1
|
Kovac M, Ignjatovic V, Orlando C, Bereczky Z, Hunt BJ. The use of direct oral anticoagulants in the secondary prevention of venous thromboembolism in patients with severe thrombophilia: communication from the ISTH SSC Subcommittee on Physiological Anticoagulants and Thrombophilia. J Thromb Haemost 2024; 22:3322-3329. [PMID: 39233011 DOI: 10.1016/j.jtha.2024.08.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 08/21/2024] [Accepted: 08/26/2024] [Indexed: 09/06/2024]
Abstract
Direct oral anticoagulants (DOACs) are the first-line anticoagulants for the secondary prevention of venous thromboembolism (VTE). However, patients with severe inherited thrombophilias represent a group in whom the efficiency and safety of DOACs is poorly studied. In this communication, we focus on the utility of DOACs in the secondary prevention of VTE in patients with severe thrombophilia. Current evidence is based only on cohort or single-center studies, and poor data are available on compliance of the patients in the studies. Analysis of the studies suggested that full-dose DOACs and vitamin K antagonists have a similar efficacy and bleeding risk in the secondary prevention of VTE in patients with thrombophilia, with a low hazard ratio for recurrent VTE calculated from cohort studies for DOAC vs warfarin, ranging from 0.3 to 0.75. We wish to highlight that treatment failure is greater in those with severe forms of protein S deficiency (below 20%) and possibly in antithrombin deficiency type II heparin-binding site homozygous Budapest 3. In summary, the current approach to using DOACs in patients with severe thrombophilia is dependent on clinical judgment and experience. Limited evidence suggests that for those with severe thrombophilias, full-dose DOACs have similar utility as vitamin K antagonists. We recommend caution in using low-dose DOACs due to lack of evidence. Ideally, large randomized multicenter studies are required to develop a reliable treatment algorithm.
Collapse
Affiliation(s)
- Mirjana Kovac
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia; Hemostasis Department, Blood Transfusion Institute of Serbia, Belgrade, Serbia.
| | - Vera Ignjatovic
- Johns Hopkins All Children's Institute for Clinical & Translational Research, St. Petersburg, Florida, USA; Johns Hopkins University School of Medicine, St. Petersburg, Florida, USA
| | - Christelle Orlando
- Department of Hematology, Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| | - Zsuzsanna Bereczky
- Division of Clinical Laboratory Science, Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Beverley J Hunt
- Haemostasis & Thrombosis Centre, Guy's and St Thomas' Hospital NHS Foundation Trust, Westminster Bridge Road, London, United Kingdom
| |
Collapse
|
2
|
Shimonishi N, Morishita E, Ogiwara K, Maruyama K, Yoshida J, Horie K, Nogami K. A novel factor V compound heterozygous mutation associated with thrombosis (Y1961C; FV-Kanazawa, together with 1982_1983del). J Thromb Haemost 2024; 22:2810-2822. [PMID: 38950780 DOI: 10.1016/j.jtha.2024.06.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 05/29/2024] [Accepted: 06/17/2024] [Indexed: 07/03/2024]
Abstract
BACKGROUND Factor (F)V is pivotal in both procoagulant and anticoagulant mechanisms. The present report describes a novel F5 mutation in a FV-deficient patient (FV activity, 6 IU/dL; FV antigen, 32 IU/dL) complicated by recurrent deep vein thrombosis. The patient demonstrated activated protein C resistance (APCR) with compound heterozygous mutations consisting of FV-Y1961C (FVKanazawa) and FV-1982_1983del. OBJECTIVES To clarify thrombotic mechanisms associated with this FV abnormality. METHODS AND RESULTS Levels of FV-1982_1983del were below the detection sensitivity in our expression experiments using human embryonic kidney 293T cells, and analyses were targeted, therefore, on the FV-Y1961C mutation. Activated partial thromboplastin time-based clotting assays demonstrated that FV-Y1961C exhibited APCR and that the reduced activated protein C (APC) susceptibility in FVa-Y1961C resulted in a marked depression of APC-catalyzed inactivation with delayed cleavage at Arg506 and little cleavage at Arg306 with or without protein S. The APC cofactor activity of FV-Y1961C in APC-catalyzed FVIIIa inactivation promoted by Arg336 cleavage in FVIII was impaired. The binding affinity of FVa-Y1961C to phospholipid membranes was reduced in reactions involving APC/protein S-catalyzed inactivation and in prothrombinase activity. Furthermore, the addition of FVa-Y1961C to plasma failed to inhibit tissue factor-induced procoagulant function. These characteristics were similar to those of FV-W1920R (FVNara) and FV-A2086D (FVBesançon). CONCLUSION We identified a compound heterozygous FV-Y1961C mutation in the C1 domain representing a novel FV mutation (FVKanazawa) resulting in not only APCR due to impaired FVa susceptibility and FV cofactor activity for APC function but also impaired inhibition of tissue factor-induced procoagulant function. These defects in anticoagulant function associated with FV in FV-Y1961C contributed to a prothrombotic state.
Collapse
Affiliation(s)
- Naruto Shimonishi
- Department of Pediatrics, Nara Medical University, Kashihara, Japan; The Course of Thrombosis and Hemostasis Molecular Pathology, Nara Medical University, Kashihara, Japan
| | - Eriko Morishita
- Department of Clinical Laboratory Science, Kanazawa University, Kanazawa, Japan
| | - Kenichi Ogiwara
- Department of Pediatrics, Nara Medical University, Kashihara, Japan.
| | - Keiko Maruyama
- Department of Clinical Laboratory Science, Kanazawa University, Kanazawa, Japan; Department of Molecular Pathogenesis, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Junko Yoshida
- Department of Physiology II, Nara Medical University, Kashihara, Japan
| | - Kyoji Horie
- Department of Physiology II, Nara Medical University, Kashihara, Japan
| | - Keiji Nogami
- Department of Pediatrics, Nara Medical University, Kashihara, Japan
| |
Collapse
|
3
|
Yakovleva E, Zhang B. Clinical, Laboratory, Molecular, and Reproductive Aspects of Combined Deficiency of Factors V and VIII. Semin Thromb Hemost 2024. [PMID: 39209292 DOI: 10.1055/s-0044-1789019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Congenital combined deficiency of factor V (FV) and factor VIII (FVIII; F5F8D, OMIM 227300) is a rare hereditary coagulopathy and accounts for approximately 3% of cases of rare coagulation disorders. The prevalence of this disease in the general population is estimated to be 1:1,000,000 and is significantly higher in regions where consanguineous marriages are permitted, such as the Mideast and South Asia. The disease has an autosomal recessive mode of inheritance and therefore occurs with an equal incidence among males and females. Heterozygous mutation carriers usually do not have clinical manifestations. The molecular basis of this disease differs from that of stand-alone congenital deficiencies of FVIII and FV. F5F8D is caused by mutations in either LMAN1 or MCFD2, which encode components of a cargo receptor complex for endoplasmic reticulum to Golgi transport of FV and FVIII, leading to defects in an intracellular transport pathway shared by these two coagulation factors. Congenital combined deficiency of FV and FVIII is characterized by decreased activities of both FV and FVIII in plasma, usually to 5 to 30% of normal. Clinical manifestations in most cases are represented by mild or moderate hemorrhagic syndrome. The simultaneous decreases of two coagulation factors present complications in the diagnosis and management of the disease. In female patients, the disease requires a special approach for family planning, pregnancy management, and parturition. This review summarizes recent progress in clinical, laboratory, and molecular understanding of this disorder.
Collapse
Affiliation(s)
- Elena Yakovleva
- Clinical and Diagnostic Department of Hematology and Hemostasis Disorders, National Medical Research Center for Hematology, Novy Zykovsky, Russia
| | - Bin Zhang
- Genomic Medicine Institute, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio
| |
Collapse
|
4
|
Mehic D, Gebhart J, Pabinger I. Bleeding Disorder of Unknown Cause: A Diagnosis of Exclusion. Hamostaseologie 2024; 44:287-297. [PMID: 38412996 DOI: 10.1055/a-2263-5706] [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: 02/29/2024] Open
Abstract
Patients with an unexplained mild to moderate bleeding tendency are diagnosed with bleeding disorder of unknown cause (BDUC), a classification reached after ruling out other mild to moderate bleeding disorders (MBD) including von Willebrand disease (VWD), platelet function defects (PFDs), coagulation factor deficiencies (CFDs), and non-hemostatic causes for bleeding. This review outlines our diagnostic approach to BDUC, a diagnosis of exclusion, drawing on current guidelines and insights from the Vienna Bleeding Biobank (VIBB). According to guidelines, we diagnose VWD based on VWF antigen and/or activity levels ≤50 IU/dL, with repeated VWF testing if VWF levels are <80 IU/dL. This has been introduced in our clinical routine after our findings of diagnostically relevant fluctuations of VWF levels in a high proportion of MBD patients. PFDs are identified through repeated abnormalities in light transmission aggregometry (LTA), flow cytometric mepacrine fluorescence, and glycoprotein expression analysis. Nevertheless, we experience diagnostic challenges with regard to reproducibility and unspecific alterations of LTA. For factor (F) VIII and FIX deficiency, a cutoff of 50% is utilized to ensure detection of mild hemophilia A or B. We apply established cutoffs for other rare CFD being aware that these do not clearly reflect the causal role of the bleeding tendency. Investigations into very rare bleeding disorders due to hyperfibrinolysis or increase in natural anticoagulants are limited to cases with a notable family history or distinct bleeding phenotypes considering cost-effectiveness. While the pathogenesis of BDUC remains unknown, further explorations of this intriguing area may reveal new mechanisms and therapeutic targets.
Collapse
Affiliation(s)
- Dino Mehic
- Clinical Division of Haematology and Haemostaseology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Johanna Gebhart
- Clinical Division of Haematology and Haemostaseology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Ingrid Pabinger
- Clinical Division of Haematology and Haemostaseology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
5
|
Arroyo-Ataz G, Yagüe AC, Breda JC, Mazzilli SA, Jones D. Transcriptional, developmental, and functional parallels of lymphatic and venous smooth muscle. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.18.604042. [PMID: 39091770 PMCID: PMC11291064 DOI: 10.1101/2024.07.18.604042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
Lymphatic muscle cells (LMCs) are indispensable for lymphatic vessel contraction and their aberrant recruitment or absence is associated with both primary and secondary lymphedema. Despite their critical role in lymphatic vessel function, the transcriptomic and developmental basis that confer the unique contractile properties to LMCs are largely undefined. In this study, we employed single-cell RNA sequencing (scRNAseq), lineage tracing and in vivo imaging to investigate the basis for the hybrid cardiomyocyte and blood vascular smooth muscle cell (SMC) characteristics that have been described for LMCs. Using scRNAseq, the transcriptomes of LMC and venous SMCs from the murine hindlimb exhibited more similarities than differences, although both were markedly distinct from that of arteriole SMCs in the same tissue. Functionally, both lymphatic vessels and blood vessels in the murine hindlimb displayed pulsatile contractility. However, despite expressing genes that overlap with the venous SMC transcriptome, through lineage tracing we show that LMCs do not originate from Myh11+ SMC progenitors. Previous studies have shown that LMCs express cardiac-related genes, whereas in our study we found that arteriole SMCs, but not LMCs, expressed cardiac-related genes. Through lineage tracing, we demonstrate that a subpopulation of LMCs and SMCs originate from WT1+ mesodermal progenitors, which are known to give rise to SMCs. LMCs, however, do not derive from Nkx2.5+ cardiomyocyte progenitors. Overall, our findings suggest that venous SMCs and LMCs and may derive from a related mesodermal progenitor and adopt a similar gene expression program that enable their contractile properties.
Collapse
Affiliation(s)
- Guillermo Arroyo-Ataz
- Department of Pathology & Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, 670 Albany Street, Boston, Massachusetts 02118, USA
| | - Alejandra Carrasco Yagüe
- Department of Pathology & Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, 670 Albany Street, Boston, Massachusetts 02118, USA
| | - Julia C. Breda
- Department of Medicine, Division of Computational Biomedicine, Boston University Chobanian & Avedisian School of Medicine, 75 E. Newton Street, Boston, Massachusetts 02118, USA
| | - Sarah A. Mazzilli
- Department of Medicine, Division of Computational Biomedicine, Boston University Chobanian & Avedisian School of Medicine, 75 E. Newton Street, Boston, Massachusetts 02118, USA
| | - Dennis Jones
- Department of Pathology & Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, 670 Albany Street, Boston, Massachusetts 02118, USA
| |
Collapse
|
6
|
Casini A, Gebhart J. How to investigate mild to moderate bleeding disorders and bleeding disorder of unknown cause. Int J Lab Hematol 2024; 46 Suppl 1:27-33. [PMID: 38454298 DOI: 10.1111/ijlh.14266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 02/28/2024] [Indexed: 03/09/2024]
Abstract
A bleeding tendency is one of the most common complaints observed by hematologists. It is challenging to differentiate a clinically insignificant bleeding from a bleeding phenotype that requires hemostatic evaluation and medical intervention. A thorough review of personal and familial history, objective assessment of bleeding severity using a bleeding assessment tool, and a focused physical examination are critical to correctly identifying suspected patients with mild to moderate bleeding disorders (MBDs). A basic laboratory work-up should be performed in all patients referred for a bleeding tendency. If a hemostatic abnormality is found such as evidence of von Willebrand disease, a platelet function disorder, or a coagulation factor deficiency, more extensive testing should be performed to further characterize the bleeding disorder. Conversely, if all results are normal the patient is considered to have bleeding disorder of unknown cause (BDUC). For patients with BDUC, further evaluation may include non-routine testing to look for rare bleeding disorders not detected by routine hemostasis tests, such as thrombomodulin-associated coagulopathy, tissue factor pathway inhibitor-related bleeding disorder, hyperfibrinolytic-bleeding disorders or impaired tissue factor production. In this review, we summarize the stepwise diagnostic procedure in MBDs and provide some insights into the biological features of BDUC.
