1
|
Moore GW. Thrombophilia Screening: Not So Straightforward. Semin Thromb Hemost 2024; 50:1131-1152. [PMID: 38733983 DOI: 10.1055/s-0044-1786807] [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: 05/13/2024]
Abstract
Although inherited thrombophilias are lifelong risk factors for a first thrombotic episode, progression to thrombosis is multifactorial and not all individuals with inherited thrombophilia develop thrombosis in their lifetimes. Consequently, indiscriminate screening in patients with idiopathic thrombosis is not recommended, since presence of a thrombophilia does not necessarily predict recurrence or influence management, and testing should be selective. It follows that a decision to undertake laboratory detection of thrombophilia should be aligned with a concerted effort to identify any significant abnormalities, because it will inform patient management. Deficiencies of antithrombin and protein C are rare and usually determined using phenotypic assays assessing biological activities, whereas protein S deficiency (also rare) is commonly detected with antigenic assays for the free form of protein S since available activity assays are considered to lack specificity. In each case, no single phenotypic assay is capable of detecting every deficiency, because the various mutations express different molecular characteristics, rendering thrombophilia screening repertoires employing one assay per potential deficiency, of limited effectiveness. Activated protein C resistance (APCR) is more common than discrete deficiencies of antithrombin, protein C, and protein S and also often detected initially with phenotypic assays; however, some centres perform only genetic analysis for factor V Leiden, as this is responsible for most cases of hereditary APCR, accepting that acquired APCR and rare F5 mutations conferring APCR will go undetected if only factor V Leiden is evaluated. All phenotypic assays have interferences and limitations, which must be factored into decisions about if, and when, to test, and be given consideration in the laboratory during assay performance and interpretation. This review looks in detail at performance and limitations of routine phenotypic thrombophilia assays.
Collapse
Affiliation(s)
- Gary W Moore
- Specialist Haemostasis Laboratory, Cambridge Haemophilia and Thrombophilia Centre, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
- Department of Natural Sciences, Middlesex University, London, United Kingdom
| |
Collapse
|
2
|
Petri A, Sasikumar P, Folgado PB, Jones D, Xu Y, Ahnström J, Salles-Crawley II, Crawley JTB. TFPIα anticoagulant function is highly dependent on protein S in vivo. SCIENCE ADVANCES 2024; 10:eadk5836. [PMID: 38306422 DOI: 10.1126/sciadv.adk5836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 01/04/2024] [Indexed: 02/04/2024]
Abstract
Tissue factor pathway inhibitor α (TFPIα) is the major physiological regulator of the initiation of blood coagulation. In vitro, TFPIα anticoagulant function is enhanced by its cofactor, protein S. To define the role of protein S enhancement in TFPIα anticoagulant function in vivo, we blocked endogenous TFPI in mice using a monoclonal antibody (14D1). This caused a profound increase in fibrin deposition using the laser injury thrombosis model. To explore the role of plasma TFPIα in regulating thrombus formation, increasing concentrations of human TFPIα were coinjected with 14D1, which dose-dependently reduced fibrin deposition. Inhibition of protein S cofactor function using recombinant C4b-binding protein β chain significantly reduced the anticoagulant function of human TFPIα in controlling fibrin deposition. We report an in vivo model that is sensitive to the anticoagulant properties of the TFPIα-protein S pathway and show the importance of protein S as a cofactor in the anticoagulant function of TFPIα in vivo.
Collapse
Affiliation(s)
- Anastasis Petri
- Centre for Haematology, Hammersmith Hospital Campus, Imperial College London, London, UK
| | - Parvathy Sasikumar
- Centre for Haematology, Hammersmith Hospital Campus, Imperial College London, London, UK
| | - Patricia Badia Folgado
- Centre for Haematology, Hammersmith Hospital Campus, Imperial College London, London, UK
| | - David Jones
- Centre for Haematology, Hammersmith Hospital Campus, Imperial College London, London, UK
| | - Yaoxian Xu
- Centre for Haematology, Hammersmith Hospital Campus, Imperial College London, London, UK
| | - Josefin Ahnström
- Centre for Haematology, Hammersmith Hospital Campus, Imperial College London, London, UK
| | - Isabelle I Salles-Crawley
- Centre for Haematology, Hammersmith Hospital Campus, Imperial College London, London, UK
- Vascular Biology Research Centre, Molecular and Clinical Sciences Research Institute, St. George's University of London, London, UK
| | - James T B Crawley
- Centre for Haematology, Hammersmith Hospital Campus, Imperial College London, London, UK
| |
Collapse
|
3
|
Dong Y, Xu Y, Lian C, Prak K, Leo HL, Tetley TD, Braga V, Emerson M, Ahnström J, Yap CH. Safe and Efficacious Near Superhydrophobic Hemostat for Reduced Blood Loss and Easy Detachment in Traumatic Wounds. ACS APPLIED MATERIALS & INTERFACES 2024; 16:4307-4320. [PMID: 38240181 PMCID: PMC10835652 DOI: 10.1021/acsami.3c12443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 12/20/2023] [Accepted: 12/20/2023] [Indexed: 02/01/2024]
Abstract
Hemorrhage is the leading cause of trauma death, and innovation in hemostatic technology is important. The strongly hydrophobic carbon nanofiber (CNF) coating has previously been shown to have excellent hemostatic properties. However, the understanding of how CNF coating guides the coagulation cascade and the biosafety of CNF as hemostatic agents has yet to be explored. Here, our thrombin generation assay investigation showed that CNF induced fast blood coagulation via factor (F) XII activation of the intrinsic pathway. We further performed studies of a rat vein injury and demonstrated that the CNF gauze enabled a substantial reduction of blood loss compared to both the plain gauze and kaolin-imbued gauze (QuikClot). Analysis of blood samples from the model revealed no acute toxicity from the CNF gauze, with no detectable CNF deposition in any organ, suggesting that the immobilization of CNF on our gauze prevented the infiltration of CNF into the bloodstream. Direct injection of CNF into the rat vein was also investigated and found not to elicit overt acute toxicity or affect animal survival or behavior. Finally, toxicity assays with primary keratinocytes revealed minimal toxicity responses to CNF. Our studies thus supported the safety and efficacy of the CNF hemostatic gauze, highlighting its potential as a promising approach in the field of hemostatic control.
Collapse
Affiliation(s)
- Yibing Dong
- Department
of Biomedical Engineering, National University
of Singapore, Singapore 117583, Singapore
| | - Yaoxian Xu
- Department
of Bioengineering, Imperial College London, London SW7 2AZ, United Kingdom
| | - Chengxing Lian
- Department
of Bioengineering, Imperial College London, London SW7 2AZ, United Kingdom
| | - Krisna Prak
- National
Heart and Lung Institute, Imperial College
London, London SW3 6LY, United Kingdom
| | - Hwa Liang Leo
- Department
of Biomedical Engineering, National University
of Singapore, Singapore 117583, Singapore
| | - Teresa D. Tetley
- National
Heart and Lung Institute, Imperial College
London, London SW3 6LY, United Kingdom
| | - Vania Braga
- National
Heart and Lung Institute, Imperial College
London, London SW3 6LY, United Kingdom
| | - Mike Emerson
- National
Heart and Lung Institute, Imperial College
London, London SW3 6LY, United Kingdom
| | - Josefin Ahnström
- Department
of Immunology and Inflammation, Imperial
College London, London W12 0NN, United Kingdom
| | - Choon Hwai Yap
- Department
of Bioengineering, Imperial College London, London SW7 2AZ, United Kingdom
| |
Collapse
|
4
|
Gierula M, Noakes VM, Salles-Crawley II, Crawley JTB, Ahnström J. The TFPIα C-terminal tail is essential for TFPIα-FV-short-protein S complex formation and synergistic enhancement of TFPIα. J Thromb Haemost 2023; 21:3568-3580. [PMID: 37739040 DOI: 10.1016/j.jtha.2023.09.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 09/01/2023] [Accepted: 09/07/2023] [Indexed: 09/24/2023]
Abstract
BACKGROUND For maximal TFPIα functionality, 2 synergistic cofactors, protein S and FV-short, are required. Both interact with TFPIα, protein S through Kunitz 3 residues Arg199/Glu226 and FV-short with the C-terminus. How these interactions impact the synergistic enhancement remains unclear. OBJECTIVES To determine the importance of the TFPIα-protein S and TFPIα-FV-short interactions for TFPIα enhancement. METHODS TFPIα variants unable to bind protein S (K3m [R199Q/E226Q]) or FV-short (ΔCT [aa 1-249]) were generated. TFPIα-FV-short binding was studied by plate-binding and co-immunoprecipitation assays; functional TFPIα enhancement by FXa inhibition and prothrombin activation. RESULTS While WT TFPIα and TFPIα K3m bound FV-short with high affinity (Kd∼2nM), TFPIα ΔCT did not. K3m, in contrast to WT, did not incorporate protein S in a TFPIα-FV-short-protein S complex while TFPIα ΔCT bound neither FV-short nor protein S. Protein S enhanced WT TFPIα-mediated FXa inhibition, but not K3m, in the absence of FV-short. However, once FV-short was present, protein S efficiently enhanced TFPIα K3m (EC50: 4.7nM vs 2.0nM for WT). FXa inhibition by ΔCT was not enhanced by protein S alone or combined with FV-short. In FXa-catalyzed prothrombin activation assays, FV-short enhanced TFPIα K3m function in the presence of protein S (5.5 vs 10.4-fold enhancement of WT) whereas ΔCT showed reduced or lack of enhancement by FV-short and protein S, respectively. CONCLUSION Full TFPIα function requires the presence of both cofactors. While synergistic enhancement can be achieved in the absence of TFPIα-protein S interaction, only TFPIα with an intact C-terminus can be synergistically enhanced by protein S and FV-short.
