1
|
Syed Mohammed RD, Gutierrez Luque L, Maurer MC. Factor XIII Activation Peptide Residues Play Important Roles in Stability, Activation, and Transglutaminase Activity. Biochemistry 2024; 63:2830-2841. [PMID: 39422351 DOI: 10.1021/acs.biochem.4c00318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
A subunit of factor XIII (FXIII-A) contains a unique activation peptide (AP) that protects the catalytic triad and prevents degradation. In plasma, FXIII is activated proteolytically (FXIII-A*) by thrombin and Ca2+ cleaving AP, while in cytoplasm, it is activated nonproteolytically (FXIII-A°) with increased Ca2+ concentrations. This study aimed to elucidate the role of individual parts of the FXIII-A AP in protein stability, thrombin activation, and transglutaminase activity. Recombinant FXIII-A AP variants were expressed, and SDS-PAGE was used to monitor thrombin hydrolysis at the AP cleavage sites R37-G38. Transglutaminase activities were assessed by cross-linking lysine mimics to Fbg αC (233-425, glutamine-substrate) and monitoring reactions by mass spectrometry and in-gel fluorescence assays. FXIII-A AP variants, S19P, E23K, and D24V, degraded during purification, indicating their vital role in FXIII-A2 stability. Mutation of P36 to L36/F36 abolished the proteolytic cleavage of AP and thus prevented activation. FXIII-A N20S and P27L exhibited slower thrombin activation, likely due to the loss of key interdomain H-bonding interactions. Except N20S and P15L/P16L, all activatable FXIII-A* variants (P15L, P16L, S19A, and P27L) showed similar cross-linking activity to WT. By contrast, FXIII-A° P15L, P16L, and P15L/P16L had significantly lower cross-linking activity than FXIII-A° WT, suggesting that loss of these prolines had a greater structural impact. In conclusion, FXIII-A AP residues that play crucial roles in FXIII-A stability, activation, and activity were identified. The interactions between these AP amino acid residues and other domains control the stability and activity of FXIII.
Collapse
Affiliation(s)
- Rameesa D Syed Mohammed
- Department of Chemistry, University of Louisville, Louisville, Kentucky 40292, United States
| | - Lianay Gutierrez Luque
- Department of Chemistry, University of Louisville, Louisville, Kentucky 40292, United States
| | - Muriel C Maurer
- Department of Chemistry, University of Louisville, Louisville, Kentucky 40292, United States
| |
Collapse
|
2
|
Feller T, McPherson HR, Connell SD, Ariëns RAS. Fibrinogen αC-region acts as a functional safety latch: Implications for a fibrin biomechanical behaviour model. Acta Biomater 2024:S1742-7061(24)00588-9. [PMID: 39393660 DOI: 10.1016/j.actbio.2024.10.005] [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: 04/17/2024] [Revised: 10/02/2024] [Accepted: 10/03/2024] [Indexed: 10/13/2024]
Abstract
Fibrin has unique biomechanical properties which are essential for its role as a scaffold for blood clots. Fibrin is highly extensible and demonstrates significant strain stiffening behaviour, which is essential for stress-distribution in the network. Yet the exact structures of fibrin at the sub-fibre level that contribute to its unique biomechanical characteristic are unknown. Here we show how truncations of the fibrinogen αC-region impact the biomechanical properties of fibrin fibres. Surprisingly, absence of the complete αC-region did not influence the low strain modulus of fibrin fibres but led to premature fibre rupture and decreased extensibility. Intermediate effects were observed with partial deletion of the αC-region, reflected by intermediate rupture stress and toughness. However, overall strain-stiffening behaviour remained even in absence of the αC-region, indicating that strain stiffening is not due to stress being transferred from the αC-region to the protofibril backbone. Upon stress-relaxation, decay constants and their relative contribution to the total relaxation remained similar at all strains, showing that a distinct relaxation process is present until fibre rupture. However, relative contribution of fast relaxation was maximal only in crosslinked fibres if the flexible αC-connector was present. These data show that the αC-region is not the main load-bearing structure within fibrin fibres and point to a critical role for the protofibril backbone instead. We present a revised structural model based on protofibril branching that fully explains the unique biomechanical behaviour of fibrin fibres, while the αC-region primarily acts as a safety latch at the highest of strains. STATEMENT OF SIGNIFICANCE: The findings presented in this paper reveal critically important details about how the molecular structure of fibrin contributes to its unique mechanical properties which are essential to fulfil its function as the scaffold of blood clots. In this work we used engineered proteins with alterations in an important but highly disordered area of the molecule called αC-region and we provide direct evidence for the first time for how the absence of either the globular αC-domain, or the complete αC-region impacts the mechanical behaviour of individual fibrin fibres. Using these results we developed a new structural model of protofibril organisation within fibrin fibres that fully explains their strain stiffening, relatively low modulus and their high, largely variable, extensibility.
Collapse
Affiliation(s)
- Tímea Feller
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, UK; Molecular and Nanoscale Physics Group, School of Physics, University of Leeds, UK.
| | - Helen R McPherson
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, UK
| | - Simon D Connell
- Molecular and Nanoscale Physics Group, School of Physics, University of Leeds, UK
| | - Robert A S Ariëns
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, UK
| |
Collapse
|
3
|
Mohammed RDS, Piell KM, Maurer MC. Identification of Factor XIII β-Sandwich Residues Mediating Glutamine Substrate Binding and Activation Peptide Cleavage. Thromb Haemost 2024; 124:408-422. [PMID: 38040030 DOI: 10.1055/a-2220-7544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2023]
Abstract
BACKGROUND Factor XIII (FXIII) forms covalent crosslinks across plasma and cellular substrates and has roles in hemostasis, wound healing, and bone metabolism. FXIII activity is implicated in venous thromboembolism (VTE) and is a target for developing pharmaceuticals, which requires understanding FXIII - substrate interactions. Previous studies proposed the β-sandwich domain of the FXIII A subunit (FXIII-A) exhibits substrate recognition sites. MATERIAL AND METHODS Recombinant FXIII-A proteins (WT, K156E, F157L, R158Q/E, R171Q, and R174E) were generated to identify FXIII-A residues mediating substrate recognition. Proteolytic (FXIII-A*) and non-proteolytic (FXIII-A°) forms were analyzed for activation and crosslinking activities toward physiological substrates using SDS-PAGE and MALDI-TOF MS. RESULTS All FXIII-A* variants displayed reduced crosslinking abilities compared to WT for Fbg αC (233 - 425), fibrin, and actin. FXIII-A* WT activity was greater than A°, suggesting the binding site is more exposed in FXIII-A*. With Fbg αC (233 - 425), FXIII-A* variants R158Q/E, R171Q, and R174E exhibited decreased activities approaching those of FXIII-A°. However, with a peptide substrate, FXIII-A* WT and variants showed similar crosslinking suggesting the recognition site is distant from the catalytic site. Surprisingly, FXIII-A R158E and R171Q displayed slower thrombin activation than WT, potentially due to loss of crucial H-bonding with neighboring activation peptide (AP) residues. CONCLUSION In conclusion, FXIII-A residues K156, F157, R158, R171, and R174 are part of a binding site for physiological substrates [fibrin (α and γ) and actin]. Moreover, R158 and R171 control AP cleavage during thrombin activation. These investigations provide new molecular details on FXIII - substrate interactions that control crosslinking abilities.
Collapse
Affiliation(s)
| | - Kellianne M Piell
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, Kentucky, United States
| | - Muriel C Maurer
- Department of Chemistry, University of Louisville, Louisville, Kentucky, United States
| |
Collapse
|
4
|
Zhang P, Zhang R, Jing C. Abnormal bleeding after lumbar vertebrae surgery because of acquired factor XIII deficiency: A case report and literature review. Medicine (Baltimore) 2024; 103:e36944. [PMID: 38215106 PMCID: PMC10783347 DOI: 10.1097/md.0000000000036944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 10/12/2023] [Indexed: 01/14/2024] Open
Abstract
RATIONALE Abnormal bleeding due to low fibrinogen (Fib) and coagulation factor XIII (FXIII) levels after lumbar vertebral surgery is exceedingly rare. Excessive bleeding is also associated with secondary hyperfibrinolysis. This report presents a case of abnormal incision bleeding caused by coagulation factor XIII deficiency (FXIIID) and secondary hyperfibrinolysis in a state of low fibrinogen after lumbar vertebral surgery. PATIENT CONCERNS A middle-aged woman experienced prolonged incision and excessive bleeding after lumbar vertebral surgery. DIAGNOSIS Combined with coagulation factors, coagulation function tests, and thromboelastography, the patient clinical presentation supported the diagnosis of FXIIID and secondary hyperfibrinolysis in a hypofibrinogenemic state. INTERVENTIONS Cryoprecipitat, Fresh Frozen Plasma, Fibrinogen Concentrate, Leukocyte-depleted Red Blood Cells, Hemostatic (Carbazochrome Sodium Sulfonate; Hemocoagulase Bothrops Atrox for Injection; Tranexamic Acid). OUTCOMES After approximately a month of replacement therapy and symptom treatment, the patient coagulation function significantly improved, and the incision healed without any hemorrhage during follow-up. LESSONS Abnormal postoperative bleeding may indicate coagulation and fibrinolysis disorders that require a full set of coagulation tests, particularly coagulation factors. Given the current lack of a comprehensive approach to detect coagulation and fibrinolysis functions, a more comprehensive understanding of hematology is imperative. The current treatment for FXIIID involves replacement therapy, which requires supplementation with both Fib and FXIII to achieve effective hemostasis.
Collapse
Affiliation(s)
- Peng Zhang
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Ruijing Zhang
- Beijing University of Traditional Chinese Medicine, Beijing, China
| | - Cheng Jing
- The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| |
Collapse
|
5
|
Simpson M, Narwal A, West E, Martin J, Bagot CN, Page AR, Watson HG, Whyte CS, Mutch NJ. Fibrinogenolysis and fibrinolysis in vaccine-induced immune thrombocytopenia and thrombosis. J Thromb Haemost 2023; 21:3589-3596. [PMID: 37734715 DOI: 10.1016/j.jtha.2023.09.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 09/01/2023] [Accepted: 09/06/2023] [Indexed: 09/23/2023]
Abstract
BACKGROUND Vaccine-induced immune thrombocytopenia and thrombosis (VITT) is a rare syndrome associated with adenoviral vector vaccines for COVID-19. The syndrome is characterized by thrombosis, anti-platelet factor 4 (PF4) antibodies, thrombocytopenia, high D-dimer, and hypofibrinogenemia. OBJECTIVES To investigate abnormalities in fibrinolysis that contribute to the clinical features of VITT. METHODS Plasma samples from 18 suspected VITT cases were tested for anti-PF4 by ELISA and characterized as meeting criteria for VITT (11/18) or deemed unlikely (7/18; non-VITT). Antigen levels of PAI-1, factor XIII (FXIII), plasmin-α2antiplasmin (PAP), and inflammatory markers were quantified. Plasmin generation was quantified by chromogenic substrate. Western blotting was performed with antibodies to fibrinogen, FXIII-A, and plasminogen. RESULTS VITT patients 10/11 had scores indicative of overt disseminated intravascular coagulation, while 0/7 non-VITT patients met the criteria. VITT patients had significantly higher levels of inflammatory markers, IL-1β, IL-6, IL-8, TNFα, and C-reactive protein. In VITT patients, both fibrinogen and FXIII levels were significantly lower, while PAP and tPA-mediated plasmin generation were higher compared to non-VITT patients. Evidence of fibrinogenolysis was observed in 9/11 VITT patients but not in non-VITT patients or healthy controls. Fibrinogen degradation products were apparent, with obvious cleavage of the fibrinogen α-chain. PAP complex was evident in those VITT patients with fibrinogenolysis, but not in non-VITT patients or healthy donors. CONCLUSION VITT patients show evidence of overt disseminated intravascular coagulation and fibrinogenolysis, mediated by dysregulated plasmin generation, as evidenced by increased PAP and plasmin generation. These observations are consistent with the clinical presentation of both thrombosis and bleeding in VITT.
