1
|
Nagata T, Huang Y. Elucidating the role of intrinsic adenosine A1 receptors in acute alcoholism using human-induced pluripotent stem cell-derived hepatocytes. Biosci Rep 2024; 44:BSR20231682. [PMID: 38419509 PMCID: PMC10958140 DOI: 10.1042/bsr20231682] [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: 10/08/2023] [Revised: 02/08/2024] [Accepted: 02/28/2024] [Indexed: 03/02/2024] Open
Abstract
Acute alcoholic hepatitis (AAH) from binge drinking is a serious disease. It is associated with a high mortality rate, especially among young adults. Apoptosis is known to be a primary cause of liver damage, and it can be induced by either intrinsic signaling pathways or by reactive oxygen species (ROS). Adenosine A1 receptors (ADORA1) are known to be involved in ethanol metabolism; however, underlying mechanism is not well understood. For investigating how the intrinsic ADORA1 function in ethanol metabolism in normal human hepatocytes without interference by extrinsic molecules, primary hepatocytes pose a challenge, due to unavoidable contamination by other kinds of cells in the liver. Also, they are difficult to culture stably. As a novel alternative, hepatocytes derived from human-induced pluripotent stem cells were employed because they display similar function to primary hepatocytes and they can be stably cultured. The dynamics and integrity of signal transduction mechanisms were investigated by following chronological changes in gene expression. This shed light on how and when the ADORA1 function and on causal relationships between the pathways and clinical symptoms. The findings of the present study shows that ADORA1 are most activated soon after exposure to ethanol, and transfection of small interfering RNA targeting ADORA1-messenger-RNA (ADORA1-siRNA) into the hepatocytes significantly suppresses production of actin protein and ROS. It suggests that ADORA1 in the liver contribute to apoptosis in acute alcoholism through both intrinsic pathway and ROS activity. Also, actin that is abundant in the cells could be an appropriate biomarker evaluating hepatic function status.
Collapse
Affiliation(s)
| | - Yuning George Huang
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, U.S.A
| |
Collapse
|
2
|
Parahuleva MS, Worsch M, Euler G, Choukeir M, Mardini A, Parviz B, Kanse SM, Portig I, Khayrutdinov E, Schieffer B, Markus B. Factor VII Activating Protease Expression in Human Platelets and Accumulation in Symptomatic Carotid Plaque. J Am Heart Assoc 2020; 9:e016445. [PMID: 32856552 PMCID: PMC7660758 DOI: 10.1161/jaha.120.016445] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Accepted: 06/24/2020] [Indexed: 01/22/2023]
Abstract
Background Factor VII activating protease (FSAP) is of interest as a marker for vascular inflammation and plaque destabilization. The aim of this study was to analyze the expression profile of FSAP in endarterectomy specimens that were taken from patients with asymptomatic and symptomatic carotid atherosclerotic plaques and to compare them with circulating FSAP levels. Methods and Results Plasma FSAP concentration, activity, and mRNA expression were measured in endarterectomy specimens and in monocytes and platelets. Plaque and plasma FSAP levels were higher in symptomatic patients (n=10) than in asymptomatic patients (n=14). Stronger FSAP immunostaining was observed in advanced symptomatic lesions, in intraplaque hemorrhage-related structures, and in lipid-rich areas within the necrotic core. FSAP was also colocalized with monocytes and macrophages (CD11b/CD68-positive cells) and platelets (CD41-positive cells) of the plaques. Moreover, human platelets expressed FSAP in vitro, at both the mRNA and protein levels. Expression is stimulated by thrombin receptor-activating peptide and ADP and reduced by acetylsalicylic acid. Conclusions Plasma FSAP levels were significantly increased in patients with symptomatic carotid stenosis and thus may be involved in plaque development This plaque-associated FSAP may be produced by platelets or macrophages or may be taken up from the circulation. To establish FSAP's utility as a circulating or plaque biomarker in patients with symptomatic carotid atherosclerotic plaques, further studies are needed.