Collapse
Affiliation(s)
- Alessandro Casini
- Division of Angiology and Hemostasis, University Hospitals of Geneva, Geneva, Switzerland
| | - Johanna Gebhart
- Department of Medicine I, Division of Hematology and Hemostaseology, Medical University Vienna, Austria
| |
Collapse
|
7
|
Gemmati D, D’Aversa E, Antonica B, Grisafi M, Salvatori F, Pizzicotti S, Pellegatti P, Ciccone M, Moratelli S, Serino ML, Tisato V. Gene Dosage of F5 c.3481C>T Stop-Codon (p.R1161Ter) Switches the Clinical Phenotype from Severe Thrombosis to Recurrent Haemorrhage: Novel Hypotheses for Readthrough Strategy. Genes (Basel) 2024; 15:432. [PMID: 38674367 PMCID: PMC11050146 DOI: 10.3390/genes15040432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 03/25/2024] [Accepted: 03/27/2024] [Indexed: 04/28/2024] Open
Abstract
Inherited defects in the genes of blood coagulation essentially express the severity of the clinical phenotype that is directly correlated to the number of mutated alleles of the candidate leader gene (e.g., heterozygote vs. homozygote) and of possible additional coinherited traits. The F5 gene, which codes for coagulation factor V (FV), plays a two-faced role in the coagulation cascade, exhibiting both procoagulant and anticoagulant functions. Thus, defects in this gene can be predisposed to either bleeding or thrombosis. A Sanger sequence analysis detected a premature stop-codon in exon 13 of the F5 gene (c.3481C>T; p.R1161Ter) in several members of a family characterised by low circulating FV levels and contrasting clinical phenotypes. The propositus, a 29 y.o. male affected by recurrent haemorrhages, was homozygous for the F5 stop-codon and for the F5 c.1691G>A (p.R506Q; FV-Leiden) inherited from the heterozygous parents, which is suggestive of combined cis-segregation. The homozygous condition of the stop-codon completely abolished the F5 gene expression in the propositus (FV:Ag < 1%; FV:C < 1%; assessed by ELISA and PT-based one-stage clotting assay respectively), removing, in turn, any chance for FV-Leiden to act as a prothrombotic molecule. His father (57 y.o.), characterised by severe recurrent venous thromboses, underwent a complete molecular thrombophilic screening, revealing a heterozygous F2 G20210A defect, while his mother (56 y.o.), who was negative for further common coagulation defects, reported fully asymptomatic anamnesis. To dissect these conflicting phenotypes, we performed the ProC®Global (Siemens Helthineers) coagulation test aimed at assessing the global pro- and anticoagulant balance of each family member, investigating the responses to the activated protein C (APC) by means of an APC-sensitivity ratio (APC-sr). The propositus had an unexpectedly poor response to APC (APC-sr: 1.09; n.v. > 2.25), and his father and mother had an APC-sr of 1.5 and 2.0, respectively. Although ProC®Global prevalently detects the anticoagulant side of FV, the exceptionally low APC-sr of the propositus and his discordant severe-moderate haemorrhagic phenotype could suggest a residual expression of mutated FV p.506QQ through a natural readthrough or possible alternative splicing mechanisms. The coagulation pathway may be physiologically rebalanced through natural and induced strategies, and the described insights might be able to track the design of novel treatment approaches and rebalancing molecules.
Collapse
Affiliation(s)
- Donato Gemmati
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
- University Strategic Centre for Studies on Gender Medicine, University of Ferrara, 44121 Ferrara, Italy
- Centre Haemostasis & Thrombosis, University of Ferrara, 44121 Ferrara, Italy
| | - Elisabetta D’Aversa
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Bianca Antonica
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Miriana Grisafi
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Francesca Salvatori
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
| | | | | | - Maria Ciccone
- Haematology Unit, Hospital-University of Ferrara, 44121 Ferrara, Italy
| | - Stefano Moratelli
- Centre Haemostasis & Thrombosis, University of Ferrara, 44121 Ferrara, Italy
| | - Maria Luisa Serino
- Centre Haemostasis & Thrombosis, University of Ferrara, 44121 Ferrara, Italy
| | - Veronica Tisato
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
- University Strategic Centre for Studies on Gender Medicine, University of Ferrara, 44121 Ferrara, Italy
- Laboratory of Technology for Advanced Therapies (LTTA) Centre, University of Ferrara, 44121 Ferrara, Italy
| |
Collapse
|
8
|
Mohammed BM, Basore K, Summers B, Pelc LA, Di Cera E. Structural architecture of the acidic region of the B domain of coagulation factor V. J Thromb Haemost 2024; 22:709-714. [PMID: 38007061 PMCID: PMC10922652 DOI: 10.1016/j.jtha.2023.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 11/03/2023] [Accepted: 11/06/2023] [Indexed: 11/27/2023]
Abstract
BACKGROUND Coagulation factor (F)V features an A1-A2-B-A3-C1-C2 domain organization and functions as the inactive precursor of FVa, a component of the prothrombinase complex required for rapid thrombin generation in the penultimate step of the coagulation cascade. An intramolecular interaction within the large B domain (residues 710-1545) involves the basic region (BR, residues 963-1008) and acidic region (AR, residues 1493-1537) and locks FV in its inactive state. However, structural information on this important regulatory interaction or on the separate architecture of the AR and BR remains elusive due to conformational disorder of the B domain. OBJECTIVES To reveal the structure of the BR-AR interaction or of its separate components. METHODS The structure of FV is solved by cryogenic electron microscopy. RESULTS A new 3.05 Å resolution cryogenic electron microscopy structure of FV confirms the overall organization of the A and C domains but resolves the segment 1507 to 1545 within a largely disordered B domain. The segment contains most of the AR and is organized as recently reported in FV short, a spliced variant of FV with a significantly shorter and less disordered B domain. CONCLUSION The similar architecture of the AR in FV and FV short provides structural context for physiologically important interactions of this region with the BR in FV and with the basic C-terminal end of tissue factor pathway inhibitor α in FV short.
Collapse
Affiliation(s)
- Bassem M Mohammed
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| | - Katherine Basore
- Washington University Center for Cellular Imaging, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Brock Summers
- Washington University Center for Cellular Imaging, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Leslie A Pelc
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| | - Enrico Di Cera
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St. Louis, Missouri, USA.
| |
Collapse
|
9
|
Zhang K, Ye L, Jin Y, Chen Y, Zeng M, Jia K, Yang L, Wang M. Clinical Characterization and Molecular Analysis of Fourteen Chinese Patients with Factor V Deficiency. Hamostaseologie 2023; 43:432-439. [PMID: 37714176 DOI: 10.1055/a-2146-9540] [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: 09/17/2023] Open
Abstract
INTRODUCTION Coagulation factor V (FV) functions as a vital cofactor that performs procoagulant roles in the coagulation system. We investigated 14 unrelated patients whose plasma FV levels were all below the reference range. METHODS FV activity (FV:C) and FV antigen were detected by one-stage clotting and ELISA, respectively. All 25 exons of the F5 gene in patients were amplified by the PCR, and they were sequenced directly. Haplotype analysis was performed with different polymorphisms on F5. Protein modeling was applied to analyze the potential molecular mechanisms. RESULTS Of five patients with higher FV levels (FV:C > 10%), only one had minor bleeding symptoms. In contrast, of the remaining eight patients with lower FV levels (FV:C < 10%), six showed various bleeding manifestations. A total of 10 mutations were detected from 14 patients (6 were novel mutations). Interestingly, the homozygous p.Phe190Ser was found in five pedigrees, and haplotype analysis showed that they shared almost the same haplotype, indicating the common origin rather than a hotspot mutation. In silico analysis preliminarily investigated the potential pathogenic mechanism of the mutation. Modeling analysis showed that all six missense mutations would lead to conformational alterations in the FV protein. Among them, three (p.Gly1715Ser, p.Ser1753Arg, and p.Asp68His) would decrease hydrogen bonds. CONCLUSION This is the largest genetic analysis of a single cohort of FV deficiency in Chinese. The study demonstrated that FV levels tended to be correlated with the probability of hemorrhage. The identification of a large number of unique FV-deficient pedigrees highlighted the screening for mutations in F5.
Collapse
Affiliation(s)
- Ke Zhang
- Department of Clinical Laboratory, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Longying Ye
- Department of Clinical Laboratory, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yanhui Jin
- Department of Clinical Laboratory, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yuan Chen
- Department of Clinical Laboratory, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Manlin Zeng
- Department of Clinical Laboratory, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Kaiqi Jia
- Department of Clinical Laboratory, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Lihong Yang
- Department of Clinical Laboratory, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Mingshan Wang
- Department of Clinical Laboratory, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
10
|
Gierula M, Noakes VM, Salles-Crawley II, Crawley JTB, Ahnström J. The TFPIα C-terminal tail is essential for TFPIα-FV-short-protein S complex formation and synergistic enhancement of TFPIα. J Thromb Haemost 2023; 21:3568-3580. [PMID: 37739040 DOI: 10.1016/j.jtha.2023.09.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 09/01/2023] [Accepted: 09/07/2023] [Indexed: 09/24/2023]
Abstract
BACKGROUND For maximal TFPIα functionality, 2 synergistic cofactors, protein S and FV-short, are required. Both interact with TFPIα, protein S through Kunitz 3 residues Arg199/Glu226 and FV-short with the C-terminus. How these interactions impact the synergistic enhancement remains unclear. OBJECTIVES To determine the importance of the TFPIα-protein S and TFPIα-FV-short interactions for TFPIα enhancement. METHODS TFPIα variants unable to bind protein S (K3m [R199Q/E226Q]) or FV-short (ΔCT [aa 1-249]) were generated. TFPIα-FV-short binding was studied by plate-binding and co-immunoprecipitation assays; functional TFPIα enhancement by FXa inhibition and prothrombin activation. RESULTS While WT TFPIα and TFPIα K3m bound FV-short with high affinity (Kd∼2nM), TFPIα ΔCT did not. K3m, in contrast to WT, did not incorporate protein S in a TFPIα-FV-short-protein S complex while TFPIα ΔCT bound neither FV-short nor protein S. Protein S enhanced WT TFPIα-mediated FXa inhibition, but not K3m, in the absence of FV-short. However, once FV-short was present, protein S efficiently enhanced TFPIα K3m (EC50: 4.7nM vs 2.0nM for WT). FXa inhibition by ΔCT was not enhanced by protein S alone or combined with FV-short. In FXa-catalyzed prothrombin activation assays, FV-short enhanced TFPIα K3m function in the presence of protein S (5.5 vs 10.4-fold enhancement of WT) whereas ΔCT showed reduced or lack of enhancement by FV-short and protein S, respectively. CONCLUSION Full TFPIα function requires the presence of both cofactors. While synergistic enhancement can be achieved in the absence of TFPIα-protein S interaction, only TFPIα with an intact C-terminus can be synergistically enhanced by protein S and FV-short.
Collapse
|
11
|
Ragni MV, Chan SY. Innovations in RNA therapy for hemophilia. Blood 2023; 142:1613-1621. [PMID: 37478403 PMCID: PMC10862240 DOI: 10.1182/blood.2022018661] [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] [Received: 01/10/2023] [Revised: 06/05/2023] [Accepted: 06/27/2023] [Indexed: 07/23/2023] Open
Abstract
Given the shortcomings of current factor-, nonfactor-, and adeno-associated virus gene-based therapies, the recent advent of RNA-based therapeutics for hemophilia is changing the fundamental approach to hemophilia management. From small interfering RNA therapeutics that knockdown clot regulators antithrombin, protein S, and heparin cofactor II, to CRISPR/Cas9 gene editing that may personalize treatment, improved technologies have the potential to reduce bleeds and factor use and avoid inhibitor formation. These novel agents, some in preclinical studies and others in early phase trials, have the potential to simplify treatment and improve hemostasis and quality of life. Furthermore, because these therapies arise from manipulation of the coagulation cascade and thrombin generation and its regulation, they will enhance our understanding of hemostasis and thrombosis and ultimately lead to better therapies for children and adults with inherited bleeding disorders. What does the future hold? With the development of novel preclinical technologies at the bench, there will be fewer joint bleeds, debilitating joint disease, orthopedic surgery, and improved physical and mental health, which were not previously possible. In this review, we identify current limitations of treatment and progress in the development of novel RNA therapeutics, including messenger RNA nanoparticle delivery and gene editing for the treatment of hemophilia.
Collapse
Affiliation(s)
- Margaret V. Ragni
- Division of Hematology Oncology, Department of Medicine, University of Pittsburgh, Hemophilia Center of Western Pennsylvania, Pittsburgh, PA
| | - Stephen Y. Chan
- Division of Cardiology, Department of Medicine, Vascular Medicine Institute, Pittsburgh, PA
| |
Collapse
|
12
|
Mehic D, Pabinger I, Gebhart J. Investigating patients for bleeding disorders when most of the "usual" ones have been ruled out. Res Pract Thromb Haemost 2023; 7:102242. [PMID: 38193045 PMCID: PMC10772891 DOI: 10.1016/j.rpth.2023.102242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 10/06/2023] [Accepted: 10/12/2023] [Indexed: 01/10/2024] Open
Abstract
A State of the Art lecture titled "Investigating Patients for Bleeding Disorders When Most of the Usual Ones Have Been Ruled Out" was presented at the International Society on Thrombosis and Haemostasis Congress in 2023. Mild to moderate bleeding disorders (MBDs) in patients in whom no diagnosis of an established disorder, such as platelet function defect, von Willebrand disease, or a coagulation factor deficiency, can be identified are classified as bleeding disorders of unknown cause (BDUCs). Prospective data from the Vienna Bleeding Biobank and other studies have revealed a high proportion of BDUCs of up to 70% among patients with MBD who have a similar bleeding phenotype as other MBDs. As BDUC is a diagnosis of exclusion, the accuracy of the diagnostic workup is essential. For example, repeated testing for von Willebrand disease should be considered if von Willebrand factor values are <80 IU/dL. Current evidence does not support the clinical use of global assays such as thromboelastography, platelet function analyzer, or thrombin generation potential. Rare and novel bleeding disorders due to genetic variants in fibrinolytic factors or natural anticoagulants are rare and should only be analyzed in patients with specific phenotypes and a clear family history. In BDUC, blood group O was identified as a risk factor for increased bleeding severity and bleeding risk after hemostatic challenges. Future studies should improve the phenotypical characterization and ideally identify novel risk factors in BDUC, as a multifactorial pathogenesis is suspected. Finally, we summarize relevant new data on this topic presented during the 2023 International Society on Thrombosis and Haemostasis Congress.