Collapse
|
5
|
High levels of factor VIII activity in patients with acquired hemophilia A in remission are associated with unusually low coagulation potentials. Int J Hematol 2023; 117:669-677. [PMID: 36607560 DOI: 10.1007/s12185-022-03528-0] [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: 10/19/2022] [Revised: 12/15/2022] [Accepted: 12/25/2022] [Indexed: 01/07/2023]
Abstract
BACKGROUND Elevated factor VIII activity (FVIII:C) is often observed in patients with acquired hemophilia A (PwAHA) in remission. However, comprehensive coagulation potentials in this patient group remain to be investigated. AIM To evaluate comprehensive coagulation potentials in PwAHA. METHODS We investigated coagulation function in eleven PwAHA with high FVIII:C (> 150 IU/dL) using thrombin generation assay (TGA) and/or rotational thromboelastometry (ROTEM), and compared findings with results obtained from contrived samples generated by spiking recombinant FVIII. RESULTS The median FVIII:C and FVIII inhibitor titers during remission in enrolled PwAHA were 206 IU/dL and 0.44 BU/mL, respectively. In all patients, lag time and time to peak were either prolonged or normal compared to contrived samples corresponding to their FVIII:C. However, higher values of peak thrombin and endogenous thrombin potentials compared to contrived samples were observed in two patients. ROTEM parameters were within normal ranges in all cases. One patient (FVIII:C 171 IU/dL) developed venous thrombosis and pulmonary embolism, but TGA parameters showed low or normal coagulation potential compared to contrived samples corresponding to his FVIII:C. CONCLUSION PwAHA with high FVIII:C could exhibit lower coagulation potentials than those corresponding to their FVIII:C.
Collapse
|
6
|
Heal SL, Hardy LJ, Wilson CL, Ali M, Ariëns RAS, Foster R, Philippou H. Novel interaction of properdin and coagulation factor XI: Crosstalk between complement and coagulation. Res Pract Thromb Haemost 2022; 6:e12715. [PMID: 35647477 PMCID: PMC9130567 DOI: 10.1002/rth2.12715] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 02/25/2022] [Accepted: 03/22/2022] [Indexed: 12/18/2022] Open
Abstract
Background Evidence of crosstalk between the complement and coagulation cascades exists, and dysregulation of either pathway can lead to serious thromboinflammatory events. Both the intrinsic pathway of coagulation and the alternative pathway of complement interact with anionic surfaces, such as glycosaminoglycans. Hitherto, there is no evidence for a direct interaction of properdin (factor P [FP]), the only known positive regulator of complement, with coagulation factor XI (FXI) or activated FXI (FXIa). Objectives The aim was to investigate crosstalk between FP and the intrinsic pathway and the potential downstream consequences. Methods Chromogenic assays were established to characterize autoactivation of FXI in the presence of dextran sulfate (DXS), enzyme kinetics of FXIa, and the downstream effects of FP on intrinsic pathway activity. Substrate specificity changes were investigated using SDS-PAGE and liquid chromatography-mass spectrometry (LC-MS). Surface plasmon resonance (SPR) was used to determine direct binding between FP and FXIa. Results/Conclusions We identified a novel interaction of FP with FXIa resulting in functional consequences. FP reduces activity of autoactivated FXIa toward S-2288. FXIa can cleave FP in the presence of DXS, demonstrated using SDS-PAGE, and confirmed by LC-MS. FXIa can cleave factor IX (FIX) and FP in the presence of DXS, determined by SDS-PAGE. DXS alone modulates FXIa activity, and this effect is further modulated by FP. We demonstrate that FXI and FXIa bind to FP with high affinity. Furthermore, FX activation downstream of FXIa cleavage of FIX is modulated by FP. These findings suggest a novel intercommunication between complement and coagulation pathways.
Collapse
Affiliation(s)
- Samantha L. Heal
- Discovery and Translational Science DepartmentLeeds Institute of Cardiovascular and Metabolic MedicineUniversity of LeedsLeedsUK
| | - Lewis J. Hardy
- Discovery and Translational Science DepartmentLeeds Institute of Cardiovascular and Metabolic MedicineUniversity of LeedsLeedsUK
| | - Clare L. Wilson
- Discovery and Translational Science DepartmentLeeds Institute of Cardiovascular and Metabolic MedicineUniversity of LeedsLeedsUK
| | - Majid Ali
- Discovery and Translational Science DepartmentLeeds Institute of Cardiovascular and Metabolic MedicineUniversity of LeedsLeedsUK
| | - Robert A. S. Ariëns
- Discovery and Translational Science DepartmentLeeds Institute of Cardiovascular and Metabolic MedicineUniversity of LeedsLeedsUK
| | | | - Helen Philippou
- Discovery and Translational Science DepartmentLeeds Institute of Cardiovascular and Metabolic MedicineUniversity of LeedsLeedsUK
| |
Collapse
|
7
|
Teraz-Orosz A, Gierula M, Petri A, Jones D, Keniyopoullos R, Folgado PB, Santamaria S, Crawley JTB, Lane DA, Ahnström J. Laminin G1 residues of protein S mediate its TFPI cofactor function and are competitively regulated by C4BP. Blood Adv 2022; 6:704-715. [PMID: 34731882 PMCID: PMC8791571 DOI: 10.1182/bloodadvances.2021005382] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 10/20/2021] [Indexed: 11/29/2022] Open
Abstract
Protein S is a cofactor in the tissue factor pathway inhibitor (TFPI) anticoagulant pathway. It enhances TFPIα-mediated inhibition of factor (F)Xa activity and generation. The enhancement is dependent on a TFPIα-protein S interaction involving TFPIα Kunitz 3 and protein S laminin G-type (LG)-1. C4b binding protein (C4BP), which binds to protein S LG1, almost completely abolishes its TFPI cofactor function. However, neither the amino acids involved in TFPIα enhancement nor the mechanisms underlying the reduced TFPI cofactor function of C4BP-bound protein S are known. To screen for functionally important regions within protein S LG1, we generated 7 variants with inserted N-linked glycosylation attachment sites. Protein S D253T and Q427N/K429T displayed severely reduced TFPI cofactor function while showing normal activated protein C (APC) cofactor function and C4BP binding. Based on these results, we designed 4 protein S variants in which 4 to 6 surface-exposed charged residues were substituted for alanine. One variant, protein S K255A/E257A/D287A/R410A/K423A/E424A, exhibited either abolished or severely reduced TFPI cofactor function in plasma and FXa inhibition assays, both in the presence or absence of FV-short, but retained normal APC cofactor function and high-affinity C4BP binding. The C4BP β-chain was expressed to determine the mechanisms behind the reduced TFPI cofactor function of C4BP-bound protein S. Like C4BP-bound protein S, C4BP β-chain-bound protein S had severely reduced TFPI cofactor function. These results show that protein S Lys255, Glu257, Asp287, Arg410, Lys423, and Glu424 are critical for protein S-mediated enhancement of TFPIα and that binding of the C4BP β-chain blocks this function.
Collapse
Affiliation(s)
| | | | | | - David Jones
- Centre for Haematology, Imperial College London, London, UK
| | | | | | | | | | - David A. Lane
- Centre for Haematology, Imperial College London, London, UK
| | | |
Collapse
|
8
|
Abstract
PURPOSE OF REVIEW Protein S (PS) is an essential natural anticoagulant. PS deficiency is a major contributor to acquired hypercoagulability. Acquired hypercoagulability causes myocardial infarction, stroke, and deep vein thrombosis in millions of individuals. Yet, despite its importance in hemostasis, PS is the least understood anticoagulant. Even after 40 years since PS was first described, we are still uncovering information about how PS functions. The purpose of this review is to highlight recent findings that advance our understanding of the functions of PS and explain hypercoagulability caused by severe PS deficiency. RECENT FINDINGS PS has long been described as a cofactor for Activated Protein C (APC) and Tissue Factor Pathway Inhibitor (TFPI). However, a recent report describes direct inhibition of Factor IXa (FIXa) by PS, an activity of PS that had been completely overlooked. Thrombophilia is becoming a more frequently reported disorder. Hereditary PS deficiency is an anticoagulant deficiency that results eventually in thrombophilia. In addition, PS deficiency is a predisposing factor for venous thromboembolism (VTE), but an effect of PS deficiency in arterial thrombosis, such as arterial ischemic stroke, is uncertain. Plasma PS concentration decreases in pregnant women. Inherited thrombophilias are important etiologies for recurrent pregnancy loss, and anticoagulation therapy is of benefit to women with recurrent pregnancy loss who had documented only PS deficiency.Hypoxia is a risk factor for VTE, and hypoxia downregulates plasma PS level. Importantly, COVID-19 can lead to hypoxemia because of lung damage from IL6-driven inflammatory responses to the viral infection. Because hypoxia decreases the abundance of the key anticoagulant PS, we surmise that the IL6-induced cytokine explosion combined with hypoxemia causes a drop in PS level that exacerbates the thrombotic risk in COVID-19 patients. SUMMARY This review is intended to advance understanding of the anticoagulant function of an important plasma protein, PS. Despite 40+ years of research, we have not had a complete description of PS biology as it pertains to control of blood coagulation. However, the picture of PS function has become sharper with the recent discovery of FIXa inhibition by PS. Hemostasis mediated by PS now includes regulation of FIXa activity alongside the cofactor activities of PS in the TFPI/APC pathways. In addition, the direct inhibition of FIXa by PS suggests that PS, particularly a small derivative of PS, could be used to treat individuals with PS deficiencies or abnormalities that cause thrombotic complications.
Collapse
Affiliation(s)
- Rinku Majumder
- Department of Biochemistry & Molecular Biology, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
| | | |
Collapse
|
9
|
Sugimoto M, Kondo M, Yasuma T, D'Alessandro-Gabazza CN, Toda M, Imai H, Nakamura M, Gabazza EC. Increased expression of Protein S in eyes with diabetic retinopathy and diabetic macular edema. Sci Rep 2021; 11:10449. [PMID: 34001977 PMCID: PMC8129118 DOI: 10.1038/s41598-021-89870-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 05/04/2021] [Indexed: 11/09/2022] Open
Abstract
Protein S (PS) is a multifunctional glycoprotein that ameliorates the detrimental effects of diabetes mellitus (DM). The aim of this study was to evaluate the distribution of PS in diabetic retinopathy (DR) and diabetic macular edema (DME). This was a study of 50 eyes with DM (37 with DME, 6 with proliferative DR, and 7 with no DR) and 19 eyes without DM. The level of PS was measured by enzyme immunoassay and was compared between eyes with or without DM, with or without DME, and with severe DME (≥ 350 μm) or mild DME (< 350 μm). We also performed immunohistopathologic evaluations of post-mortem eyes and the cystoid lesions excised during surgery. The aqueous free PS was significantly higher with DM (7.9 ± 1.2 ng/ml, P < 0.01) than without DM (6.1 ± 0.7). The aqueous free PS was significantly elevated with DME (8.2 ± 1.2, P < 0.05) compared to proliferative DR (7.0 ± 1.0) and no DR (7.0 ± 0.7). Eyes with severe DME had significantly higher aqueous free PS than mild DME (8.5 ± 1.3 vs. 7.7 ± 1.0, P < 0.05). Immunohistochemistry showed PS in the outer plexiform layer of the retina and cystoid lesion. The higher expression of PS with DR and DME suggests that PS is involved in their pathogenesis.