Collapse
Affiliation(s)
| | - Michael Worsch
- Department of Internal Medicine I/Cardiology and AngiologyUKGMGiessenGermany
| | - Gerhild Euler
- Department of Internal Medicine I/Cardiology and AngiologyUKGMGiessenGermany
| | - Maryana Choukeir
- Department of Cardiology, Angiology and Internal Intensive CareUKGMMarburgGermany
| | - Amar Mardini
- Department of Cardiology, Angiology and Internal Intensive CareUKGMMarburgGermany
| | - Behnoush Parviz
- Department of Internal Medicine I/Cardiology and AngiologyUKGMGiessenGermany
| | - Sandip M. Kanse
- Institute for Basic Medical SciencesUniversity of OsloNorway
| | - Irene Portig
- Department of Cardiology, Angiology and Internal Intensive CareUKGMMarburgGermany
| | - Evgeny Khayrutdinov
- Department of Cardiology, Angiology and Internal Intensive CareUKGMMarburgGermany
| | - Bernhard Schieffer
- Department of Cardiology, Angiology and Internal Intensive CareUKGMMarburgGermany
| | - Birgit Markus
- Department of Cardiology, Angiology and Internal Intensive CareUKGMMarburgGermany
| |
Collapse
|
3
|
Kara E, Nielsen NV, Eggertsdottir B, Thiede B, Kanse SM, Løset GÅ. Design and Characterization of a New pVII Combinatorial Phage Display Peptide Library for Protease Substrate Mining Using Factor VII Activating Protease (FSAP) as Model. Chembiochem 2020; 21:1875-1884. [PMID: 32180321 PMCID: PMC7383712 DOI: 10.1002/cbic.201900705] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 02/07/2020] [Indexed: 12/18/2022]
Abstract
We describe a novel, easy and efficient combinatorial phage display peptide substrate-mining method to map the substrate specificity of proteases. The peptide library is displayed on the pVII capsid of the M13 bacteriophage, which renders pIII necessary for infectivity and efficient retrieval, in an unmodified state. As capture module, the 3XFLAG was chosen due to its very high binding efficiency to anti-FLAG mAbs and its independency of any post-translational modification. This library was tested with Factor-VII activating protease (WT-FSAP) and its single-nucleotide polymorphism variant Marburg-I (MI)-FSAP. The WT-FSAP results confirmed the previously reported Arg/Lys centered FSAP cleavage site consensus as dominant, as well as reinforcing MI-FSAP as a loss-of-function mutant. Surprisingly, rare substrate clones devoid of basic amino acids were also identified. Indeed one of these peptides was cleaved as free peptide, thus suggesting a broader range of WT-FSAP substrates than previously anticipated.
Collapse
Affiliation(s)
- Emrah Kara
- Institute of Basal Medical Sciences Oslo University HospitalUniversity of OsloOsloNorway
| | - Nis Valentin Nielsen
- Institute of Basal Medical Sciences Oslo University HospitalUniversity of OsloOsloNorway
| | | | - Bernd Thiede
- Department of BiosciencesUniversity of Oslo0316OsloNorway
| | - Sandip M. Kanse
- Institute of Basal Medical Sciences Oslo University HospitalUniversity of OsloOsloNorway
| | - Geir Åge Løset
- Department of BiosciencesUniversity of Oslo0316OsloNorway
- Nextera ASOsloNorway
| |
Collapse
|
4
|
Olsson M, Stanne TM, Pedersen A, Lorentzen E, Kara E, MartinezâPalacian A, RÃnnow Sand NP, Jacobsen AF, Sandset PM, Sidelmann JJ, EngstrÃm G, Melander O, Kanse SM, Jern C. Genome-wide analysis of genetic determinants of circulating factor VII-activating protease (FSAP) activity. J Thromb Haemost 2018; 16:2024-2034. [PMID: 30070759 PMCID: PMC6485504 DOI: 10.1111/jth.14258] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Indexed: 01/17/2023]
Abstract
Essentials Knowledge of genetic regulators of plasma factor VII activating protease (FSAP) levels is limited. We performed a genome-wide analysis of variants influencing FSAP activity in Scandinavian cohorts. We replicated an association for Marburg-1 and identified an association for a HABP2 stop variant. We identified a novel locus near ADCY2 as a potential additional regulator of FSAP activity. SUMMARY Background Factor VII-activating protease (FSAP) has roles in both coagulation and fibrinolysis. Recent data indicate its involvement in several other processes, such as vascular remodeling and inflammation. Plasma FSAP activity is highly variable among healthy individuals and, apart from the low-frequency missense variant Marburg-I (rs7080536) in the FSAP-encoding gene HABP2, determinants of this variation are unclear. Objectives To identify novel genetic variants within and outside of the HABP2 locus that influence circulating FSAP activity. Patients/Methods We performed an exploratory genome-wide association study (GWAS) on plasma FSAP activity amongst 3230 Swedish subjects. Directly genotyped rare variants were also analyzed with gene-based tests. Using GWAS, we confirmed the strong association between the Marburg-I variant and FSAP activity. HABP2 was also significant in the gene-based analysis, and remained significant after exclusion of Marburg-I carriers. This was attributable to a rare HABP2 stop variant (rs41292628). Carriers of this stop variant showed a similar reduction in FSAP activity as Marburg-I carriers, and this finding was replicated. A secondary genome-wide significant locus was identified at a 5p15 locus (rs35510613), and this finding requires future replication. This common variant is located upstream of ADCY2, which encodes a protein catalyzing the formation of cAMP. Results and Conclusions This study verified the Marburg-I variant to be a strong regulator of FSAP activity, and identified an HABP2 stop variant with a similar impact on FSAP activity. A novel locus near ADCY2 was identified as a potential additional regulator of FSAP activity.