Collapse
Affiliation(s)
- Dino Mehic
- Clinical Division of Hematology and Hemostaseology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Ingrid Pabinger
- Clinical Division of Hematology and Hemostaseology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Johanna Gebhart
- Clinical Division of Hematology and Hemostaseology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
13
|
Shimonishi N, Ogiwara K, Yoshida J, Horie K, Nakajima Y, Furukawa S, Takeyama M, Nogami K. Impaired factor V-related anticoagulant mechanisms and deep vein thrombosis associated with A2086D and W1920R mutations. Blood Adv 2023; 7:2831-2842. [PMID: 36780344 PMCID: PMC10279549 DOI: 10.1182/bloodadvances.2022008918] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 02/08/2023] [Accepted: 02/08/2023] [Indexed: 02/14/2023] Open
Abstract
Factor V (FV) plays pivotal roles in both procoagulant and anticoagulant mechanisms. Genetic mutations, FV-W1920R (FVNara) and FV-A2086D (FVBesançon), in the C1 and C2 domains of FV light chain, respectively, seem to be associated with deep vein thrombosis. However, the detailed mechanism(s) through which these mutations are linked to thrombophilia remains to be fully explored. The aim of this study was to clarify thrombotic mechanism(s) in the presence of these FV abnormalities. Full-length wild-type (WT) and mutated FV were prepared using stable, human cell lines (HEK293T) and the piggyBac transposon system. Susceptibility of FVa-A2086D to activated protein C (APC) was reduced, resulting in significant inhibition of APC-catalyzed inactivation with limited cleavage at Arg306 and delayed cleavage at Arg506. Furthermore, APC cofactor activity of FV-A2086D in APC-catalyzed inactivation of FVIIIa through cleavage at Arg336 was impaired. Surface plasmon resonance-based assays demonstrated that FV-A2086D bound to Glu-Gly-Arg-chloromethylketone active site-blocked APC and protein S (P) with similar affinities to that of FV-WT. However, weakened interaction between FVa-A2086D and phospholipid membranes was evident through the prothrombinase assay. Moreover, addition of FVa-A2086D to plasma failed to inhibit tissue factor (TF)-induced thrombin generation and reduce prothrombin times. This inhibitory effect was independent of PC, PS, and antithrombin. The coagulant and anticoagulant characteristics of FV(a)-W1920R were similar to those of FV(a)-A2086D. FV-A2086D presented defects in the APC mechanisms associated with FVa inactivation and FV cofactor activity, similar to FV-W1920R. Moreover, both FV proteins that were mutated in the light chain impaired inhibition of TF-induced coagulation reactions. These defects were consistent with congenital thrombophilia.
Collapse
Affiliation(s)
- Naruto Shimonishi
- Department of Pediatrics, Nara Medical University, Kashihara, Japan
- The Course of Thrombosis and Hemostasis Molecular Pathology, Nara Medical University, Kashihara, Japan
| | - Kenichi Ogiwara
- Department of Pediatrics, Nara Medical University, Kashihara, Japan
| | - Junko Yoshida
- Department of Physiology II, Nara Medical University, Kashihara, Japan
| | - Kyoji Horie
- Department of Physiology II, Nara Medical University, Kashihara, Japan
| | - Yuto Nakajima
- Department of Pediatrics, Nara Medical University, Kashihara, Japan
- Advanced Medical Science of Thrombosis and Hemostasis, Nara Medical University, Kashihara, Japan
| | - Shoko Furukawa
- Department of Pediatrics, Nara Medical University, Kashihara, Japan
| | | | - Keiji Nogami
- Department of Pediatrics, Nara Medical University, Kashihara, Japan
| |
Collapse
|
14
|
Badescu MC, Butnariu LI, Costache AD, Gheorghe L, Seritean Isac PN, Chetran A, Leancă SA, Afrăsânie I, Duca ȘT, Gorduza EV, Costache II, Rezus C. Acute Myocardial Infarction in Patients with Hereditary Thrombophilia-A Focus on Factor V Leiden and Prothrombin G20210A. Life (Basel) 2023; 13:1371. [PMID: 37374153 DOI: 10.3390/life13061371] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 05/21/2023] [Accepted: 06/07/2023] [Indexed: 06/29/2023] Open
Abstract
Factor V (FV) Leiden and prothrombin G20210A are the most common hereditary thrombophilias. While their role in venous thromboembolism is well known, there are still uncertainties regarding their relationship with arterial thrombotic events, especially coronary ones. Our research, based on an in-depth analysis of the available literature, provides up-to-date information on the relationship between FV Leiden and prothrombin G20210A and acute myocardial infarction. FV Leiden and prothrombin G20210A screening should be implemented only in select cases, such as acute coronary syndrome in young individuals and/or in the absence of traditional cardiovascular risk factors and/or in the absence of significant coronary artery stenosis at angiography. Their identification should be followed by the implementation of optimal control of modifiable traditional cardiovascular risk factors to reduce the risk of recurrent events and genotyping and genetic counseling of all family members of affected cases for proper prophylaxis. An extended dual antiplatelet therapy (DAPT) may be considered, given the lower risk of bleeding under DAPT conferred by FV Leiden.
Collapse
Affiliation(s)
- Minerva Codruta Badescu
- Department of Internal Medicine, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iasi, Romania
- III Internal Medicine Clinic, "St. Spiridon" County Emergency Clinical Hospital, 700111 Iasi, Romania
| | - Lăcrămioara Ionela Butnariu
- Department of Mother and Child Medicine, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Alexandru Dan Costache
- Department of Internal Medicine, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iasi, Romania
- Cardiovascular Rehabilitation Clinic, Clinical Rehabilitation Hospital, 700661 Iasi, Romania
| | - Liliana Gheorghe
- Department of Radiology, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iasi, Romania
- Radiology Clinic "St. Spiridon" County Emergency Clinical Hospital, 700111 Iasi, Romania
| | - Petronela Nicoleta Seritean Isac
- Department of Internal Medicine, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iasi, Romania
- III Internal Medicine Clinic, "St. Spiridon" County Emergency Clinical Hospital, 700111 Iasi, Romania
| | - Adriana Chetran
- Department of Internal Medicine, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iasi, Romania
- Cardiology Clinic, "St. Spiridon" County Emergency Clinical Hospital, 700111 Iasi, Romania
| | - Sabina Andreea Leancă
- Department of Internal Medicine, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iasi, Romania
- Cardiology Clinic, "St. Spiridon" County Emergency Clinical Hospital, 700111 Iasi, Romania
| | - Irina Afrăsânie
- Department of Internal Medicine, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iasi, Romania
- Cardiology Clinic, "St. Spiridon" County Emergency Clinical Hospital, 700111 Iasi, Romania
| | - Ștefania-Teodora Duca
- Department of Internal Medicine, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iasi, Romania
- Cardiology Clinic, "St. Spiridon" County Emergency Clinical Hospital, 700111 Iasi, Romania
| | - Eusebiu Vlad Gorduza
- Department of Mother and Child Medicine, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Irina Iuliana Costache
- Department of Internal Medicine, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iasi, Romania
- Cardiology Clinic, "St. Spiridon" County Emergency Clinical Hospital, 700111 Iasi, Romania
| | - Ciprian Rezus
- Department of Internal Medicine, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iasi, Romania
- III Internal Medicine Clinic, "St. Spiridon" County Emergency Clinical Hospital, 700111 Iasi, Romania
| |
Collapse
|
15
|
Fenclova T, Matyskova M, Provaznikova D, Marecek F, Geierova V, Kovarova-Kudrnova Z, Hrachovinova I. The impact of PROS1 mutation position on thrombotic risk in protein S-deficient patients. Res Pract Thromb Haemost 2023; 7:100194. [PMID: 37384225 PMCID: PMC10293767 DOI: 10.1016/j.rpth.2023.100194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 05/16/2023] [Indexed: 06/30/2023] Open
Abstract
Background Inherited protein S deficiency is a thrombophilic risk factor associated with venous thromboembolism. However, there is not much data on the impact of mutation position on thrombotic risk. Objectives The aim of this study was to evaluate the risk of thrombosis due to mutations located in the sex hormone-binding globulin (SHBG)-like region as opposed to the rest of the protein. Methods Genetic analysis of PROS1 was performed in 76 patients with suspected inherited protein S deficiency, and the effect of missense mutations present in the SHBG region on thrombosis risk was analyzed by statistical methods. Results We found 30 unique mutations (13 of them novel), of which 17 were missense mutations, in 70 patients. Patients with missense mutations were then divided into 2 groups: the "SHBG-region" mutation group (27 patients) and the "non-SHBG" group (24 patients). The multivariable binary logistic regression analysis showed that mutation position in the SHBG region of protein S is an independent risk factor for thrombosis in deficient patients (OR, 5.17; 95% CI, 1.29-20.65; P = .02). The patients with a mutation in the SHBG-like region also developed a thrombotic event at a younger age compared to the "non-SHBG" group in the Kaplan-Meier analysis (median thrombosis-free survival of 33 vs 47 years, respectively; P = .018). Conclusion Our findings show that a missense mutation located in the SHBG-like region may contribute to higher thrombotic risk rather than a missense mutation located elsewhere in the protein. However, as our cohort was relatively small, these findings should be taken with this limitation.
Collapse
Affiliation(s)
- Tereza Fenclova
- First Faculty of Medicine, Charles University, Prague, Czech Republic
- Institute of Hematology and Blood Transfusion, National Reference Laboratory for Disorders in Hemostasis, Prague, Czech Republic
| | | | - Dana Provaznikova
- Institute of Hematology and Blood Transfusion, National Reference Laboratory for Disorders in Hemostasis, Prague, Czech Republic
| | - Frantisek Marecek
- Institute of Hematology and Blood Transfusion, National Reference Laboratory for Disorders in Hemostasis, Prague, Czech Republic
| | - Vera Geierova
- Institute of Hematology and Blood Transfusion, Centre for Thrombosis and Hemostasis, Prague, Czech Republic
| | - Zuzana Kovarova-Kudrnova
- Thrombotic Centre of Institute of Medical Biochemistry and Laboratory Diagnostics, General University Hospital, Prague, Czech Republic
| | - Ingrid Hrachovinova
- Institute of Hematology and Blood Transfusion, National Reference Laboratory for Disorders in Hemostasis, Prague, Czech Republic
| |
Collapse
|
16
|
Maroney SA, Siebert AE, Martinez ND, Rasmussen M, Peterson JA, Weiler H, Lincoln J, Mast AE. Platelet tissue factor pathway inhibitor-α dampens cardiac thrombosis and associated fibrosis in mice. J Thromb Haemost 2023; 21:639-651. [PMID: 36696221 PMCID: PMC10200073 DOI: 10.1016/j.jtha.2022.11.034] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 11/17/2022] [Accepted: 11/28/2022] [Indexed: 01/21/2023]
Abstract
BACKGROUND Tissue factor pathway inhibitor (TFPI) is the primary inhibitor of events initiating the blood coagulation pathway. Tfpi-/- mice die during embryonic development. The absence of protease-activated receptor (PAR) 4, the major thrombin receptor on mouse platelets, rescues Tfpi-/-mice to adulthood. Among the 3 TFPI isoforms in mice, TFPIα is the only isoform within platelets (pltTFPIα) and the only isoform that inhibits prothrombinase, the enzymatic complex that converts prothrombin to thrombin. OBJECTIVES To determine biological functions of pltTFPIα. METHODS Tfpi-/-/Par4-/- mice were irradiated and transplanted with bone marrow from mice lacking or containing pltTFPIα. Thus, PAR4 expression was restored in the recipient mice, which differed selectively by the presence or absence of pltTFPIα and lacked other forms of TFPI. RESULTS Recipient mice lacking pltTFPIα had reduced survival over the 200-day posttransplant period. Necropsy revealed radiation injury associated with large intraventricular platelet-rich thrombi, whereas other organs were not affected. Thrombi were associated with fibrotic presentations, including increased collagen deposition, periostin-positive activated fibroblasts, myofibroblasts, and macrophage infiltrates. Recipient mice containing pltTFPIα showed evidence of radiation injury but lacked heart pathology. CONCLUSIONS Tfpi-/-/Par4-/- mice develop severe cardiac fibrosis following irradiation and transplantation with bone marrow lacking pltTFPIα. This pathology is markedly reduced when the mice are transplanted with bone marrow containing pltTFPIα. Thus, in this model system pltTFPIα has an important physiological role in dampening pathological responses mediated by activated platelets within the heart tissue.
Collapse
Affiliation(s)
- Susan A Maroney
- Thrombosis and Hemostasis Program, Versiti Blood Research Institute, Milwaukee, WI, USA
| | - Amy E Siebert
- Thrombosis and Hemostasis Program, Versiti Blood Research Institute, Milwaukee, WI, USA
| | - Nicholas D Martinez
- Thrombosis and Hemostasis Program, Versiti Blood Research Institute, Milwaukee, WI, USA
| | - Mark Rasmussen
- Thrombosis and Hemostasis Program, Versiti Blood Research Institute, Milwaukee, WI, USA
| | - Julie A Peterson
- Thrombosis and Hemostasis Program, Versiti Blood Research Institute, Milwaukee, WI, USA
| | - Hartmut Weiler
- Thrombosis and Hemostasis Program, Versiti Blood Research Institute, Milwaukee, WI, USA; Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Joy Lincoln
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, USA; Division of Pediatric Cardiology, The Herma Heart Institute, Children's Wisconsin, Milwaukee, WI, USA
| | - Alan E Mast
- Thrombosis and Hemostasis Program, Versiti Blood Research Institute, Milwaukee, WI, USA; Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA.
| |
Collapse
|
17
|
Dahlbäck B. Natural anticoagulant discovery, the gift that keeps on giving: finding FV-Short. J Thromb Haemost 2023; 21:716-727. [PMID: 36746318 DOI: 10.1016/j.jtha.2023.01.033] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 01/24/2023] [Accepted: 01/24/2023] [Indexed: 02/05/2023]
Abstract
The complex reactions of blood coagulation are balanced by several natural anticoagulants resulting in tuned hemostasis. During several decades, the knowledge base of the natural anticoagulants has greatly increased and we have also learned about antiinflammatory and cytoprotective activities expressed by antithrombin and activated protein C (APC). Some coagulation proteins have also been found to function as anticoagulants; e.g., thrombin when bound to thrombomodulin activates protein C. Another example is factor V (FV), which in addition to being a procofactor to FVa has emerged as an anticoagulant. The discovery of APC resistance, caused by FVLeiden, as a thrombosis risk factor resulted in the identification of FV as an APC cofactor working in synergy with protein S in the regulation of FVIIIa in the Xase complex. More recently, a natural anticoagulant FV splice isoform (FV-Short) was discovered when investigating the East Texas bleeding disorder. In FV-Short, the truncated B domain exposes a high-affinity binding site for tissue factor pathway inhibitor alpha (TFPIα), and together with protein S a high-affinity trimolecular complex is generated. The FXa-inhibitory activity of TFPIα is synergistically stimulated by FV-Short and protein S. The circulating FV-Short/protein S/TFPIα complex concentration is normally low (≈0.2 nM) but provides an anticoagulant threshold. In the East Texas bleeding, the concentration of the complex, and thus the threshold, is increased 10-fold, which results in bleeding manifestations. The anticoagulant properties of FV were discovered during investigations of individual patients and follow the great tradition of bed-to-bench and bench-to-bed research in the coagulation field.
