allow sufficient coagulant activity to achieve haemostasis but not to cause thrombosis. It is achieved largely by selective localization of TF in the body and encryption of TF at the cell surface. A vast majority of TF on resting cells exists in an encrypted state with minimal pro-coagulant activity but becomes pro-thrombotic following cell injury or activation. At present, the mechanisms that are responsible for TF encryption and activation (decryption) are not entirely clear, but recent studies provide important mechanistic insights into these processes. To date, externalization of phosphatidylserine to the outer leaflet and thiol-disulphide exchange pathways that either turn on and off the allosteric disulphide bond in TF are shown to play a major role in regulating TF pro-coagulant activity on cell surfaces. Recent studies showed that sphingomyelin, a major phospholipid in the outer leaflet of plasma membrane, plays a critical role in the encryption of TF in resting cells. The present review provides an overview of recent literature on the above-described mechanisms of TF encryption and decryption with a particular emphasis on our recent findings.
Collapse
Affiliation(s)
- Shabbir A Ansari
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, Texas, United States
| | - Usha R Pendurthi
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, Texas, United States
| | - L Vijaya Mohan Rao
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, Texas, United States
| |
Collapse
|
36
|
Hemostasis based on a novel 'two-path unifying theory' and classification of hemostatic disorders. Blood Coagul Fibrinolysis 2019; 29:573-584. [PMID: 30063477 DOI: 10.1097/mbc.0000000000000765] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
: Hemostasis is the most important protective mechanism for human survival following harmful vascular damage caused by internal disease or external injury. Physiological mechanism of hemostasis is partially understood. Hemostasis can be initiated by either intravascular injury or external bodily injury involving two different levels of damage [i.e., limited to the endothelium or combined with extravascular tissue (EVT)]. In intravascular injury, traumatic damage limited to local endothelium typically is of no consequence, but disease-induced endothelial damage associated with systemic endothelial injury seen in sepsis and other critical illnesses could cause generalized 'endotheliopathy'. It triggers no bleeding but promotes serious endothelial molecular response. If intravascular local trauma extends beyond the endothelium and into EVT, it causes intravascular 'bleeding' and initiate 'clotting' via normal hemostasis. In external bodily injury, local traumatic damage always extends to the endothelium and EVT, and triggers 'bleeding' and 'clotting'. Systemic endotheliopathy activates only unusually large von Willebrand factor multimers (ULVWF) path and mediates 'microthrombogenesis', producing 'microthrombi' strings. This partial activation of hemostasis with ULVWF path leads to vascular microthrombotic disease. But localized traumatic injury extending to the endothelium and EVT activates both ULVWF and tissue factor paths. Combined activation of ULVWF and tissue factor paths provides normal hemostasis in external bodily injury, but causes 'macrothrombus' formation in intravascular injury. This 'two-path unifying theory' concept succinctly elucidates simplified nature of hemostasis in intravascular and external bodily injuries. It also clarifies different pathogenesis of every hemorrhagic disease and thrombotic disorder related to internal vascular disease and external vascular injury.