Collapse
Affiliation(s)
- Megan Simpson
- Aberdeen Cardiovascular & Diabetes Centre, Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, UK. https://twitter.com/SimpsonMegan8
| | - Anuj Narwal
- Aberdeen Cardiovascular & Diabetes Centre, Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, UK
| | - Eric West
- Aberdeen Cardiovascular & Diabetes Centre, Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, UK
| | - Jill Martin
- Department of Haematology Laboratory, Aberdeen Royal Infirmary, Aberdeen, UK
| | | | - Andrew R Page
- Department of Haematology, Royal Infirmary of Edinburgh, Edinburgh, UK
| | - Henry G Watson
- Aberdeen Cardiovascular & Diabetes Centre, Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, UK
| | - Claire S Whyte
- Aberdeen Cardiovascular & Diabetes Centre, Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, UK. https://twitter.com/ClaireW63108369
| | - Nicola J Mutch
- Aberdeen Cardiovascular & Diabetes Centre, Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, UK.
| |
Collapse
|
6
|
Ablan FDO, Maurer MC. Fbg αC 389-402 Enhances Factor XIII Cross-Linking in the Fibrinogen αC Region Via Electrostatic and Hydrophobic Interactions. Biochemistry 2023; 62:2170-2181. [PMID: 37410946 PMCID: PMC10583745 DOI: 10.1021/acs.biochem.3c00066] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/08/2023]
Abstract
Coagulation Factor XIII (FXIII) stabilizes blood clots by cross-linking glutamines and lysines in fibrin and other proteins. FXIII activity in the fibrinogen αC region (Fbg αC 221-610) is critical for clot stability and growth. Fbg αC 389-402 is a binding site for thrombin-activated FXIII, (FXIII-A*), with αC E396 promoting FXIII-A* binding and activity in αC. The current study aimed to discover additional residues within Fbg αC 389-402 that accelerate transglutaminase activity toward αC. Electrostatic αC residues (E395, E396, and D390), hydrophobic αC residues (W391 and F394), and residues αC 328-425 were studied by mutations to recombinant Fbg αC 233-425. FXIII activity was monitored through MS-based glycine ethyl ester (GEE) cross-linking and gel-based fluorescence monodansylcadaverine (MDC) cross-linking assays. Truncation mutations 403 Stop (Fbg αC 233-402), 389 Stop (Fbg αC 233-388), and 328 Stop (Fbg αC 233-327) reduced Q237-GEE and MDC cross-linking compared to wild-type (WT). Comparable cross-linking between 389 Stop and 328 Stop showed that FXIII is mainly affected by the loss of Fbg αC 389-402. Substitution mutations E396A, D390A, W391A, and F394A decreased cross-linking relative to WT, whereas E395A, E395S, E395K, and E396D had no effect. Similar FXIII-A* activities were observed for double mutants (D390A, E396A) and (W391A, E396A), relative to D390A and W391A, respectively. In contrast, cross-linking was reduced in (F394A, E396A), relative to F394A. In conclusion, Fbg αC 389-402 boosts FXIII activity in Fbg αC, with D390, W391, and F394 identified as key contributors in enhancing αC cross-linking.
Collapse
Affiliation(s)
- Francis D. O. Ablan
- Department of Chemistry, University of Louisville, Louisville, KY 40292, USA
| | - Muriel C. Maurer
- Department of Chemistry, University of Louisville, Louisville, KY 40292, USA
| |
Collapse
|
7
|
Syed Mohammed RD, Ablan FDO, McCann NM, Hindi MM, Maurer MC. Transglutaminase Activities of Blood Coagulant Factor XIII Are Dependent on the Activation Pathways and on the Substrates. Thromb Haemost 2023; 123:380-392. [PMID: 36473493 PMCID: PMC10719020 DOI: 10.1055/a-1993-4193] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Factor XIII (FXIII) catalyzes formation of γ-glutamyl-ε-lysyl crosslinks between reactive glutamines (Q) and lysines (K). In plasma, FXIII is activated proteolytically (FXIII-A*) by the concerted action of thrombin and Ca2+. Cellular FXIII is activated nonproteolytically (FXIII-A°) by elevation of physiological Ca2+ concentrations. FXIII-A targets plasmatic and cellular substrates, but questions remain on correlating FXIII activation, resultant conformational changes, and crosslinking function to different physiological substrates. To address these issues, the characteristics of FXIII-A* versus FXIII-A° that contribute to transglutaminase activity and substrate specificities were investigated. Crosslinking of lysine mimics into a series of Q-containing substrates were measured using in-gel fluorescence, mass spectrometry, and UV-Vis spectroscopy. Covalent incorporation of fluorescent monodansylcadaverine revealed that FXIII-A* exhibits greater activity than FXIII-A° toward Q residues within Fbg αC (233-425 WT, Q328P Seoul II, and Q328PQ366N) and actin. FXIII-A* and FXIII-A° displayed similar activities toward α2-antiplasmin (α2AP), fibronectin, and Fbg αC (233-388, missing FXIII-binding site αC 389-402). Furthermore, the N-terminal α2AP peptide (1-15) exhibited similar kinetic properties for FXIII-A* and FXIII-A°. MALDI-TOF mass spectrometry assays with glycine ethyl ester and Fbg αC (233-425 WT, αC E396A, and truncated αC (233-388) further documented that FXIII-A* exerts greater benefit from the αC 389-402 binding site than FXIII-A°. Conformational properties of FXIII-A* versus A° are proposed to help promote transglutaminase function toward different substrates. A combination of protein substrate disorder and secondary FXIII-binding site exposure are utilized to control activity and specificity. From these studies, greater understandings of how FXIII-A targets different substrates are achieved.
Collapse
Affiliation(s)
| | | | | | - Mohammed M. Hindi
- Department of Chemistry, University of Louisville, Louisville, KY, USA
| | - Muriel C. Maurer
- Department of Chemistry, University of Louisville, Louisville, KY, USA
| |
Collapse
|
8
|
Javed H, Singh S, Urs SUR, Oldenburg J, Biswas A. Genetic landscape in coagulation factor XIII associated defects – Advances in coagulation and beyond. Blood Rev 2022; 59:101032. [PMID: 36372609 DOI: 10.1016/j.blre.2022.101032] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 10/26/2022] [Accepted: 10/28/2022] [Indexed: 11/06/2022]
Abstract
Coagulation factor XIII (FXIII) acts as a fine fulcrum in blood plasma that maintains the balance between bleeding and thrombosis by covalently crosslinking the pre-formed fibrin clot into an insoluble one that is resistant to premature fibrinolysis. In plasma, FXIII circulates as a pro-transglutaminase complex composed of the dimeric catalytic FXIII-A encoded by the F13A1 gene and dimeric carrier/regulatory FXIII-B subunits encoded by the F13B gene. Growing evidence accumulated over decades of exhaustive research shows that not only does FXIII play major roles in both pathological extremes of hemostasis i.e. bleeding and thrombosis, but that it is, in fact, a pleiotropic protein with physiological roles beyond coagulation. However, the current FXIII genetic-epidemiological literature is overwhelmingly derived from the bleeding pathology associated with its deficiency. In this article we review the current clinical, functional, and molecular understanding of this fascinating multifaceted protein, especially putting into the same perspective its genetic landscape.
Collapse
|
9
|
Wolberg AS, Sang Y. Fibrinogen and Factor XIII in Venous Thrombosis and Thrombus Stability. Arterioscler Thromb Vasc Biol 2022; 42:931-941. [PMID: 35652333 PMCID: PMC9339521 DOI: 10.1161/atvbaha.122.317164] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
As the third most common vascular disease, venous thromboembolism is associated with significant mortality and morbidity. Pathogenesis underlying venous thrombosis is still not fully understood. Accumulating data suggest fibrin network structure and factor XIII-mediated crosslinking are major determinants of venous thrombus mass, composition, and stability. Understanding the cellular and molecular mechanisms mediating fibrin(ogen) and factor XIII production and function and their ability to influence venous thrombogenesis and resolution may inspire new anticoagulant strategies that target these proteins to reduce or prevent venous thrombosis in certain at-risk patients. This article summarizes fibrinogen and factor XIII biology and current knowledge of their function during venous thromboembolism.
Collapse
Affiliation(s)
- Alisa S Wolberg
- Department of Pathology and UNC Blood Research Center, University of North Carolina, Chapel Hill
| | - Yaqiu Sang
- Department of Pathology and UNC Blood Research Center, University of North Carolina, Chapel Hill
| |
Collapse
|
10
|
Xu RG, Gauer JS, Baker SR, Slater A, Martin EM, McPherson HR, Duval C, Manfield IW, Bonna AM, Watson SP, Ariëns RAS. GPVI (Glycoprotein VI) Interaction With Fibrinogen Is Mediated by Avidity and the Fibrinogen αC-Region. Arterioscler Thromb Vasc Biol 2021; 41:1092-1104. [PMID: 33472402 PMCID: PMC7901536 DOI: 10.1161/atvbaha.120.315030] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Supplemental Digital Content is available in the text. Objective: GPVI (glycoprotein VI) is a key molecular player in collagen-induced platelet signaling and aggregation. Recent evidence indicates that it also plays important role in platelet aggregation and thrombus growth through interaction with fibrin(ogen). However, there are discrepancies in the literature regarding whether the monomeric or dimeric form of GPVI binds to fibrinogen at high affinity. The mechanisms of interaction are also not clear, including which region of fibrinogen is responsible for GPVI binding. We aimed to gain further understanding of the mechanisms of interaction at molecular level and to identify the regions on fibrinogen important for GPVI binding. Approach and Results: Using multiple surface- and solution-based protein-protein interaction methods, we observe that dimeric GPVI binds to fibrinogen with much higher affinity and has a slower dissociation rate constant than the monomer due to avidity effects. Moreover, our data show that the highest affinity interaction of GPVI is with the αC-region of fibrinogen. We further show that GPVI interacts with immobilized fibrinogen and fibrin variants at a similar level, including a nonpolymerizing fibrin variant, suggesting that GPVI binding is independent of fibrin polymerization. Conclusions: Based on the above findings, we conclude that the higher affinity of dimeric GPVI over the monomer for fibrinogen interaction is achieved by avidity. The αC-region of fibrinogen appears essential for GPVI binding. We propose that fibrin polymerization into fibers during coagulation will cluster GPVI through its αC-region, leading to downstream signaling, further activation of platelets, and potentially stimulating clot growth.
Collapse
Affiliation(s)
- Rui-Gang Xu
- Discovery and Translational Science Department, Institute of Cardiovascular and Metabolic Medicine (R.-G.X., J.S.G., S.R.B., H.R.M., C.D., R.A.S.A.)
| | - Julia S Gauer
- Discovery and Translational Science Department, Institute of Cardiovascular and Metabolic Medicine (R.-G.X., J.S.G., S.R.B., H.R.M., C.D., R.A.S.A.)
| | - Stephen R Baker
- Discovery and Translational Science Department, Institute of Cardiovascular and Metabolic Medicine (R.-G.X., J.S.G., S.R.B., H.R.M., C.D., R.A.S.A.).,Department of Physics, Wake Forest University, Winston Salem, NC (S.R.B.)
| | - Alexandre Slater
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, United Kingdom (A.S., E.M.M., S.P.W.)
| | - Eleyna M Martin
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, United Kingdom (A.S., E.M.M., S.P.W.)
| | - Helen R McPherson
- Discovery and Translational Science Department, Institute of Cardiovascular and Metabolic Medicine (R.-G.X., J.S.G., S.R.B., H.R.M., C.D., R.A.S.A.)
| | - Cédric Duval
- Discovery and Translational Science Department, Institute of Cardiovascular and Metabolic Medicine (R.-G.X., J.S.G., S.R.B., H.R.M., C.D., R.A.S.A.)
| | - Iain W Manfield
- School of Molecular and Cellular Biology, Faculty of Biological Sciences (I.W.M.), University of Leeds, United Kingdom
| | - Arkadiusz M Bonna
- Department of Biochemistry, University of Cambridge, United Kingdom (A.M.B.)
| | - Steve P Watson
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, United Kingdom (A.S., E.M.M., S.P.W.)
| | - Robert A S Ariëns
- Discovery and Translational Science Department, Institute of Cardiovascular and Metabolic Medicine (R.-G.X., J.S.G., S.R.B., H.R.M., C.D., R.A.S.A.)
| |
Collapse
|
11
|
Clinical Validation of an Automated Fluorogenic Factor XIII Activity Assay Based on Isopeptidase Activity. Int J Mol Sci 2021; 22:ijms22031002. [PMID: 33498248 PMCID: PMC7863959 DOI: 10.3390/ijms22031002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 01/14/2021] [Accepted: 01/18/2021] [Indexed: 02/06/2023] Open
Abstract
Hereditary factor XIII (FXIII) deficiency is a rare autosomal bleeding disorder which can cause life-threatening bleeding. Acquired deficiency can be immune-mediated or due to increased consumption or reduced synthesis. The most commonly used screening test is insensitive, and widely used quantitative assays have analytical limitations. The present study sought to validate Technofluor FXIII Activity, the first isopeptidase-based assay available on a routine coagulation analyser, the Ceveron s100. Linearity was evidenced throughout the measuring range, with correlation coefficients of >0.99, and coefficients of variation for repeatability and reproducibility were <5% and <10%, respectively. A normally distributed reference range of 47.0–135.5 IU/dL was derived from 154 normal donors. Clinical samples with Technofluor FXIII Activity results between 0 and 167.0 IU/dL were assayed with Berichrom® FXIII Activity, a functional ammonia release assay, and the HemosIL™ FXIII antigen assay, generating correlations of 0.950 and 0.980, respectively. Experiments with a transglutaminase inhibitor showed that Technofluor FXIII Activity can detect inhibition of enzymatic activity. No interference was exhibited by high levels of haemolysis and lipaemia, and interference by bilirubin was evident at 18 mg/dL, a level commensurate with severe liver disease. Technofluor FXIII Activity is a rapid, accurate and precise assay suitable for routine diagnostic use with fewer interferents than ammonia release FXIII activity assays.