Collapse
Affiliation(s)
- Björn Dahlbäck
- Department of Translational Medicine, University Hospital, Lund University, 21428 Malmö, Sweden.
| |
Collapse
|
18
|
Abstract
INTRODUCTION Hemophilia A (HA) or B (HB) is an X-linked recessive disorder caused by a defect in the factor VIII (FVIII) or factor IX (FIX) gene which leads to the dysfunction of blood coagulation. Protein replacement therapy (PRT) uses recombinant proteins and plasma-derived products, which incurs high cost and inconvenience requiring routine intravenous infusions and life-time treatment. Understanding of detailed molecular mechanisms on FVIII gene function could provide innovative solutions to amend this disorder. In recent decades, gene therapeutics have advanced rapidly and a one-time cure solution has been proposed. AREAS COVERED This review summarizes current understanding of molecular pathways involved in blood coagulation, with emphasis on FVIII's functional role. The existing knowledge and challenges on FVIII gene expression, from transcription, translation, post-translational modification including glycosylation to protein processing and secretion, and co-factor interactions are deciphered and potential molecular interventions discussed. EXPERT OPINION This article reviews the potential treatment targets for HA and HB, including antibodies, small molecules and gene therapeutics, based on molecular mechanisms of FVIII biosynthesis, and further, assessing the pros and cons of these various treatment strategies. Understanding detailed FVIII protein synthesis and secretory pathways could provide exciting opportunities in identifying novel therapeutics to ameliorate hemophilia state.
Collapse
Affiliation(s)
- Jie Gong
- School of Medicine, University of Electronic Science and Technology of China, Sichuan, China
| | - Hao-Lin Wang
- School of Medicine, University of Electronic Science and Technology of China, Sichuan, China
| | - Lung-Ji Chang
- School of Medicine, University of Electronic Science and Technology of China, Sichuan, China.,Geno-Immune Medical Institute, Shenzhen, China
| |
Collapse
|
19
|
Dahlbäck B, Tran S. A hydrophobic patch (PLVIVG; 1481-1486) in the B-domain of factor V-short is crucial for its synergistic TFPIα-cofactor activity with protein S and for the formation of the FXa-inhibitory complex comprising FV-short, TFPIα, and protein S. J Thromb Haemost 2022; 20:1146-1157. [PMID: 35247027 PMCID: PMC9313797 DOI: 10.1111/jth.15690] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 02/09/2022] [Accepted: 02/25/2022] [Indexed: 11/28/2022]
Abstract
BACKGROUND Factor V-short (FV756-1458) is a natural splice variant functioning in synergy with protein S as tissue factor pathway inhibitor alpha (TFPIα)-cofactor in inhibition of factor Xa (FXa). An exposed acid region (AR2; 1493-1537) in the B domain binds TFPIα. The preAR2 (1458-1492) is crucial for the synergistic TFPIα-cofactor activity between FV-short and protein S and for assembly of a trimolecular FXa-inhibitory complex among FV-short, protein S, and TFPIα. OBJECTIVE To identify which part of preAR2 is required for the synergistic TFPIα-cofactor activity between FV-short and protein S. METHODS A FXa-inhibition assay was used to test the synergistic TFPIα cofactor activity between protein S and new FV-short variants FV709-1476, FV712-1478, FV712-1481, FV712-1484, FV712-1487, and FV712-1490. A microtiter-based assay analyzed binding among FV-short variants, protein S, and TFPIα. RESULTS FV709-1476, FV712-1478, and FV712-1481 were fully active as synergistic TFPIα cofactors with protein S; FV712-1484 showed intermediate activity; and FV712-1487 and FV712-1490 were inactive. TFPIα interacted with all variants in the absence of protein S but FV712-1478 and FV712-1481 bound TFPIα with highest affinity. None of the FV-short variants bound directly to protein S in the absence of TFPIα. In the presence of TFPIα, efficient cooperative binding was demonstrated between protein S, TFPIα, and FV709-1476, FV712-1478, or FV712-1481. In contrast, no cooperativity among TFPIα, protein S, and FV712-1484, FV712-1487, or FV712-1490 was seen. CONCLUSION A short hydrophobic patch in preAR2 (PLVIVG, 1481-1486) in FV-short is crucial for the synergistic TFPIα-cofactor activity between FV-short and protein S and for the assembly of a trimolecular FXa-inhibitory complex among FV-short, protein S, and TFPIα.
Collapse
Affiliation(s)
- Björn Dahlbäck
- Department of Translational MedicineUniversity HospitalLund UniversityMalmöSweden
| | - Sinh Tran
- Department of Translational MedicineUniversity HospitalLund UniversityMalmöSweden
| |
Collapse
|
20
|
Ma J, Sun W, Tang L, Yang D. Case Report and Literature Review: Behçet's Disease With a Novel TFPI Gene Mutation. Front Med (Lausanne) 2022; 9:873600. [PMID: 35514752 PMCID: PMC9063658 DOI: 10.3389/fmed.2022.873600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 03/23/2022] [Indexed: 11/13/2022] Open
Abstract
We report a case of Behçet's disease (BD) with a newly identified tissue factor pathway inhibitor (TFPI) gene mutation. The patient suffered from recurrent deep vein thrombosis and dural sinus thrombosis which could not be relieved by constant anticoagulation therapy. Slight relapsing oral lesion was the initial manifestation of BD but was neglected. Genital ulcers and ocular symptoms were manifest 8-month later than vascular involvement. The patient was diagnosed with BD at last and a novel mutation in TFPI was identified simultaneously. After administration with azathioprine and dexamethasone, the clinical symptoms were quickly gone and no relapse was found during 7-month follow-up.
Collapse
Affiliation(s)
| | | | | | - Di Yang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
21
|
Cavalcante JDS, de Almeida CAS, Clasen MA, da Silva EL, de Barros LC, Marinho AD, Rossini BC, Marino CL, Carvalho PC, Jorge RJB, Dos Santos LD. A fingerprint of plasma proteome alteration after local tissue damage induced by Bothrops leucurus snake venom in mice. J Proteomics 2022; 253:104464. [PMID: 34954398 DOI: 10.1016/j.jprot.2021.104464] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 11/30/2021] [Accepted: 12/19/2021] [Indexed: 12/21/2022]
Abstract
Bothrops spp. is responsible for about 70% of snakebites in Brazil, causing a diverse and complex pathophysiological condition. Bothrops leucurus is the main species of medical relevance found in the Atlantic coast in the Brazilian Northeast region. The pathophysiological effects involved B. leucurus snakebite as well as the organism's reaction in response to this envenoming, it has not been explored yet. Thus, edema was induced in mice paw using 1.2, 2.5, and 5.0 μg of B. leucurus venom, the percentage of edema was measured 30 min after injection and the blood plasma was collected and analyzed by shotgun proteomic strategy. We identified 80 common plasma proteins with differential abundance among the experimental groups and we can understand the early aspects of this snake envenomation, regardless of the suggestive severity of an ophidian accident. The results showed B. leucurus venom triggers a thromboinflammation scenario where family's proteins of the Serpins, Apolipoproteins, Complement factors and Component subunits, Cathepsins, Kinases, Oxidoreductases, Proteases inhibitors, Proteases, Collagens, Growth factors are related to inflammation, complement and coagulation systems, modulators platelets and neutrophils, lipid and retinoid metabolism, oxidative stress and tissue repair. Our findings set precedents for future studies in the area of early diagnosis and/or treatment of snakebites. SIGNIFICANCE: The physiopathological effects that the snake venoms can cause have been investigated through classical and reductionist tools, which allowed, so far, the identification of action mechanisms of individual components associated with specific tissue damage. The currently incomplete limitations of this knowledge must be expanded through new approaches, such as proteomics, which may represent a big leap in understanding the venom-modulated pathological process. The exploration of the complete protein set that suffer modifications by the simultaneous action of multiple toxins, provides a map of the establishment of physiopathological phenotypes, which favors the identification of multiple toxin targets, that may or may not act in synergy, as well as favoring the discovery of biomarkers and therapeutic targets for manifestations that are not neutralized by the antivenom.
Collapse
Affiliation(s)
- Joeliton Dos Santos Cavalcante
- Graduate Program in Tropical Diseases, Botucatu Medical School (FMB), São Paulo State University (UNESP), Botucatu, SP, Brazil
| | | | - Milan Avila Clasen
- Laboratory for Structural and Computational Proteomics, ICC, Oswaldo Cruz Foundation (FIOCRUZ), Curitiba, PR, Brazil
| | - Emerson Lucena da Silva
- Drug Research and Development Center, Federal University of Ceará (UFC), Fortaleza, CE, Brazil; Department of Physiology and Pharmacology, School of Medicine, Federal University of Ceará (UFC), Fortaleza, CE, Brazil
| | - Luciana Curtolo de Barros
- Center for the Study of Venoms and Venomous Animals (CEVAP), São Paulo State University (UNESP), Botucatu, SP, Brazil
| | - Aline Diogo Marinho
- Drug Research and Development Center, Federal University of Ceará (UFC), Fortaleza, CE, Brazil; Department of Physiology and Pharmacology, School of Medicine, Federal University of Ceará (UFC), Fortaleza, CE, Brazil
| | - Bruno Cesar Rossini
- Biotechnology Institute (IBTEC), São Paulo State University (UNESP), Botucatu, SP, Brazil; Department of Chemical and Biological Sciences, São Paulo State University (UNESP), Botucatu, SP, Brazil
| | - Celso Luís Marino
- Biotechnology Institute (IBTEC), São Paulo State University (UNESP), Botucatu, SP, Brazil; Department of Chemical and Biological Sciences, São Paulo State University (UNESP), Botucatu, SP, Brazil
| | - Paulo Costa Carvalho
- Laboratory for Structural and Computational Proteomics, ICC, Oswaldo Cruz Foundation (FIOCRUZ), Curitiba, PR, Brazil
| | - Roberta Jeane Bezerra Jorge
- Drug Research and Development Center, Federal University of Ceará (UFC), Fortaleza, CE, Brazil; Department of Physiology and Pharmacology, School of Medicine, Federal University of Ceará (UFC), Fortaleza, CE, Brazil
| | - Lucilene Delazari Dos Santos
- Graduate Program in Tropical Diseases, Botucatu Medical School (FMB), São Paulo State University (UNESP), Botucatu, SP, Brazil; Biotechnology Institute (IBTEC), São Paulo State University (UNESP), Botucatu, SP, Brazil.
| |
Collapse
|
22
|
Maag A, van Rein N, Schuijt TJ, Kopatz WF, Kruijswijk D, Thomassen S, Hackeng TM, Camire RM, van der Poll T, Meijers JCM, Bos MHA, van ’t Veer C. Major bleeding during oral anticoagulant therapy associated with factor V activation by factor Xa. J Thromb Haemost 2022; 20:328-338. [PMID: 34773381 PMCID: PMC9299225 DOI: 10.1111/jth.15589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 10/29/2021] [Accepted: 11/10/2021] [Indexed: 12/03/2022]
Abstract
OBJECTIVE Plasma thrombin generation (TG) provides important information on coagulation status; however, current TG output parameters do not predict major bleeding of patients on anticoagulants. We recently reported that factor V (FV) activation by factor X (FX)a contributes importantly to the initiation phase of TG. Here we investigated how this pathway varies in the normal population and whether FXa-mediated activation of FV is associated with major bleeding in patients on anticoagulant therapy. APPROACH We employed TIX-5, a specific inhibitor of FV activation by FXa, to estimate the contribution of FXa-mediated FV activation to tissue factor (TF)-initiated TG. RESULTS We show that the contribution of this pathway to plasma TG varies considerably in the normal population, as measured by the time needed to form the first traces of thrombin (TG lag time; mean prolongation by TIX-5 40%, range 0%-116%). Comparing patients on vitamin K antagonists (VKA) of the BLEED study (263 patients with and 538 patients without major bleeding), showed a marked prolongation in the median TG lag time in the presence of TIX-5 in cases (12.83 versus 11.00 minutes, P = 0.0030), while the TG lag time without TIX-5 only showed a minor although significant difference (5.83 vs. 5.67 minutes, P = 0.0198). The TIX-5 sensitivity (lag time + TIX-5/lag time + vehicle) in the upper quartile was associated with a 1.62-fold (95% confidence interval 1.04-2.52) increased risk of major bleeding compared to the lowest quartile. CONCLUSION A greater dependence on FXa-mediated activation of FV of TG is associated with increased risk of major bleeding during VKA therapy.