Collapse
|
37
|
Benelhaj NE, Maraveyas A, Featherby S, Collier MEW, Johnson MJ, Ettelaie C. Alteration in endothelial permeability occurs in response to the activation of PAR2 by factor Xa but not directly by the TF-factor VIIa complex. Thromb Res 2019; 175:13-20. [PMID: 30677622 DOI: 10.1016/j.thromres.2019.01.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 01/04/2019] [Accepted: 01/15/2019] [Indexed: 01/09/2023]
Abstract
Alterations in the endothelial permeability occur in response to the activation of coagulation mechanisms in order to control clot formation. The activation of the protease activated receptors (PAR) can induce signals that regulate such cellular responses. PAR2 is a target for the coagulation factor Xa (fXa) and tissue factor-factor VIIa (TF-fVIIa) complex. By measuring the permeability of dextran blue across endothelial monolayer, we examined the mechanisms linking coagulation and endothelial permeability. Activation of PAR2 using the agonist peptide (PAR2-AP) resulted in increased permeability across the monolayer and was comparable to that obtained with VEGF at 60 min. Incubation of cells with activated factor Xa (fXa) resulted in an initial decrease in permeability by 30 min, but then significantly increased at 60 min. These responses required fXa activity, and were abrogated by incubation of the cells with a PAR2-blocking antibody (SAM11). Activation of PAR2 alone, or inhibition of PAR1, abrogated the initial reduction in permeability. Additionally, inclusion of Rivaroxaban (0.6 μg/ml) significantly inhibited the response to fXa. Finally, incubation of the endothelial monolayers up to 2 h with TF-containing microvesicles derived from MDA-MB-231 cells, in the presence or absence of fVIIa, did not influence the permeability across the monolayers. In conclusion, fXa but not TF-fVIIa is a noteworthy mediator of endothelial permeability. The rapid initial decrease in permeability requires PAR2 and PAR1 which may act to constrain bleeding. The longer-term response is mediated by PAR2 with increased permeability, presumably to enhance clot formation at the site of damage.
Collapse
Affiliation(s)
- Naima E Benelhaj
- Division of Cancer-Hull York Medical School, University of Hull, Cottingham Road, Hull HU6 7RX, UK
| | - Anthony Maraveyas
- Division of Cancer-Hull York Medical School, University of Hull, Cottingham Road, Hull HU6 7RX, UK
| | - Sophie Featherby
- Biomedical Section, School of Life Sciences, University of Hull, Cottingham Road, Hull HU6 7RX, UK
| | - Mary E W Collier
- Department of Cardiovascular Sciences, University of Leicester, Glenfield General Hospital, Leicester LE3 9QP, UK
| | - Miriam J Johnson
- Division of Cancer-Hull York Medical School, University of Hull, Cottingham Road, Hull HU6 7RX, UK
| | - Camille Ettelaie
- Biomedical Section, School of Life Sciences, University of Hull, Cottingham Road, Hull HU6 7RX, UK.
| |
Collapse
|
38
|
Abstract
Biochemical and structural data reveal important aspects of the properties and function of a protein disulphide bond. Molecular dynamics simulations can complement this experimental data and can yield valuable insights into the dynamical behavior of the disulphide bond within the protein environment. Due to the increasing accuracy of the underlying energetic description and the increasing computational power at hand, such simulations have now reached a level, at which they can also make quantitative and experimentally testable predictions. We here give an overview of the computational methods used to predict functional aspects of protein disulphides, including the prestress, protein allosteric effects upon thiol/disulphide exchange, and disulphide redox potentials. We then outline in detail the use of free-energy perturbation methods to calculate the redox potential of a protein disulphide bond of interest. In a step-by-step protocol, we describe the workflow within the MD suite Gromacs, including practical advice on the simulation setup and choice of parameters. For other disulphide-related simulation methods, we refer to resources available online.
Collapse
Affiliation(s)
- Frauke Gräter
- Heidelberg Institute for Theoretical Studies, Heidelberg, Germany. .,Interdisciplinary Center for Scientific Computing, Heidelberg University, Heidelberg, Germany.