Collapse
|
12
|
Leitner M, Büchold C, Pasternack R, Binder NB. Feasibility of an automated coagulation factor XIIIa test using its isopeptidase activity. Anal Biochem 2020; 600:113757. [PMID: 32422134 DOI: 10.1016/j.ab.2020.113757] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 03/27/2020] [Accepted: 04/06/2020] [Indexed: 11/29/2022]
Abstract
Plasma transglutaminase FXIII provides mechanical and biochemical stability to blood clots. Congenital or acquired deficiency may be associated with bleeding diathesis and requires therefore careful monitoring. The precise automated measurement of a large number of plasma samples can provide new insights regarding the clinical relevance of certain FXIII levels. There is still the unmet diagnostic need for a reliable high-throughput method. Here we report the development and feasibility study of a promising prototype, adapting the precise FXIIIa isopeptidase assay principle on the optimized automated Ceveron s100 platform.
Collapse
Affiliation(s)
- Martina Leitner
- Technoclone Herstellung von Diagnostika und Arzneimitteln GmbH, Brunner Str. 67, 1230, Vienna, Austria
| | | | - Ralf Pasternack
- Zedira GmbH, Roesslerstrasse 83, D 64293, Darmstadt, Germany.
| | - Nikolaus B Binder
- Technoclone Herstellung von Diagnostika und Arzneimitteln GmbH, Brunner Str. 67, 1230, Vienna, Austria.
| |
Collapse
|
13
|
Pasternack R, Büchold C, Jähnig R, Pelzer C, Sommer M, Heil A, Florian P, Nowak G, Gerlach U, Hils M. Novel inhibitor ZED3197 as potential drug candidate in anticoagulation targeting coagulation FXIIIa (F13a). J Thromb Haemost 2020; 18:191-200. [PMID: 31578814 PMCID: PMC6973046 DOI: 10.1111/jth.14646] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 09/02/2019] [Accepted: 09/03/2019] [Indexed: 12/15/2022]
Abstract
BACKGROUND Factor XIII (FXIII) is the final enzyme of the coagulation cascade. While the other enzymatic coagulation factors are proteases, FXIII belongs to the transglutaminase family. FXIIIa covalently crosslinks the fibrin clot and represents a promising target for drug development to facilitate fibrinolysis. However, no FXIII-inhibiting compound has entered clinical trials. Here, we introduce the features of a peptidomimetic inhibitor of FXIIIa (ZED3197) as a potential drug candidate. METHODS The potency of ZED3197 against FXIIIa and the selectivity against other human transglutaminases were characterized using transamidation and isopeptidase assays. The inhibition of fibrin crosslinking was evaluated by biochemical methods and thromboelastometry. Further, the pharmacology of the compound was explored in a rabbit model of venous stasis and reperfusion. RESULTS ZED3197 proved to be a potent and selective inhibitor of human FXIIIa. Further, the compound showed broad inhibitory activity against cellular FXIIIA from various animal species. Rotational thromboelastometry in whole human blood indicated that the inhibitor, in a dose-dependent manner, prolonged clot formation, reduced clot firmness, and facilitated clot lysis without affecting the clotting time, indicating minimal impact on hemostasis. In vivo, the novel FXIIIa inhibitor effectively decreased the weight of clots and facilitated flow restoration without prolongation of the bleeding time. CONCLUSIONS ZED3197 is the first drug-like potent compound targeting FXIIIa, a yet untapped target in anticoagulation. Due to the function of FXIII downstream of thrombin the approach provides minimal impact on hemostasis. In vivo data imply that the inhibitor dissociates an antithrombotic effect from increased bleeding tendency.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Uwe Gerlach
- Sanofi‐Aventis Deutschland GmbHFrankfurtGermany
| | | |
Collapse
|
14
|
Supsrisunjai C, Hsu CK, Michael M, Duval C, Lee JYW, Yang HS, Huang HY, Chaikul T, Onoufriadis A, Steiner RA, Ariëns RAS, Sarig O, Sprecher E, Eskin-Schwartz M, Samlaska C, Simpson MA, Calonje E, Parsons M, McGrath JA. Coagulation Factor XIII-A Subunit Missense Mutation in the Pathobiology of Autosomal Dominant Multiple Dermatofibromas. J Invest Dermatol 2019; 140:624-635.e7. [PMID: 31493396 DOI: 10.1016/j.jid.2019.08.441] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 07/26/2019] [Accepted: 08/06/2019] [Indexed: 01/16/2023]
Abstract
Dermatofibromas are common benign skin lesions, the etiology of which is poorly understood. We identified two unrelated pedigrees in which there was autosomal dominant transmission of multiple dermatofibromas. Whole exome sequencing revealed a rare shared heterozygous missense variant in the F13A1 gene encoding factor XIII subunit A (FXIII-A), a transglutaminase involved in hemostasis, wound healing, tumor growth, and apoptosis. The variant (p.Lys679Met) has an allele frequency of 0.0002 and is predicted to be a damaging mutation. Recombinant human Lys679Met FXIII-A demonstrated reduced fibrin crosslinking activity in vitro. Of note, the treatment of fibroblasts with media containing Lys679Met FXIII-A led to enhanced adhesion, proliferation, and type I collagen synthesis. Immunostaining revealed co-localization between FXIII-A and α4β1 integrins, more prominently for Lys679Met FXIII-A than the wild type. In addition, both the α4β1 inhibitors and the mutation of the FXIII-A Isoleucine-Leucine-Aspartate-Threonine (ILDT) motif prevented Lys679Met FXIII-A-dependent proliferation and collagen synthesis of fibroblasts. Our data suggest that the Lys679Met mutation may lead to a conformational change in the FXIII-A protein that enhances α4-integrin binding and provides insight into an unexpected role for FXIII-A in the pathobiology of familial dermatofibroma.
Collapse
Affiliation(s)
- Chavalit Supsrisunjai
- St John's Institute of Dermatology, School of Basic and Medical Biosciences, King's College London, Guy's Hospital, London, United Kingdom; Institute of Dermatology, Ministry of Public Health, Bangkok, Thailand
| | - Chao-Kai Hsu
- St John's Institute of Dermatology, School of Basic and Medical Biosciences, King's College London, Guy's Hospital, London, United Kingdom; Department of Dermatology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan; International Center for Wound Repair and Regeneration, National Cheng Kung University, Tainan, Taiwan
| | - Magdalene Michael
- Randall Centre for Cell and Molecular Biophysics, School of Basic and Medical Biosciences, King's College London, United Kingdom
| | - Cédric Duval
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom
| | - John Y W Lee
- St John's Institute of Dermatology, School of Basic and Medical Biosciences, King's College London, Guy's Hospital, London, United Kingdom
| | - Hsing-San Yang
- Department of Dermatology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Hsin-Yu Huang
- School of Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Thitiwat Chaikul
- St John's Institute of Dermatology, School of Basic and Medical Biosciences, King's College London, Guy's Hospital, London, United Kingdom
| | - Alexandros Onoufriadis
- St John's Institute of Dermatology, School of Basic and Medical Biosciences, King's College London, Guy's Hospital, London, United Kingdom
| | - Roberto A Steiner
- Randall Centre for Cell and Molecular Biophysics, School of Basic and Medical Biosciences, King's College London, United Kingdom
| | - Robert A S Ariëns
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom
| | - Ofer Sarig
- Division of Dermatology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Eli Sprecher
- Division of Dermatology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; Department of Human Molecular Genetics & Biochemistry, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | | | - Curt Samlaska
- Academic Dermatology of Nevada, University of Nevada School of Medicine, Reno, Nevada
| | - Michael A Simpson
- Department of Genetics, School of Basic and Medical Biosciences, King's College London, Guy's Hospital, London, United Kingdom
| | - Eduardo Calonje
- St John's Institute of Dermatology, School of Basic and Medical Biosciences, King's College London, Guy's Hospital, London, United Kingdom; Department of Dermatopathology, St John's Institute of Dermatology, Guy's and St Thomas' NHS Foundation Trust, London, United Kingdom
| | - Maddy Parsons
- Randall Centre for Cell and Molecular Biophysics, School of Basic and Medical Biosciences, King's College London, United Kingdom
| | - John A McGrath
- St John's Institute of Dermatology, School of Basic and Medical Biosciences, King's College London, Guy's Hospital, London, United Kingdom.
| |
Collapse
|
15
|
Anokhin BA, Dean WL, Smith KA, Flick MJ, Ariëns RAS, Philippou H, Maurer MC. Proteolytic and nonproteolytic activation mechanisms result in conformationally and functionally different forms of coagulation factor XIII A. FEBS J 2019; 287:452-464. [PMID: 31407850 DOI: 10.1111/febs.15040] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 06/21/2019] [Accepted: 08/12/2019] [Indexed: 12/17/2022]
Abstract
Factor XIIIA (FXIIIA) is a transglutaminase that cross-links intra- and extracellular protein substrates. FXIIIA is expressed as an inactive zymogen, and during blood coagulation, it is activated by removal of an activation peptide by the protease thrombin. No such proteolytic FXIIIA activation is known to occur in other tissues or the intracellular form of FXIIIA. For those locations, FXIIIA is assumed instead to undergo activation by Ca2+ ions. Previously, we demonstrated a monomeric state for active FXIIIA. Current analytical ultracentrifugation and kinetic experiments revealed that thrombin-activated FXIIIA has a higher conformational flexibility and a stronger affinity toward glutamine substrate than does nonproteolytically activated FXIIIA. The proteolytic activation of FXIIIA was further investigated in a context of fibrin clotting. In a series of fibrin cross-linking assays and scanning electron microscopy studies of plasma clots, the activation rates of FXIIIA V34X variants were correlated with the extent of fibrin cross-linking and incorporation of nonfibrous protein into the clot. Overall, the results suggest conformational and functional differences between active FXIIIA forms, thus expanding the understanding of FXIIIA function. Those differences may serve as a basis for developing therapeutic strategies to target FXIIIA in different physiological environments. ENZYMES: Factor XIIIA ( EC 2.3.2.13).
Collapse
Affiliation(s)
| | - William L Dean
- Brown Cancer Center, University of Louisville School of Medicine, KY, USA.,Department of Medicine, University of Louisville, KY, USA.,Department of Biochemistry and Molecular Genetics, University of Louisville, KY, USA
| | - Kerrie A Smith
- Leeds Thrombosis Collective, Department of Discovery and Translational Science, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, UK
| | - Matthew J Flick
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, OH, USA
| | - Robert A S Ariëns
- Leeds Thrombosis Collective, Department of Discovery and Translational Science, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, UK
| | - Helen Philippou
- Leeds Thrombosis Collective, Department of Discovery and Translational Science, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, UK
| | | |
Collapse
|
16
|
Kattula S, Byrnes JR, Wolberg AS. Fibrinogen and Fibrin in Hemostasis and Thrombosis. Arterioscler Thromb Vasc Biol 2019; 37:e13-e21. [PMID: 28228446 DOI: 10.1161/atvbaha.117.308564] [Citation(s) in RCA: 243] [Impact Index Per Article: 48.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Sravya Kattula
- From the Department of Pathology and Laboratory Medicine, McAllister Heart Institute, University of North Carolina, Chapel Hill
| | - James R Byrnes
- From the Department of Pathology and Laboratory Medicine, McAllister Heart Institute, University of North Carolina, Chapel Hill
| | - Alisa S Wolberg
- From the Department of Pathology and Laboratory Medicine, McAllister Heart Institute, University of North Carolina, Chapel Hill.
| |
Collapse
|
17
|
Mouapi KN, Wagner LJ, Stephens CA, Hindi MM, Wilkey DW, Merchant ML, Maurer MC. Evaluating the Effects of Fibrinogen αC Mutations on the Ability of Factor XIII to Crosslink the Reactive αC Glutamines (Q237, Q328, Q366). Thromb Haemost 2019; 119:1048-1057. [PMID: 31055797 DOI: 10.1055/s-0039-1687875] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Fibrinogen (Fbg) levels and extent of fibrin polymerization have been associated with various pathological conditions such as cardiovascular disease, arteriosclerosis, and coagulation disorders. Activated factor XIII (FXIIIa) introduces γ-glutamyl-ε-lysinyl isopeptide bonds between reactive glutamines and lysines in the fibrin network to form a blood clot resistant to fibrinolysis. FXIIIa crosslinks the γ-chains and at multiple sites in the αC region of Fbg. Fbg αC contains a FXIII binding site involving αC (389-402) that is located near three glutamines whose reactivities rank Q237 >> Q366 ≈ Q328. Mass spectrometry and two-dimensional heteronuclear single-quantum correlation nuclear magnetic resonance assays were used to probe the anchoring role that αC E396 may play in controlling FXIII function and characterize the effects of Q237 on the reactivities of Q328 and Q366. Studies with αC (233-425) revealed that the E396A mutation does not prevent the transglutaminase function of FXIII A2 or A2B2. Other residues must play a compensatory role in targeting FXIII to αC. Unlike full Fbg, Fbg αC (233-425) did not promote thrombin cleavage of FXIII, an event contributing to activation. With the αC (233-425) E396A mutant, Q237 exhibited slower reactivities compared with αC wild-type (WT) consistent with difficulties in directing this N-terminal segment toward an anchored FXIII interacting at a weaker binding region. Q328 and Q366 became less reactive when Q237 was replaced with inactive N237. Q237 crosslinking is proposed to promote targeting of Q328 and Q366 to the FXIII active site. FXIII thus uses Fbg αC anchoring sites and distinct Q environments to regulate substrate specificity.