Collapse
Affiliation(s)
- M. Olsson
- Department of Pathology and GeneticsInstitute of BiomedicineThe Sahlgrenska Academy at University of GothenburgGothenburgSweden
| | - T. M. Stanne
- Department of Pathology and GeneticsInstitute of BiomedicineThe Sahlgrenska Academy at University of GothenburgGothenburgSweden
| | - A. Pedersen
- Department of Pathology and GeneticsInstitute of BiomedicineThe Sahlgrenska Academy at University of GothenburgGothenburgSweden
| | - E. Lorentzen
- Bioinformatics Core FacilityUniversity of GothenburgGothenburgSweden
| | - E. Kara
- Institute of Basic Medical SciencesFaculty of MedicineUniversity of OsloOsloNorway
| | - A. MartinezâPalacian
- Institute of Basic Medical SciencesFaculty of MedicineUniversity of OsloOsloNorway
| | - N. P. RÃnnow Sand
- Department of CardiologyHospital of South West DenmarkEsbjerg and Department of Regional Health ResearchFaculty of Health ScienceUniversity of Southern DenmarkEsbjergDenmark
| | - A. F. Jacobsen
- Department of ObstetricsOslo University Hospital and University of OsloOsloNorway
| | - P. M. Sandset
- Department of HematologyOslo University Hospital and University of OsloOsloNorway
| | - J. J. Sidelmann
- Unit for Thrombosis ResearchDepartment of Regional Health ResearchFaculty of Health ScienceUniversity of Southern DenmarkEsbjergDenmark
| | - G. EngstrÃm
- Department of Clinical Sciences, MalmÃLund UniversityLundSweden
| | - O. Melander
- Department of Clinical Sciences, MalmÃLund UniversityLundSweden
| | - S. M. Kanse
- Institute of Basic Medical SciencesFaculty of MedicineUniversity of OsloOsloNorway
| | - C. Jern
- Department of Pathology and GeneticsInstitute of BiomedicineThe Sahlgrenska Academy at University of GothenburgGothenburgSweden
| |
Collapse
|
5
|
Sperling C, Maitz MF, Grasso S, Werner C, Kanse SM. A Positively Charged Surface Triggers Coagulation Activation Through Factor VII Activating Protease (FSAP). ACS APPLIED MATERIALS & INTERFACES 2017; 9:40107-40116. [PMID: 29091393 DOI: 10.1021/acsami.7b14281] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Contact between biomedical materials and blood often initiates undesirable pro-coagulant and pro-inflammatory processes. On negatively charged materials, blood coagulation is known to be triggered through autoactivation of Factor XII, while activation on cationic surfaces follows a distinct and so far enigmatic mechanism. Because Factor VII activating protease (FSAP) is known to be activated on positively and on negatively charged macromolecules in plasma, we have investigated its interaction with charged biomaterials and its consequences for coagulation. Several activation processes in blood and plasma were characterized after contact with material surfaces with varied charge. FSAP was found to be exclusively activated by the positively charged surfaces polyethylenimine (PEI) and poly-l-lysine (PLL), not by the negatively charged glass or self-assembled monolayer with carboxyl group termination (SAM-COOH), as well as uncharged (Teflon AF) surfaces. Whole blood incubation on PEI showed that this activation was concomitant with coagulation as determined by thrombin and fibrin formation, which was high for glass (F1+2, 138 nM) and PEI (F1+2, 44 nM) but low for Teflon AF (F1+2, 3.3 nM) and SAM COOH (F1+2, 5.8 nM). Contact phase inhibitor diminished coagulation to background levels for all surfaces except PEI (F1+2: ^PEI 43 to 25 nM; glass, 58 to 1.5 nM) indicating that coagulation activation is not dependent on FXII activation on the PEI surface. A decisive role of endogenous FSAP for coagulation however was confirmed with the use of FSAP inhibitory antibodies which showed no influence on Teflon AF, glass and SAM COOH but diminished F1+2 on PEI to less than 50%. We propose that FSAP activation could be a novel mechanism of surface-driven coagulation. An inhibition of this protease might improve hemocompatibility of cationic surfaces and therefore facilitate the application of polycationic surfaces in blood.
Collapse
Affiliation(s)
- Claudia Sperling
- Institute Biofunctional Polymer Materials, Max Bergmann Center of Biomaterials, Leibniz-Institut für Polymerforschung Dresden e.V. Hohe Strasse 6, 01069 Dresden, Germany
| | - Manfred F Maitz
- Institute Biofunctional Polymer Materials, Max Bergmann Center of Biomaterials, Leibniz-Institut für Polymerforschung Dresden e.V. Hohe Strasse 6, 01069 Dresden, Germany
| | - Simona Grasso
- Oslo University Hospital and University of Oslo , 0372 Oslo, Norway
| | - Carsten Werner
- Institute Biofunctional Polymer Materials, Max Bergmann Center of Biomaterials, Leibniz-Institut für Polymerforschung Dresden e.V. Hohe Strasse 6, 01069 Dresden, Germany
| | - Sandip M Kanse
- Oslo University Hospital and University of Oslo , 0372 Oslo, Norway
| |
Collapse
|
6
|
Whole-exome sequencing in evaluation of patients with venous thromboembolism. Blood Adv 2017; 1:1224-1237. [PMID: 29296762 DOI: 10.1182/bloodadvances.2017005249] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Accepted: 05/26/2017] [Indexed: 01/05/2023] Open
Abstract
Genetics play a significant role in venous thromboembolism (VTE), yet current clinical laboratory-based testing identifies a known heritable thrombophilia (factor V Leiden, prothrombin gene mutation G20210A, or a deficiency of protein C, protein S, or antithrombin) in only a minority of VTE patients. We hypothesized that a substantial number of VTE patients could have lesser-known thrombophilia mutations. To test this hypothesis, we performed whole-exome sequencing (WES) in 64 patients with VTE, focusing our analysis on a novel 55-gene extended thrombophilia panel that we compiled. Our extended thrombophilia panel identified a probable disease-causing genetic variant or variant of unknown significance in 39 of 64 study patients (60.9%), compared with 6 of 237 control patients without VTE (2.5%) (P < .0001). Clinical laboratory-based thrombophilia testing identified a heritable thrombophilia in only 14 of 54 study patients (25.9%). The majority of WES variants were either associated with thrombosis based on prior reports in the literature or predicted to affect protein structure based on protein modeling performed as part of this study. Variants were found in major thrombophilia genes, various SERPIN genes, and highly conserved areas of other genes with established or potential roles in coagulation or fibrinolysis. Ten patients (15.6%) had >1 variant. Sanger sequencing performed in family members of 4 study patients with and without VTE showed generally concordant results with thrombotic history. WES and extended thrombophilia testing are promising tools for improving our understanding of VTE pathogenesis and identifying inherited thrombophilias.