Collapse
Affiliation(s)
- Anja Maag
- Center for Experimental and Molecular MedicineAmsterdam Infection and Immunity Institute, Amsterdam UMCUniversity of AmsterdamAmsterdamthe Netherlands
- Division of Thrombosis and HemostasisLeiden University Medical CenterLeidenthe Netherlands
| | - Nienke van Rein
- Division of Thrombosis and HemostasisLeiden University Medical CenterLeidenthe Netherlands
- Department of Clinical EpidemiologyLeiden University Medical CenterLeidenthe Netherlands
- Department of Clinical Pharmacy and ToxicologyLeiden University Medical CenterLeidenthe Netherlands
| | - Tim J. Schuijt
- Clinical Chemistry and Hematology LaboratoryHospital Gelderse Vallei EdeEdethe Netherlands
| | - Wil F. Kopatz
- Department of Experimental Vascular MedicineAmsterdam Cardiovascular Sciences, Amsterdam UMCUniversity of AmsterdamAmsterdamthe Netherlands
| | - Danielle Kruijswijk
- Center for Experimental and Molecular MedicineAmsterdam Infection and Immunity Institute, Amsterdam UMCUniversity of AmsterdamAmsterdamthe Netherlands
| | - Stella Thomassen
- Department of BiochemistryCardiovascular Research Institute MaastrichtMaastricht UniversityMaastrichtthe Netherlands
| | - Tilman M. Hackeng
- Department of BiochemistryCardiovascular Research Institute MaastrichtMaastricht UniversityMaastrichtthe Netherlands
| | - Rodney M. Camire
- Division of Hematology and the Perelman Center for Cellular and Molecular TherapeuticsChildren’s Hospital of PhiladelphiaPhiladelphiaPennsylvaniaUSA
- Department of PediatricsPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Tom van der Poll
- Center for Experimental and Molecular MedicineAmsterdam Infection and Immunity Institute, Amsterdam UMCUniversity of AmsterdamAmsterdamthe Netherlands
| | - Joost C. M. Meijers
- Department of Experimental Vascular MedicineAmsterdam Cardiovascular Sciences, Amsterdam UMCUniversity of AmsterdamAmsterdamthe Netherlands
- Department of Molecular and Cellular HemostasisSanquin ResearchAmsterdamthe Netherlands
| | - Mettine H. A. Bos
- Division of Thrombosis and HemostasisLeiden University Medical CenterLeidenthe Netherlands
| | - Cornelis van ’t Veer
- Center for Experimental and Molecular MedicineAmsterdam Infection and Immunity Institute, Amsterdam UMCUniversity of AmsterdamAmsterdamthe Netherlands
| |
Collapse
|
23
|
Dahlbäck B, Tran S. The preAR2 region (1458-1492) in factor V-Short is crucial for the synergistic TFPIα-cofactor activity with protein S and the assembly of a trimolecular factor Xa-inhibitory complex comprising FV-Short, protein S, and TFPIα. J Thromb Haemost 2022; 20:58-68. [PMID: 34623729 DOI: 10.1111/jth.15547] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 10/05/2021] [Indexed: 01/22/2023]
Abstract
BACKGROUND Factor V-Short (FV756-1458) is a natural splice variant in which 702 residues are deleted from the B domain. It exposes an acid region (AR2; 1493-1537) that binds tissue factor pathway inhibitor alpha (TFPIα). Protein S also interacts with TFPIα and serves as TFPIα-cofactor in factor Xa (FXa) inhibition. FV-Short and protein S function as synergistic TFPIα-cofactors in inhibition of FXa. FV810-1492 is an artificial FV-Short variant that cannot synergize with protein S as TFPIα cofactor even though it contains AR2 and binds TFPIα. OBJECTIVE To elucidate the mechanisms for the synergism between FV756-1458 and protein S as TFPIα cofactors. METHODS Four FV-Short variants were created, FV756-1458 and FV712-1458 contained the preAR2 region (1458-1492), whereas FV810-1492 and FV713-1492 lacked this region. The synergistic TFPIα cofactor activity between FV-Short variants and protein S was analyzed by FXa-inhibition. A microtiter-based assay tested binding between FV-Short variants, protein S, and TFPIα. RESULTS The two preAR2-containing FV-Short variants were active as synergistic TFPIα cofactors, whereas the other two were inactive. All variants bound to TFPIα. None of the FV-Short variants bound directly to protein S. The combination of TFPIα and preAR2-containing FV-Short variants bound protein S, whereas TFPIα together with the preAR2-minus variants did not. Protein S potentiated TFPIα-binding to the preAR2-containing variants and binding between TFPIα and protein S was stimulated only by the preAR2-containing variants. CONCLUSION The preAR2 region is demonstrated to be crucial for the synergistic TFPIα-cofactor activity between FV-Short and protein S and for the assembly of a trimolecular FXa-inhibitory complex comprising FV-Short, protein S, and TFPIα.
Collapse
Affiliation(s)
- Björn Dahlbäck
- Department of Translational Medicine, Lund University, University Hospital, Malmö, Sweden
| | - Sinh Tran
- Department of Translational Medicine, Lund University, University Hospital, Malmö, Sweden
| |
Collapse
|
24
|
Ichinose A, Osaki T, Souri M. A Review of Coagulation Abnormalities of Autoimmune Acquired Factor V Deficiency with a Focus on Japan. Semin Thromb Hemost 2021; 48:206-218. [PMID: 34942668 DOI: 10.1055/s-0041-1740149] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Coagulation factor V (or FV for the purpose of medical safety) is an essential cofactor of coagulation factor X in the common pathway of coagulation; severe FV deficiency leads to a bleeding tendency. Although both congenital and acquired FV deficiencies are widely recognized, FV deficiency also presents as an autoimmune disorder. A nationwide survey on autoimmune coagulation factor deficiencies (AiCFDs) conducted in Japan by our Japanese Collaborative Research Group identified 24 new patients with autoimmune FV deficiency (AiFVD) in the past 5 years. Furthermore, our extensive literature search confirmed that 177 AiFVD cases have been reported in previous articles published from Japan. Patients with AiFVD in Japan were predominantly men, with age similar to those with other AiCFDs. AiFVD was confirmed as a relatively mild type of bleeding diathesis, associated with lower mortality rate than that for AiFVD and other AiCFDs reported in previous studies. Patients with AiFVD had variable FV inhibitor titers and both neutralizing anti-FV autoantibodies and nonneutralizing counterparts. Although spontaneous resolution occurs in some patients, timely initiation of hemostatic and immunosuppressive therapies helps arrest the bleeding and eliminate anti-FV antibodies, resulting in a high cumulative recovery rate. Immunological anti-FV antibody detection is recommended to avoid missing AiFVD cases for the presence of nonneutralizing anti-FV autoantibodies. Further investigation is necessary to clarify the long-term prognosis and optimal management of AiFVD.
Collapse
Affiliation(s)
- Akitada Ichinose
- Department of Molecular Pathobiochemistry and Pathobiology, Yamagata University School of Medicine, Yamagata, Japan.,The Japanese Collaborative Research Group (JCRG) on Autoimmune Acquired Coagulation Factor Deficiencies supported by the Japanese Ministry of Health, Labor and Welfare (MHLW), Yamagata, Japan
| | - Tsukasa Osaki
- Department of Molecular Pathobiochemistry and Pathobiology, Yamagata University School of Medicine, Yamagata, Japan.,The Japanese Collaborative Research Group (JCRG) on Autoimmune Acquired Coagulation Factor Deficiencies supported by the Japanese Ministry of Health, Labor and Welfare (MHLW), Yamagata, Japan.,Department of Public Health and Hygiene, Yamagata University Graduate School of Medical Science, Iida-Nishi, Yamagata, Japan
| | - Masayoshi Souri
- Department of Molecular Pathobiochemistry and Pathobiology, Yamagata University School of Medicine, Yamagata, Japan.,The Japanese Collaborative Research Group (JCRG) on Autoimmune Acquired Coagulation Factor Deficiencies supported by the Japanese Ministry of Health, Labor and Welfare (MHLW), Yamagata, Japan.,Department of Public Health and Hygiene, Yamagata University Graduate School of Medical Science, Iida-Nishi, Yamagata, Japan
| |
Collapse
|
25
|
Zimowski KL, Petrillo T, Ho MD, Wechsler J, Shields JE, Denning G, Jhita N, Rivera AA, Escobar MA, Kempton CL, Camire RM, Doering CB. F5-Atlanta: A novel mutation in F5 associated with enhanced East Texas splicing and FV-short production. J Thromb Haemost 2021; 19:1653-1665. [PMID: 33773040 DOI: 10.1111/jth.15314] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Revised: 03/21/2021] [Accepted: 03/22/2021] [Indexed: 01/14/2023]
Abstract
BACKGROUND Elucidating the molecular pathogenesis underlying East Texas bleeding disorder (ET) led to the discovery of alternatively spliced F5 transcripts harboring large deletions within exon 13. These alternatively spliced transcripts produce a shortened form of coagulation factor V (FV) in which a large portion of its B-domain is deleted. These FV isoforms bind tissue factor pathway inhibitor alpha (TFPIα) with high affinity, prolonging its circulatory half-life and enhancing its anticoagulant effects. While two missense pathogenic variants highlighted this alternative splicing event, similar internally deleted FV proteins are found in healthy controls. OBJECTIVE We identified a novel heterozygous 832 base pair deletion within F5 exon 13, termed F5-Atlanta (F5-ATL), in a patient with severe bleeding. Our objective is to investigate the effect of this deletion on F5 and FV expression. METHODS & RESULTS Assessment of patient plasma revealed markedly elevated levels of total and free TFPI and a FV isoform similar in size to the FV-short described in ET. Sequencing analyses of cDNA revealed the presence of a transcript alternatively spliced using the ET splice sites, thereby removing the F5-ATL deletion. This alternative splicing pattern was recapitulated by heterologous expression in mammalian cells. CONCLUSIONS These findings support a mechanistic model consisting of cis-acting regulatory sequences encoded within F5 exon 13 that control alternative splicing at the ET splice sites and thereby regulate circulating FV-short and TFPIα levels.
Collapse
Affiliation(s)
- Karen L Zimowski
- Aflac Cancer and Blood Disorders Center, Emory University/Children's Healthcare of Atlanta, Atlanta, Georgia, USA
| | - Teodolinda Petrillo
- The Children's Hospital of Philadelphia, The Raymond G. Perelman Center for Cellular and Molecular Therapeutics, Philadelphia, Pennsylvania, USA
| | - Michelle D Ho
- The Children's Hospital of Philadelphia, The Raymond G. Perelman Center for Cellular and Molecular Therapeutics, Philadelphia, Pennsylvania, USA
| | - Julie Wechsler
- Aflac Cancer and Blood Disorders Center, Emory University/Children's Healthcare of Atlanta, Atlanta, Georgia, USA
| | - Jordan E Shields
- Aflac Cancer and Blood Disorders Center, Emory University/Children's Healthcare of Atlanta, Atlanta, Georgia, USA
| | | | | | | | - Miguel A Escobar
- University of Texas Houston Health Science Center, Houston, Texas, USA
| | - Christine L Kempton
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Rodney M Camire
- The Children's Hospital of Philadelphia, The Raymond G. Perelman Center for Cellular and Molecular Therapeutics, Philadelphia, Pennsylvania, USA
- Division of Hematology, Department of Pediatrics, Perelman School of Medicine, The University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Christopher B Doering
- Aflac Cancer and Blood Disorders Center, Emory University/Children's Healthcare of Atlanta, Atlanta, Georgia, USA
| |
Collapse
|
26
|
Abstract
The serine protease thrombin, a naturally derived enzyme, plays a key role in hemostasis by converting fibrinogen to fibrin and activating coagulation factor XIII whereby the fibrin clot is stabilized. Furthermore, thrombin activates platelets through protease-activated receptors on the platelet surface. Conversely, thrombin also exerts anticoagulant effects, enhancing the protein C activity while complexed with thrombomodulin. During recent years, it has become evident that thrombin has significant effects beyond hemostasis, as it contributes also to modulation of the endothelium, promotes inflammation and angiogenesis, and plays a role in tumor progression. Yet, due to the very short half-life and almost immediate inhibition in fluid phase by antithrombin, thrombin itself remains elusive, and only indirect measurement of thrombin generation is possible. This review provides a description of structure and mechanisms of action of thrombin both in physiological and pathological processes. Furthermore, it summarizes laboratory tests that measure in vivo or ex vivo thrombin generation, and presents knowledge on the value of these biomarkers in bleeding disorders, cardiopulmonary bypass surgery, and thromboembolic risk assessment in different patient populations. Finally, this review outlines further perspectives on using thrombin generation biomarkers for research purposes and in clinical practice.
Collapse
Affiliation(s)
- Julie Brogaard Larsen
- Department of Clinical Biochemistry, Thrombosis and Hemostasis Research Unit, Aarhus University Hospital, Aarhus, Denmark
| | - Anne-Mette Hvas
- Department of Clinical Biochemistry, Thrombosis and Hemostasis Research Unit, Aarhus University Hospital, Aarhus, Denmark.,Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
27
|
Mehic D, Colling M, Pabinger I, Gebhart J. Natural anticoagulants: A missing link in mild to moderate bleeding tendencies. Haemophilia 2021; 27:701-709. [PMID: 34110661 PMCID: PMC8518679 DOI: 10.1111/hae.14356] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 05/22/2021] [Accepted: 05/28/2021] [Indexed: 12/15/2022]
Abstract
Introduction There is a growing interest in natural anticoagulants as a cause of mild to moderate bleeding disorders (MBDs), particularly in patients with bleeding of unknown cause (BUC), which is defined as having a mild to moderate bleeding phenotype without a definite diagnosis despite exhaustive and repeated laboratory investigations. Recently, abnormalities in two natural anticoagulant pathways, thrombomodulin (TM), and tissue factor pathway inhibitor (TFPI), were identified in single patients or families as the underlying cause for a bleeding tendency. Aim The objective of this review is to discuss the current understanding of the role of natural anticoagulants in MBDs using available clinical and translational data. Methods A Cochrane Library and PubMed (MEDLINE) search focusing on selected natural anticoagulants and their role in MBDs was conducted. Results Data on the influence of natural anticoagulants including protein C, protein S, antithrombin, TM, and TFPI or factors with anticoagulant properties like fibrinogen gamma prime (γ’) on MBDs are scarce. Observations from sepsis treatment and from translational research highlight their importance as regulators of the haemostatic balance, especially via the activated protein C‐related pathway, and suggest a role in some MBDs. Conclusion Similar to the distinct genetic variants of natural anticoagulants linked to thrombosis, we hypothesize that novel variants may be associated with a bleeding tendency and could be identified using next generation sequencing.