| | - Wenjin Li
- Institute for Advanced Study, Shenzhen University, Shenzhen, Guangdong, People's Republic of China
| |
Collapse
|
39
|
Phosphatidylserine-exposing blood cells and microparticles induce procoagulant activity in non-valvular atrial fibrillation. Int J Cardiol 2018; 258:138-143. [PMID: 29544920 DOI: 10.1016/j.ijcard.2018.01.116] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 01/21/2018] [Accepted: 01/26/2018] [Indexed: 11/23/2022]
Abstract
BACKGROUND The definitive role of phosphatidylserine (PS) in the prothrombotic state of non-valvular atrial fibrillation (NVAF) remains unclear. Our objectives were to study the PS exposure on blood cells and microparticles (MPs) in NVAF, and evaluate their procoagulant activity (PCA). METHODS NVAF patients without (n = 60) and with left atrial thrombi (n = 18) and controls (n = 36) were included in our study. Exposed PS was analyzed with flow cytometry and confocal microscopy. PCA was evaluated using clotting time, factor Xa (FXa), thrombin and fibrin formation. RESULTS PS+ blood cells and MPs were significantly higher in NVAF patients without and with left atrial thrombi (both P < 0.01) than in controls. Patients with left atrial thrombi showed increased PS+ platelets, neutrophils, erythrocytes and MPs compared with patients without thrombi (all P < 0.05). Moreover, in patients with left atrial thrombi, MPs primarily originated from platelets (56.1%) followed by leukocytes (21.9%, including MPs from neutrophils, monocytes and lymphocytes), erythrocytes (12.2%) and endothelial cells (8.9%). Additionally, PS+ blood cells and MPs contributed to markedly shortened coagulation time and dramatically increased FXa/thrombin/fibrin (all P < 0.001) generation in both NVAF groups. Furthermore, blockade of exposed PS on blood cells and MPs with lactadherin inhibited PCA by approximately 80%. Lastly, we found that the amount of PS+ platelets and MPs was positively correlated with thrombus diameter (all p < 0.005). CONCLUSIONS Our results suggest that exposed PS on blood cells and MPs play a procoagulant role in NVAF patients. Blockade of PS prior to thrombus formation might be a novel therapeutic approach in these patients.
Collapse
|
40
|
Abstract
: 'Disseminated intravascular coagulation (DIC)' occurs commonly in critical illnesses such as sepsis, trauma, cancer, and complications of surgery and pregnancy. Mortality is very high. The pathogenesis has been ascribed to tissue factor-initiated coagulation disorder, resulting in disseminated microblood clots that are made of platelets, plasma factors, fibrins, and blood cells. True DIC depletes coagulation factors and consumes platelets, and activates fibrinolysis. 'DIC' is assumed to orchestrate thrombocytopenia, microangiopathic hemolytic anemia and hypoxic multiorgan dysfunction syndrome, and causes hemorrhagic disorder due to depleted coagulation factors. In contrast, disseminated intravascular microthrombosis (DIT) occurs in thrombotic thrombocytopenic purpura (TTP) and TTP-like syndrome due to ADAMTS13 deficiency or insufficiency. The pathogenesis is due to formation of intravascular 'microthrombi' composed of complexes of platelets and unusually large von Willebrand factor multimers. Interestingly, DIT also occurs in the same critically ill patients as 'DIC' does. Following activation of complement system, the terminal complex C5b-9 causes endotheliopathy via channel formation to the endothelial cell membrane. Endotheliopathy activates microthrombotic pathway and initiates microthrombogenesis, leading to endotheliopathy-associated DIT. DIT results in TTP-like syndrome with hematologic phenotype of consumptive thrombocytopenia, microangiopathic hemolytic anemia, and multiorgan dysfunction syndrome. In reinterpretation of 'DIC', the true lesion is 'microthrombi' but not microblood clots. Thus, 'DIC' is endotheliopathy-associated DIT. This concept reconciles all the clinical features of 'DIC', and dramatically changes our understanding of pathophysiological mechanism in hemostasis and thrombosis. This new paradigm should assist the physician with correct diagnostic evaluation and treatment intervention.