Collapse
|
7
|
Stavenuiter F, Ebberink EHTM, Mertens K, Meijer AB. Role of glycine 221 in catalytic activity of hyaluronan-binding protein 2. J Biol Chem 2017; 292:6381-6388. [PMID: 28246168 DOI: 10.1074/jbc.m116.757849] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Revised: 02/25/2017] [Indexed: 11/06/2022] Open
Abstract
HABP2 (hyaluronan-binding protein 2) is a Ca2+-dependent serine protease with putative roles in blood coagulation and fibrinolysis. A G221E substitution, known as the Marburg I polymorphism, reportedly affects HABP2 function and has been associated with increased risk for cardiovascular disease. However, the importance of Gly-221 for HABP2 activity is unclear. Here, we used G221E, G221A, and G221S mutants to assess the role of Gly-221 in HABP2 catalysis. The G221E variant failed to activate the single-chain urokinase-type plasminogen activator, and the G221A and G221S variants displayed moderately reduced single-chain urokinase-type plasminogen activator activation. Activity toward the peptide substrate S-2288 was markedly decreased in all HABP2 variants, with G221E being the most defective and G221A being the least defective. In the absence of Ca2+, S-2288 cleavage by wild-type HABP2 was Na+-dependent, with Km decreasing from 3.0 to 0.6 mm upon titration from 0 to 0.3 m Na+ In the presence of 5 mm Ca2+, Km was further reduced to 0.05 mm, but without an appreciable contribution of Na+ At physiological concentrations of Na+ and Ca2+, the three HABP2 variants, and particularly G221E, displayed a major Km increase for S-2288. Chemical footprinting revealed that Ile-16 is significantly less protected from chemical modification in G221E than in wild-type HABP2, suggesting impaired insertion of the N terminus into the G221E protease domain, with a concomitant impact on catalytic activity. Homology modeling suggested that the Glu-221 side chain could sterically hinder insertion of the N terminus into the HABP2 protease domain, helping to explain the detrimental effects of Glu-221 substitution on HABP2 activity.
Collapse
Affiliation(s)
- Fabian Stavenuiter
- From the Department of Plasma Proteins, Sanquin Research, 1066 CX Amsterdam, The Netherlands and
| | - Eduard H T M Ebberink
- From the Department of Plasma Proteins, Sanquin Research, 1066 CX Amsterdam, The Netherlands and
| | - Koen Mertens
- From the Department of Plasma Proteins, Sanquin Research, 1066 CX Amsterdam, The Netherlands and.,the Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands
| | - Alexander B Meijer
- From the Department of Plasma Proteins, Sanquin Research, 1066 CX Amsterdam, The Netherlands and .,the Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands
| |
Collapse
|
8
|
Gara SK, Jia L, Merino MJ, Agarwal SK, Zhang L, Cam M, Patel D, Kebebew E. Germline HABP2 Mutation Causing Familial Nonmedullary Thyroid Cancer. N Engl J Med 2015; 373. [PMID: 26222560 PMCID: PMC4562406 DOI: 10.1056/nejmoa1502449] [Citation(s) in RCA: 110] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Familial nonmedullary thyroid cancer accounts for 3 to 9% of all cases of thyroid cancer, but the susceptibility genes are not known. Here, we report a germline variant of HABP2 in seven affected members of a kindred with familial nonmedullary thyroid cancer and in 4.7% of 423 patients with thyroid cancer. This variant was associated with increased HABP2 protein expression in tumor samples from affected family members, as compared with normal adjacent thyroid tissue and samples from sporadic cancers. Functional studies showed that HABP2 has a tumor-suppressive effect, whereas the G534E variant results in loss of function.