Collapse
Affiliation(s)
- Masahiko Sugimoto
- Department of Ophthalmology, Mie University Graduate School of Medicine, 2-174, Edobashi, Tsu, Mie, 514-8507, Japan.
| | - Mineo Kondo
- Department of Ophthalmology, Mie University Graduate School of Medicine, 2-174, Edobashi, Tsu, Mie, 514-8507, Japan
| | - Taro Yasuma
- Department of Immunology, Mie University Graduate School of Medicine, Tsu, Mie, Japan
| | | | - Masaaki Toda
- Department of Immunology, Mie University Graduate School of Medicine, Tsu, Mie, Japan
| | - Hisanori Imai
- Division of Ophthalmology, Department of Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Makoto Nakamura
- Division of Ophthalmology, Department of Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Esteban C Gabazza
- Department of Immunology, Mie University Graduate School of Medicine, Tsu, Mie, Japan
| |
Collapse
|
10
|
Kallikrein directly interacts with and activates Factor IX, resulting in thrombin generation and fibrin formation independent of Factor XI. Proc Natl Acad Sci U S A 2021; 118:2014810118. [PMID: 33397811 PMCID: PMC7826336 DOI: 10.1073/pnas.2014810118] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Prekallikrein (PK) is a zymogen that is converted to kallikrein (PKa) by factor (F)XIIa. PK and FXII reciprocally activate each other; the resulting FXIIa initiates activation of the coagulation system via the cleavage of FXI to FXIa, which then activates FIX. This manuscript describes a novel high-affinity binding interaction between FIX(a) and PK(a) and reports that PKa can dose- and time-dependently activate FIX to generate FIXa, resulting in thrombin generation and clot formation independent of FXIa. Characterization of the kinetics of FIX activation reveal that PKa is a more significant activator of FIX than previously considered. This work highlights a new amendment to the coagulation cascade where PKa can directly activate FIX. Kallikrein (PKa), generated by activation of its precursor prekallikrein (PK), plays a role in the contact activation phase of coagulation and functions in the kallikrein-kinin system to generate bradykinin. The general dogma has been that the contribution of PKa to the coagulation cascade is dependent on its action on FXII. Recently this dogma has been challenged by studies in human plasma showing thrombin generation due to PKa activity on FIX and also by murine studies showing formation of FIXa-antithrombin complexes in FXI deficient mice. In this study, we demonstrate high-affinity binding interactions between PK(a) and FIX(a) using surface plasmon resonance and show that these interactions are likely to occur under physiological conditions. Furthermore, we directly demonstrate dose- and time-dependent cleavage of FIX by PKa in a purified system by sodium dodecyl sulfate-polyacrylamide gel electrophoresis analysis and chromogenic assays. By using normal pooled plasma and a range of coagulation factor-deficient plasmas, we show that this action of PKa on FIX not only results in thrombin generation, but also promotes fibrin formation in the absence of FXII or FXI. Comparison of the kinetics of either FXIa- or PKa-induced activation of FIX suggest that PKa could be a significant physiological activator of FIX. Our data indicate that the coagulation cascade needs to be redefined to indicate that PKa can directly activate FIX. The circumstances that drive PKa substrate specificity remain to be determined.
Collapse
|
11
|
Eliwan H, Omer M, McKenna E, Kelly LA, Nolan B, Regan I, Molloy EJ. Protein C Pathway in Paediatric and Neonatal Sepsis. Front Pediatr 2021; 9:562495. [PMID: 35186813 PMCID: PMC8849213 DOI: 10.3389/fped.2021.562495] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 12/20/2021] [Indexed: 11/24/2022] Open
Abstract
Protein C plays a major role in the physiological regulation of coagulation pathways through inactivation of factor Va, factor VIIIa, and plasminogen activator inhibitor. Protein C is involved in the control of inflammation during sepsis, by inhibiting release of pro-inflammatory cytokines, thereby controlling neutrophil, and monocyte effects on injured tissue. Recombinant human activated protein C (rhAPC) reduced mortality in adult sepsis in earlier studies but had no significant benefit in more recent trials. Protein C levels are reduced during paediatric and neonatal sepsis, which may play a major role in the development of disseminated intravascular thrombosis, purpura fulminans, and multiorgan dysfunction. The role of protein C in paediatric sepsis requires further clinical and immunological evaluation to define the patient subgroups who may benefit from this therapy. Newer versions of rhAPC are under development with less risk of haemorrhage potentially broadening the scope of this intervention.
Collapse
Affiliation(s)
- Hassan Eliwan
- National Children's Research Centre, Dublin, Ireland.,Department of Paediatrics, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland
| | - Murwan Omer
- Department of Paediatrics, Children's Health Ireland at Tallaght, Dublin, Ireland
| | - Ellen McKenna
- Department of Paediatrics, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland
| | - Lynne A Kelly
- National Children's Research Centre, Dublin, Ireland.,Department of Paediatrics, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland.,Trinity Research in Childhood Centre, Dublin, Ireland
| | - Beatrice Nolan
- Department of Haematology, Children's Health Ireland at Crumlin, Dublin, Ireland
| | - Irene Regan
- National Children's Research Centre, Dublin, Ireland.,Department of Haematology, Children's Health Ireland at Crumlin, Dublin, Ireland
| | - Eleanor J Molloy
- National Children's Research Centre, Dublin, Ireland.,Department of Paediatrics, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland.,Department of Paediatrics, Children's Health Ireland at Tallaght, Dublin, Ireland.,Trinity Research in Childhood Centre, Dublin, Ireland.,Department of Neonatology, Children's Health Ireland at Crumlin, Dublin, Ireland.,Department of Paediatrics, Coombe Women's and Infant's University Hospital, Dublin, Ireland
| |
Collapse
|
12
|
Gierula M, Ahnström J. Anticoagulant protein S-New insights on interactions and functions. J Thromb Haemost 2020; 18:2801-2811. [PMID: 32702208 DOI: 10.1111/jth.15025] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 06/25/2020] [Accepted: 07/10/2020] [Indexed: 01/21/2023]
Abstract
Protein S is a critical regulator of coagulation that functions as a cofactor for the activated protein C (APC) and tissue factor pathway inhibitor (TFPI) pathways. It also has direct anticoagulant functions, inhibiting the intrinsic tenase and prothrombinase complexes. Through these functions, protein S regulates coagulation during both its initiation and its propagation phases. The importance of protein S in hemostatic regulation is apparent from the strong association between protein S deficiencies and increased risk for venous thrombosis. This is most likely because both APC and TFPIα are inefficient anticoagulants in the absence of any cofactors. The detailed molecular mechanisms involved in protein S cofactor functions remain to be fully clarified. However, recent advances in the field have greatly improved our understanding of these functions. Evidence suggests that protein S anticoagulant properties often depend on the presence of synergistic cofactors and the formation of multicomponent complexes on negatively charged phospholipid surfaces. Their high affinity binding to negatively charged phospholipids helps bring the anticoagulant proteins to the membranes, resulting in efficient and targeted regulation of coagulation. In this review, we provide an update on protein S and how it functions as a critical hemostatic regulator.
Collapse
|
13
|
Yamashita A, Zhang Y, Sanner MF, Griffin JH, Mosnier LO. C-terminal residues of activated protein C light chain contribute to its anticoagulant and cytoprotective activities. J Thromb Haemost 2020; 18:1027-1038. [PMID: 32017367 PMCID: PMC7380734 DOI: 10.1111/jth.14756] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 01/23/2020] [Accepted: 01/28/2020] [Indexed: 12/18/2022]
Abstract
BACKGROUND Activated protein C (APC) is an important homeostatic blood coagulation protease that conveys anticoagulant and cytoprotective activities. Proteolytic inactivation of factors Va and VIIIa facilitated by cofactor protein S is responsible for APC's anticoagulant effects, whereas cytoprotective effects of APC involve primarily the endothelial protein C receptor (EPCR), protease activated receptor (PAR)1 and PAR3. OBJECTIVE To date, several binding exosites in the protease domain of APC have been identified that contribute to APC's interaction with its substrates but potential contributions of the C-terminus of the light chain have not been studied in detail. METHODS Site-directed Ala-scanning mutagenesis of six positively charged residues within G142-L155 was used to characterize their contributions to APC's anticoagulant and cytoprotective activities. RESULTS AND CONCLUSIONS K151 was involved in protein S dependent-anticoagulant activity of APC with some contribution of K150. 3D structural analysis supported that these two residues were exposed in an extended protein S binding site on one face of APC. Both K150 and K151 were important for PAR1 and PAR3 cleavage by APC, suggesting that this region may also mediate interactions with PARs. Accordingly, APC's cytoprotective activity as determined by endothelial barrier protection was impaired by Ala substitutions of these residues. Thus, both K150 and K151 are involved in APC's anticoagulant and cytoprotective activities. The differential contribution of K150 relative to K151 for protein S-dependent anticoagulant activity and PAR cleavage highlights that binding exosites for protein S binding and for PAR cleavage in the C-terminal region of APC's light chain overlap.
Collapse
Affiliation(s)
- Atsuki Yamashita
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA
| | - Yuqi Zhang
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla
| | - Michel F. Sanner
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla
| | - John H. Griffin
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA
| | - Laurent O. Mosnier
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA
| |
Collapse
|
14
|
Ahnström J, Gierula M, Temenu J, Laffan MA, Lane DA. Partial rescue of naturally occurring active site factor X variants through decreased inhibition by tissue factor pathway inhibitor and antithrombin. J Thromb Haemost 2020; 18:136-150. [PMID: 31466141 DOI: 10.1111/jth.14627] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 08/22/2019] [Accepted: 08/26/2019] [Indexed: 12/19/2022]
Abstract
BACKGROUND Activated coagulation factor X (FXa) is the serine protease component of prothrombinase, the physiological activator of prothrombin. Factor X Nottingham (A404T) and Taunton (R405G) are two naturally occurring mutations, identified in families with a bleeding phenotype. OBJECTIVE To characterize these FX variants functionally. METHODS The activity and inhibition of recombinant FX variants were quantified in plasma-based and pure component assays. RESULTS The prothrombin times in FX-depleted plasma supplemented with FX Nottingham and Taunton were greatly increased compared to that of wild-type (WT) FX. Kinetic investigations of activated variants in the prothrombinase complex showed kcat /Km reduced ~50-fold and ~5-fold, respectively, explaining the prolonged prothrombin time (PT). The substituted residues are located in the protease domain Na+ -binding loop, important for the activity of FXa, as well as its inhibition. Both FXa Nottingham and Taunton showed reduced affinity for Na+ . Plasma-based thrombin generation assays triggered with 1 pmol/L tissue factor (TF) demonstrated only small differences in activities compared to WT FX, but large reductions at 10 pmol/L TF. Severely reduced inhibition of both FXa Nottingham and Taunton by tissue factor pathway inhibitor (TFPI) and antithrombin (AT), was shown in pure-component FXa inhibition assays. Factor Xa Nottingham and Taunton produced higher amounts of thrombin than WT FXa in pure-component prothrombinase assays in the presence of TFPI and AT, explaining the results from the plasma-based assay. CONCLUSIONS Factor X Nottingham and Taunton both display decreased proteolytic activity. However, their reduced activity in plasma triggered by low TF can be rescued by decreased inhibition by the natural FXa inhibitors, TFPI and AT.