Collapse
Affiliation(s)
- Kelly Njine Mouapi
- Chemistry Department, University of Louisville, Louisville, Kentucky, United States
| | - Lucille J Wagner
- Chemistry Department, University of Louisville, Louisville, Kentucky, United States
| | - Chad A Stephens
- Chemistry Department, University of Louisville, Louisville, Kentucky, United States
| | - Mohammed M Hindi
- Chemistry Department, University of Louisville, Louisville, Kentucky, United States
| | - Daniel W Wilkey
- Division of Nephrology and Hypertension, Department of Medicine, University of Louisville, Kentucky, United States
| | - Michael L Merchant
- Division of Nephrology and Hypertension, Department of Medicine, University of Louisville, Kentucky, United States
| | - Muriel C Maurer
- Chemistry Department, University of Louisville, Louisville, Kentucky, United States
| |
Collapse
|
18
|
Ismail AE, Fabian FM, Wang O, Lei Y, Carlson MA, Burgess WH, Velander WH. The isolation of a plasma-derived γγ’ fibrinogen: Fibronectin mixture that forms a novel polymeric matrix. Process Biochem 2018. [DOI: 10.1016/j.procbio.2018.09.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
19
|
Wolberg AS. Fibrinogen and factor XIII: newly recognized roles in venous thrombus formation and composition. Curr Opin Hematol 2018; 25:358-364. [PMID: 29994896 PMCID: PMC6215450 DOI: 10.1097/moh.0000000000000445] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
PURPOSE OF REVIEW In spite of significant morbidity and mortality associated with venous thromboembolism, the underlying pathogenesis remains poorly understood. RECENT FINDINGS Clues to operant pathogenic mechanisms are found in the unique morphology and composition of these thrombi, which have substantial red blood cell and fibrin content. Recent studies have revealed biochemical and biophysical mechanisms that dictate fibrin structure in venous thrombi and promote retention of red blood cells within the contracted clots. These mechanisms include newly recognized contributions of fibrin network structure and factor XIII(a)-mediated fibrin crosslinking to venous thrombus composition, size, and stability. SUMMARY Continued work to elucidate mechanisms by which fibrin(ogen), factor XIII, and red blood cells contribute to venous thrombus formation, structure, and stability may expose novel molecular targets and strategies for reducing thrombosis and thrombotic complications in certain at-risk patients.
Collapse
Affiliation(s)
- Alisa S Wolberg
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
20
|
Hethershaw EL, Adamson PJ, Smith KA, Goldsberry WN, Pease RJ, Radford SE, Grant PJ, Ariëns RAS, Maurer MC, Philippou H. The role of β-barrels 1 and 2 in the enzymatic activity of factor XIII A-subunit. J Thromb Haemost 2018; 16:1391-1401. [PMID: 29675848 PMCID: PMC6175083 DOI: 10.1111/jth.14128] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Indexed: 01/04/2023]
Abstract
Essentials The roles of β-barrels 1 and 2 in factor XIII (FXIII) are currently unknown. FXIII truncations lacking β-barrel 2, both β-barrels, or full length FXIII, were made. Removing β-barrel 2 caused total loss of activity, removing both β-barrels returned 30% activity. β-barrel 2 is necessary for exposure of the active site cysteine during activation. SUMMARY Background Factor XIII is composed of an activation peptide segment, a β-sandwich domain, a catalytic core, and, finally, β-barrels 1 and 2. FXIII is activated following cleavage of its A-subunits by thrombin. The resultant transglutaminase activity leads to increased resistance of fibrin clots to fibrinolysis. Objectives To assess the functional roles of β-barrels 1 and 2 in FXIII, we expressed and characterized the full-length FXIII A-subunit (FXIII-A) and variants truncated to residue 628 (truncated to β-barrel 1 [TB1]), residue 515 (truncated to catalytic core [TCC]), and residue 184 (truncated to β-sandwich). Methods Proteins were analyzed by gel electrophoresis, circular dichroism, fluorometric assays, and colorimetric activity assays, clot structure was analyzed by turbidity measurements and confocal microscopy, and clot formation was analyzed with a Chandler loop system. Results and Conclusions Circular dichroism spectroscopy and tryptophan fluorometry indicated that full-length FXIII-A and the truncation variants TCC and TB1 retain their secondary and tertiary structure. Removal of β-barrel 2 (TB1) resulted in total loss of transglutaminase activity, whereas the additional removal of β-barrel 1 (TCC) restored enzymatic activity to ~ 30% of that of full-length FXIII-A. These activity trends were observed with physiological substrates and smaller model substrates. Our data suggest that the β-barrel 1 domain protects the active site cysteine in the FXIII protransglutaminase, whereas the β-barrel 2 domain is necessary for exposure of the active site cysteine during activation. This study demonstrates the importance of individual β-barrel domains in modulating access to the FXIII active site region.
Collapse
Affiliation(s)
- E. L. Hethershaw
- Discovery and Translational Science DepartmentLeeds Institute for Cardiovascular and Metabolic MedicineFaculty of Medicine and HealthUniversity of LeedsLeedsUK
| | - P. J. Adamson
- Discovery and Translational Science DepartmentLeeds Institute for Cardiovascular and Metabolic MedicineFaculty of Medicine and HealthUniversity of LeedsLeedsUK
| | - K. A. Smith
- Discovery and Translational Science DepartmentLeeds Institute for Cardiovascular and Metabolic MedicineFaculty of Medicine and HealthUniversity of LeedsLeedsUK
| | | | - R. J. Pease
- Discovery and Translational Science DepartmentLeeds Institute for Cardiovascular and Metabolic MedicineFaculty of Medicine and HealthUniversity of LeedsLeedsUK
| | - S. E. Radford
- Astbury Centre for Structural Molecular BiologySchool of Molecular and Cellular BiologyUniversity of LeedsLeedsUK
| | - P. J. Grant
- Discovery and Translational Science DepartmentLeeds Institute for Cardiovascular and Metabolic MedicineFaculty of Medicine and HealthUniversity of LeedsLeedsUK
| | - R. A. S. Ariëns
- Discovery and Translational Science DepartmentLeeds Institute for Cardiovascular and Metabolic MedicineFaculty of Medicine and HealthUniversity of LeedsLeedsUK
| | - M. C. Maurer
- Chemistry DepartmentUniversity of LouisvilleLouisvilleKYUSA
| | - H. Philippou
- Discovery and Translational Science DepartmentLeeds Institute for Cardiovascular and Metabolic MedicineFaculty of Medicine and HealthUniversity of LeedsLeedsUK
| |
Collapse
|
21
|
Spronk HMH, Padro T, Siland JE, Prochaska JH, Winters J, van der Wal AC, Posthuma JJ, Lowe G, d'Alessandro E, Wenzel P, Coenen DM, Reitsma PH, Ruf W, van Gorp RH, Koenen RR, Vajen T, Alshaikh NA, Wolberg AS, Macrae FL, Asquith N, Heemskerk J, Heinzmann A, Moorlag M, Mackman N, van der Meijden P, Meijers JCM, Heestermans M, Renné T, Dólleman S, Chayouâ W, Ariëns RAS, Baaten CC, Nagy M, Kuliopulos A, Posma JJ, Harrison P, Vries MJ, Crijns HJGM, Dudink EAMP, Buller HR, Henskens YMC, Själander A, Zwaveling S, Erküner O, Eikelboom JW, Gulpen A, Peeters FECM, Douxfils J, Olie RH, Baglin T, Leader A, Schotten U, Scaf B, van Beusekom HMM, Mosnier LO, van der Vorm L, Declerck P, Visser M, Dippel DWJ, Strijbis VJ, Pertiwi K, Ten Cate-Hoek AJ, Ten Cate H. Atherothrombosis and Thromboembolism: Position Paper from the Second Maastricht Consensus Conference on Thrombosis. Thromb Haemost 2018; 118:229-250. [PMID: 29378352 DOI: 10.1160/th17-07-0492] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Atherothrombosis is a leading cause of cardiovascular mortality and long-term morbidity. Platelets and coagulation proteases, interacting with circulating cells and in different vascular beds, modify several complex pathologies including atherosclerosis. In the second Maastricht Consensus Conference on Thrombosis, this theme was addressed by diverse scientists from bench to bedside. All presentations were discussed with audience members and the results of these discussions were incorporated in the final document that presents a state-of-the-art reflection of expert opinions and consensus recommendations regarding the following five topics: 1. Risk factors, biomarkers and plaque instability: In atherothrombosis research, more focus on the contribution of specific risk factors like ectopic fat needs to be considered; definitions of atherothrombosis are important distinguishing different phases of disease, including plaque (in)stability; proteomic and metabolomics data are to be added to genetic information. 2. Circulating cells including platelets and atherothrombosis: Mechanisms of leukocyte and macrophage plasticity, migration, and transformation in murine atherosclerosis need to be considered; disease mechanism-based biomarkers need to be identified; experimental systems are needed that incorporate whole-blood flow to understand how red blood cells influence thrombus formation and stability; knowledge on platelet heterogeneity and priming conditions needs to be translated toward the in vivo situation. 3. Coagulation proteases, fibrin(ogen) and thrombus formation: The role of factor (F) XI in thrombosis including the lower margins of this factor related to safe and effective antithrombotic therapy needs to be established; FXI is a key regulator in linking platelets, thrombin generation, and inflammatory mechanisms in a renin-angiotensin dependent manner; however, the impact on thrombin-dependent PAR signaling needs further study; the fundamental mechanisms in FXIII biology and biochemistry and its impact on thrombus biophysical characteristics need to be explored; the interactions of red cells and fibrin formation and its consequences for thrombus formation and lysis need to be addressed. Platelet-fibrin interactions are pivotal determinants of clot formation and stability with potential therapeutic consequences. 4. Preventive and acute treatment of atherothrombosis and arterial embolism; novel ways and tailoring? The role of protease-activated receptor (PAR)-4 vis à vis PAR-1 as target for antithrombotic therapy merits study; ongoing trials on platelet function test-based antiplatelet therapy adjustment support development of practically feasible tests; risk scores for patients with atrial fibrillation need refinement, taking new biomarkers including coagulation into account; risk scores that consider organ system differences in bleeding may have added value; all forms of oral anticoagulant treatment require better organization, including education and emergency access; laboratory testing still needs rapidly available sensitive tests with short turnaround time. 5. Pleiotropy of coagulation proteases, thrombus resolution and ischaemia-reperfusion: Biobanks specifically for thrombus storage and analysis are needed; further studies on novel modified activated protein C-based agents are required including its cytoprotective properties; new avenues for optimizing treatment of patients with ischaemic stroke are needed, also including novel agents that modify fibrinolytic activity (aimed at plasminogen activator inhibitor-1 and thrombin activatable fibrinolysis inhibitor.