Collapse
Affiliation(s)
- Sudheer Kumar Gara
- From the Endocrine Oncology Branch (S.K.G., L.Z., D.P., E.K.), Bioinformatics Core (L.J.) and Laboratory of Pathology (M.J.M.), Center for Cancer Research, and the Office of Science and Technology Resources (M.C.), National Cancer Institute, and the Metabolic Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases (S.K.A.) - both in Bethesda, MD
| | | | | | | | | | | | | | | |
Collapse
|
9
|
Parahuleva MS, Kanse S, Hölschermann H, Zheleva K, Zandt D, Worsch M, Parviz B, Güttler N, Tillmanns H, Böning A, Erdogan A. Association of circulating factor seven activating protease (FSAP) and of oral Omega-3 fatty acids supplements with clinical outcome in patients with atrial fibrillation: the OMEGA-AF study. J Thromb Thrombolysis 2015; 37:317-25. [PMID: 23575879 DOI: 10.1007/s11239-013-0921-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Factor VII Activating Protease (FSAP) activates factor VII (FVII) as well as pro-urokinase (uPA). Our goal was to evaluate the relation between plasma levels of FSAP and clinical instability in atrial fibrillation (AF) and possible effects of oral omega-3 fatty acids (FA) supplements. 101 patients with persistent AF were analyzed in the OMEGA-AF Study. Plasma FSAP levels were measured at baseline and after 12 weeks of treatment with omega-3 FA. The median FSAP antigen concentration, in contrast to FSAP activity, was higher in patients with persistent AF. The maintenance of SR after successful cardioversion (CV) did not lead to a normalization of FSAP concentration. Supplementation with omega-3 FA but not placebo significantly reduced elevated FSAP concentration. Furthermore, elevated FSAP levels did not indicate a significantly increased risk of recurrence of AF after electrical CV or cardiovascular clinical events during 1 year of follow-up. Plasma FSAP concentration was increased in patients with AF and may be involved in the pathogenesis of this condition. The possible effects of omega-3 FA on clinical AF potential could be linked with modulation of circulating FSAP levels.
Collapse
Affiliation(s)
- Mariana S Parahuleva
- Internal Medicine I/Cardiology and Angiology, University Hospital of Giessen and Marburg, Klinikstr. 36, 35392, Giessen, Germany,
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Inhibition of Plasma Hyaluronan-Binding Protein Autoactivation by Laccaic Acid. Biosci Biotechnol Biochem 2014; 74:2320-2. [DOI: 10.1271/bbb.100373] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
11
|
Bacterial plasminogen receptors utilize host plasminogen system for effective invasion and dissemination. J Biomed Biotechnol 2012; 2012:482096. [PMID: 23118509 PMCID: PMC3477821 DOI: 10.1155/2012/482096] [Citation(s) in RCA: 102] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2012] [Revised: 07/24/2012] [Accepted: 08/13/2012] [Indexed: 01/06/2023] Open
Abstract
In order for invasive pathogens to migrate beyond the site of infection, host physiological barriers such as the extracellular matrix, the basement membrane, and encapsulating fibrin network must be degraded. To circumvent these impediments, proteolytic enzymes facilitate the dissemination of the microorganism. Recruitment of host proteases to the bacterial surface represents a particularly effective mechanism for enhancing invasiveness. Plasmin is a broad spectrum serine protease that degrades fibrin, extracellular matrices, and connective tissue. A large number of pathogens express plasminogen receptors which immobilize plasmin(ogen) on the bacterial surface. Surface-bound plasminogen is then activated by plasminogen activators to plasmin through limited proteolysis thus triggering the development of a proteolytic surface on the bacteria and eventually assisting the spread of bacteria. The host hemostatic system plays an important role in systemic infection. The interplay between hemostatic processes such as coagulation and fibrinolysis and the inflammatory response constitutes essential components of host defense and bacterial invasion. The goal of this paper is to highlight mechanisms whereby pathogenic bacteria, by engaging surface receptors, utilize and exploit the host plasminogen and fibrinolytic system for the successful dissemination within the host.
Collapse
|
12
|
Parahuleva MS, Langanke E, Hölschermann H, Parviz B, Abdallah Y, Stracke S, Tillmanns H, Kanse SM. Nicotine modulation of factor VII activating protease (FSAP) expression in human monocytes. J Atheroscler Thromb 2012; 19:962-9. [PMID: 22878700 DOI: 10.5551/jat.9589] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
AIM Factor VII activating protease (FSAP) is a plasma serine protease involved in hemostasis and remodeling processes. Increased levels of circulating FSAP during pregnancy and in women using oral contraceptives (OCs) indicate that the hormonal status critically influences FSAP expression. In this respect, the aim of this study was to quantify nicotine modulation of FSAP expression in human monocytes/macrophages isolated from healthy female smokers and non-smokers, and from women who use OCs and smoke. METHODS FSAP concentration and activity were measured in plasma samples obtained from healthy non-pregnant, pre-menopausal, non-smoking women who did not use OCs (n=69), non-pregnant, pre-menopausal women who currently smoke and use OCs (n=43), and women who are only smokers (n=40) or currently use OCs (n=48). Expressions of FSAP mRNA and protein in monocytes isolated from healthy non-pregnant female or healthy male donors were analyzed. RESULTS Strongest circulating FSAP concentration and activity occurred in women with combined smoking and use of OCs compared to the control group. Enhanced FSAP levels were also observed in smoking women when compared to non-smokers. Ex vivo experiments demonstrated enhanced FSAP expression in monocytes isolated from women using OCs and currently smoking. Nicotine enhanced FSAP mRNA and protein levels in monocytes. CONCLUSIONS Monocytes from healthy female smokers show a constitutively enhanced FSAP expression and this effect could be replicated in vitro by stimulating monocytes with nicotine. The upregulation of FSAP due to nicotine and OC usage may be linked to a higher incidence of arteriothromboembolic diseases related to their usage.