Collapse
Affiliation(s)
- Josefin Ahnström
- Centre for Haematology, Faculty of Medicine, Imperial College London, London, UK
| | - Magdalena Gierula
- Centre for Haematology, Faculty of Medicine, Imperial College London, London, UK
| | - Joseph Temenu
- Centre for Haematology, Faculty of Medicine, Imperial College London, London, UK
| | - Michael A Laffan
- Centre for Haematology, Faculty of Medicine, Imperial College London, London, UK
| | - David A Lane
- Centre for Haematology, Faculty of Medicine, Imperial College London, London, UK
| |
Collapse
|
15
|
Gierula M, Salles‐Crawley II, Santamaria S, Teraz‐Orosz A, Crawley JTB, Lane DA, Ahnström J. The roles of factor Va and protein S in formation of the activated protein C/protein S/factor Va inactivation complex. J Thromb Haemost 2019; 17:2056-2068. [PMID: 31364267 PMCID: PMC6916587 DOI: 10.1111/jth.14594] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 07/26/2019] [Indexed: 12/22/2022]
Abstract
BACKGROUND Activated protein C (APC)-mediated inactivation of factor (F)Va is greatly enhanced by protein S. For inactivation to occur, a trimolecular complex among FVa, APC, and protein S must form on the phospholipid membrane. However, direct demonstration of complex formation has proven elusive. OBJECTIVES To elucidate the nature of the phospholipid-dependent interactions among APC, protein S, and FVa. METHODS We evaluated binding of active site blocked APC to phospholipid-coated magnetic beads in the presence and absence of protein S and/or FVa. The importance of protein S and FV residues were evaluated functionally. RESULTS Activated protein C alone bound weakly to phospholipids. Protein S mildly enhanced APC binding to phospholipid surfaces, whereas FVa did not. However, FVa together with protein S enhanced APC binding (>14-fold), demonstrating formation of an APC/protein S/FVa complex. C4b binding protein-bound protein S failed to enhance APC binding, agreeing with its reduced APC cofactor function. Protein S variants (E36A and D95A) with reduced APC cofactor function exhibited essentially normal augmentation of APC binding to phospholipids, but diminished APC/protein S/FVa complex formation, suggesting involvement in interactions dependent upon FVa. Similarly, FVaNara (W1920R), an APC-resistant FV variant, also did not efficiently incorporate into the trimolecular complex as efficiently as wild-type FVa. FVa inactivation assays suggested that the mutation impairs its affinity for phospholipid membranes and with protein S within the complex. CONCLUSIONS FVa plays a central role in the formation of its inactivation complex. Furthermore, membrane proximal interactions among FVa, APC, and protein S are essential for its cofactor function.
Collapse
Affiliation(s)
| | | | | | | | | | - David A. Lane
- Centre for HaematologyImperial College LondonLondonUK
| | | |
Collapse
|
16
|
Peghaire C, Dufton NP, Lang M, Salles-Crawley II, Ahnström J, Kalna V, Raimondi C, Pericleous C, Inuabasi L, Kiseleva R, Muzykantov VR, Mason JC, Birdsey GM, Randi AM. The transcription factor ERG regulates a low shear stress-induced anti-thrombotic pathway in the microvasculature. Nat Commun 2019; 10:5014. [PMID: 31676784 PMCID: PMC6825134 DOI: 10.1038/s41467-019-12897-w] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Accepted: 09/30/2019] [Indexed: 12/30/2022] Open
Abstract
Endothelial cells actively maintain an anti-thrombotic environment; loss of this protective function may lead to thrombosis and systemic coagulopathy. The transcription factor ERG is essential to maintain endothelial homeostasis. Here, we show that inducible endothelial ERG deletion (ErgiEC-KO) in mice is associated with spontaneous thrombosis, hemorrhages and systemic coagulopathy. We find that ERG drives transcription of the anticoagulant thrombomodulin (TM), as shown by reporter assays and chromatin immunoprecipitation. TM expression is regulated by shear stress (SS) via Krüppel-like factor 2 (KLF2). In vitro, ERG regulates TM expression under low SS conditions, by facilitating KLF2 binding to the TM promoter. However, ERG is dispensable for TM expression in high SS conditions. In ErgiEC-KO mice, TM expression is decreased in liver and lung microvasculature exposed to low SS but not in blood vessels exposed to high SS. Our study identifies an endogenous, vascular bed-specific anticoagulant pathway in microvasculature exposed to low SS.
Collapse
Affiliation(s)
- C Peghaire
- National Heart and Lung Institute, Imperial College London, London, UK
| | - N P Dufton
- National Heart and Lung Institute, Imperial College London, London, UK
| | - M Lang
- National Heart and Lung Institute, Imperial College London, London, UK
| | - I I Salles-Crawley
- Centre for Haematology, Hammersmith Hospital Campus, Imperial College London, London, UK
| | - J Ahnström
- Centre for Haematology, Hammersmith Hospital Campus, Imperial College London, London, UK
| | - V Kalna
- National Heart and Lung Institute, Imperial College London, London, UK
| | - C Raimondi
- National Heart and Lung Institute, Imperial College London, London, UK
| | - C Pericleous
- National Heart and Lung Institute, Imperial College London, London, UK
| | - L Inuabasi
- National Heart and Lung Institute, Imperial College London, London, UK
| | - R Kiseleva
- Department of Pharmacology, Institute for Translational Medicine and Therapeutics, University of Pennsylvania School of Medicine, Philadelphia, USA
| | - V R Muzykantov
- Department of Pharmacology, Institute for Translational Medicine and Therapeutics, University of Pennsylvania School of Medicine, Philadelphia, USA
| | - J C Mason
- National Heart and Lung Institute, Imperial College London, London, UK
| | - G M Birdsey
- National Heart and Lung Institute, Imperial College London, London, UK
| | - A M Randi
- National Heart and Lung Institute, Imperial College London, London, UK.
| |
Collapse
|
17
|
Crawley JTB, Zalli A, Monkman JH, Petri A, Lane DA, Ahnstrӧm J, Salles‐Crawley II. Defective fibrin deposition and thrombus stability in Bambi -/- mice are mediated by elevated anticoagulant function. J Thromb Haemost 2019; 17:1935-1949. [PMID: 31351019 PMCID: PMC6899896 DOI: 10.1111/jth.14593] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 07/22/2019] [Indexed: 01/22/2023]
Abstract
BACKGROUND Bone morphogenetic and activin membrane-bound inhibitor (BAMBI) is a transmembrane protein related to the type I transforming growth factor- β (TGF-β) receptor family that is present on both platelets and endothelial cells (ECs). Bambi-deficient mice exhibit reduced hemostatic function and thrombus stability characterized by an increased embolization. OBJECTIVE We aimed to delineate how BAMBI influences endothelial function and thrombus stability. METHODS Bambi-deficient mice were subjected to the laser-induced thrombosis model where platelet and fibrin accumulation was evaluated. Expression of thrombomodulin and tissue factor pathway inhibitor (TFPI) was also assessed in these mice. RESULTS Thrombus instability in Bambi-/- mice was associated with a profound defect in fibrin deposition. Injection of hirudin into Bambi+/+ mice prior to thrombus formation recapitulated the Bambi-/- thrombus instability phenotype. In contrast, hirudin had no additional effect upon thrombus formation in Bambi-/- mice. Deletion of Bambi in ECs resulted in mice with defective thrombus stability caused by decreased fibrin accumulation. Increased levels of the anticoagulant proteins TFPI and thrombomodulin were detected in Bambi-/- mouse lung homogenates. Endothelial cells isolated from Bambi-/- mouse lungs exhibited enhanced ability to activate protein C due to elevated thrombomodulin levels. Blocking thrombomodulin and TFPI in vivo fully restored fibrin accumulation and thrombus stability in Bambi-/- mice. CONCLUSIONS We demonstrate that endothelial BAMBI influences fibrin generation and thrombus stability by modulating thrombomodulin and TFPI anticoagulant function of the endothelium; we also highlight the importance of these anticoagulant proteins in the laser-induced thrombosis model.
Collapse
Affiliation(s)
- James T. B. Crawley
- Centre for HaematologyHammersmith Hospital CampusImperial College LondonLondonUK
| | - Argita Zalli
- Centre for HaematologyHammersmith Hospital CampusImperial College LondonLondonUK
| | - James H. Monkman
- Centre for HaematologyHammersmith Hospital CampusImperial College LondonLondonUK
| | - Anastasis Petri
- Centre for HaematologyHammersmith Hospital CampusImperial College LondonLondonUK
| | - David A. Lane
- Centre for HaematologyHammersmith Hospital CampusImperial College LondonLondonUK
| | - Josefin Ahnstrӧm
- Centre for HaematologyHammersmith Hospital CampusImperial College LondonLondonUK
| | | |
Collapse
|
18
|
Zhou J, Shen W, Gu Y, Li M, Shen W. Compound heterozygous mutations identified in severe type I protein S deficiency impaired the secretion of protein S. J Clin Pathol 2019; 73:7-13. [PMID: 31422373 DOI: 10.1136/jclinpath-2019-205956] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 08/06/2019] [Accepted: 08/07/2019] [Indexed: 11/04/2022]
Abstract
AIMS Hereditary protein S (PS) deficiency is one of the natural anticoagulant deficiencies causing thrombophilia. We herein described a young male with recurrent deep venous thrombosis, who was diagnosed as type I PS deficiency with compound heterozygous mutations of PROS1 gene. We aimed to analyse the relationship between the genotype and phenotype detection and investigate the pathological mechanisms of PROS1 mutations causing PS deficiency. METHODS Genetic analysis of PROS1 gene was carried out by direct sequencing. Thrombin generation potential and the inhibition function of thrombin generation by plasma PS were detected by thrombin generation test (TGT). The mRNA transcription level of mutant PS in vitro was measured by real-time PCR, while the protein level was evaluated by western blot and ELISA. Cellular distribution of the protein was further analysed by immunofluorescence. RESULTS Compound heterozygous mutations (PROS1 c.1551_1552delinsG, p.Thr518Argfs*39 and PROS1 c.1681C>T, p.Arg561Trp) were identified in the propositus, and the former one was a novel small indel mutation. TGT results showed impaired inhibition of thrombin generation with the addition of activated protein C in his parents with certain heterozygous mutations. In vitro expression study, p.Thr518Argfs*39 mutant produced truncated protein retained in the cytoplasm, while p.Arg561Trp mutant partially affected the secretion of PS. Both mutations are located in C-terminal sex hormone-binding globulin (SHBG)-like domain of PS. CONCLUSIONS Compound heterozygous mutations identified in the study have strong detrimental effect, causing severe type I PS deficiency in the propositus. SHBG-like domain of PS might play an important role in PS secretion system.