Collapse
Affiliation(s)
- H M H Spronk
- Laboratory for Clinical Thrombosis and Haemostasis, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, The Netherlands
| | - T Padro
- Cardiovascular Research Center (ICCC), Hospital Sant Pau, Barcelona, Spain
| | - J E Siland
- Department of Cardiology, University Medical Center Groningen, Groningen, The Netherlands
| | - J H Prochaska
- Center for Cardiology/Center for Thrombosis and Hemostasis/DZHK, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - J Winters
- Department of Physiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - A C van der Wal
- Department of Pathology, Academic Medical Center (AMC), Amsterdam, The Netherlands
| | - J J Posthuma
- Laboratory for Clinical Thrombosis and Haemostasis, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, The Netherlands
| | - G Lowe
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, Scotland
| | - E d'Alessandro
- Department of Physiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands.,Department of Pathology, Academic Medical Center (AMC), Amsterdam, The Netherlands
| | - P Wenzel
- Department of Cardiology, Universitätsmedizin Mainz, Mainz, Germany
| | - D M Coenen
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - P H Reitsma
- Einthoven Laboratory, Leiden University Medical Center, Leiden, The Netherlands
| | - W Ruf
- Center for Cardiology/Center for Thrombosis and Hemostasis/DZHK, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - R H van Gorp
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - R R Koenen
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - T Vajen
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - N A Alshaikh
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - A S Wolberg
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, North Carolina, United States
| | - F L Macrae
- Thrombosis and Tissue Repair Group, Division of Cardiovascular and Diabetes Research, Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, UK
| | - N Asquith
- Thrombosis and Tissue Repair Group, Division of Cardiovascular and Diabetes Research, Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, UK
| | - J Heemskerk
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - A Heinzmann
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - M Moorlag
- Synapse, Maastricht, The Netherlands
| | - N Mackman
- Department of Medicine, UNC McAllister Heart Institute, University of North Carolina, Chapel Hill, North Carolina, United States
| | - P van der Meijden
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - J C M Meijers
- Department of Plasma Proteins, Sanquin, Amsterdam, The Netherlands
| | - M Heestermans
- Einthoven Laboratory, Leiden University Medical Center, Leiden, The Netherlands
| | - T Renné
- Department of Molecular Medicine and Surgery, Karolinska Institutet and University Hospital, Stockholm, Sweden.,Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - S Dólleman
- Department of Nephrology, Leiden University Medical Centre, Leiden, The Netherlands
| | - W Chayouâ
- Synapse, Maastricht, The Netherlands
| | - R A S Ariëns
- Thrombosis and Tissue Repair Group, Division of Cardiovascular and Diabetes Research, Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, UK
| | - C C Baaten
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - M Nagy
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - A Kuliopulos
- Tufts University School of Graduate Biomedical Sciences, Biochemistry/Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, Boston, Massachusetts
| | - J J Posma
- Laboratory for Clinical Thrombosis and Haemostasis, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, The Netherlands
| | - P Harrison
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
| | - M J Vries
- Laboratory for Clinical Thrombosis and Haemostasis, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, The Netherlands
| | - H J G M Crijns
- Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, The Netherlands
| | - E A M P Dudink
- Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, The Netherlands
| | - H R Buller
- Department of Vascular Medicine, Academic Medical Center (AMC), Amsterdam, The Netherlands
| | - Y M C Henskens
- Laboratory for Clinical Thrombosis and Haemostasis, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, The Netherlands
| | - A Själander
- Department of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
| | - S Zwaveling
- Laboratory for Clinical Thrombosis and Haemostasis, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, The Netherlands.,Synapse, Maastricht, The Netherlands
| | - O Erküner
- Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, The Netherlands
| | - J W Eikelboom
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - A Gulpen
- Laboratory for Clinical Thrombosis and Haemostasis, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, The Netherlands
| | - F E C M Peeters
- Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, The Netherlands
| | - J Douxfils
- Department of Pharmacy, Thrombosis and Hemostasis Center, Faculty of Medicine, Namur University, Namur, Belgium
| | - R H Olie
- Laboratory for Clinical Thrombosis and Haemostasis, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, The Netherlands
| | - T Baglin
- Department of Haematology, Addenbrookes Hospital Cambridge, Cambridge, United Kingdom
| | - A Leader
- Laboratory for Clinical Thrombosis and Haemostasis, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, The Netherlands.,Davidoff Cancer Center, Rabin Medical Center, Institute of Hematology, Sackler Faculty of Medicine, Tel Aviv University, Petah Tikva, Tel Aviv, Israel
| | - U Schotten
- Center for Cardiology/Center for Thrombosis and Hemostasis/DZHK, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - B Scaf
- Laboratory for Clinical Thrombosis and Haemostasis, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, The Netherlands.,Department of Physiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - H M M van Beusekom
- Department of Experimental Cardiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - L O Mosnier
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, United States
| | | | - P Declerck
- Department of Pharmaceutical and Pharmacological Sciences, University of Leuven, Leuven, Belgium
| | | | - D W J Dippel
- Department of Neurology, Erasmus MC, Rotterdam, The Netherlands
| | | | - K Pertiwi
- Department of Cardiovascular Pathology, University of Amsterdam, Academic Medical Center, Amsterdam, The Netherlands
| | - A J Ten Cate-Hoek
- Laboratory for Clinical Thrombosis and Haemostasis, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, The Netherlands
| | - H Ten Cate
- Laboratory for Clinical Thrombosis and Haemostasis, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, The Netherlands
| |
Collapse
|
22
|
Jadhav MA, Goldsberry WN, Zink SE, Lamb KN, Simmons KE, Riposo CM, Anokhin BA, Maurer MC. Screening cleavage of Factor XIII V34X Activation Peptides by thrombin mutants: A strategy for controlling fibrin architecture. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2017; 1865:1246-1254. [PMID: 28687225 DOI: 10.1016/j.bbapap.2017.07.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2017] [Revised: 06/26/2017] [Accepted: 07/02/2017] [Indexed: 10/19/2022]
Abstract
In blood coagulation, thrombin converts fibrinogen into fibrin monomers that polymerize into a clot network. Thrombin also activates Factor XIII by cleaving the R37-G38 peptide bond of the Activation Peptide (AP) segment. The resultant transglutaminase introduces covalent crosslinks into the fibrin clot. A strategy to modify clot architecture would be to design FXIII AP sequences that are easier or more difficult to be thrombin-cleaved thus controlling initiation of crosslinking. To aid in this design process, FXIII V34X (28-41) Activation Peptides were kinetically ranked for cleavage by wild-type thrombin and several anticoagulant mutants. Thrombin-catalyzed hydrolysis of aromatic FXIII F34, W34, and Y34 APs was compared with V34 and L34. Cardioprotective FXIII L34 remained the variant most readily cleaved by wild-type thrombin. The potent anticoagulant thrombins W215A and W215A/E217A (missing a key substrate platform for binding fibrinogen) were best able to hydrolyze FXIII F34 and W34 APs. Thrombin I174A and L99A could effectively accommodate FXIII W34 and Y34 APs yielding kinetic parameters comparable to FXIII AP L34 with wild-type thrombin. None of the aromatic FXIII V34X APs could be hydrolyzed by thrombin Y60aA. FXIII F34 and W34 are promising candidates for FXIII - anticoagulant thrombin systems that could permit FXIII-catalyzed crosslinking in the presence of reduced fibrin formation. By contrast, FXIII Y34 with thrombin (Y60aA or W215A/E217A) could help assure that both fibrin clot formation and protein crosslinking are hindered. Regulating the activation of FXIII is predicted to be a strategy for helping to control fibrin clot architecture and its neighboring environments.
Collapse
Affiliation(s)
- Madhavi A Jadhav
- Chemistry Department, University of Louisville, 2320 South Brook Street, Louisville, KY 40292, USA
| | - Whitney N Goldsberry
- Chemistry Department, University of Louisville, 2320 South Brook Street, Louisville, KY 40292, USA
| | - Sara E Zink
- Chemistry Department, University of Louisville, 2320 South Brook Street, Louisville, KY 40292, USA
| | - Kelsey N Lamb
- Chemistry Department, University of Louisville, 2320 South Brook Street, Louisville, KY 40292, USA
| | - Katelyn E Simmons
- Chemistry Department, University of Louisville, 2320 South Brook Street, Louisville, KY 40292, USA
| | - Carmela M Riposo
- Chemistry Department, University of Louisville, 2320 South Brook Street, Louisville, KY 40292, USA
| | - Boris A Anokhin
- Chemistry Department, University of Louisville, 2320 South Brook Street, Louisville, KY 40292, USA
| | - Muriel C Maurer
- Chemistry Department, University of Louisville, 2320 South Brook Street, Louisville, KY 40292, USA.
| |
Collapse
|
23
|
Hudson NE. Biophysical Mechanisms Mediating Fibrin Fiber Lysis. BIOMED RESEARCH INTERNATIONAL 2017; 2017:2748340. [PMID: 28630861 PMCID: PMC5467299 DOI: 10.1155/2017/2748340] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Accepted: 04/30/2017] [Indexed: 01/19/2023]
Abstract
The formation and dissolution of blood clots is both a biochemical and a biomechanical process. While much of the chemistry has been worked out for both processes, the influence of biophysical properties is less well understood. This review considers the impact of several structural and mechanical parameters on lytic rates of fibrin fibers. The influences of fiber and network architecture, fiber strain, FXIIIa cross-linking, and particle transport phenomena will be assessed. The importance of the mechanical aspects of fibrinolysis is emphasized, and future research avenues are discussed.
Collapse
Affiliation(s)
- Nathan E. Hudson
- Department of Physics, East Carolina University, N304 Howell Science Complex, Greenville, NC 27858, USA
| |
Collapse
|
24
|
Tsai JYC, Loh JMS, Clow F, Lorenz N, Proft T. The Group A Streptococcus serotype M2 pilus plays a role in host cell adhesion and immune evasion. Mol Microbiol 2016; 103:282-298. [PMID: 27741558 DOI: 10.1111/mmi.13556] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/11/2016] [Indexed: 01/22/2023]
Abstract
Group A Streptococcus (GAS), or Streptococcus pyogenes, is a human pathogen that causes diseases ranging from skin and soft tissue infections to severe invasive diseases, such as toxic shock syndrome. Each GAS strain carries a particular pilus type encoded in the variable fibronectin-binding, collagen-binding, T antigen (FCT) genomic region. Here, we describe the functional analysis of the serotype M2 pilus encoded in the FCT-6 region. We found that, in contrast to other investigated GAS pili, the ancillary pilin 1 lacks adhesive properties. Instead, the backbone pilin is important for host cell adhesion and binds several host factors, including fibronectin and fibrinogen. Using a panel of recombinant pilus proteins, GAS gene deletion mutants and Lactococcus lactis gain-of-function mutants we show that, unlike other GAS pili, the FCT-6 pilus also contributes to immune evasion. This was demonstrated by a delay in blood clotting, increased intracellular survival of the bacteria in macrophages, higher bacterial survival rates in human whole blood and greater virulence in a Galleria mellonella infection model in the presence of fully assembled FCT-6 pili.
Collapse
Affiliation(s)
- Jia-Yun C Tsai
- Department of Molecular Medicine & Pathology, School of Medical Sciences.,Maurice Wilkins Centre, The University of Auckland, Auckland, New Zealand
| | - Jacelyn M S Loh
- Department of Molecular Medicine & Pathology, School of Medical Sciences.,Maurice Wilkins Centre, The University of Auckland, Auckland, New Zealand
| | - Fiona Clow
- Department of Molecular Medicine & Pathology, School of Medical Sciences
| | - Natalie Lorenz
- Department of Molecular Medicine & Pathology, School of Medical Sciences
| | - Thomas Proft
- Department of Molecular Medicine & Pathology, School of Medical Sciences.,Maurice Wilkins Centre, The University of Auckland, Auckland, New Zealand
| |
Collapse
|
25
|
South K, Freitas MO, Lane DA. Conformational quiescence of ADAMTS-13 prevents proteolytic promiscuity. J Thromb Haemost 2016; 14:2011-2022. [PMID: 27514025 PMCID: PMC5111603 DOI: 10.1111/jth.13445] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Indexed: 11/29/2022]
Abstract
Essentials Recently, ADAMTS-13 has been shown to undergo substrate induced conformation activation. Conformational quiescence of ADAMTS-13 may serve to prevent off-target proteolysis in plasma. Conformationally active ADAMTS-13 variants are capable of proteolysing the Aα chain of fibrinogen. This should be considered as ADAMTS-13 variants are developed as potential therapeutic agents. Click to hear Dr Zheng's presentation on structure function and cofactor-dependent regulation of ADAMTS-13 SUMMARY: Background Recent work has revealed that ADAMTS-13 circulates in a 'closed' conformation, only fully interacting with von Willebrand factor (VWF) following a conformational change. We hypothesized that this conformational quiescence also maintains the substrate specificity of ADAMTS-13 and that the 'open' conformation of the protease might facilitate proteolytic promiscuity. Objectives To identify a novel substrate for a constitutively active gain of function (GoF) ADAMTS-13 variant (R568K/F592Y/R660K/Y661F/Y665F). Methods Fibrinogen proteolysis was characterized using SDS PAGE and liquid chromatography-tandem mass spectrometry (LC-MS/MS). Fibrin formation was monitored by turbidity measurements and fibrin structure visualized by confocal microscopy. Results ADAMTS-13 exhibits proteolytic activity against the Aα chain of human fibrinogen, but this is only manifest on its conformational activation. Accordingly, the GoF ADAMTS-13 variant and truncated variants such as MDTCS exhibit this activity. The cleavage site has been determined by LC-MS/MS to be Aα chain Lys225-Met226. Proteolysis of fibrinogen by GoF ADAMTS-13 impairs fibrin formation in plasma-based assays, alters clot structure and increases clot permeability. Although GoF ADAMTS-13 does not appear to proteolyse preformed cross-linked fibrin, its proteolytic activity against fibrinogen increases the susceptibility of fibrin to tissue-type plasminogen activator (t-PA)-induced lysis by plasmin and increases the fibrin clearance rate more than 8-fold compared with wild-type (WT) ADAMTS-13 (EC50 values of 3.0 ± 1.7 nm and 25.2 ± 9.7 nm, respectively) in in vitro thrombosis models. Conclusion The 'closed' conformation of ADAMTS-13 restricts its specificity and protects against fibrinogenolysis. Induced substrate promiscuity will be important as ADAMTS-13 variants are developed as potential therapeutic agents against thrombotic thrombocytopenic purpura (TTP) and other cardiovascular diseases.