Collapse
|
13
|
Kemmochi S, Shimamoto K, Shiraki A, Onda N, Hasumi K, Suzuki K, Mitsumori K, Shibutani M. Promoting effects of carminic acid-enriched cochineal extracts on capsular invasive thyroid carcinomas through targeting activation of angiogenesis in rats. J Toxicol Sci 2012; 37:475-82. [PMID: 22687987 DOI: 10.2131/jts.37.475] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Cochineal extracts (CE) is a coccid-derived natural food colorant containing carminic acid (CA) as an active ingredient that potentiates inhibition of tissue proteolysis mediated by activation of plasma hyaluronan-binding protein (PHBP). In our previous study, dietary administered CE (CA: 28.5% in CE) has shown to promote the macroscopic development of capsular invasive carcinomas (CICs) associated with up-regulation of angiogenesis-related genes in an intracapsular invasion model of experimental thyroid cancers using rats. However, the promoting effect of CE could not be confirmed histopathologically. The purpose of the present study was to confirm the promoting effect of CE through direct injections to animals on the development of CICs using this cancer invasion model. One week after initiation with N-bis(hydroxypropyl)nitrosamine, male F344/NSlc rats were administered CA-enriched CE (CA: 52.6% in CE) by intraperitoneal injections every other day (10 mg/kg body weight) during the promotion with 0.15% sulfadimethoxine in the drinking water for 8 weeks. The multiplicities of macroscopical CICs and the mean area of early capsular invasive foci estimated by Tenascin (TN)-C-immunoreactivity in the thyroid significantly increased with CE-treatment, while the number of TN-C-positive foci did not change with CE. Transcript level of Phbp and downstream genes unchanged; however, transcript level of angiogenesis-related genes, i.e, Vegfb and its transcription factor gene, Hif1a, those being downstream of phosphatase and tensin homolog (PTEN)/Akt signaling, up-regulated in the thyroid tissue with CE-administration. These results suggest that CE potentiates promotion activity by facilitating angiogenesis through activation of PTEN/Akt signaling without accompanying modification of PHBP-related proteolysis.
Collapse
Affiliation(s)
- Sayaka Kemmochi
- Laboratory of Veterinary Pathology,Tokyo University of Agriculture and Technology, Tokyo, Japan
| | | | | | | | | | | | | | | |
Collapse
|
14
|
Stephan F, Dienava-Verdoold I, Bulder I, Wouters D, Mast AE, Te Velthuis H, Aarden LA, Zeerleder S. Tissue factor pathway inhibitor is an inhibitor of factor VII-activating protease. J Thromb Haemost 2012; 10:1165-71. [PMID: 22449009 PMCID: PMC3574557 DOI: 10.1111/j.1538-7836.2012.04712.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
BACKGROUND Factor VII-activating protease (FSAP) is a serine protease that circulates in plasma in its inactive single-chain form and can be activated upon contact with dead cells. When activated by apoptotic cells, FSAP leads to the release of nucleosomes. The serpins C1-inhibitor and α(2) -antiplasmin are reported to be the major inhibitors of FSAP. However, regulation of FSAP activity by Kunitz-type inhibitors is not well studied. OBJECTIVES To compare the inhibition of FSAP activity and FSAP-induced nucleosome release from apoptotic cells by tissue factor pathway inhibitor (TFPI) with that of C1-inhibitor and α(2) -antiplasmin. METHODS Apoptotic cells were incubated with plasma or FSAP in presence of the inhibitor, and nucleosome release was analyzed with flow cytometry. Monoclonal antibodies against TFPI and altered forms of TFPI were used to investigate which domains of TFPI contribute to FSAP inhibition. RESULTS AND CONCLUSIONS We show that TFPI abrogates FSAP activity and nucleosome release from apoptotic cells. TFPI is a much more efficient inhibitor than C1-inhibitor or α(2) -antiplasmin. The active site of K2 is required for inhibition of FSAP. A direct binding interaction between FSAP and the C-terminal domain of TFPI is also required for efficient inhibition. Inhibition of FSAP-induced nucleosome release by recombinant TFPI might, in part, explain the anti-inflammatory effects of recombinant TFPI infusion observed in animal and human sepsis.