Collapse
Affiliation(s)
- Jingyi Zhou
- Department of Laboratory Medicine, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Wenyan Shen
- Department of Laboratory Medicine, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yi Gu
- Department of Laboratory Medicine, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Min Li
- Department of Laboratory Medicine, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Wei Shen
- Department of Laboratory Medicine, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
19
|
Lu Y, Mehta-D'souza P, Biswas I, Villoutreix BO, Wang X, Ding Q, Rezaie AR. Ile73Asn mutation in protein C introduces a new N-linked glycosylation site on the first EGF-domain of protein C and causes thrombosis. Haematologica 2019; 105:1712-1722. [PMID: 31399531 PMCID: PMC7271577 DOI: 10.3324/haematol.2019.227033] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 08/07/2019] [Indexed: 12/24/2022] Open
Abstract
Activated protein C exerts its anticoagulant activity by protein S-dependent inactivation of factors Va and VIIIa by limited proteolysis. We identified a venous thrombosis patient who has plasma protein C antigen level of 63% and activity levels of 44% and 23%, as monitored by chromogenic and clotting assays. Genetic analysis revealed the proband carries compound heterozygous mutations (c.344T>A, p.I73N and c.1181G>A, p.R352Q) in PROC. We individually expressed protein C mutations and discovered that thrombin-thrombomodulin activates both variants normally and the resulting activated protein C mutants exhibit normal amidolytic and proteolytic activities. However, while protein S-dependent catalytic activity of activated protein C-R352Q toward factor Va was normal, it was significantly impaired for activated protein C-I73N. These results suggest that the Ile to Asn substitution impairs interaction of activated protein C-I73N with protein S. This conclusion was supported by a normal anticoagulant activity for activated protein C-I73N in protein S-deficient but not in normal plasma. Further analysis revealed Ile to Asn substitution introduces a new glycosylation site on first EGF-like domain of protein C, thereby adversely affecting interaction of activated protein C with protein S. Activated protein C-R352Q only exhibited reduced activity in sub-physiological concentrations of Na+ and Ca2+, suggesting that this residue contributes to metal ion-binding affinity of the protease, with no apparent adverse effect on its function in the presence of physiological levels of metal ions. These results provide insight into the mechanism by which I73N/R352Q mutations in activated protein C cause thrombosis in proband carrying this compound heterozygous mutation.
Collapse
Affiliation(s)
- Yeling Lu
- Department of Laboratory Medicine, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.,Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Padmaja Mehta-D'souza
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Indranil Biswas
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Bruno O Villoutreix
- Université Lille, Inserm, Institut Pasteur de Lille, U1177 - Drugs and Molecules for Living Systems, F-59000 Lille, France
| | - Xuefeng Wang
- Department of Laboratory Medicine, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Qiulan Ding
- Department of Laboratory Medicine, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Alireza R Rezaie
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA .,Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| |
Collapse
|
20
|
Alshaikh NA, Rosing J, Thomassen MCLGD, Castoldi E, Simioni P, Hackeng TM. New functional assays to selectively quantify the activated protein C- and tissue factor pathway inhibitor-cofactor activities of protein S in plasma. J Thromb Haemost 2017; 15:950-960. [PMID: 28211163 DOI: 10.1111/jth.13657] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Indexed: 11/28/2022]
Abstract
Essentials Protein S is a cofactor of activated protein C (APC) and tissue factor pathway inhibitor (TFPI). There are no assays to quantify separate APC and TFPI cofactor activities of protein S in plasma. We developed assays to measure the APC- and TFPI-cofactor activities of protein S in plasma. The assays were sensitive to protein S deficiency, and not affected by the Factor V Leiden mutation. SUMMARY Background Protein S plays an important role in the down-regulation of coagulation as cofactor for activated protein C (APC) and tissue factor pathway inhibitor (TFPI). Aim To develop functional assays to quantify the APC- and TFPI-cofactor activities of protein S in plasma. Methods APC- and TFPI-cofactor activities of protein S in plasma were measured using calibrated automated thrombography in protein S-depleted plasma supplemented with a small amount of sample plasma either in the presence of anti-TFPI antibodies and APC (APC-cofactor activity) or at excess full-length TFPI without APC (TFPI-cofactor activity). Total and free protein S levels in plasma were measured by ELISAs. Results Average APC-cofactor activities of protein S were 113%, 108% and 89% in plasma from normal individuals (n = 15), FV Leiden heterozygotes (n = 14) and FV Leiden homozygotes (n = 7), respectively, whereas the average APC-cofactor activity of protein S in plasma from heterozygous protein S-deficient individuals (n = 21) was significantly lower (55%). Similar trends were observed for the TFPI-cofactor activity of protein S, with averages of 109%, 115% and 124% in plasma from individuals with normal protein S levels and different FV Leiden genotypes, and 64% in plasma from protein S-deficient patients. APC-cofactor activities of protein S correlated significantly with free and total protein S antigen levels, whereas TFPI-cofactor activities correlated less with protein S antigen levels. Conclusion We have developed functional protein S assays that measure both the APC- and TFPI-cofactor activities of protein S in plasma, which are hardly if at all affected by the FV Leiden mutation.
Collapse
Affiliation(s)
- N A Alshaikh
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, the Netherlands
| | - J Rosing
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, the Netherlands
| | - M C L G D Thomassen
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, the Netherlands
| | - E Castoldi
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, the Netherlands
| | - P Simioni
- Thrombotic and Haemorrhagic Unit, Department of Medicine (DIMED), 5th Chair of Internal Medicine, University of Padua Medical School, Padua, Italy
| | - T M Hackeng
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, the Netherlands
| |
Collapse
|
21
|
Santamaria S, Reglińska-Matveyev N, Gierula M, Camire RM, Crawley JTB, Lane DA, Ahnström J. Factor V has an anticoagulant cofactor activity that targets the early phase of coagulation. J Biol Chem 2017; 292:9335-9344. [PMID: 28420729 PMCID: PMC5454113 DOI: 10.1074/jbc.m116.769570] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Revised: 04/11/2017] [Indexed: 11/21/2022] Open
Abstract
Tissue factor pathway inhibitor (TFPI), the main inhibitor of initiation of coagulation, exerts an important anticoagulant role through the factor Xa (FXa)-dependent inhibition of tissue factor/factor VIIa. Protein S is a TFPI cofactor, enhancing the efficiency of FXa inhibition. TFPI can also inhibit prothrombinase assembly by directly interacting with coagulation factor V (FV), which has been activated by FXa. Because full-length TFPI associates with FV in plasma, we hypothesized that FV may influence TFPI inhibitory function. Using pure component FXa inhibition assays, we found that although FV alone did not influence TFPI-mediated FXa inhibition, it further enhanced TFPI in the presence of protein S, resulting in an ∼8-fold reduction in Ki compared with TFPI alone. A FV variant (R709Q/R1018Q/R1545Q, FVΔIIa) that cannot be cleaved/activated by thrombin or FXa also enhanced TFPI-mediated inhibition of FXa ∼12-fold in the presence of protein S. In contrast, neither activated FV nor recombinant B-domain-deleted FV could enhance TFPI-mediated inhibition of FXa in the presence of protein S, suggesting a functional contribution of the B domain. Using TFPI and protein S variants, we show further that the enhancement of TFPI-mediated FXa inhibition by protein S and FV depends on a direct protein S/TFPI interaction and that the TFPI C-terminal tail is not essential for this enhancement. In FXa-catalyzed prothrombin activation assays, both FV and FVΔIIa (but not activated FV) enhanced TFPI function in the presence of protein S. These results demonstrate a new anticoagulant (cofactor) function of FV that targets the early phase of coagulation before prothrombinase assembly.
Collapse
Affiliation(s)
- Salvatore Santamaria
- From the Centre for Haematology, Faculty of Medicine, Imperial College London, London W12 0NN, United Kingdom
| | - Natalia Reglińska-Matveyev
- From the Centre for Haematology, Faculty of Medicine, Imperial College London, London W12 0NN, United Kingdom
| | - Magdalena Gierula
- From the Centre for Haematology, Faculty of Medicine, Imperial College London, London W12 0NN, United Kingdom
| | - Rodney M Camire
- the Division of Hematology, Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, and.,the Center for Cell and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104
| | - James T B Crawley
- From the Centre for Haematology, Faculty of Medicine, Imperial College London, London W12 0NN, United Kingdom
| | - David A Lane
- From the Centre for Haematology, Faculty of Medicine, Imperial College London, London W12 0NN, United Kingdom
| | - Josefin Ahnström
- From the Centre for Haematology, Faculty of Medicine, Imperial College London, London W12 0NN, United Kingdom,
| |
Collapse
|
22
|
Chen C, Yang L, Villoutreix BO, Wang X, Ding Q, Rezaie AR. Gly74Ser mutation in protein C causes thrombosis due to a defect in protein S-dependent anticoagulant function. Thromb Haemost 2017; 117:1358-1369. [PMID: 28405673 DOI: 10.1160/th17-01-0043] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 03/25/2017] [Indexed: 12/30/2022]
Abstract
Protein C is a vitamin K-dependent serine protease zymogen in plasma which upon activation by thrombin in complex with thrombomodulin (TM) down-regulates the clotting cascade by a feedback loop inhibition mechanism. Activated protein C (APC) exerts its anticoagulant function through protein S-dependent degradation of factors Va and VIIIa. We recently identified a venous thrombosis patient whose plasma level of protein C antigen is normal, but its anticoagulant activity is only 34 % of the normal level. Genetic analysis revealed that the proband and her younger brother carry a novel heterozygous mutation c.346G>A, p.Gly74Ser (G74S) in PROC. Thrombin generation assay indicated that the TM-dependent anticoagulant activity of the proband's plasma has been significantly impaired. We expressed protein C-G74S in mammalian cells and characterised its properties in established coagulation assays. We demonstrate that the protein C variant can be normally activated by the thrombin-TM complex and the resulting APC mutant also exhibits normal amidolytic and proteolytic activities toward both FVa and FVIIIa. However, it was discovered the protein S-dependent catalytic activity of APC variant toward both procoagulant cofactors has been significantly impaired. Protein S concentration-dependence of FVa degradation revealed that the capacity of APC variant to interact with the cofactor has been markedly impaired. The same results were obtained for inactivation of FVa-Leiden suggesting that the protein S-dependent activity of APC variant toward cleavage of Arg-306 site has been adversely affected. These results provide insight into the mechanism through which G74S substitution in APC causes thrombosis in the proband carrying this mutation.