Collapse
Affiliation(s)
- K South
- Centre for Haematology, Imperial College London, London, UK.
| | - M O Freitas
- Centre for Haematology, Imperial College London, London, UK
| | - D A Lane
- Centre for Haematology, Imperial College London, London, UK
| |
Collapse
|
26
|
Hsieh CL, Chang E, Tseng A, Ptak C, Wu LC, Su CL, McDonough SP, Lin YP, Chang YF. Leptospira Immunoglobulin-Like Protein B (LigB) Binds to Both the C-Terminal 23 Amino Acids of Fibrinogen αC Domain and Factor XIII: Insight into the Mechanism of LigB-Mediated Blockage of Fibrinogen α Chain Cross-Linking. PLoS Negl Trop Dis 2016; 10:e0004974. [PMID: 27622634 PMCID: PMC5021285 DOI: 10.1371/journal.pntd.0004974] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Accepted: 08/11/2016] [Indexed: 12/20/2022] Open
Abstract
The coagulation system provides a primitive but effective defense against hemorrhage. Soluble fibrinogen (Fg) monomers, composed of α, β and γ chains, are recruited to provide structural support for the formation of a hemostatic plug. Fg binds to platelets and is processed into a cross-linked fibrin polymer by the enzymatic clotting factors, thrombin and Factor XIII (FXIII). The newly formed fibrin-platelet clot can act as barrier to protect against pathogens from entering the bloodstream. Further, injuries caused by bacterial infections can be confined to the initial wound site. Many pathogenic bacteria have Fg-binding adhesins that can circumvent the coagulation pathway and allow the bacteria to sidestep containment. Fg expression is upregulated during lung infection providing an attachment surface for bacteria with the ability to produce Fg-binding adhesins. Fg binding by leptospira might play a crucial factor in Leptospira-associated pulmonary hemorrhage, the main factor contributing to lethality in severe cases of leptospirosis. The 12th domain of Leptospira immunoglobulin-like protein B (LigB12), a leptospiral adhesin, interacts with the C-terminus of FgαC (FgαCC). In this study, the binding site for LigB12 was mapped to the final 23 amino acids at the C-terminal end of FgαCC (FgαCC8). The association of FgαCC8 with LigB12 (ELISA, KD = 0.76 μM; SPR, KD = 0.96 μM) was reduced by mutations of both charged residues (R608, R611 and H614 from FgαCC8; D1061 from LigB12) and hydrophobic residues (I613 from FgαCC8; F1054 and A1065 from LigB12). Additionally, LigB12 bound strongly to FXIII and also inhibited fibrin formation, suggesting that LigB can disrupt coagulation by suppressing FXIII activity. Here, the detailed binding mechanism of a leptospiral adhesin to a host hemostatic factor is characterized for the first time and should provide better insight into the pathogenesis of leptospirosis. Leptospirosis, caused by pathogenic Leptospira spp., has been increasingly recognized as an emerging zoonosis worldwide. In human cases, clinical presentation can vary from a mild flu-like syndrome to severe multi-organ failure including hepatitis, nephritis and occasionally meningitis. Particularly, pulmonary hemorrhage has become one of the major factors leading to fatality. The host coagulation system normally can be activated to confine damage caused by bacteria. However, this spirochete has developed several virulence proteins to manipulate hemostatic factors including fibrinogen (Fg). Previously, we had observed that Leptospira immunoglobulin-like protein B (LigB) can bind to Fg and inhibit fibrin clot formation. In this study, the LigB binding site on fibrinogen was fine-mapped. The key amino acids contributing to this strong pathogen-host interaction were also identified. In addition, LigB bound to factor XIII and further interfered with the cross-linking of Fg. For the first time, a potential mechanism of leptospiral adhesin binding to fibrinogen was revealed, which should provide a better understanding of the pathogenesis of leptospirosis.
Collapse
Affiliation(s)
- Ching-Lin Hsieh
- Department of Population Medicine and Diagnostic Sciences, College of Veterinary Medicine, Cornell University, Ithaca, New York, United States of America
| | - Eric Chang
- Department of Population Medicine and Diagnostic Sciences, College of Veterinary Medicine, Cornell University, Ithaca, New York, United States of America
| | - Andrew Tseng
- Department of Population Medicine and Diagnostic Sciences, College of Veterinary Medicine, Cornell University, Ithaca, New York, United States of America
| | - Christopher Ptak
- Department of Population Medicine and Diagnostic Sciences, College of Veterinary Medicine, Cornell University, Ithaca, New York, United States of America
| | - Li-Chen Wu
- Department of Population Medicine and Diagnostic Sciences, College of Veterinary Medicine, Cornell University, Ithaca, New York, United States of America
| | - Chun-Li Su
- Department of Population Medicine and Diagnostic Sciences, College of Veterinary Medicine, Cornell University, Ithaca, New York, United States of America
| | - Sean P. McDonough
- Department of Biomedical Science, College of Veterinary Medicine, Cornell University, Ithaca, New York, United States of America
| | - Yi-Pin Lin
- Division of Infectious Disease, Wadsworth Center, New York State Department of Health, Albany, New York, United States of America
| | - Yung-Fu Chang
- Department of Population Medicine and Diagnostic Sciences, College of Veterinary Medicine, Cornell University, Ithaca, New York, United States of America
- * E-mail:
| |
Collapse
|
27
|
The interaction between fibrinogen and zymogen FXIII-A2B2 is mediated by fibrinogen residues γ390-396 and the FXIII-B subunits. Blood 2016; 128:1969-1978. [PMID: 27561317 DOI: 10.1182/blood-2016-04-712323] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Accepted: 08/22/2016] [Indexed: 01/07/2023] Open
Abstract
Coagulation transglutaminase factor XIII (FXIII) exists in circulation as heterotetrameric proenzyme FXIII-A2B2 Effectively all FXIII-A2B2 circulates bound to fibrinogen, and excess FXIII-B2 circulates in plasma. The motifs that mediate interaction of FXIII-A2B2 with fibrinogen have been elusive. We recently detected reduced binding of FXIII-A2B2 to murine fibrinogen that has γ-chain residues 390-396 mutated to alanines (Fibγ390-396A). Here, we evaluated binding features using human components, including recombinant fibrinogen variants, FXIII-A2B2, and isolated FXIII-A2 and -B2 homodimers. FXIII-A2B2 coprecipitated with wild-type (γA/γA), alternatively-spliced (γ'/γ'), and αC-truncated (Aα251) fibrinogens, whereas coprecipitation with human Fibγ390-396A was reduced by 75% (P <0001). Surface plasmon resonance showed γA/γA, γ'/γ', and Aα251 fibrinogens bound FXIII-A2B2 with high affinity (nanomolar); however, Fibγ390-396A did not bind FXIII-A2B2 These data indicate fibrinogen residues γ390-396 comprise the major binding motif for FXIII-A2B2 Compared with γA/γA clots, FXIII-A2B2 activation peptide release was 2.7-fold slower in Fibγ390-396A clots (P < .02). Conversely, activation of recombinant FXIII-A2 (lacking FXIII-B2) was similar in γA/γA and Fibγ390-396A clots, suggesting fibrinogen residues γ390-396 accelerate FXIII-A2B2 activation in a FXIII-B2-dependent mechanism. Recombinant FXIII-B2 bound γA/γA, γ'/γ', and Aα251 with similar affinities as FXIII-A2B2, but did not bind or coprecipitate with Fibγ390-396A FXIII-B2 also coprecipitated with fibrinogen from FXIII-A-deficient mouse and human plasmas. Collectively, these data indicate that FXIII-A2B2 binds fibrinogen residues γ390-396 via the B subunits, and that excess plasma FXIII-B2 is not free, but rather circulates bound to fibrinogen. These findings provide insight into assembly of the fibrinogen/FXIII-A2B2 complex in both physiologic and therapeutic situations.
Collapse
|
28
|
Abstract
Arterial and venous thromboses are major contributors to coagulation-associated morbidity and mortality. Greater understanding of mechanisms leading to thrombus formation and stability is expected to lead to improved treatment strategies. Factor XIII (FXIII) is a transglutaminase found in plasma and platelets. During thrombosis, activated FXIII cross-links fibrin and promotes thrombus stability. Recent studies have provided new information about FXIII activity during coagulation and its effects on clot composition and function. These findings reveal newly-recognized roles for FXIII in thrombosis. Herein, we review published literature on FXIII biology and effects on fibrin structure and stability, epidemiologic data associating FXIII with thrombosis, and evidence from animal models indicating FXIII has an essential role in determining thrombus stability, composition, and size.
Collapse
Affiliation(s)
- James R Byrnes
- Department of Pathology and Laboratory Medicine, McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Alisa S Wolberg
- Department of Pathology and Laboratory Medicine, McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| |
Collapse
|
29
|
|
30
|
Ranking reactive glutamines in the fibrinogen αC region that are targeted by blood coagulant factor XIII. Blood 2016; 127:2241-8. [PMID: 26951791 DOI: 10.1182/blood-2015-09-672303] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2015] [Accepted: 02/24/2016] [Indexed: 01/09/2023] Open
Abstract
Factor XIIIa (FXIIIa) introduces covalent γ-glutamyl-ε-lysyl crosslinks into the blood clot network. These crosslinks involve both the γ and α chains of fibrin. The C-terminal portion of the fibrin α chain extends into the αC region (210-610). Crosslinks within this region help generate a stiffer clot, which is more resistant to fibrinolysis. Fibrinogen αC (233-425) contains a binding site for FXIIIa and three glutamines Q237, Q328, and Q366 that each participate in physiological crosslinking reactions. Although these glutamines were previously identified, their reactivities toward FXIIIa have not been ranked. Matrix-assisted laser desorption/ionization time of flight (MALDI-TOF) mass spectrometry and nuclear magnetic resonance (NMR) methods were thus used to directly characterize these three glutamines and probe for sources of FXIIIa substrate specificity. Glycine ethyl ester (GEE) and ammonium chloride served as replacements for lysine. Mass spectrometry and 2D heteronuclear single quantum coherence NMR revealed that Q237 is rapidly crosslinked first by FXIIIa followed by Q366 and Q328. Both (15)NH4Cl and (15)N-GEE could be crosslinked to the three glutamines in αC (233-425) with a similar order of reactivity as observed with the MALDI-TOF mass spectrometry assay. NMR studies using the single αC mutants Q237N, Q328N, and Q366N demonstrated that no glutamine is dependent on another to react first in the series. Moreover, the remaining two glutamines of each mutant were both still reactive. Further characterization of Q237, Q328, and Q366 is important because they are located in a fibrinogen region susceptible to physiological truncations and mutation. The current results suggest that these glutamines play distinct roles in fibrin crosslinking and clot architecture.