Collapse
Affiliation(s)
- Dino Mehic
- Clinical Division of Hematology and Hemostaseology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Meaghan Colling
- Clinical Division of Hematology and Hemostaseology, Department of Medicine I, Medical University of Vienna, Vienna, Austria.,Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Ingrid Pabinger
- Clinical Division of Hematology and Hemostaseology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Johanna Gebhart
- Clinical Division of Hematology and Hemostaseology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
28
|
FV/FVa revealed. Blood 2021; 137:3011-3013. [PMID: 34081120 DOI: 10.1182/blood.2021011573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
29
|
Elevated levels of tissue factor pathway inhibitor in patients with mild to moderate bleeding tendency. Blood Adv 2021; 5:391-398. [PMID: 33496735 DOI: 10.1182/bloodadvances.2020003464] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 12/05/2020] [Indexed: 12/26/2022] Open
Abstract
High levels of tissue factor pathway inhibitor (TFPI), caused by a longer TFPIα half-life after binding to a factor V splice variant and variants in the F5 gene, were recently identified in 2 families with an as-yet-unexplained bleeding tendency. This study aimed to investigate free TFPIα in a well-characterized cohort of 620 patients with mild to moderate bleeding tendencies and its association to genetic alterations in the F5 gene. TFPIα levels were higher in patients with bleeding compared with healthy controls (median [interquartile range], 8.2 [5.5-11.7] vs 7.8 [4.3-11.1]; P = .026). A higher proportion of patients had free TFPIα levels more than or equal to the 95th percentile compared with healthy controls (odds ratio [OR] [95% confidence interval (CI)], 2.82 [0.98-8.13]). This was pronounced in the subgroup of patients in whom no bleeding disorder could be identified (bleeding of unknown cause [BUC; n = 420]; OR [95% CI], 3.03 [1.02-8.98]) and in platelet function defects (PFDs) (n = 121; OR [95% CI], 3.47 [1.09-11.08]). An increase in free TFPIα was associated with a mild delay in thrombin generation (prolonged lag time and time to peak), but not with alterations in routinely used global clotting tests. We could neither identify new or known genetic variations in the F5 gene that are associated with free TFPIα levels, nor an influence of the single-nucleotide variant rs10800453 on free TFPIα levels in our patient cohort. An imbalance of natural coagulation inhibitors such as TFPIα could be an underlying cause or contributor for unexplained bleeding, which is most probably multifactorial in a majority of patients.
Collapse
|
30
|
Tong N, Li C. Changes of fibrinolytic system in thrombolytic resuscitation of pulmonary thromboembolism-induced cardiac arrest model. Int Wound J 2021; 18:874-880. [PMID: 33942504 PMCID: PMC8613376 DOI: 10.1111/iwj.13589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 03/08/2021] [Accepted: 03/09/2021] [Indexed: 12/04/2022] Open
Abstract
The objective of this study is to explore the changes in the coagulation and fibrinolysis system in an animal model with pulmonary embolism after cardiopulmonary bypass and to provide a theoretical basis for clinical practice. An animal model of cardiac arrest due to pulmonary embolism was established for venous thrombus (10‐15 mL) in the left external jugular vein of 21 pigs. Computed tomography (CT) pulmonary arteriography was performed after the recovery of the underlying state, cardiac arrest state and spontaneous circulation, and then thrombolysis and cardiopulmonary resuscitation (recombinant tissue plasminogen activator [t‐PA] 50 mg) were performed immediately. The changes of tissue factor (TF), tissue factor pathway inhibitor (TFPI), t‐PA and plasminogen activator inhibitor‐1 (PAI‐1) in the blood were detected by ELISA. The blood samples were collected immediately, 1, 2, 4 and 6 hours after the recovery of spontaneous circulation. Data from animals that were successfully resuscitated at different time points were compared using a repeated measures one‐way analysis of variance. Seventeen pigs had cardiac arrest after 10 to 15 mL of thrombus injection, and the other four had cardiac arrest after 5 to 8 mL of additional thrombus. Nine pigs survived 6 hours of cardiopulmonary resuscitation. CT pulmonary angiogram showed pulmonary artery obstruction. TF levels were increased compared with basal status, but there was no statistical difference (P > .05). TFPI levels were higher at 1, 2, 4 and 6 hours after recovery of spontaneous circulation compared with basal state (P < .05); t‐PA levels were higher at cardiac arrest, and immediately after recovery of spontaneous circulation compared with basal state. There was a statistical difference in PAI‐1 level at 1, 2, 4 and 6 hours after recovery of spontaneous circulation (P < .05). There was no statistical difference in PAI‐1 level at each stage compared with basal state (P > .05). TFPI has a certain influence on the coagulation and thrombosis regulation of the body, and the increase in fibrinolytic activity has a positive promoting effect on the thrombolysis. It provided the theoretical basis of clinical treatment of thrombotic diseases.
Collapse
Affiliation(s)
- Nan Tong
- Department of Emergency, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Chunsheng Li
- Department of Emergency, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
31
|
Zhao Y, Weyand AC, Shavit JA. Novel treatments for hemophilia through rebalancing of the coagulation cascade. Pediatr Blood Cancer 2021; 68:e28934. [PMID: 33577709 PMCID: PMC8751759 DOI: 10.1002/pbc.28934] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 12/19/2020] [Accepted: 01/12/2021] [Indexed: 11/11/2022]
Abstract
Hemophilia A and B are inherited hemorrhagic disorders that result from alterations in the coagulation cascade. Aside from spontaneous bleeding, the main complication of hemophilia is hemarthrosis. Progress over the last three decades, specifically prophylaxis using recombinant factor, has prevented hemarthrosis and lengthened patient life expectancies. However, many treatments require frequent dosing up to three times a week, and alloantibodies (inhibitors) against replacement factor continues to be an issue. These problems call for novel treatments for patients with hemophilia. Although there has been progress in extended half-life factors and mimetics of factor VIII, an alternative treatment methodology is to rebalance the activities of pro- and anticoagulant factors through inhibition of the natural anticoagulants: antithrombin, tissue factor pathway inhibitor, protein C, and protein S. This review will explore the efficacy of targeting these inhibitory pathways from preclinical development through clinical trials, and delve into concerns of thrombotic risk.
Collapse
Affiliation(s)
- Yakun Zhao
- Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, MI
| | - Angela C. Weyand
- Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, MI
| | - Jordan A. Shavit
- Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, MI
| |
Collapse
|
32
|
Obermann WMJ, Brockhaus K, Eble JA. Platelets, Constant and Cooperative Companions of Sessile and Disseminating Tumor Cells, Crucially Contribute to the Tumor Microenvironment. Front Cell Dev Biol 2021; 9:674553. [PMID: 33937274 PMCID: PMC8085416 DOI: 10.3389/fcell.2021.674553] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 03/29/2021] [Indexed: 12/12/2022] Open
Abstract
Although platelets and the coagulation factors are components of the blood system, they become part of and contribute to the tumor microenvironment (TME) not only within a solid tumor mass, but also within a hematogenous micrometastasis on its way through the blood stream to the metastatic niche. The latter basically consists of blood-borne cancer cells which are in close association with platelets. At the site of the primary tumor, the blood components reach the TME via leaky blood vessels, whose permeability is increased by tumor-secreted growth factors, by incomplete angiogenic sprouts or by vasculogenic mimicry (VM) vessels. As a consequence, platelets reach the primary tumor via several cell adhesion molecules (CAMs). Moreover, clotting factor VII from the blood associates with tissue factor (TF) that is abundantly expressed on cancer cells. This extrinsic tenase complex turns on the coagulation cascade, which encompasses the activation of thrombin and conversion of soluble fibrinogen into insoluble fibrin. The presence of platelets and their release of growth factors, as well as fibrin deposition changes the TME of a solid tumor mass substantially, thereby promoting tumor progression. Disseminating cancer cells that circulate in the blood stream also recruit platelets, primarily by direct cell-cell interactions via different receptor-counterreceptor pairs and indirectly by fibrin, which bridges the two cell types via different integrin receptors. These tumor cell-platelet aggregates are hematogenous micrometastases, in which platelets and fibrin constitute a particular TME in favor of the cancer cells. Even at the distant site of settlement, the accompanying platelets help the tumor cell to attach and to grow into metastases. Understanding the close liaison of cancer cells with platelets and coagulation factors that change the TME during tumor progression and spreading will help to curb different steps of the metastatic cascade and may help to reduce tumor-induced thrombosis.
Collapse
Affiliation(s)
| | | | - Johannes A. Eble
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Münster, Germany
| |
Collapse
|
33
|
Petrillo T, Ayombil F, Van't Veer C, Camire RM. Regulation of factor V and factor V-short by TFPIα: Relationship between B-domain proteolysis and binding. J Biol Chem 2021; 296:100234. [PMID: 33376137 PMCID: PMC7948760 DOI: 10.1074/jbc.ra120.016341] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 12/17/2020] [Accepted: 12/29/2020] [Indexed: 12/25/2022] Open
Abstract
Coagulation factor V (FV) plays an anticoagulant role but serves as a procoagulant cofactor in the prothrombinase complex once activated to FVa. At the heart of these opposing effects is the proteolytic removal of its central B-domain, including conserved functional landmarks (basic region, BR; 963–1008 and acidic region 2, AR2; 1493–1537) that enforce the inactive FV procofactor state. Tissue factor pathway inhibitor α (TFPIα) has been associated with FV as well as FV-short, a physiologically relevant isoform with a shortened B-domain missing the BR. However, it is unclear which forms of FV are physiologic ligands for TFPIα. Here, we characterize the binding and regulation of FV and FV-short by TFPIα via its positively charged C-terminus (TFPIα-BR) and examine how bond cleavage in the B-domain influences these interactions. We show that FV-short is constitutively active and functions in prothrombinase like FVa. Unlike FVa, FV-short binds with high affinity (Kd ∼1 nM) to TFPIα-BR, which blocks procoagulant function unless FV-short is cleaved at Arg1545, removing AR2. Importantly, we do not observe FV binding (μM detection limit) to TFPIα. However, cleavage at Arg709 and Arg1018 displaces the FV BR, exposing AR2 and allowing TFPIα to bind via its BR. We conclude that for full-length FV, the detachment of FV BR from AR2 is necessary and sufficient for TFPIα binding and regulation. Our findings pinpoint key forms of FV, including FV-short, that act as physiologic ligands for TFPIα and establish a mechanistic framework for assessing the functional connection between these proteins.
Collapse
Affiliation(s)
- Teodolinda Petrillo
- Division of Hematology and the Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Francis Ayombil
- Division of Hematology and the Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Cornelis Van't Veer
- Center of Experimental and Molecular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Rodney M Camire
- Division of Hematology and the Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA; Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
| |
Collapse
|
34
|
Abstract
Human factor Xa (FXa) is a serine protease of the common coagulation pathway. FXa is known to activate prothrombin to thrombin, which eventually leads to the formation of cross-linked blood clots. While this process is important in maintaining hemostasis, excessive thrombin generation results in a host of thrombotic conditions. FXa has also been linked to inflammation via protease-activated receptors. Together, coagulopathy and inflammation have been implicated in the pathogenesis of viral infections, including the current coronavirus pandemic. Direct FXa inhibitors have been shown to possess anti-inflammatory and antiviral effects, in addition to their established anticoagulant activity. This review summarizes the pharmacological activities of direct FXa inhibitors, their pharmacokinetics, potential drug–drug interactions and adverse effects, and the details of clinical trials involving direct FXa inhibitors in coronavirus disease 2019 (COVID-19) patients.
Collapse
Affiliation(s)
- Rami A Al-Horani
- Division of Basic Pharmaceutical Sciences, College of Pharmacy, Xavier University of Louisiana, 1 Drexel Drive, New Orleans, LA, 70125-1089, USA.
| |
Collapse
|
35
|
Gierula M, Ahnström J. Anticoagulant protein S-New insights on interactions and functions. J Thromb Haemost 2020; 18:2801-2811. [PMID: 32702208 DOI: 10.1111/jth.15025] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 06/25/2020] [Accepted: 07/10/2020] [Indexed: 01/21/2023]
Abstract
Protein S is a critical regulator of coagulation that functions as a cofactor for the activated protein C (APC) and tissue factor pathway inhibitor (TFPI) pathways. It also has direct anticoagulant functions, inhibiting the intrinsic tenase and prothrombinase complexes. Through these functions, protein S regulates coagulation during both its initiation and its propagation phases. The importance of protein S in hemostatic regulation is apparent from the strong association between protein S deficiencies and increased risk for venous thrombosis. This is most likely because both APC and TFPIα are inefficient anticoagulants in the absence of any cofactors. The detailed molecular mechanisms involved in protein S cofactor functions remain to be fully clarified. However, recent advances in the field have greatly improved our understanding of these functions. Evidence suggests that protein S anticoagulant properties often depend on the presence of synergistic cofactors and the formation of multicomponent complexes on negatively charged phospholipid surfaces. Their high affinity binding to negatively charged phospholipids helps bring the anticoagulant proteins to the membranes, resulting in efficient and targeted regulation of coagulation. In this review, we provide an update on protein S and how it functions as a critical hemostatic regulator.
Collapse
|
36
|
Brunet JG, Sharma T, Tasneem S, Liang M, Wilson MD, Rivard GE, Hayward CPM. Thrombin generation abnormalities in Quebec platelet disorder. Int J Lab Hematol 2020; 42:801-809. [PMID: 32761872 PMCID: PMC7754340 DOI: 10.1111/ijlh.13302] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 07/13/2020] [Accepted: 07/15/2020] [Indexed: 01/07/2023]
Abstract
INTRODUCTION Calibrated automated thrombograms (CAT) with platelet-poor (PPP) and platelet-rich plasma (PRP) have provided useful insights on bleeding disorders. We used CAT to assess thrombin generation (TG) in Quebec platelet disorder (QPD)-a bleeding disorder caused by a PLAU duplication mutation that increases platelet (but not plasma) urokinase plasminogen activator (uPA), leading to intraplatelet (but not systemic) plasmin generation that degrades α-granule proteins and causes platelet (but not plasma) factor V (FV) deficiency. METHODS Calibrated automated thrombograms was used to test QPD (n = 7) and control (n = 22) PPP and PRP, with or without added tranexamic acid (TXA). TG endpoints were evaluated for relationships to platelet FV and uPA, plasma FV and tissue factor pathway inhibitor (TFPI) levels, and bleeding scores. RESULTS Quebec platelet disorder PPP TG was normal whereas QPD PRP had reduced endogenous thrombin potential and peak thrombin concentrations (P values < .01), proportionate to the platelet FV deficiency (R2 ≥ 0.81), but unrelated to platelet uPA, plasma FV, or bleeding scores. QPD TG abnormalities were not associated with TFPI abnormalities and were not reproduced by adding uPA to control PRP. TXA increased QPD and control PRP TG more than PPP TG, but it did not fully correct QPD PRP TG abnormalities or improve TG by plasminogen-deficient plasma. CONCLUSION Quebec platelet disorder results in a platelet-specific TG defect, proportionate to the loss of platelet FV, that is improved but not fully corrected by TXA. Our study provides an interesting example of why it is important to assess both PRP and PPP TG in bleeding disorders.