Collapse
Affiliation(s)
- F Stephan
- Departments of Immunopathology, University of Amsterdam, Amsterdam, the Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
15
|
Kanse SM, Declerck PJ, Ruf W, Broze G, Etscheid M. Factor VII-activating protease promotes the proteolysis and inhibition of tissue factor pathway inhibitor. Arterioscler Thromb Vasc Biol 2011; 32:427-33. [PMID: 22116096 DOI: 10.1161/atvbaha.111.238394] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Factor VII-activating protease (FSAP) activates both factor VII and pro-urokinase and inhibits platelet-derived growth factor-BB, thus regulating hemostasis- and remodeling-associated processes in the vasculature. A genetic variant of FSAP (Marburg I polymorphism) results in low enzymatic activity and is associated with an enhanced risk of carotid stenosis and stroke. We postulate that there are additional substrates for FSAP that will help to explain its role in vascular biology and have searched for such a substrate. METHODS AND RESULTS Using screening procedures to determine the influence of FSAP on various hemostasis-related processes on endothelial cells, we discovered that FSAP inhibited tissue factor pathway inhibitor (TFPI), a major anticoagulant secreted by these cells. Proteolytic degradation of TFPI by FSAP could also be demonstrated by Western blotting, and the exact cleavage sites were determined by N-terminal sequencing. The Marburg I variant of FSAP had a diminished ability to inhibit TFPI. A monoclonal antibody to FSAP that specifically inhibited FSAP binding to TFPI reversed the inhibitory effect of FSAP on TFPI. CONCLUSIONS The identification of TFPI as a sensitive substrate for FSAP increases our understanding of its role in regulating hemostasis and proliferative remodeling events in the vasculature.
Collapse
Affiliation(s)
- Sandip M Kanse
- Institute for Biochemistry, Justus-Liebig-University, Friedrichstrasse 24, D-35392 Giessen, Germany.
| | | | | | | | | |
Collapse
|
16
|
Elucidation of crucial structures for a catechol-based inhibitor of plasma hyaluronan-binding protein (factor VII activating protease) autoactivation. Biosci Biotechnol Biochem 2011; 75:2070-2. [PMID: 21979097 DOI: 10.1271/bbb.110515] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Plasma hyaluronan-binding protein (PHBP) is a serine protease the activation of which is implicated in inflammation. Previous investigations have suggested the presence of catechol-binding sites in its proenzyme form, pro-PHBP. Here we found that compounds with plural catechol groups conjugated with strong electron-withdrawing groups, such as tyrphostin AG 537 (IC(50)=18 nM), were potent inhibitors of pro-PHBP activation.
Collapse
|
17
|
Trompet S, Pons D, Kanse SM, de Craen AJM, Ikram MA, Verschuren JJW, Zwinderman AH, Doevendans PAFM, Tio RA, de Winter RJ, Slagboom PE, Westendorp RGJ, Jukema JW. Factor VII Activating Protease Polymorphism (G534E) Is Associated with Increased Risk for Stroke and Mortality. Stroke Res Treat 2011; 2011:424759. [PMID: 21789270 PMCID: PMC3140705 DOI: 10.4061/2011/424759] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2010] [Revised: 04/28/2011] [Accepted: 05/06/2011] [Indexed: 11/20/2022] Open
Abstract
Introduction. The FSAP-Marburg I polymorphism (1704G > A), which reduces FSAP activity, is associated with late complications of carotid stenosis in humans. Therefore, this study examines the influence of the Marburg I polymorphism and the closely linked Marburg II polymorphism (1280G > C) on various cardiovascular outcomes in two large independent study populations. Methods. The two Marburg polymorphisms in the HABP2 gene encoding FSAP were genotyped in a large population of elderly patients at risk for vascular disease (the PROSPER-study, n = 5804) and in a study population treated with a percutaneous coronary intervention (the GENDER-study, n = 3104). Results. In the PROSPER study, the Marburg I polymorphism was associated with an increased risk of clinical stroke (HR: 1.60, 95% CI: 1.13-2.28) and all-cause mortality (HR: 1.33, 95% CI: 1.04-1.71). In the GENDER study carriers of this variant seemed at lower risk of developing restenosis (HR: 0.59, 95% CI: 0.34-1.01). The Marburg II polymorphism showed similar but weaker results. Conclusion. The increase in stroke risk in Marburg I carriers could be due to differential effects on smooth muscle cells and on matrix metalloproteinases, thereby influencing plaque stability. The possible protective effect on restenosis could be the result of reduced activation of zymogens, which are involved in hemostasis and matrix remodeling.