Collapse
|
31
|
Fries D, Bachler M, Hermann M. Fibrinogen (FI). TRANSFUSIONSASSOZIIERTE PHARMAKOTHERAPIE 2016. [PMCID: PMC7121223 DOI: 10.1007/978-3-662-47258-3_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Das Hauptsubstrat der Gerinnung ist Fibrinogen (FI). Bei akuter Blutung ist es zumeist der erste Gerinnungsfaktor, der kritische Grenzwerte erreicht (150–200 mg/dl). FI kann hervorragend mittels point-of-care-tauglicher viskoelastischer Methoden (Thrombelstographie oder Thrombelastometrie) monitiert werden. Die Substitution kann mittels Frischplasma, Cryopräzipitat oder Fibrinogenkonzentrat erfolgen. Frischplasma ist nicht besonders effektiv, mit einer erhöhten Morbidität, insbesondere bei kritisch Kranken, sowie mit Volumenbelastung assoziiert. Cryopräzipitat wird in einigen europäischen Ländern nicht angeboten. Die Gabe von Fibrinogenkonzentrat wird in verschiedenen Leitlinien empfohlen. Als Akut-Phase-Protein kann FI physiologischerweise bei Entzündungsprozessen, schweren Verletzungen sowie nach großen Operationen in kurzer Zeit auf über 1000 mg/dl ansteigen; wobei hier Fibrinogenspaltprodukte anti-inflammatorische und sogar antibakterielle Eigenschaften haben.
Collapse
|
32
|
Tiwari PB, Üren A, He J, Darici Y, Wang X. Note: Model identification and analysis of bivalent analyte surface plasmon resonance data. THE REVIEW OF SCIENTIFIC INSTRUMENTS 2015; 86:106107. [PMID: 26521004 PMCID: PMC4617740 DOI: 10.1063/1.4933318] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
Surface plasmon resonance (SPR) is a widely used, affinity based, label-free biophysical technique to investigate biomolecular interactions. The extraction of rate constants requires accurate identification of the particular binding model. The bivalent analyte model involves coupled non-linear differential equations. No clear procedure to identify the bivalent analyte mechanism has been established. In this report, we propose a unique signature for the bivalent analyte model. This signature can be used to distinguish the bivalent analyte model from other biphasic models. The proposed method is demonstrated using experimentally measured SPR sensorgrams.
Collapse
Affiliation(s)
| | - Aykut Üren
- Department of Oncology, Georgetown University, Washington, District of Columbia 20057, USA
| | - Jin He
- Department of Physics, Florida International University, Miami, Florida 33199, USA
| | - Yesim Darici
- Department of Physics, Florida International University, Miami, Florida 33199, USA
| | - Xuewen Wang
- Department of Physics, Florida International University, Miami, Florida 33199, USA
| |
Collapse
|
33
|
Walton BL, Byrnes JR, Wolberg AS. Fibrinogen, red blood cells, and factor XIII in venous thrombosis. J Thromb Haemost 2015; 13 Suppl 1:S208-15. [PMID: 26149026 PMCID: PMC5975093 DOI: 10.1111/jth.12918] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Cardiovascular disease is the leading cause of death and disability worldwide. Among cardiovascular causes of death, venous thrombosis (VT) is ranked third most common in the world. Venous thrombi have high red blood cell and fibrin content; however, the pathophysiologic mechanisms that contribute to venous thrombus composition and stability are still poorly understood. This article reviews biological, biochemical, and biophysical contributions of fibrinogen, factor XIII, and red blood cells to VT, and new evidence suggesting interactions between these components mediate venous thrombus composition and size.
Collapse
Affiliation(s)
- B L Walton
- Department of Pathology and Laboratory Medicine and McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - J R Byrnes
- Department of Pathology and Laboratory Medicine and McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - A S Wolberg
- Department of Pathology and Laboratory Medicine and McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
34
|
Cordell PA, Newell LM, Standeven KF, Adamson PJ, Simpson KR, Smith KA, Jackson CL, Grant PJ, Pease RJ. Normal Bone Deposition Occurs in Mice Deficient in Factor XIII-A and Transglutaminase 2. Matrix Biol 2015; 43:85-96. [DOI: 10.1016/j.matbio.2015.02.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Revised: 02/03/2015] [Accepted: 02/03/2015] [Indexed: 11/29/2022]
|
35
|
Aleman MM, Byrnes JR, Wang JG, Tran R, Lam WA, Di Paola J, Mackman N, Degen JL, Flick MJ, Wolberg AS. Factor XIII activity mediates red blood cell retention in venous thrombi. J Clin Invest 2014; 124:3590-600. [PMID: 24983320 DOI: 10.1172/jci75386] [Citation(s) in RCA: 154] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Accepted: 05/21/2014] [Indexed: 11/17/2022] Open
Abstract
Venous thrombi, fibrin- and rbc-rich clots triggered by inflammation and blood stasis, underlie devastating, and sometimes fatal, occlusive events. During intravascular fibrin deposition, rbc are thought to become passively trapped in thrombi and therefore have not been considered a modifiable thrombus component. In the present study, we determined that activity of the transglutaminase factor XIII (FXIII) is critical for rbc retention within clots and directly affects thrombus size. Compared with WT mice, mice carrying a homozygous mutation in the fibrinogen γ chain (Fibγ390-396A) had a striking 50% reduction in thrombus weight due to reduced rbc content. Fibrinogen from mice harboring the Fibγ390-396A mutation exhibited reduced binding to FXIII, and plasma from these mice exhibited delayed FXIII activation and fibrin crosslinking, indicating these residues mediate FXIII binding and activation. FXIII-deficient mice phenocopied mice carrying Fibγ390-396A and produced smaller thrombi with fewer rbc than WT mice. Importantly, FXIII-deficient human clots also exhibited reduced rbc retention. The addition of FXIII to FXIII-deficient clots increased rbc retention, while inhibition of FXIII activity in normal blood reduced rbc retention and produced smaller clots. These findings establish the FXIII-fibrinogen axis as a central determinant in venous thrombogenesis and identify FXIII as a potential therapeutic target for limiting venous thrombosis.
Collapse
|
36
|
Galanakis DK, Neerman-Arbez M, Brennan S, Rafailovich M, Hyder L, Travlou O, Papadakis E, Manco-Johnson MJ, Henschen A, Scharrer I. Thromboelastographic phenotypes of fibrinogen and its variants: clinical and non-clinical implications. Thromb Res 2014; 133:1115-23. [PMID: 24679643 DOI: 10.1016/j.thromres.2014.03.026] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Revised: 02/20/2014] [Accepted: 03/04/2014] [Indexed: 10/25/2022]
Abstract
INTRODUCTION Thromboelastography (TEG), a widely used clinical point of care coagulation test, is poorly understood. To investigate its fibrin determinants we used normal and variant fibrinogen isolates. MATERIALS AND METHODS We focused mainly on the TEG maximum signal amplitude (MA), a shear modulus and clot stiffness indicator. Isolates included normal des-αC, cord, and abnormal congenital variants with amino acid substitutions or deletions that impaired fibrin polymerization. Heterophenotypic congenital isolates were from cryoprecipitate-depleted plasma owing to their more diminished clot MA than their cryoprecipitate counterparts. By colorimetric assay, the amount of fibrinogen adsorbed by untreated TEG cups was 83.5±12.4 pM/cm(2), n=18. Thrombin-induced clots were obtained at pH6.4 or 7.4, the latter containing 8mM CaCl2, and 14% afibrinogenemic plasma with and without gel-sieved platelets. RESULTS AND CONCLUSIONS Measured by the water droplet contact angle, >90% reduction of surface hydrophobicity by exposure of TEG cup and pin to ozone plasma decreased MA by 74%. Increasing normal fibrinogen or thrombin concentrations progressively increased MA. Platelets increased MA further ~2 fold, except for ≥10 fold for des-αC clots. Examined in the absence of platelets, MA of heterophenotypic fibrin variants averaged 21%, n=15. The results imply that essential MA determinants include hydrophobic fibrinogen/fibrin adsorption and each polymerization contact site, with substantial enhancement by platelets. Also, cryoprecipitate-harvested soluble fibrinogen/fibrin complexes contained mostly normal molecules, while cryoprecipitate-depleted plasma contained mostly variant molecules. Moreover, significantly decreased MA by fibrinogen anomalies and/or low level thrombin generation can potentially impact clinical interpretation of MA.
Collapse
Affiliation(s)
| | | | | | | | - Luke Hyder
- Stony Brook University, Stony Brook, USA
| | | | | | | | | | | |
Collapse
|
37
|
Biswas A, Ivaskevicius V, Thomas A, Oldenburg J. Coagulation factor XIII deficiency. Diagnosis, prevalence and management of inherited and acquired forms. Hamostaseologie 2014; 34:160-6. [PMID: 24503678 DOI: 10.5482/hamo-13-08-0046] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2013] [Accepted: 01/28/2014] [Indexed: 11/05/2022] Open
Abstract
The plasma circulating zymogenic coagulation factor XIII (FXIII) is a protransglutaminase, which upon activation by thrombin and calcium cross-links preformed fibrin clots/fibrinolytic inhibitors making them mechanically stable and less susceptible to fibrinolysis. The zymogenic plasma FXIII molecule is a heterotetramer composed of two catalytic FXIII-A and two protective FXIII-B subunits. Factor XIII deficiency resulting from inherited or acquired causes can result in pathological bleeding episodes. A diverse spectrum of mutations have been reported in the F13A1 and F13B genes which cause inherited severe FXIII deficiency. The inherited severe FXIII deficiency, which is a rare coagulation disorder with a prevalence of 1 in 4 million has been the prime focus of clinical and genetic investigations owing to the severity of the bleeding phenotype associated with it. Recently however, with a growing understanding into the pleiotropic roles of FXIII, the fairly frequent milder form of FXIII deficiency caused by heterozygous mutations has become one of the subjects of investigative research. The acquired form of FXIII deficiency is usually caused by generation of autoantibodies or hyperconsumption in other disease states such as disseminated intravascular coagulation. Here, we update the knowledge about the pathophysiology of factor XIII deficiency and its therapeutic options.
Collapse
Affiliation(s)
- A Biswas
- Arijit Biswas Institute of Experimental Haematology and Transfusion Medicine, University Clinic Bonn, Sigmund Freud Str. 25 53127 Bonn, Germany, Tel. +49/(0)228/28 71 94 28, Fax +49/(0)228/28 71 43 20, E-mail:
| | | | | | | |
Collapse
|
38
|
Westbury SK, Duval C, Philippou H, Brown R, Lee KR, Murden SL, Phillips E, Reilly-Stitt C, Whalley D, Ariëns RA, Mumford AD. Partial deletion of the αC-domain in the Fibrinogen Perth variant is associated with thrombosis, increased clot strength and delayed fibrinolysis. Thromb Haemost 2013; 110:1135-44. [PMID: 24048413 DOI: 10.1160/th13-05-0408] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2013] [Accepted: 08/07/2013] [Indexed: 12/17/2022]
Abstract
Genetic fibrinogen (FGN) variants that are associated with bleeding or thrombosis may be informative about fibrin polymerisation, structure and fibrinolysis. We report a four generation family with thrombosis and heritable dysfibrinogenaemia segregating with a c.[1541delC];[=] variation in FGA (FGN-Perth). This deletion predicts a truncated FGN αC-domain with an unpaired terminal Cys at residue 517 of FGN-Aα. In keeping with this, SDS-PAGE of purified FGN-Perth identified a truncated FGN-Aα chain with increased co-purification of albumin, consistent with disulphide bonding to the terminal Cys of the variant FGN-Aα. Clot visco-elastic strength in whole blood containing FGN-Perth was greater than controls and tPA-mediated fibrinolysis was delayed. In FGN-Perth plasma and in purified FGN-Perth, there was markedly reduced final turbidity after thrombin-mediated clot generation. Consistent with this, FGN-Perth formed tighter, thinner fibrin fibres than controls indicating defective lateral aggregation of protofibrils. Clots generated with thrombin in FGN-Perth plasma were resistant to tPA-mediated fibrinolysis. FGN-Perth clot also displayed impaired tPA-mediated plasmin generation but incorporated α2-antiplasmin at a similar rate to control. Impaired fibrinolysis because of defective plasmin generation potentially explains the FGN-Perth clinical phenotype. These findings highlight the importance of the FGN αC-domain in the regulation of clot formation and fibrinolysis.