Collapse
|
41
|
Chang JC. TTP-like syndrome: novel concept and molecular pathogenesis of endotheliopathy-associated vascular microthrombotic disease. Thromb J 2018; 16:20. [PMID: 30127669 PMCID: PMC6087012 DOI: 10.1186/s12959-018-0174-4] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 06/07/2018] [Indexed: 02/07/2023] Open
Abstract
TTP is characterized by microangiopathic hemolytic anemia and thrombocytopenia associated with brain and kidney dysfunction. It occurs due to ADAMTS13 deficiency. TTP-like syndrome occurs in critically ill patients with the similar hematologic changes and additional organ dysfunction syndromes. Vascular microthrombotic disease (VMTD) includes both TTP and TTP-like syndrome because their underlying pathology is the same disseminated intravascular microthrombosis (DIT). Microthrombi are composed of platelet-unusually large von Willebrand factor multimers (ULVWF) complexes. TTP occurs as a result of accumulation of circulating ULVWF secondary to ADAMTS13 deficiency. This protease deficiency triggers microthrombogenesis, leading to "microthrombi" formation in microcirculation. Unlike TTP, TTP-like syndrome occurs in critical illnesses due to complement activation. Terminal C5b-9 complex causes channel formation to endothelial membrane, leading to endotheliopathy, which activates two different molecular pathways (i.e., inflammatory and microthrombotic). Activation of inflammatory pathway triggers inflammation. Activation of microthrombotic pathway promotes platelet activation and excessive endothelial exocytosis of ULVWF from endothelial cells (ECs). Overexpressed and uncleaved ULVWF become anchored to ECs as long elongated strings to recruit activated platelets, and assemble "microthrombi". In TTP, circulating microthrombi typically be lodged in microvasculature of the brain and kidney, but in TTP-like syndrome, microthrombi anchored to ECs of organs such as the lungs and liver as well as the brain and kidneys, leading to multiorgan dysfunction syndrome. TTP occurs as hereditary or autoimmune disease and is the phenotype of ADAMTS13 deficiency-associated VMTD. But TTP-like syndrome is hemostatic disorder occurring in critical illnesses and is the phenotype of endotheliopathy-associated VMTD. Thus, this author's contention is TTP and TTP-like syndrome are two distinctly different disorders with dissimilar underlying pathology and pathogenesis.
Collapse
Affiliation(s)
- Jae C. Chang
- Department of Medicine, University of California Irvine School of Medicine, Irvine, CA USA
| |
Collapse
|
42
|
Thulin Å, Yan J, Åberg M, Christersson C, Kamali-Moghaddam M, Siegbahn A. Sensitive and Specific Detection of Platelet-Derived and Tissue Factor-Positive Extracellular Vesicles in Plasma Using Solid-Phase Proximity Ligation Assay. TH OPEN 2018; 2:e250-e260. [PMID: 31276087 PMCID: PMC6602879 DOI: 10.1055/s-0038-1667204] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Accepted: 06/13/2018] [Indexed: 12/17/2022] Open
Abstract
Extracellular vesicles (EVs) derived from blood cells are promising biomarkers for various diseases. However, they are difficult to measure accurately in plasma due to their small size. Here, we demonstrate that platelet-derived EVs in plasma can be measured using solid-phase proximity ligation assay with high sensitivity and specificity using very small sample volume of biological materials. The results correlate well with high-sensitivity flow cytometry with the difference that the smallest EVs are detected. Briefly, the EVs are first captured on a solid phase, using lactadherin binding, and detection requires recognition with two antibodies followed by qPCR. The assay, using cholera toxin subunit-B or lactadherin as capture agents, also allowed detection of the more rare population of tissue factor (TF)-positive EVs at a concentration similar to sensitive TF activity assays. Thus, this assay can detect different types of EVs with high specificity and sensitivity, and has the potential to be an attractive alternative to flow cytometric analysis of preclinical and clinical samples. Improved techniques for measuring EVs in plasma will hopefully contribute to the understanding of their role in several diseases.