Collapse
Affiliation(s)
| | | | | | | | | | - Alireza R Rezaie
- Alireza R. Rezaie, PhD, Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA, Tel: +1 405 271 4711, E-mail: , or, Qiulan Ding, PhD, Department of Laboratory Medicine, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, No. 197 Ruijin Second Road, Shanghai, 200025 China, Tel.: +86 21 54667770, Fax: +86 21 64333548, E-mail:
| |
Collapse
|
23
|
Yasuma T, Yano Y, D'Alessandro-Gabazza CN, Toda M, Gil-Bernabe P, Kobayashi T, Nishihama K, Hinneh JA, Mifuji-Moroka R, Roeen Z, Morser J, Cann I, Motoh I, Takei Y, Gabazza EC. Amelioration of Diabetes by Protein S. Diabetes 2016; 65:1940-51. [PMID: 27207541 DOI: 10.2337/db15-1404] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2015] [Accepted: 04/04/2016] [Indexed: 11/13/2022]
Abstract
Protein S is an anticoagulant factor that also regulates inflammation and cell apoptosis. The effect of protein S on diabetes and its complications is unknown. This study compared the development of diabetes between wild-type and transgenic mice overexpressing human protein S and the development of diabetic glomerulosclerosis between mice treated with and without human protein S and between wild-type and protein S transgenic mice. Mice overexpressing protein S showed significant improvements in blood glucose level, glucose tolerance, insulin sensitivity, and insulin secretion compared with wild-type counterparts. Exogenous protein S improved insulin sensitivity in adipocytes, skeletal muscle, and liver cell lines in db/db mice compared with controls. Significant inhibition of apoptosis with increased expression of BIRC3 and Bcl-2 and enhanced activation of Akt/PKB was induced by protein S in islet β-cells compared with controls. Diabetic wild-type mice treated with protein S and diabetic protein S transgenic mice developed significantly less severe diabetic glomerulosclerosis than controls. Patients with type 2 diabetes had significantly lower circulating free protein S than healthy control subjects. This study shows that protein S attenuates diabetes by inhibiting apoptosis of β-cells and the development of diabetic nephropathy.
Collapse
Affiliation(s)
- Taro Yasuma
- Department of Diabetes, Metabolism and Endocrinology, Mie University Graduate School of Medicine, Edobashi, Japan
| | - Yutaka Yano
- Department of Diabetes, Metabolism and Endocrinology, Mie University Graduate School of Medicine, Edobashi, Japan
| | | | - Masaaki Toda
- Department of Immunology, Mie University Graduate School of Medicine, Edobashi, Japan
| | - Paloma Gil-Bernabe
- Department of Immunology, Mie University Graduate School of Medicine, Edobashi, Japan
| | - Tetsu Kobayashi
- Department of Pulmonary and Critical Care Medicine, Mie University Graduate School of Medicine, Edobashi, Japan
| | - Kota Nishihama
- Department of Diabetes, Metabolism and Endocrinology, Mie University Graduate School of Medicine, Edobashi, Japan
| | - Josephine A Hinneh
- Department of Immunology, Mie University Graduate School of Medicine, Edobashi, Japan
| | - Rumi Mifuji-Moroka
- Department of Gastroenterology and Hepatology, Mie University Graduate School of Medicine, Edobashi, Japan
| | - Ziaurahman Roeen
- Department of Immunology, Mie University Graduate School of Medicine, Edobashi, Japan
| | - John Morser
- Division of Hematology, Stanford School of Medicine, Stanford, CA
| | - Isaac Cann
- Carl R. Woese Institute for Genomic Biology Institute for Genomic Biology and Department of Animal Sciences and Department of Microbiology, University of Illinois at Urbana-Champaign, Urbana, IL
| | - Iwasa Motoh
- Department of Immunology, Mie University Graduate School of Medicine, Edobashi, Japan Department of Gastroenterology and Hepatology, Mie University Graduate School of Medicine, Edobashi, Japan
| | - Yoshiyuki Takei
- Department of Diabetes, Metabolism and Endocrinology, Mie University Graduate School of Medicine, Edobashi, Japan Department of Pulmonary and Critical Care Medicine, Mie University Graduate School of Medicine, Edobashi, Japan Department of Gastroenterology and Hepatology, Mie University Graduate School of Medicine, Edobashi, Japan
| | - Esteban C Gabazza
- Department of Immunology, Mie University Graduate School of Medicine, Edobashi, Japan
| |
Collapse
|
24
|
Ogbechi J, Ruf MT, Hall BS, Bodman-Smith K, Vogel M, Wu HL, Stainer A, Esmon CT, Ahnström J, Pluschke G, Simmonds RE. Mycolactone-Dependent Depletion of Endothelial Cell Thrombomodulin Is Strongly Associated with Fibrin Deposition in Buruli Ulcer Lesions. PLoS Pathog 2015; 11:e1005011. [PMID: 26181660 PMCID: PMC4504485 DOI: 10.1371/journal.ppat.1005011] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 06/07/2015] [Indexed: 01/21/2023] Open
Abstract
A well-known histopathological feature of diseased skin in Buruli ulcer (BU) is coagulative necrosis caused by the Mycobacterium ulcerans macrolide exotoxin mycolactone. Since the underlying mechanism is not known, we have investigated the effect of mycolactone on endothelial cells, focussing on the expression of surface anticoagulant molecules involved in the protein C anticoagulant pathway. Congenital deficiencies in this natural anticoagulant pathway are known to induce thrombotic complications such as purpura fulimans and spontaneous necrosis. Mycolactone profoundly decreased thrombomodulin (TM) expression on the surface of human dermal microvascular endothelial cells (HDMVEC) at doses as low as 2ng/ml and as early as 8hrs after exposure. TM activates protein C by altering thrombin’s substrate specificity, and exposure of HDMVEC to mycolactone for 24 hours resulted in an almost complete loss of the cells’ ability to produce activated protein C. Loss of TM was shown to be due to a previously described mechanism involving mycolactone-dependent blockade of Sec61 translocation that results in proteasome-dependent degradation of newly synthesised ER-transiting proteins. Indeed, depletion from cells determined by live-cell imaging of cells stably expressing a recombinant TM-GFP fusion protein occurred at the known turnover rate. In order to determine the relevance of these findings to BU disease, immunohistochemistry of punch biopsies from 40 BU lesions (31 ulcers, nine plaques) was performed. TM abundance was profoundly reduced in the subcutis of 78% of biopsies. Furthermore, it was confirmed that fibrin deposition is a common feature of BU lesions, particularly in the necrotic areas. These findings indicate that there is decreased ability to control thrombin generation in BU skin. Mycolactone’s effects on normal endothelial cell function, including its ability to activate the protein C anticoagulant pathway are strongly associated with this. Fibrin-driven tissue ischemia could contribute to the development of the tissue necrosis seen in BU lesions. Buruli ulcer (BU) is a neglected tropical disease that is most common in West Africa and parts of Australia, but has been reported from over 30 countries worldwide. The symptoms are painless ulcers of the skin caused by a bacterial infection. The bacteria, Mycobacterium ulcerans, produce a macrolide toxin called mycolactone. In this manuscript, we have studied the effects of mycolactone on endothelial cells, specialised cells that line blood vessels and form capillaries. One of the most important functions of these cells is to prevent blood from clotting inside the vessels. We show that mycolactone reduces the ability of cultured endothelial cells to anticoagulate blood, by blocking the expression of a protein called thrombomodulin. We went on to examine samples of BU patient skin and found that thrombomodulin is also reduced here, and that in contrast to normal skin large amounts of fibrin (one of the main constituents of blood clots) were present. This means that it may be useful to consider whether anticoagulants might improve the response to antibiotics and thereby improve treatment outcomes for BU patients.
Collapse
Affiliation(s)
- Joy Ogbechi
- Department of Microbial and Cellular Sciences, School of Biosciences and Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom
| | - Marie-Thérèse Ruf
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Belinda S. Hall
- Department of Microbial and Cellular Sciences, School of Biosciences and Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom
| | - Katherine Bodman-Smith
- Department of Microbial and Cellular Sciences, School of Biosciences and Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom
| | - Moritz Vogel
- Section Clinical Tropical Medicine, Heidelberg University Hospital, Heidelberg, Germany
| | - Hua-Lin Wu
- Department of Biochemistry and Molecular Biology, National Cheng Kung University, Tainan, Taiwan
| | - Alexander Stainer
- Institute for Cardiovascular and Metabolic Research, School of Biological Sciences, University of Reading, Reading, United Kingdom
| | - Charles T. Esmon
- Coagulation Biology Laboratory, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, United States of America
| | - Josefin Ahnström
- Centre for Haematology, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Gerd Pluschke
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Rachel E. Simmonds
- Department of Microbial and Cellular Sciences, School of Biosciences and Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom
- * E-mail:
| |
Collapse
|
25
|
Studer RA, Opperdoes FR, Nicolaes GAF, Mulder AB, Mulder R. Understanding the functional difference between growth arrest-specific protein 6 and protein S: an evolutionary approach. Open Biol 2015; 4:rsob.140121. [PMID: 25339693 PMCID: PMC4221892 DOI: 10.1098/rsob.140121] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Although protein S (PROS1) and growth arrest-specific protein 6 (GAS6) proteins are homologous with a high degree of structural similarity, they are functionally different. The objectives of this study were to identify the evolutionary origins from which these functional differences arose. Bioinformatics methods were used to estimate the evolutionary divergence time and to detect the amino acid residues under functional divergence between GAS6 and PROS1. The properties of these residues were analysed in the light of their three-dimensional structures, such as their stability effects, the identification of electrostatic patches and the identification potential protein-protein interaction. The divergence between GAS6 and PROS1 probably occurred during the whole-genome duplications in vertebrates. A total of 78 amino acid sites were identified to be under functional divergence. One of these sites, Asn463, is involved in N-glycosylation in GAS6, but is mutated in PROS1, preventing this post-translational modification. Sites experiencing functional divergence tend to express a greater diversity of stabilizing/destabilizing effects than sites that do not experience such functional divergence. Three electrostatic patches in the LG1/LG2 domains were found to differ between GAS6 and PROS1. Finally, a surface responsible for protein-protein interactions was identified. These results may help researchers to analyse disease-causing mutations in the light of evolutionary and structural constraints, and link genetic pathology to clinical phenotypes.