Collapse
Affiliation(s)
- Justin G Brunet
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Tanmya Sharma
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Subia Tasneem
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Minggao Liang
- Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Michael D Wilson
- Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada.,Molecular Genetics, University of Toronto, Toronto, ON, Canada.,Heart & Stroke Richard Lewar Centre of Excellence in Cardiovascular Research, Toronto, ON, Canada
| | - Georges E Rivard
- Centre Hospitalier Universitaire Sainte Justine, Montreal, QC, Canada
| | - Catherine P M Hayward
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada.,Department of Medicine, McMaster University, Hamilton, ON, Canada.,Hamilton Regional Laboratory Medicine Program, Hamilton, ON, Canada
| |
Collapse
|
37
|
Ariens R, Becattini C, Bender M, Bergmeier W, Castoldi E, Devreese K, Ellis M, Gailani D, Ignjatovic V, James PD, Kerrigan S, Lambert M, Lee LH, Levi M, Maugeri N, Meijers J, Melero-Martin J, Michelson AD, Mingozzi F, Neeves K, Ni H, Olsson AK, Prohászka Z, Ranson M, Riva N, Senis Y, van Ommen CH, Vaughan DE, Weisel J. Illustrated State-of-the-Art Capsules of the ISTH 2020 Congress. Res Pract Thromb Haemost 2020; 4:680-713. [PMID: 32685876 PMCID: PMC7354406 DOI: 10.1002/rth2.12368] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 03/25/2020] [Accepted: 05/08/2020] [Indexed: 01/19/2023] Open
Abstract
The 2020 Congress of the International Society of Thrombosis and Haemostasis (ISTH) was held virtually July 12-15, 2019, due to the coronavirus disease 2019 pandemic. The congress convenes annually to discuss clinical and basic topics in hemostasis and thrombosis. Each year, the program includes State of Art (SOA) lectures given by prominent scientists. Presenters are asked to create Illustrated Capsules of their talks, which are concise illustrations with minimal explanatory text. Capsules cover major themes of the presentation, and these undergo formal peer review for inclusion in this article. Owing to the shift to a virtual congress this year, organizers reduced the program size. There were 39 SOA lectures virtually presented, and 29 capsules (9 from talks omitted from the virtual congress) were both submitted and successful in peer review, and are included in this article. Topics include the roles of the hemostatic system in inflammation, infection, immunity, and cancer, platelet function and signaling, platelet function disorders, megakaryocyte biology, hemophilia including gene therapy, phenotype tests in hemostasis, von Willebrand factor, anticoagulant factor V, computational driven discovery, endothelium, clinical and basic aspects of thrombotic microangiopathies, fibrinolysis and thrombolysis, antithrombotics in pediatrics, direct oral anticoagulant management, and thrombosis and hemostasis in pregnancy. Capsule authors invite virtual congress attendees to refer to these capsules during the live presentations and participate on Twitter in discussion. Research and Practice in Haemostasis and Thrombosis will release 2 tweets from @RPTHJournal during each presentation, using #IllustratedReview, #CoagCapsule and #ISTH2020. Readers are also welcome to utilize capsules for teaching and ongoing education.
Collapse
Affiliation(s)
- Robert Ariens
- Discovery and Translational Science Department Leeds Institute of Cardiovascular and Metabolic Medicine University of Leeds Leeds UK
| | - Cecilia Becattini
- Internal and Cardiovascular Medicine - Stroke Unit University of Perugia Perugia Italy
| | - Markus Bender
- Institute of Experimental Biomedicine - Chair I University Hospital and Rudolf Virchow Center Würzburg Germany
| | - Wolfgang Bergmeier
- Department of Biochemistry and Biophysics UNC Blood Research Center University of North Carolina at Chapel Hill Chapel Hill NC USA
| | - Elisabetta Castoldi
- Cardiovascular Research Institute Maastricht (CARIM) Maastricht University Maastricht The Netherlands
| | - Katrien Devreese
- Coagulation Laboratory Department of Laboratory Medicine Ghent University Hospital Ghent University Ghent Belgium
- Coagulation Laboratory Department of Diagnostic Sciences Ghent University Hospital Ghent University Ghent Belgium
| | - Martin Ellis
- Hematology Institute and Blood Bank Meir Medical Center and Sackler School of Medicine Tel Aviv University Tel Aviv Israel
| | - David Gailani
- Department of Pathology, Microbiology and Immunology Vanderbilt University Medical Center Nashville TN USA
| | - Vera Ignjatovic
- Haematology Research Team Murdoch Children's Research Institute Department of Paediatrics The University of Melbourne Parkville Vic. Australia
| | | | - Steven Kerrigan
- Royal College of Surgeons in Ireland School of Pharmacy and Biomolecular Sciences Irish Centre for Vascular Biology Dublin Ireland
| | - Michele Lambert
- Department of Pediatrics Perelman School of Medicine at the University of Pennsylvania Philadelphia PA USA
| | - Lai Heng Lee
- Department of Haematology Singapore General Hospital SingHealth Singapore City Singapore
| | - Marcel Levi
- University College London Hospitals London UK
| | - Norma Maugeri
- San Raffaele Scientific Institute and Vita-Salute San Raffaele University Milano Italy
| | - Joost Meijers
- Department of Experimental Vascular Medicine Amsterdam University Medical Centers University of Amsterdam Amsterdam The Netherlands
- Department of Molecular and Cellular Hemostasis Sanquin Research Amsterdam The Netherlands
| | | | - Alan D Michelson
- Boston Children's Hospital and Harvard Medical School Boston MA USA
| | | | - Keith Neeves
- Department of Chemical and Biological Engineering Colorado School of Mines Golden CO USA
| | - Heyu Ni
- Department of Laboratory Medicine and Keenan Research Centre for Biomedical Science of St. Michael's Hospital University of Toronto Toronto ON Canada
| | - Anna-Karin Olsson
- Department of Medical Biochemistry and Microbiology Uppsala University Uppsala Sweden
| | - Zoltán Prohászka
- Research Laboratory 3rd Department of Internal Medicine MTA-SE Research Group of Immunology and Hematology Hungarian Academy of Sciences and Semmelweis University Budapest Hungary
| | - Marie Ranson
- School of Chemistry and Molecular Bioscience University of Wollongong Wollongong NSW Australia
| | - Nicoletta Riva
- Department of Pathology Faculty of Medicine and Surgery University of Malta Msida Malta
| | - Yotis Senis
- Directeur de Recherche Etablissement Français du Sang Grand Est Inserm UMR-S1255 Université de Strasbourg Strasbourg France
| | - Cornelia H van Ommen
- Department of Pediatric Hematology Oncology Erasmus MC Sophia Children's Hospital Rotterdam The Netherlands
| | | | - John Weisel
- Department of Cell and Developmental Biology Perelman School of Medicine University of Pennsylvania Philadelphia PA USA
| |
Collapse
|
38
|
Gierula M, Salles‐Crawley II, Santamaria S, Teraz‐Orosz A, Crawley JTB, Lane DA, Ahnström J. The roles of factor Va and protein S in formation of the activated protein C/protein S/factor Va inactivation complex. J Thromb Haemost 2019; 17:2056-2068. [PMID: 31364267 PMCID: PMC6916587 DOI: 10.1111/jth.14594] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 07/26/2019] [Indexed: 12/22/2022]
Abstract
BACKGROUND Activated protein C (APC)-mediated inactivation of factor (F)Va is greatly enhanced by protein S. For inactivation to occur, a trimolecular complex among FVa, APC, and protein S must form on the phospholipid membrane. However, direct demonstration of complex formation has proven elusive. OBJECTIVES To elucidate the nature of the phospholipid-dependent interactions among APC, protein S, and FVa. METHODS We evaluated binding of active site blocked APC to phospholipid-coated magnetic beads in the presence and absence of protein S and/or FVa. The importance of protein S and FV residues were evaluated functionally. RESULTS Activated protein C alone bound weakly to phospholipids. Protein S mildly enhanced APC binding to phospholipid surfaces, whereas FVa did not. However, FVa together with protein S enhanced APC binding (>14-fold), demonstrating formation of an APC/protein S/FVa complex. C4b binding protein-bound protein S failed to enhance APC binding, agreeing with its reduced APC cofactor function. Protein S variants (E36A and D95A) with reduced APC cofactor function exhibited essentially normal augmentation of APC binding to phospholipids, but diminished APC/protein S/FVa complex formation, suggesting involvement in interactions dependent upon FVa. Similarly, FVaNara (W1920R), an APC-resistant FV variant, also did not efficiently incorporate into the trimolecular complex as efficiently as wild-type FVa. FVa inactivation assays suggested that the mutation impairs its affinity for phospholipid membranes and with protein S within the complex. CONCLUSIONS FVa plays a central role in the formation of its inactivation complex. Furthermore, membrane proximal interactions among FVa, APC, and protein S are essential for its cofactor function.
Collapse
Affiliation(s)
| | | | | | | | | | - David A. Lane
- Centre for HaematologyImperial College LondonLondonUK
| | | |
Collapse
|
39
|
Thibord F, Hardy L, Ibrahim-Kosta M, Saut N, Pulcrano-Nicolas AS, Goumidi L, Civelek M, Eriksson P, Deleuze JF, Le Goff W, Trégouët DA, Morange PE. A Genome Wide Association Study on plasma FV levels identified PLXDC2 as a new modifier of the coagulation process. J Thromb Haemost 2019; 17:1808-1814. [PMID: 31271701 DOI: 10.1111/jth.14562] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 07/01/2019] [Indexed: 12/23/2022]
Abstract
BACKGROUND Factor V (FV) is a circulating protein primarily synthesized in the liver, and mainly present in plasma. It is a major component of the coagulation process. OBJECTIVE To detect novel genetic loci participating to the regulation of FV plasma levels. METHODS We conducted the first Genome Wide Association Study on FV plasma levels in a sample of 510 individuals and replicated the main findings in an independent sample of 1156 individuals. RESULTS In addition to genetic variations at the F5 locus, we identified novel associations at the PLXDC2 locus, with the lead PLXDC2 rs927826 polymorphism explaining ~3.7% (P = 7.5 × 10-15 in the combined discovery and replication samples) of the variability of FV plasma levels. In silico transcriptomic analyses in various cell types confirmed that PLXDC2 expression is positively correlated to F5 expression. SiRNA experiments in human hepatocellular carcinoma cell line confirmed the role of PLXDC2 in modulating factor F5 gene expression, and revealed further influences on F2 and F10 expressions. CONCLUSION Our study identified PLXDC2 as a new molecular player of the coagulation process.