Collapse
Affiliation(s)
- Åsa Thulin
- Department of Medical Sciences, Clinical Chemistry and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Junhong Yan
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden.,Department of Biomedical Engineering, Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Mikael Åberg
- Department of Medical Sciences, Clinical Chemistry and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | | | - Masood Kamali-Moghaddam
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Agneta Siegbahn
- Department of Medical Sciences, Clinical Chemistry and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| |
Collapse
|
43
|
Guo L, Tong D, Yu M, Zhang Y, Li T, Wang C, Zhou P, Jin J, Li B, Liu Y, Liu R, Novakovic VA, Dong Z, Tian Y, Kou J, Bi Y, Zhou J, Shi J. Phosphatidylserine-exposing cells contribute to the hypercoagulable state in patients with multiple myeloma. Int J Oncol 2018; 52:1981-1990. [PMID: 29620266 DOI: 10.3892/ijo.2018.4354] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 03/06/2018] [Indexed: 11/06/2022] Open
Abstract
Multiple myeloma (MM) is characterized by an increased incidence of thromboembolic events, particularly when treated with immunomodulatory drugs (IMiDs) in combination with dexamethasone. The optimal prophylactic strategy to prevent the hypercoagulable state of patients with MM is still debated. The aim of the current study was to investigate the definitive role of phosphatidylserine (PS) in supporting procoagulant activity (PCA) in patients with MM. Patients with MM (n=20) and healthy subjects (n=15) were recruited for the present study. PS analyses were performed by flow cytometry and confocal microscopy. The PCA was evaluated by clotting time, purified coagulation complex assays and fibrin production assays. The percentage of PS+ blood cells was significantly higher in patients with MM than in healthy subjects. Additionally, the patient serum induced more PS exposure on endothelial cells (ECs) in vitro than serum from healthy subjects. Isolated blood cells from patients with MM and ECs cultured with patient serum in vitro demonstrated significantly shortened coagulation time, greatly intrinsic/extrinsic factor Xa generation and increased thrombin formation. In addition, the levels of PS+ erythrocytes, platelets, leukocytes, and ECs incubated with IMiDs and dexamethasone were higher than with IMiDs alone. The findings support the hypothesis that increased PS exposure on blood cells and ECs participates in the hypercoagulable state in patients with MM. Thus, blocking PS may be a novel therapeutic target for the prevention of thrombosis in these patients.
Collapse
Affiliation(s)
- Li Guo
- Department of Hematology, The First Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Dongxia Tong
- Department of Hematology, The First Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Muxin Yu
- Department of Hematology, The First Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Yan Zhang
- Department of Hematology, The First Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Tao Li
- Department of Hematology, The First Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Chunxu Wang
- Department of Hematology, The First Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Peng Zhou
- Department of Neurosurgery, The Second Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Jiaqi Jin
- Department of Neurosurgery, The Second Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Baorong Li
- Department of Stomatology, The First Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Yingmiao Liu
- Department of Stomatology, The First Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Ruipeng Liu
- Department of Cardiology, The Second Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Valerie A Novakovic
- Department of Research and Surgery, VA Boston Healthcare System, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02132, USA
| | - Zengxiang Dong
- Department of Cardiology, The First Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Ye Tian
- Department of Cardiology, The First Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Junjie Kou
- Department of Cardiology, The Second Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Yayan Bi
- Department of Cardiology, The First Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Jin Zhou
- Department of Hematology, The First Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Jialan Shi
- Department of Hematology, The First Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| |
Collapse
|
44
|
Phosphatidylserine-exposing blood and endothelial cells contribute to the hypercoagulable state in essential thrombocythemia patients. Ann Hematol 2018; 97:605-616. [PMID: 29332224 DOI: 10.1007/s00277-018-3228-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Accepted: 12/28/2017] [Indexed: 12/27/2022]
Abstract
The mechanisms of thrombogenicity in essential thrombocythemia (ET) are complex and not well defined. Our objective was to explore whether phosphatidylserine (PS) exposure on blood cells and endothelial cells (ECs) can account for the increased thrombosis and distinct thrombotic risks among mutational subtypes in ET. Using flow cytometry and confocal microscopy, we found that the levels of PS-exposing erythrocytes, platelets, leukocytes, and serum-cultured ECs were significantly higher in each ET group [JAK2, CALR, and triple-negative (TN) (all P < 0.001)] than those in controls. Among ET patients, those with JAK2 mutations showed higher levels of PS-positive erythrocytes, platelets, neutrophils, and serum-cultured ECs than TN patients or those with CALR mutations, which show similar levels. Coagulation function assays showed that higher levels of PS-positive blood cells and serum-cultured ECs led to markedly shortened coagulation time and dramatically increased levels of FXa, thrombin, and fibrin production. This procoagulant activity could be largely blocked by addition of lactadherin (approx. 70% inhibition). Confocal microscopy showed that the FVa/FXa complex and fibrin fibrils colocalized with PS on ET serum-cultured ECs. Additionally, we found a relationship between D-dimer, prothrombin fragment F1 + 2, and PS exposure. Our study reveals a previously unrecognized link between hypercoagulability and exposed PS on cells, which might also be associated with distinct thrombotic risks among mutational subtypes in ET. Thus, blocking PS-binding sites may represent a new therapeutic target for preventing thrombosis in ET.