Collapse
Affiliation(s)
- Romain A Studer
- European Molecular Biology Laboratory-European Bioinformatics Institute (EMBL-EBI), Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| | - Fred R Opperdoes
- Laboratory of Biochemistry, de Duve Institute and Université catholique de Louvain, Brussels 1200, Belgium
| | - Gerry A F Nicolaes
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands
| | - André B Mulder
- Department of Laboratory Medicine, University Medical Centre Groningen, Groningen, The Netherlands
| | - René Mulder
- Department of Laboratory Medicine, University Medical Centre Groningen, Groningen, The Netherlands
| |
Collapse
|
26
|
Andreou AP, Efthymiou M, Yu Y, Watts HR, Noormohamed FH, Ma D, Lane DA, Crawley JTB. Protective effects of non-anticoagulant activated protein C variant (D36A/L38D/A39V) in a murine model of ischaemic stroke. PLoS One 2015; 10:e0122410. [PMID: 25830552 PMCID: PMC4382112 DOI: 10.1371/journal.pone.0122410] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Accepted: 02/20/2015] [Indexed: 11/30/2022] Open
Abstract
Ischaemic stroke is caused by occlusive thrombi in the cerebral vasculature. Although tissue-plasminogen activator (tPA) can be administered as thrombolytic therapy, it has major limitations, which include disruption of the blood-brain barrier and an increased risk of bleeding. Treatments that prevent or limit such deleterious effects could be of major clinical importance. Activated protein C (APC) is a natural anticoagulant that regulates thrombin generation, but also confers endothelial cytoprotective effects and improved endothelial barrier function mediated through its cell signalling properties. In murine models of stroke, although APC can limit the deleterious effects of tPA due to its cell signalling function, its anticoagulant actions can further elevate the risk of bleeding. Thus, APC variants such as APC(5A), APC(Ca-ins) and APC(36-39) with reduced anticoagulant, but normal signalling function may have therapeutic benefit. Human and murine protein C (5A), (Ca-ins) and (36-39) variants were expressed and characterised. All protein C variants were secreted normally, but 5-20% of the protein C (Ca-ins) variants were secreted as disulphide-linked dimers. Thrombin generation assays suggested reductions in anticoagulant function of 50- to 57-fold for APC(36-39), 22- to 27-fold for APC(Ca-ins) and 14- to 17-fold for APC(5A). Interestingly, whereas human wt APC, APC(36-39) and APC(Ca-ins) were inhibited similarly by protein C inhibitor (t½ - 33 to 39 mins), APC(5A) was inactivated ~9-fold faster (t½ - 4 mins). Using the murine middle cerebral artery occlusion ischaemia/repurfusion injury model, in combination with tPA, APC(36-39), which cannot be enhanced by its cofactor protein S, significantly improved neurological scores, reduced cerebral infarct area by ~50% and reduced oedema ratio. APC(36-39) also significantly reduced bleeding in the brain induced by administration of tPA, whereas wt APC did not. If our data can be extrapolated to clinical settings, then APC(36-39) could represent a feasible adjunctive therapy for ischaemic stroke.
Collapse
Affiliation(s)
- Anna P. Andreou
- Centre for Haematology, Imperial College London, London, United Kingdom
- Section of Anaesthetics, Pain Medicine & Intensive Care, Imperial College London, London, United Kingdom
- * E-mail:
| | - Maria Efthymiou
- Centre for Haematology, Imperial College London, London, United Kingdom
| | - Yao Yu
- Centre for Haematology, Imperial College London, London, United Kingdom
| | - Helena R. Watts
- Section of Anaesthetics, Pain Medicine & Intensive Care, Imperial College London, London, United Kingdom
| | - Faruq H. Noormohamed
- Section of Anaesthetics, Pain Medicine & Intensive Care, Imperial College London, London, United Kingdom
| | - Daqing Ma
- Section of Anaesthetics, Pain Medicine & Intensive Care, Imperial College London, London, United Kingdom
| | - David A. Lane
- Centre for Haematology, Imperial College London, London, United Kingdom
| | - James TB Crawley
- Centre for Haematology, Imperial College London, London, United Kingdom
| |
Collapse
|
27
|
Somajo S, Ahnström J, Fernandez-Recio J, Gierula M, Villoutreix BO, Dahlbäck B. Amino acid residues in the laminin G domains of protein S involved in tissue factor pathway inhibitor interaction. Thromb Haemost 2015; 113:976-87. [PMID: 25716664 DOI: 10.1160/th14-09-0803] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Accepted: 01/05/2015] [Indexed: 11/05/2022]
Abstract
Protein S functions as a cofactor for tissue factor pathway inhibitor (TFPI) and activated protein C (APC). The sex hormone binding globulin (SHBG)-like region of protein S, consisting of two laminin G-like domains (LG1 and LG2), contains the binding site for C4b-binding protein (C4BP) and TFPI. Furthermore, the LG-domains are essential for the TFPI-cofactor function and for expression of full APC-cofactor function. The aim of the current study was to localise functionally important interaction sites in the protein S LG-domains using amino acid substitutions. Four protein S variants were created in which clusters of surface-exposed amino acid residues within the LG-domains were substituted. All variants bound normally to C4BP and were fully functional as cofactors for APC in plasma and in pure component assays. Two variants, SHBG2 (E612A, I614A, F265A, V393A, H453A), involving residues from both LG-domains, and SHBG3 (K317A, I330A, V336A, D365A) where residues in LG1 were substituted, showed 50-60 % reduction in enhancement of TFPI in FXa inhibition assays. For SHBG3 the decreased TFPI cofactor function was confirmed in plasma based thrombin generation assays. Both SHBG variants bound to TFPI with decreased affinity in surface plasmon resonance experiments. The TFPI Kunitz 3 domain is known to contain the interaction site for protein S. Using in silico analysis and protein docking exercises, preliminary models of the protein S SHBG/TFPI Kunitz domain 3 complex were created. Based on a combination of experimental and in silico data we propose a binding site for TFPI on protein S, involving both LG-domains.
Collapse
Affiliation(s)
| | | | | | | | | | - Björn Dahlbäck
- Björn Dahlbäck MD, PhD, Professor of Blood Coagulation Research, Lund University, Department of Translational Medicine, Division of Clinical Chemistry, Wallenberg laboratory, floor 6, University Hospital, Malmö, S-20502 Malmö, Sweden, E-mail:
| |
Collapse
|
28
|
Livaja Koshiar R, Somajo S, Norström E, Dahlbäck B. Erythrocyte-derived microparticles supporting activated protein C-mediated regulation of blood coagulation. PLoS One 2014; 9:e104200. [PMID: 25136857 PMCID: PMC4138094 DOI: 10.1371/journal.pone.0104200] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Accepted: 07/07/2014] [Indexed: 12/30/2022] Open
Abstract
Elevated levels of erythrocyte-derived microparticles are present in the circulation in medical conditions affecting the red blood cells. Erythrocyte-derived microparticles expose phosphatidylserine thus providing a suitable surface for procoagulant reactions leading to thrombin formation via the tenase and prothrombinase complexes. Patients with elevated levels of circulating erythrocyte-derived microparticles have increased thrombin generation in vivo. The aim of the present study was to investigate whether erythrocyte-derived microparticles are able to support the anticoagulant reactions of the protein C system. Erythrocyte-derived microparticles were isolated using ultracentrifugation after incubation of freshly prepared erythrocytes with the ionophore A23187 or from outdated erythrocyte concentrates, the different microparticles preparations yielding similar results. According to flow cytometry analysis, the microparticles exposed phoshatidylserine and bound lactadherin, annexin V, and protein S, which is a cofactor to activated protein C. The microparticles were able to assemble the tenase and prothrombinase complexes and to stimulate the formation of thrombin in plasma-based thrombin generation assay both in presence and absence of added tissue factor. The addition of activated protein C in the thrombin generation assay inhibited thrombin generation in a dose-dependent fashion. The anticoagulant effect of activated protein C in the thrombin generation assay was inhibited by a monoclonal antibody that prevents binding of protein S to microparticles and also attenuated by anti-TFPI antibodies. In the presence of erythrocyte-derived microparticles, activated protein C inhibited tenase and prothrombinase by degrading the cofactors FVIIIa and FVa, respectively. Protein S stimulated the Arg306-cleavage in FVa, whereas efficient inhibition of FVIIIa depended on the synergistic cofactor activity of protein S and FV. In summary, the erythrocyte-derived microparticle surface is suitable for the anticoagulant reactions of the protein C system, which may be important to balance the initiation and propagation of coagulation in vivo.