Collapse
Affiliation(s)
- Florian Thibord
- Pierre Louis Doctoral School of Public Health, Sorbonne-Université, Paris, France
- Institut National pour la Santé et la Recherche Médicale (INSERM) Unité Mixte de Recherche en Santé (UMR_S) 1219, Bordeaux Population Health Research Center, University of Bordeaux, Bordeaux, France
- INSERM UMR_S 1166, Université Pierre et Marie Curie (UPMC Univ Paris 06), Sorbonne Université, Paris, France
| | - Lise Hardy
- INSERM UMR_S 1166, Université Pierre et Marie Curie (UPMC Univ Paris 06), Sorbonne Université, Paris, France
- ICAN Institute of Cardiometabolism and Nutrition, Paris, France
| | - Manal Ibrahim-Kosta
- Laboratory of Haematology, La Timone Hospital, Marseille, France
- C2VN, Aix Marseille Univ, INSERM, INRA, Marseille, France
| | - Noémie Saut
- Laboratory of Haematology, La Timone Hospital, Marseille, France
- C2VN, Aix Marseille Univ, INSERM, INRA, Marseille, France
| | - Anne-Sophie Pulcrano-Nicolas
- Pierre Louis Doctoral School of Public Health, Sorbonne-Université, Paris, France
- INSERM UMR_S 1166, Université Pierre et Marie Curie (UPMC Univ Paris 06), Sorbonne Université, Paris, France
- ICAN Institute of Cardiometabolism and Nutrition, Paris, France
| | - Louisa Goumidi
- C2VN, Aix Marseille Univ, INSERM, INRA, Marseille, France
| | - Mete Civelek
- Department of Biomedical Engineering, Center for Public Health Genomics, University of Virginia, Charlottesville, Virginia
| | - Per Eriksson
- Department of Medicine, Cardiovascular Medicine Unit, BioClinicum, Karolinska Institutet, Stockholm, Sweden
- Karolinska University Hospital, Solna, Sweden
| | - Jean-François Deleuze
- Centre National de Recherche en Génomique Humaine, Direction de la Recherche Fondamentale, CEA, Evry, France
- CEPH, Fondation Jean Dausset, Paris, France
| | - Wilfried Le Goff
- INSERM UMR_S 1166, Université Pierre et Marie Curie (UPMC Univ Paris 06), Sorbonne Université, Paris, France
- ICAN Institute of Cardiometabolism and Nutrition, Paris, France
| | - David-Alexandre Trégouët
- Institut National pour la Santé et la Recherche Médicale (INSERM) Unité Mixte de Recherche en Santé (UMR_S) 1219, Bordeaux Population Health Research Center, University of Bordeaux, Bordeaux, France
- INSERM UMR_S 1166, Université Pierre et Marie Curie (UPMC Univ Paris 06), Sorbonne Université, Paris, France
| | - Pierre-Emmanuel Morange
- Laboratory of Haematology, La Timone Hospital, Marseille, France
- C2VN, Aix Marseille Univ, INSERM, INRA, Marseille, France
- CRB Assistance Publique - Hôpitaux de Marseille, HemoVasc (CRB AP-HM HemoVasc), Marseille, France
| |
Collapse
|
40
|
Sachetto A, Mackman N. Modulation of the mammalian coagulation system by venoms and other proteins from snakes, arthropods, nematodes and insects. Thromb Res 2019; 178:145-154. [DOI: 10.1016/j.thromres.2019.04.019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 04/04/2019] [Accepted: 04/15/2019] [Indexed: 12/22/2022]
|
41
|
Dahlbäck B, Guo LJ, Zöller B, Tran S. New functional test for the TFPIα cofactor activity of Protein S working in synergy with FV-Short. J Thromb Haemost 2019; 17:585-595. [PMID: 30740865 DOI: 10.1111/jth.14405] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 01/30/2019] [Indexed: 08/31/2023]
Abstract
Essentials Protein S and FV-Short are synergistic cofactors to Tissue Factor Pathway Inhibitor α (TFPIα). An assay for the TFPIα synergistic cofactor activity of protein S with FV-Short was developed. The assay was specific for the synergistic TFPIα-cofactor activity of free protein S. Protein S deficient individuals with known mutations were correctly distinguished from controls. SUMMARY: Background Protein S is an anticoagulant cofactor to both activated protein C and tissue factor pathway inhibitor (TFPIα). The TFPIα-cofactor activity of protein S is stimulated by a short isoform of factor V (FV-Short), the two proteins functioning in synergy. Objective Using the synergistic TFPIα-cofactor activity between protein S and FV-Short to develop a functional test for plasma protein S. Patients/Methods TFPIα-mediated inhibition of FXa in the presence of FV-Short, protein S and negatively charged phospholipid vesicles was monitored in time by synthetic substrate S2765. TFPIα, FXa and FV-Short were purified proteins, whereas diluted plasma from protein S deficient patients or controls were used as source for protein S. Results The assay was specific for free protein S demonstrating good correlation to free protein S plasma levels (r = 0.92) with a Y-axis intercept of -5%. Correlation to concentrations of total protein S (free and C4BPβ+-bound) was lower (r = 0.88) and the Y-axis intercept was +46%, which is consistent with the specificity for free protein S. The test distinguished protein S-deficient individuals from 6 families with known ProS1 mutations from family members having no mutation. Protein S levels of warfarin-treated protein S deficient cases were lower than protein S in cases treated with warfarin for other causes. Conclusions We describe a new assay measuring the TFPIα-cofactor activity of plasma protein S. The test identifies type I/III protein S deficiencies and will be a useful tool to detect type II protein S deficiency having defective TFPIα-cofactor activity.
Collapse
Affiliation(s)
- Björn Dahlbäck
- Department of Translational Medicine, Lund University, Skåne University Hospital, Malmö, Sweden
| | - Li Jun Guo
- Department of Translational Medicine, Lund University, Skåne University Hospital, Malmö, Sweden
| | - Bengt Zöller
- Department of Clinical Sciences, Lund University, Skåne University Hospital, Malmö, Sweden
| | - Sinh Tran
- Department of Translational Medicine, Lund University, Skåne University Hospital, Malmö, Sweden
| |
Collapse
|
42
|
Verleih M, Borchel A, Rebl A, Brenmoehl J, Kühn C, Goldammer T. A molecular survey of programmed cell death in rainbow trout: Structural and functional specifications of apoptotic key molecules. Comp Biochem Physiol B Biochem Mol Biol 2019; 230:57-69. [DOI: 10.1016/j.cbpb.2019.01.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 01/11/2019] [Accepted: 01/18/2019] [Indexed: 12/24/2022]
|
43
|
O'Donnell JS, O'Sullivan JM, Preston RJS. Advances in understanding the molecular mechanisms that maintain normal haemostasis. Br J Haematol 2019; 186:24-36. [DOI: 10.1111/bjh.15872] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- James S. O'Donnell
- Haemostasis Research Group Department of Molecular and Cellular Therapeutics Irish Centre for Vascular Biology Royal College of Surgeons in Ireland Dublin Ireland
| | - Jamie M. O'Sullivan
- Haemostasis Research Group Department of Molecular and Cellular Therapeutics Irish Centre for Vascular Biology Royal College of Surgeons in Ireland Dublin Ireland
| | - Roger J. S. Preston
- Haemostasis Research Group Department of Molecular and Cellular Therapeutics Irish Centre for Vascular Biology Royal College of Surgeons in Ireland Dublin Ireland
| |
Collapse
|
44
|
Post-transcriptional, post-translational and pharmacological regulation of tissue factor pathway inhibitor. Blood Coagul Fibrinolysis 2018; 29:668-682. [PMID: 30439766 DOI: 10.1097/mbc.0000000000000775] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
: Tissue factor (TF) pathway inhibitor (TFPI) is an endogenous natural anticoagulant that readily inhibits the extrinsic coagulation initiation complex (TF-FVIIa-Xa) and prothrombinase (FXa, FVa and calcium ions). Alternatively, spliced TFPI isoforms (α, β and δ) are expressed by vascular and extravascular cells and regulate thrombosis and haemostasis, as well as cell signalling functions of TF complexes via protease-activated receptors (PARs). Proteolysis of TFPI plays an important role in regulating physiological roles of the TF pathway in host defense and possibly haemostasis. Elimination of TFPI inhibition has therefore been proposed as an approach to improve haemostasis in haemophilia patients. In this review, we focus on posttranscription and translational modification of TFPI and its function in thrombosis and how pharmacological inhibitors and endogenous proteases interfere with TFPI and alter haemostasis.
Collapse
|
45
|
A high titer of acquired factor V inhibitor in a hemodialysis patient who developed arterial thrombosis. Int J Hematol 2018; 109:214-220. [PMID: 30446942 DOI: 10.1007/s12185-018-2561-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2018] [Revised: 11/12/2018] [Accepted: 11/13/2018] [Indexed: 10/27/2022]
Abstract
An 87-year-old man with diabetes mellitus was admitted to control recurrent bleeding from hemodialysis puncture sites. He was a smoker and had been diagnosed with arteriosclerosis obliterans. His PT and APTT were markedly prolonged, and all coagulation factors were markedly decreased (factor V [FV] activity < 1%) or below the measurement threshold, with the exception of fibrinogen and factor XIII. Neither PT nor APTT were corrected upon mixing with normal plasma. A high titer of FV inhibitor was found at 415 BU/mL, and anti-FV autoantibody was detected by both immunoblot assay and ELISA. Prednisolone administration and plasma exchange partially improved prolonged PT and APTT and decreased the FV inhibitor level. Five months later, he manifested symptoms of severe ischemia in both legs. Angiography revealed diffuse stenosis downstream of both common iliac arteries. Endovascular therapy was repeated four times, the prednisolone dose was reduced, and low-dose antiplatelet therapy was initiated. After the final successful endovascular therapy, arterial thrombosis was detected using ultrasound and angiography. Aspiration thrombectomy and thrombolytic therapy failed to achieve recanalization, and necrosis of the legs worsened. Despite the severe coagulation abnormalities, vascular interventions should have been performed with regular-dose antiplatelet therapy, as the patient exhibited multiple risk factors for atherothrombosis.
Collapse
|
46
|
Romero-Gavilán F, Araújo-Gomes N, García-Arnáez I, Martínez-Ramos C, Elortza F, Azkargorta M, Iloro I, Gurruchaga M, Suay J, Goñi I. The effect of strontium incorporation into sol-gel biomaterials on their protein adsorption and cell interactions. Colloids Surf B Biointerfaces 2018; 174:9-16. [PMID: 30408675 DOI: 10.1016/j.colsurfb.2018.10.075] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 10/11/2018] [Accepted: 10/25/2018] [Indexed: 01/06/2023]
Abstract
It is known strontium can both inhibit the osteoclast formation and stimulate the osteoblast maturation, so biomaterials containing this element can favour bone structure stabilisation. The addition of Sr to biomaterials could affect their interactions with proteins and cells. Here, a silica-hybrid sol-gel network doped with different amounts of SrCl2 and applied as coatings on titanium discs was examined. in vitro analysis was performed to determine the potential effect of Sr in the coatings, showing enhanced gene expression of osteogenic markers (alkaline phosphatase and transforming growth factor-β) in MC3T3-E1 incubated with Sr-doped biomaterials. The examination of inflammatory markers (tumour necrosis factor-α and interleukin 10) in RAW 264.7 macrophages revealed an anti-inflammatory potential of these materials. Proteins adsorbed onto the coatings incubated with human serum (3 h at 37 °C) were also analysed; mass spectrometry was used to characterise the proteins adhering to materials with different Sr content. Adding Sr to the coatings increased their affinity to APOE and VTNC proteins (associated with anti-inflammatory and osteogenic functions). Moreover, the proteins involved in coagulation processes, such as prothrombin, were more abundant on the coatings containing Sr than on the base sol-gel surfaces. Correlations between gene expression and proteomic results were also examined.
Collapse
Affiliation(s)
- F Romero-Gavilán
- Departamento de Ingeniería de Sistemas Industriales y Diseño, Universitat Jaume I, Av. Vicent-Sos Baynat s/n. Castellón 12071. Spain
| | - N Araújo-Gomes
- Departamento de Ingeniería de Sistemas Industriales y Diseño, Universitat Jaume I, Av. Vicent-Sos Baynat s/n. Castellón 12071. Spain; Department of Medicine. Universitat Jaume I, Av. Vicent-Sos Baynat s/n. Castellón 12071. Spain.
| | - I García-Arnáez
- Facultad de Ciencias Químicas. Universidad del País Vasco (UPV/EHU). P. M. de Lardizábal, 3. San Sebastián 20018. Spain
| | - C Martínez-Ramos
- Department of Medicine. Universitat Jaume I, Av. Vicent-Sos Baynat s/n. Castellón 12071. Spain
| | - F Elortza
- Proteomics Platform, CIC bioGUNE, CIBERehd, ProteoRed-ISCIII, Bizkaia Science and Technology Park, 48160 Derio, Spain
| | - M Azkargorta
- Proteomics Platform, CIC bioGUNE, CIBERehd, ProteoRed-ISCIII, Bizkaia Science and Technology Park, 48160 Derio, Spain
| | - I Iloro
- Proteomics Platform, CIC bioGUNE, CIBERehd, ProteoRed-ISCIII, Bizkaia Science and Technology Park, 48160 Derio, Spain
| | - M Gurruchaga
- Facultad de Ciencias Químicas. Universidad del País Vasco (UPV/EHU). P. M. de Lardizábal, 3. San Sebastián 20018. Spain
| | - J Suay
- Departamento de Ingeniería de Sistemas Industriales y Diseño, Universitat Jaume I, Av. Vicent-Sos Baynat s/n. Castellón 12071. Spain
| | - I Goñi
- Facultad de Ciencias Químicas. Universidad del País Vasco (UPV/EHU). P. M. de Lardizábal, 3. San Sebastián 20018. Spain
| |
Collapse
|
47
|
De Maertelaere E, Castoldi E, Van Haute I, Deeren D, Devreese KM. The interaction of factor V and tissue factor pathway inhibitor in a myeloma patient with acquired factor V deficiency. Haemophilia 2018; 24:e160-e163. [PMID: 29665188 DOI: 10.1111/hae.13492] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/11/2018] [Indexed: 11/30/2022]
Affiliation(s)
| | - E Castoldi
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, the Netherlands
| | - I Van Haute
- Clinical Laboratory, AZ Delta Hospital, Roeselare, Belgium
| | - D Deeren
- Clinical Haematology, AZ Delta Hospital, Roeselare, Belgium
| | - K M Devreese
- Coagulation Laboratory, Department of Clinical Chemistry, Microbiology and Immunology, Ghent University Hospital, Ghent, Belgium
| |
Collapse
|
48
|
|
49
|
Zöller B, Melander O, Svensson PJ, Engström G. Factor V Leiden paradox in a middle-aged Swedish population: A prospective study. Vasc Med 2018; 23:52-59. [DOI: 10.1177/1358863x17745591] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Few prospective studies have examined the factor V paradox: factor V Leiden (FVL) is a stronger risk factor for deep venous thrombosis (DVT) than for pulmonary embolism (PE). The present study, to the best of our knowledge, is the first population-based study aimed to examine the relationship between FVL and incidence of venous thromboembolism (VTE), DVT and PE in a prospective cohort study of middle-aged Swedish individuals. FVL was determined in 4890 subjects (aged 46–68 years, 57% women) from the general population without previous VTE or cancer, who participated in the Malmö Diet and Cancer study between 1991 and 1994. Incident cases of VTE were identified from the Swedish patient register during a mean follow-up of 15.6 years. Of 4890 subjects with determination of FVL (10.2% carriers), 220 had VTE during follow-up (113 DVT, 78 PE, 29 both). Incidence of VTE was significantly higher in subjects with heterozygous and homozygous FVL: adjusted hazard ratios (HR) were 1.8 (95% CI 1.3–2.6, p=0.001) and 6.5 (2.1–21, p=0.001), respectively. The population attributable fraction was 8.7% for FVL. Adjusted HRs for DVT were 2.2 (1.4–3.3, p<0.001) for heterozygotes and 3.3 (0.5–24, p=0.233) for homozygotes. Adjusted HRs for PE were 1.2 (0.65–2.2, p=0.582) for heterozygotes and 8.7 (2.1–36, p=0.003) for homozygotes. The FVL paradox was confirmed for heterozygotes for FVL. However, homozygotes for FVL had a high risk for PE, suggesting that the FVL paradox is related to the carriership of one wild type and one mutated factor V allele.
Collapse
Affiliation(s)
- Bengt Zöller
- Center for Primary Health Care Research, Lund University/Region Skåne, Malmö, Sweden
| | - Olle Melander
- Department of Clinical Sciences, Lund University, Malmö, Sweden
| | | | - Gunnar Engström
- Department of Clinical Sciences, Lund University, Malmö, Sweden
| |
Collapse
|