Collapse
|
45
|
Canzano P, Rossetti L, Ferri N, Balduini A, Abbonante V, Boselli D, De Marco L, Di Minno M, Toschi V, Corsini A, Tremoli E, Brambilla M, Facchinetti L, Camera M. Human megakaryocytes confer tissue factor to a subset of shed platelets to stimulate thrombin generation. Thromb Haemost 2017; 114:579-92. [DOI: 10.1160/th14-10-0830] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2014] [Accepted: 04/11/2015] [Indexed: 11/05/2022]
Abstract
SummaryTissue factor (TF), the main activator of the blood coagulation cascade, has been shown to be expressed by platelets. Despite the evidence that both megakaryocytes and platelets express TF mRNA, and that platelets can make de novo protein synthesis, the main mechanism thought to be responsible for the presence of TF within platelets is through the uptake of TF positive microparticles. In this study we assessed 1) whether human megakaryocytes synthesise TF and transfer it to platelets and 2) the contribution of platelet-TF to the platelet hemostatic capacity. In order to avoid the cross-talk with circulating microparticles, we took advantage from an in vitro cultured megakaryoblastic cell line (Meg-01) able to differentiate into megakaryocytes releasing platelet-like particles. We show that functionally active TF is expressed in human megakaryoblasts, increased in megakaryocytes, and is transferred to a subset of shed platelets where it contributes to clot formation. These data were all confirmed in human CD34pos- derived megakaryocytes and in their released platelets. The effect of TF silencing in Meg-megakaryoblasts resulted in a significant reduction of TF expression in these cells and also in Meg-megakaryocytes and Meg-platelets. Moreover, the contribution of platelet-TF to the platelet hemostatic capacity was highlighted by the significant delay in the kinetic of thrombin formation observed in platelets released by TF-silenced megakaryocytes. These findings provide evidences that TF is an endogenously synthesised protein that characterises megakaryocyte maturation and that it is transferred to a subset of newly-released platelets where it is functionally active and able to trigger thrombin generation.
Collapse
|
46
|
Platelet-neutrophil interactions as drivers of inflammatory and thrombotic disease. Cell Tissue Res 2017; 371:567-576. [PMID: 29178039 PMCID: PMC5820397 DOI: 10.1007/s00441-017-2727-4] [Citation(s) in RCA: 147] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Accepted: 10/27/2017] [Indexed: 02/07/2023]
Abstract
Neutrophils are well known for their role in infection and inflammatory disease and are first responders at sites of infection or injury. Platelets have an established role in hemostasis and thrombosis and are first responders at sites of vascular damage. However, neutrophils are increasingly recognized for their role in thrombosis, while the immunemodulatory properties of platelets are being increasingly studied. Platelets and neutrophils interact during infection, inflammation and thrombosis and modulate each other’s functions. This review will discuss the consequences of platelet–neutrophil interactions in infection, thrombosis, atherosclerosis and tissue injury and repair.