Collapse
Affiliation(s)
- Ruzica Livaja Koshiar
- Department of Laboratory Medicine, Division of Clinical Chemistry, Lund University, Skåne University Hospital, Malmö, Sweden
| | - Sofia Somajo
- Department of Laboratory Medicine, Division of Clinical Chemistry, Lund University, Skåne University Hospital, Malmö, Sweden
| | - Eva Norström
- Department of Laboratory Medicine, Division of Clinical Chemistry, Lund University, Skåne University Hospital, Malmö, Sweden
| | - Björn Dahlbäck
- Department of Laboratory Medicine, Division of Clinical Chemistry, Lund University, Skåne University Hospital, Malmö, Sweden
- * E-mail:
| |
Collapse
|
29
|
Reglińska-Matveyev N, Andersson HM, Rezende SM, Dahlbäck B, Crawley JTB, Lane DA, Ahnström J. TFPI cofactor function of protein S: essential role of the protein S SHBG-like domain. Blood 2014; 123:3979-87. [PMID: 24740810 PMCID: PMC4064334 DOI: 10.1182/blood-2014-01-551812] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Accepted: 04/09/2014] [Indexed: 01/17/2023] Open
Abstract
Protein S is a cofactor for tissue factor pathway inhibitor (TFPI), accelerating the inhibition of activated factor X (FXa). TFPI Kunitz domain 3 residue Glu226 is essential for enhancement of TFPI by protein S. To investigate the complementary functional interaction site on protein S, we screened 44 protein S point, composite or domain swap variants spanning the whole protein S molecule for their TFPI cofactor function using a thrombin generation assay. Of these variants, two protein S/growth arrest-specific 6 chimeras, with either the whole sex hormone-binding globulin (SHBG)-like domain (Val243-Ser635; chimera III) or the SHBG laminin G-type 1 subunit (Ser283-Val459; chimera I), respectively, substituted by the corresponding domain in growth arrest-specific 6, were unable to enhance TFPI. The importance of the protein S SHBG-like domain (and its laminin G-type 1 subunit) for binding and enhancement of TFPI was confirmed in FXa inhibition assays and using surface plasmon resonance. In addition, protein S bound to C4b binding protein showed greatly reduced enhancement of TFPI-mediated inhibition of FXa compared with free protein S. We show that binding of TFPI to the protein S SHBG-like domain enables TFPI to interact optimally with FXa on a phospholipid membrane.
Collapse
Affiliation(s)
| | - Helena M Andersson
- Centre for Haematology, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Suely M Rezende
- Department of Internal Medicine, Faculty of Medicine, Universidade Federal de Minas Gerais, Minas Gerais, Brazil; and
| | - Björn Dahlbäck
- Department of Laboratory Medicine, Faculty of Medicine, Lund University, Malmö, Sweden
| | - James T B Crawley
- Centre for Haematology, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - David A Lane
- Centre for Haematology, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Josefin Ahnström
- Centre for Haematology, Faculty of Medicine, Imperial College London, London, United Kingdom
| |
Collapse
|
30
|
Abstract
Bone morphogenetic protein and activin membrane-bound inhibitor (BAMBI) is a transmembrane protein related to the transforming growth factor-β superfamily, and is highly expressed in platelets and endothelial cells. We previously demonstrated its positive role in thrombus formation using a zebrafish thrombosis model. In the present study, we used Bambi-deficient mice and radiation chimeras to evaluate the function of this receptor in the regulation of both hemostasis and thrombosis. We show that Bambi(-/-) and Bambi(+/-) mice exhibit mildly prolonged bleeding times compared with Bambi(+/+) littermates. In addition, using 2 in vivo thrombosis models in mesenterium or cremaster muscle arterioles, we demonstrate that Bambi-deficient mice form unstable thrombi compared with Bambi(+/+) mice. No defects in thrombin generation in Bambi(-/-) mouse plasma could be detected ex vivo. Moreover, the absence of BAMBI had no effect on platelet counts, platelet activation, aggregation, or platelet procoagulant function. Similar to Bambi(-/-) mice, Bambi(-/-) transplanted with Bambi(+/+) bone marrow formed unstable thrombi in the laser-induced thrombosis model that receded more rapidly than thrombi that formed in Bambi(+/+) mice receiving Bambi(-/-) bone marrow transplants. Taken together, these results provide strong evidence for an important role of endothelium rather than platelet BAMBI as a positive regulator of both thrombus formation and stability.
Collapse
|
31
|
Yu Y, Millar CM. Measurement of factor IX activity in plasma-derived and recombinant concentrates: insights from thrombin generation and activation-based assays. J Thromb Haemost 2014; 12:62-70. [PMID: 24215160 DOI: 10.1111/jth.12452] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2013] [Indexed: 11/30/2022]
Abstract
BACKGROUND Hemophilia B, resulting from a deficiency of coagulation factor IX, is treated effectively with either recombinant FIX (r-FIX) or plasma-derived FIX (pd-FIX) concentrates, although differences in pharmacokinetics are observed. FIX is activated in vivo by both activated FXI (FXIa) and tissue factor (TF)-activated FVII (FVIIa); however, conventional activated partial thromboplastin time (APTT)-based assays assess only activation by FXIa. OBJECTIVES To examine the differences between pd-FIX and r-FIX concentrates with respect to their thrombogenicity and activation. METHODS AND RESULTS FIX ELISA was used to quantify antigenic FIX. Calibrated automated thrombography was performed to evaluate the effect of FIX on thrombin generation. FIXa was quantified by the cleavage of FIXa-specific chromogenic substrate. FIX activation was studied in a purified system. RESULTS We found that r-FIX had ~ 1.6-fold greater specific activity than pd-FIX. r-FIX generated a markedly higher thrombin peak than pd-FIX at an equivalent antigen level when coagulation was initiated by TF, but this was not seen in contact activation-triggered thrombin generation (TG). Interestingly, the amount of FIXa in r-FIX concentrate was 10 times higher than that in pd-FIX concentrate. In a purified system, the amount of r-FIXa generated by FXIa in the first 10 min of activation was 1.37-fold that of pd-FIXa, whereas no difference between the concentrates was observed when triggered by TF-FVIIa. CONCLUSIONS Clear differences were observed between pd-FIX and r-FIX concentrates, including the proportion of FIXa and the activation by FXIa. These may explain some of the discrepancies observed clinically, and suggest that the APTT may not reflect their resultant in vivo properties.
Collapse
Affiliation(s)
- Y Yu
- Centre for Haematology, Department of Medicine, Imperial College London, London, UK
| | | |
Collapse
|
32
|
Heeb MJ, Mesters RM, Fernández JA, Hackeng TM, Nakasone RK, Griffin JH. Plasma protein S residues 37-50 mediate its binding to factor Va and inhibition of blood coagulation. Thromb Haemost 2013; 110:275-82. [PMID: 23892573 DOI: 10.1160/th12-12-0953] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2012] [Accepted: 05/04/2013] [Indexed: 11/05/2022]
Abstract
Protein S (PS) is an anticoagulant plasma protein whose deficiency is associated with increased risk of venous thrombosis. PS directly inhibits thrombin generation by the blood coagulation pathways by several mechanisms, including by binding coagulation factors (F) Va and Xa. To identify PS sequences that mediate inhibition of FVa activity, antibodies and synthetic peptides based on PS sequence were prepared and employed in plasma coagulation assays, purified component prothrombinase assays, binding assays, and immunoblots. In the absence of activated protein C, monoclonal antibody (Mab) S4 shortened FXa-induced clotting in normal plasma but not in PS-depleted plasma. Mab S4 also blocked PS inhibition of FVa-dependent prothrombinase activity in purified component assays in the absence or presence of phospholipids and inhibited binding of PS to immobilised FVa. Epitope mapping identified N-terminal region residues 37-67 of PS as this antibody's epitope. A peptide representing PS residues 37-50 inhibited FVa-dependent prothrombinase activity in a non-competitive manner, with 50% inhibition observed at 11 µM peptide, whereas a peptide with a D-amino acid sequence of 37-50 was ineffective. FVa, but not FXa, bound specifically to the immobilised peptide representing residues 37-50, and the peptide inhibited binding of FVa to immobilised PS. These data implicate PS residues 37-50 as a binding site for FVa that mediates, at least in part, the direct inhibition of FVa-dependent procoagulant activity by PS.
Collapse
Affiliation(s)
- Mary J Heeb
- Department of Molecular and Experimental Medicine, Scripps Research Institute, La Jolla, CA 92037, USA.
| | | | | | | | | | | |
Collapse
|
33
|
Stavenuiter F, Gale AJ, Heeb MJ. Phosphorylation of protein S by platelet kinases enhances its activated protein C cofactor activity. FASEB J 2013; 27:2918-25. [PMID: 23580615 DOI: 10.1096/fj.12-225961] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Protein S (PS) is a multifunctional plasma protein of the hemostatic and inflammatory pathways, although mechanisms for its regulation are poorly understood. Since certain plasma proteins are regulated through extracellular phosphorylation, we investigated whether the anticoagulant activity of PS is regulated through phosphorylation by platelet-secreted kinases. PS was phosphorylated on exposure to activated platelets or their releasates, as judged by immunoblotting for phospho-amino acids and PS. PS phosphorylation was reduced by specific inhibitors of casein kinase 1 (CK1) and casein kinase 2 (CK2) (10 μM D4476, 100 μM CK2-inhibitory peptide YNLKSKSSEDIDESS). Involvement of CKs in PS phosphorylation was confirmed using purified CK1/CK2. Phosphorylation of PS by purified CK1 did not affect its activated protein C (APC) cofactor activity in activated partial thromboplastin time assays in PS-depleted plasma. However, phosphorylation of PS by CK2 or by CK1/CK2 increased PS cofactor activity ∼1.5-fold (158.7±4.8%, P<0.01) or ∼2-fold (191.5±6.4%, P<0.0001), respectively. The APC cofactor activity of PS in PS-depleted plasma exposed to platelet-secreted kinases was enhanced, while CK2 but not CK1 inhibitors reduced APC cofactor activity. Mass spectrometry revealed a phosphorylated CK2 site at Thr37 within the N-terminal Gla-domain. Thus, platelet-mediated extracellular phosphorylation of PS is a potential mechanism by which its activity is regulated.
Collapse
Affiliation(s)
- Fabian Stavenuiter
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, 10550 N. Torrey Pines Rd, La Jolla, CA 92037, USA
| | | | | |
Collapse
|
34
|
Identification of functionally important residues in TFPI Kunitz domain 3 required for the enhancement of its activity by protein S. Blood 2012; 120:5059-62. [DOI: 10.1182/blood-2012-05-432005] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
Protein S is a cofactor for tissue factor pathway inhibitor (TFPI) that critically reduces the inhibition constant for FXa to below the plasma concentration of TFPI. TFPI Kunitz domain 3 is required for this enhancement to occur. To delineate the molecular mechanism underlying enhancement of TFPI function, in the present study, we produced a panel of Kunitz domain 3 variants of TFPI encompassing all 12 surface-exposed charged residues. Thrombin-generation assays in TFPI-depleted plasma identified a novel variant, TFPI E226Q, which exhibited minimal enhancement by protein S. This was confirmed in purified FXa inhibition assays in which no protein S enhancement of TFPI E226Q was detected. Surface plasmon resonance demonstrated concentration-dependent binding of protein S to wild-type TFPI, but almost no binding to TFPI E226Q. We conclude that the TFPI Kunitz domain 3 residue Glu226 is essential for TFPI enhancement by protein S.
Collapse
|