Collapse
Affiliation(s)
- Sarah K Westbury
- Dr. Sarah Westbury, University of Bristol, Level 7 Bristol Royal Infirmary, Bristol, BS2 8HW, United Kingdom, Tel +44 117 3423152; Fax +44 117 3424036, E-mail
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Ariëns RAS. Fibrin(ogen) and thrombotic disease. J Thromb Haemost 2013; 11 Suppl 1:294-305. [PMID: 23809133 DOI: 10.1111/jth.12229] [Citation(s) in RCA: 122] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2013] [Accepted: 03/08/2013] [Indexed: 01/21/2023]
Abstract
Fibrinogen is an abundant plasma protein that, when converted to fibrin by thrombin, provides the main building blocks for the clot. Dys-, a-, and hypo-fibrinogenemias have been variably linked to a normal phenotype, bleeding or even thrombosis. Meanwhile, increased fibrinogen concentrations in the blood have been associated with risk for thrombosis. More recently, studies have focussed on abnormal fibrin structure as a cause for thrombosis. Fibrin clots that have high fiber density and increased resistance to fibrinolysis have been consistently associated with risk for thrombosis. Fibrin structure measurements can (i) provide an overall assessment of hemostatic capacity of a sample, (ii) include effects of thrombin generation and fibrinogen concentrations, (iii) include effects of fibrinogen mutations, polymorphisms, and modifications, and (iv) give an indication of clot mechanical strength and resistance to fibrinolysis. A fibrinogen splice variation of the γ-chain (γ') is discussed as a model for changes in fibrin structure in relation to thrombosis. Results from prospective studies on fibrin structure are awaited. Studies of fibrin formation under flow, interactions of fibrin with blood cells, the mechanical properties of the fibrin clot, and nanoscale/molecular characterization of fibrin formation are likely to expose new causal mechanisms for the role of fibrin in thrombotic disease. Future studies into the causality and mechanisms may lead to new opportunities using fibrin structure in the diagnosis or treatment of thrombosis.
Collapse
Affiliation(s)
- R A S Ariëns
- Division of Cardiovascular and Diabetes Research, Leeds Institute for Genetics, Health and Therapeutics, Multidisciplinary Cardiovascular Research Centre, University of Leeds, Leeds, UK.
| |
Collapse
|
40
|
Abstract
In this issue of Blood, Smith and colleagues report on the functional role of the interaction between these 2 proteins by studying the involved binding sites responsible for clot stabilization.(1)
Collapse
|
41
|
Soendergaard C, Kvist PH, Seidelin JB, Nielsen OH. Tissue-regenerating functions of coagulation factor XIII. J Thromb Haemost 2013; 11:806-16. [PMID: 23406195 DOI: 10.1111/jth.12169] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2012] [Accepted: 02/04/2013] [Indexed: 12/13/2022]
Abstract
The protransglutaminase factor XIII (FXIII) has recently attracted attention within the field of tissue regeneration, as it has been found that FXIII significantly influences wound healing by exerting a multitude of functions. It supports hemostasis by enhancing platelet adhesion to damaged endothelium, and by its cross-linking activity it stabilizes the formed fibrin clot. Furthermore, FXIII limits bacterial dissemination from the wound and incorporates macromolecules of importance for cellular infiltration, supporting cell migration and survival. FXIII-mediated complex formation of the vascular endothelial growth factor receptor 2 and αV β3 integrin is important for angiogenesis, supporting the formation of granulation tissue. Chronic inflammatory conditions involving bleeding and activation of the coagulation cascade have been shown to lead to reduced FXIII levels in plasma. Of particular importance for this review is the fact that patients suffering from inflammatory bowel disease (IBD) have reduced FXIII antigen levels and activity. Furthermore, these patients show impaired mucosal healing, which supports the inflammatory state of the disease. This review summarizes the role of FXIII in the healing of wounds, and briefly summarizes the previous use of FXIII in clinical settings. Moreover, it addresses the potential role for FXIII as a therapeutic agent in the healing of persistent wounds during chronic conditions, with an emphasis on IBD.
Collapse
Affiliation(s)
- C Soendergaard
- Department of Gastroenterology, Medical Section, Herlev Hospital, University of Copenhagen, Copenhagen, Denmark.
| | | | | | | |
Collapse
|
42
|
Smith E, Cardinali B, Ping L, Ariëns R, Philippou H. Elimination of coagulation factor XIII from fibrinogen preparations. J Thromb Haemost 2013; 11:993-5. [PMID: 23413924 PMCID: PMC3656969 DOI: 10.1111/jth.12174] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Accepted: 02/12/2013] [Indexed: 11/28/2022]
Affiliation(s)
- E.L. Smith
- Division of Cardiovascular and Diabetes Research, MCRC, The LIGHT Laboratories, University of Leeds, Leeds, UK
| | - B. Cardinali
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, North Carolina 27599-7525, USA
| | - L. Ping
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, North Carolina 27599-7525, USA
| | - R.A.S. Ariëns
- Division of Cardiovascular and Diabetes Research, MCRC, The LIGHT Laboratories, University of Leeds, Leeds, UK
| | - H. Philippou
- Division of Cardiovascular and Diabetes Research, MCRC, The LIGHT Laboratories, University of Leeds, Leeds, UK
| |
Collapse
|
43
|
The activation peptide cleft exposed by thrombin cleavage of FXIII-A(2) contains a recognition site for the fibrinogen α chain. Blood 2013; 121:2117-26. [PMID: 23303819 DOI: 10.1182/blood-2012-07-446393] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Formation of a stable fibrin clot is dependent on interactions between factor XIII and fibrin. We have previously identified a key residue on the αC of fibrin(ogen) (Glu396) involved in binding activated factor XIII-A(2) (FXIII-A(2)*); however, the functional role of this interaction and binding site(s) on FXIII-A(2)* remains unknown. Here we (1) characterized the functional implications of this interaction; (2) identified by liquid-chromatography-tandem mass spectrometry the interacting residues on FXIII-A(2)* following chemical cross-linking of fibrin(ogen) αC389-402 peptides to FXIII-A(2)*; and (3) carried out molecular modeling of the FXIII-A(2)*/peptide complex to identify contact site(s) involved. Results demonstrated that inhibition of the FXIII-A(2)*/αC interaction using αC389-402 peptide (Pep1) significantly decreased incorporation of biotinamido-pentylamine and α2-antiplasmin to fibrin, and fibrin cross-linking, in contrast to Pep1-E396A and scrambled peptide controls. Pep1 did not inhibit transglutaminase-2 activity, and incorporation of biotinyl-TVQQEL to fibrin was only weakly inhibited. Molecular modeling predicted that Pep1 binds the activation peptide cleft (AP-cleft) within the β-sandwich domain of FXIII-A(2)* localizing αC cross-linking Q366 to the FXIII-A(2)* active site. Our findings demonstrate that binding of fibrin αC389-402 to the AP-cleft is fundamental to clot stabilization and presents this region of FXIII-A(2)* as a potential site involved in glutamine-donor substrate recognition.
Collapse
|
44
|
|
45
|
Levy JH, Greenberg C. Biology of Factor XIII and clinical manifestations of Factor XIII deficiency. Transfusion 2012; 53:1120-31. [DOI: 10.1111/j.1537-2995.2012.03865.x] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
46
|
Howes JM, Richardson VR, Smith KA, Schroeder V, Somani R, Shore A, Hess K, Ajjan R, Pease RJ, Keen JN, Standeven KF, Carter AM. Complement C3 is a novel plasma clot component with anti-fibrinolytic properties. Diab Vasc Dis Res 2012; 9:216-25. [PMID: 22253322 DOI: 10.1177/1479164111432788] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND AND METHOD Increased plasma clot density and prolonged lysis times are associated with cardiovascular disease. In this study, we employed a functional proteomics approach to identify novel clot components which may influence clot phenotypes. RESULTS Analysis of perfused, solubilised plasma clots identified inflammatory proteins, including complement C3, as novel clot components. Analysis of paired plasma and serum samples confirmed concentration-dependent incorporation of C3 into clots. Surface plasmon resonance indicated high-affinity binding interactions between C3 and fibrinogen and fibrin. Turbidimetric clotting and lysis assays indicated C3 impaired fibrinolysis in a concentration-dependent manner, both in vitro and ex vivo. CONCLUSION These data indicate functional interactions between complement C3 and fibrin leading to prolonged fibrinolysis. These interactions are physiologically relevant in the context of protection following injury and suggest a mechanistic link between increased plasma C3 concentration and acute cardiovascular thrombotic events.
Collapse
Affiliation(s)
- Joanna-Marie Howes
- Division of Cardiovascular & Diabetes Research, Leeds Institute of Genetics Health & Therapeutics, University of Leeds, UK
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Hess K, Alzahrani SH, Mathai M, Schroeder V, Carter AM, Howell G, Koko T, Strachan MWJ, Price JF, Smith KA, Grant PJ, Ajjan RA. A novel mechanism for hypofibrinolysis in diabetes: the role of complement C3. Diabetologia 2012; 55:1103-13. [PMID: 21918806 DOI: 10.1007/s00125-011-2301-7] [Citation(s) in RCA: 77] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2011] [Accepted: 08/12/2011] [Indexed: 12/14/2022]
Abstract
AIMS/HYPOTHESIS Impaired fibrin clot lysis is a key abnormality in diabetes and complement C3 is one protein identified in blood clots. This work investigates the mechanistic pathways linking C3 and hypofibrinolysis in diabetes using ex vivo/in vitro studies. METHODS Fibrinolysis and C3 plasma levels were determined in type 1 diabetic patients and healthy controls, and the effects of glycaemia investigated. C3 incorporation into fibrin clots and modulation of fibrinolysis were analysed by ELISA, immunoblotting, turbidimetric assays and electron and confocal microscopy. RESULTS Clot lysis time was longer in diabetic children than in controls (599 ± 18 and 516 ± 12 s respectively; p < 0.01), C3 levels were higher in diabetic children (0.55 ± 0.02 and 0.43 ± 0.02 g/l respectively; p < 0.01) and both were affected by improving glycaemia. An interaction between C3 and fibrin was confirmed by the presence of lower protein levels in sera compared with corresponding plasma and C3 detection in plasma clots by immunoblot. In a purified system, C3 was associated with thinner fibrin fibres and more prolongation of lysis time of clots made from fibrinogen from diabetic participants compared with controls (244 ± 64 and 92 ± 23 s respectively; p < 0.05). Confocal microscopy showed higher C3 incorporation into diabetic clots compared with controls, and fully formed clot lysis was prolonged by 764 ± 76 and 428 ± 105 s respectively (p < 0.05). Differences in lysis, comparing diabetes and controls, were not related to altered plasmin generation or C3-fibrinogen binding assessed by plasmon resonance. CONCLUSIONS/INTERPRETATION C3 incorporation into clots from diabetic fibrinogen is enhanced and adversely affects fibrinolysis. This may be one novel mechanism for compromised clot lysis in diabetes, potentially offering a new therapeutic target.
Collapse
Affiliation(s)
- K Hess
- University of Leeds, Leeds, UK
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Tsurupa G, Pechik I, Litvinov RI, Hantgan RR, Tjandra N, Weisel JW, Medved L. On the mechanism of αC polymer formation in fibrin. Biochemistry 2012; 51:2526-38. [PMID: 22397628 DOI: 10.1021/bi2017848] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Our previous studies revealed that the fibrinogen αC-domains undergo conformational changes and adopt a physiologically active conformation upon their self-association into αC polymers in fibrin. In the present study, we analyzed the mechanism of αC polymer formation and tested our hypothesis that self-association of the αC-domains occurs through the interaction between their N-terminal subdomains and may include β-hairpin swapping. Our binding experiments performed by size-exclusion chromatography and optical trap-based force spectroscopy revealed that the αC-domains self-associate exclusively through their N-terminal subdomains, while their C-terminal subdomains were found to interact with the αC-connectors that tether the αC-domains to the bulk of the molecule. This interaction should reinforce the structure of αC polymers and provide the proper orientation of their reactive residues for efficient cross-linking by factor XIIIa. Molecular modeling of self-association of the N-terminal subdomains confirmed that the hypothesized β-hairpin swapping does not impose any steric hindrance. To "freeze" the conformation of the N-terminal subdomain and prevent the hypothesized β-hairpin swapping, we introduced by site-directed mutagenesis an extra disulfide bond between two β-hairpins of the bovine Aα406-483 fragment corresponding to this subdomain. The experiments performed by circular dichroism revealed that Aα406-483 mutant containing Lys429Cys/Thr463Cys mutations preserved its β-sheet structure. However, in contrast to wild-type Aα406-483, this mutant had lower tendency for oligomerization, and its structure was not stabilized upon oligomerization, in agreement with the above hypothesis. On the basis of the results obtained and our previous findings, we propose a model of fibrin αC polymer structure and molecular mechanism of assembly.
Collapse
Affiliation(s)
- Galina Tsurupa
- Center for Vascular and Inflammatory Diseases and the Department of Biochemistry, University of Maryland School of Medicine, Baltimore, Maryland 21201, United States
| | | | | | | | | | | | | |
Collapse
|
49
|
|