Collapse
|
47
|
Meng H, Kou J, Ma R, Ding W, Kou Y, Cao M, Dong Z, Bi Y, Thatte HS, Shi J. Prognostic implications and procoagulant activity of phosphatidylserine exposure of blood cells and microparticles in patients with atrial fibrillation treated with pulmonary vein isolation. Mol Med Rep 2017; 16:8579-8588. [PMID: 29039531 PMCID: PMC5779910 DOI: 10.3892/mmr.2017.7763] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Accepted: 01/27/2017] [Indexed: 11/21/2022] Open
Abstract
The present study aimed to evaluate the procoagulant effects of phosphatidylserine (PS) exposure on blood cells and microparticles (MPs), and examine its role in predicting early recurrence atrial fibrillation (ERAF) in patients with atrial fibrillation (AF) treated with pulmonary vein isolation (PVI). Blood samples were obtained from 40 healthy controls and 56 patients with AF at baseline (prior to PVI), and 0, 1 h, 1 day, 3 days and 7 days following PVI. The exposure of PS (PS+) to blood cells (platelets, erythrocytes and leukocytes) and MPs was detected using flow cytometry. The procoagulant activity was evaluated by coagulation time, and the formation of factor Xa (FXa) and thrombin. In addition, independent factors associated with PS+ blood cells and MPs, and significant predictors of ERAF following PVI were investigated by statistical analyses. The numbers of PS+ blood cells and MPs were significantly increased by PVI (P<0.01). A significant decrease in coagulation time, and increases in FXa and thrombin were exhibited in the PS+ blood cells and MPs from patients with AF treated with PVI, whereas these alterations were inhibited by either lactadherin or anti-tissue factor (P<0.01). The maximum power of the PVI was significantly associated with platelet-derived MPs, and high-sensitivity C-reactive protein (hs-CRP) was closely associated with leukocyte-derived MPs and endothelial-derived MPs (EMPs) (P<0.01). In addition, hs-CRP and EMPs >355/µl were identified as independent predictors of ERAF (P<0.05). The increased numbers of PS+ platelets, erythrocytes, leukocytes and MPs contributed to the procoagulant state of AF, and hs-CRP and EMPs were able to predict ERAF following PVI.
Collapse
Affiliation(s)
- Huan Meng
- Department of Cardiology, The Second Hospital, Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Junjie Kou
- Department of Cardiology, The Second Hospital, Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Ruishuang Ma
- Department of Hematology, The First Hospital, Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Wenbo Ding
- Department of Cardiology, The Second Hospital, Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Yan Kou
- Department of Cardiology, The First Hospital, Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Muhua Cao
- Department of Hematology, The First Hospital, Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Zengxiang Dong
- Department of Cardiology, The First Hospital, Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Yayan Bi
- Department of Cardiology, The First Hospital, Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Hemant S Thatte
- Department of Surgery, VA Boston Healthcare System, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Jialan Shi
- Department of Surgery, VA Boston Healthcare System, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
48
|
Grape intake reduces thrombin generation and enhances plasma fibrinolysis. Potential role of circulating procoagulant microparticles. J Nutr Biochem 2017; 50:66-73. [PMID: 29040837 DOI: 10.1016/j.jnutbio.2017.08.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Revised: 07/17/2017] [Accepted: 08/28/2017] [Indexed: 12/23/2022]
Abstract
Phytochemicals contained in grapes down-regulate several prothrombotic pathways in vitro. We evaluated the effect of grape consumption on coagulation and fibrinolysis in healthy volunteers. Thirty subjects were enrolled: 20 were given grape (5 g/kg body weight/day for 3 weeks), while 10 served as controls. Blood samples were taken at baseline (T0), at the end of the grape diet (T1) and after 4-week wash-out (T2). Grape intake caused a significant decrease of the procoagulant and inflammatory responses of whole blood and/or mononuclear cells to bacterial lipopolysaccharide at both T1 and T2. At plasma level, grape diet decreased thrombin generation at T1 and T2, largely through a reduction in the number and/or activity of procoagulant microparticles. This anticoagulant effect resulted in the formation of clots that were more susceptible to fibrinolysis, mainly because of a lesser activation of thrombin activatable fibrinolysis inhibitor. No difference in any variables was detected in controls at the time points considered. In conclusion, chronic grape consumption induces sustained anticoagulant and profibrinolytic effects with potential benefits for human health.
Collapse
|
49
|
Hudák R, Debreceni IB, Deák I, Szabó GG, Hevessy Z, Antal-Szalmás P, Osterud B, Kappelmayer J. Laboratory characterization of leukemic cell procoagulants. Clin Chem Lab Med 2017; 55:1215-1223. [PMID: 28593927 DOI: 10.1515/cclm-2017-0021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 03/30/2017] [Indexed: 12/15/2022]
Abstract
BACKGROUND In acute myeloid leukemias, there is an increased chance to develop thrombotic disorders. We hypothesized that in addition to leukemic promyelocytes, monocytic leukemia cells may also have a higher procoagulant activity. METHODS Fibrin formation was assessed by a one-stage clotting assay using a magnetic coagulometer. The thrombin generation test (TGT) of magnetically isolated normal human monocytes, intact leukemic cells and their isolated microparticles was performed by a fluorimetric assay. Phosphatidylserine (PS) expression of leukemic cells and microparticle number determinations were carried out by flow cytometry. RESULTS All cell lines displayed a significant procoagulant potential compared to isolated normal human monocytes. In the TGT test, the mean of lagtime and the time to peak parameters were significantly shorter in leukemic cells (3.9-4.7 and 9.9-10.3 min) compared to monocytes (14.9 and 26.5 min). The mean of peak thrombin in various monocytic leukemia cell lines was 112.1-132.9 nM vs. 75.1 nM in monocytes; however, no significant difference was observed in the ETP parameter. Factor VII-deficient plasma abolished all procoagulant activity, whereas factor XII-deficient plasma did not affect the speed of fibrin formation and thrombin generation but modulated the amount of thrombin. Factor XI-deficient plasma affected the time to peak values in one leukemic cell line and also attenuated peak thrombin. Leukemia cell-derived microparticles from all three cell lines exerted a procoagulant effect by significantly shortening the lagtime in TGT; there was a nonsignificant difference in case of ETP parameter. CONCLUSIONS All investigated monocytic leukemia cell lines exhibited significant thrombin generation. This phenomenon was achieved by the procoagulants on the surface of leukemic cells as well as by their microparticles.
Collapse
|
50
|
Wang J, Pendurthi UR, Rao LVM. Sphingomyelin encrypts tissue factor: ATP-induced activation of A-SMase leads to tissue factor decryption and microvesicle shedding. Blood Adv 2017; 1:849-862. [PMID: 28758160 PMCID: PMC5531194 DOI: 10.1182/bloodadvances.2016003947] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2016] [Accepted: 04/09/2017] [Indexed: 11/20/2022] Open
Abstract
A majority of tissue factor (TF) on cell surfaces exists in an encrypted state with minimal to no procoagulant activity. At present, it is unclear whether limited availability of phosphatidylserine (PS) and/or a specific membrane lipid in the outer leaflet of the plasma membrane contributes to TF encryption. Sphingomyelin (SM) is a major phospholipid in the outer leaflet, and SM metabolism is shown to be altered in many disease settings that cause thrombotic disorders. The present study is carried out to investigate the effect of SM metabolism on TF activity and TF+ microvesicles (MVs) release. In vitro studies using TF reconstituted into liposomes containing varying molar ratios of SM showed that a high molar ratio of SM in the proteoliposomes inhibits TF coagulant activity. Treatment of macrophages with sphingomyelinase (SMase) that hydrolyzes SM in the outer leaflet results in increased TF activity at the cell surface and TF+ MVs release without increasing PS externalization. Adenosine triphosphate (ATP) stimulation of macrophages that activates TF and induces MV shedding also leads to translocation of acid-sphingomyelinase (A-SMase) to the plasma membrane. ATP stimulation increases the hydrolysis of SM in the outer leaflet. Inhibition of A-SMase expression or activity not only attenuates ATP-induced SM hydrolysis, but also inhibits ATP-induced TF decryption and TF+ MVs release. Overall, our novel findings show that SM plays a role in maintaining TF in an encrypted state in resting cells and hydrolysis of SM following cell injury removes the inhibitory effect of SM on TF activity, thus leading to TF decryption.
Collapse
Affiliation(s)
- Jue Wang
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, TX
| | - Usha R Pendurthi
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, TX
| | - L Vijaya Mohan Rao
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, TX
| |
Collapse
|