Collapse
Affiliation(s)
- Stella Trompet
- Department of Cardiology, Leiden University Medical Center, C5-P, P.O. Box 9600, 2300 RC Leiden, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Stephan S, Schwarz H, Haude-Barten A, Sidelmann JJ, Fischer B, Althaus H, Hahn M, Kappel A, Ehm M, Vitzthum F. Direct chromogenic substrate immuno-capture activity assay for testing of factor VII-activating protease. Clin Chem Lab Med 2011; 49:1199-204. [PMID: 21663569 DOI: 10.1515/cclm.2011.199] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND The Marburg I (MRI) single nucleotide polymorphism (SNP) of the factor VII-activating protease (FSAP) gene has been associated with thrombophilia and atherosclerotic disease. PCR is used to detect the SNP. Also, the specific FSAP activity to cleave single-chain urokinase-type plasminogen activator (scu-PA) serves as a surrogate for PCR testing. Development of further assays is indicated in order to increase testing opportunities for future studies. METHODS A direct chromogenic substrate immuno-capture activity assay for FSAP (FSAP dcs activity assay) was established. Performance characteristics of the FSAP dcs activity assay were compared to the FSAP scu-PA activity assay. RESULTS The FSAP dcs activity assay detects FSAP activity from 25% to 150% of the norm. Total CVs ranged from 6% to 10% for FSAP wild type samples and 9%-18% for MRI samples. Correlation between the FSAP dcs and scu-PA activity assays was low (R=0.7). The FSAP dcs activity determined the presence of the MRI FSAP alloenzyme with a diagnostic sensitivity and specificity of 100% [95% confidence interval (CI): 89.6%-100%] and 96.2% (95% CI: 93.2%-97.4%), respectively, whereas the specific FSAP dcs activity increased specificity to 99.0% (95% CI: 97.2%-99.6%). CONCLUSIONS The specific FSAP dcs activity represents a reliable method for the detection of the FSAP MRI alloenzyme. Due to the limited correlation between the FSAP dcs and scu-PA activity assays, these different measurands may exhibit different utility in research and clinical applications. Thus, the FSAP dcs activity assay can represent a valuable complement or alternative for FSAP testing in future studies.
Collapse
Affiliation(s)
- Sina Stephan
- Siemens Healthcare Diagnostics Products GmbH, Marburg, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Stephan F, Hazelzet JA, Bulder I, Boermeester MA, van Till JO, van der Poll T, Wuillemin WA, Aarden LA, Zeerleder S. Activation of factor VII-activating protease in human inflammation: a sensor for cell death. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2011; 15:R110. [PMID: 21466697 PMCID: PMC3219388 DOI: 10.1186/cc10131] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/17/2011] [Accepted: 04/05/2011] [Indexed: 01/26/2023]
Abstract
INTRODUCTION Cell death is a central event in the pathogenesis of sepsis and is reflected by circulating nucleosomes. Circulating nucleosomes were suggested to play an important role in inflammation and were demonstrated to correlate with severity and outcome in sepsis patients. We recently showed that plasma can release nucleosomes from late apoptotic cells. Factor VII-activating protease (FSAP) was identified to be the plasma serine protease responsible for nucleosome release. The aim of this study was to investigate FSAP activation in patients suffering from various inflammatory diseases of increasing severity. METHODS We developed ELISAs to measure FSAP-C1-inhibitor and FSAP-α2-antiplasmin complexes in plasma. FSAP-inhibitor complexes were measured in the plasma of 20 adult patients undergoing transhiatal esophagectomy, 32 adult patients suffering from severe sepsis and 8 from septic shock and 38 children suffering from meningococcal sepsis. RESULTS We demonstrate plasma FSAP to be activated upon contact with apoptotic and necrotic cells by an assay detecting complexes between FSAP and its target serpins α2-antiplasmin and C1-inhibitor, respectively. By means of that assay we demonstrate FSAP activation in post-surgery patients, patients suffering from severe sepsis, septic shock and meningococcal sepsis. Levels of FSAP-inhibitor complexes correlate with nucleosome levels and correlate with severity and mortality in these patients. CONCLUSIONS These results suggest FSAP activation to be a sensor for cell death in the circulation and that FSAP activation in sepsis might be involved in nucleosome release, thereby contributing to lethality.
Collapse
Affiliation(s)
- Femke Stephan
- Department of Immunopathology, Sanquin Research at CLB and Landsteiner Laboratory of AMC, Plesmanlaan 125, 1066 CX Amsterdam, The Netherlands
| | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Yamamoto E, Nishimura N, Okada K, Sekido C, Yamamichi S, Hasumi K. Inhibitors of Autoactivation of Plasma Hyaluronan-Binding Protein (Factor VII Activating Protease). Biol Pharm Bull 2011; 34:462-70. [DOI: 10.1248/bpb.34.462] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Eisaku Yamamoto
- Department of Applied Biological Science, Tokyo Noko University
| | - Naoko Nishimura
- Department of Applied Biological Science, Tokyo Noko University
| | - Ken Okada
- Department of Applied Biological Science, Tokyo Noko University
| | - Chikako Sekido
- Department of Applied Biological Science, Tokyo Noko University
| | | | - Keiji Hasumi
- Department of Applied Biological Science, Tokyo Noko University
| |
Collapse
|
21
|
Baeten KM, Richard MC, Kanse SM, Mutch NJ, Degen JL, Booth NA. Activation of single-chain urokinase-type plasminogen activator by platelet-associated plasminogen: a mechanism for stimulation of fibrinolysis by platelets. J Thromb Haemost 2010; 8:1313-22. [PMID: 20180903 DOI: 10.1111/j.1538-7836.2010.03813.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
BACKGROUND AND OBJECTIVE Platelets are essential for hemostasis, and they cause resistance to fibrinolysis by tissue-type plasminogen activator. In contrast, platelets enhance fibrinolysis mediated by single-chain urokinase-type plasminogen activator (scu-PA). This study investigated the mechanism behind this profibrinolytic role of platelets. METHODS AND RESULTS Platelets enhanced scu-PA activity, but not urokinase-type plasminogen activator (u-PA) activity, in plasma clot lysis and chromogenic assays. We established, using the non-cleavable scu-PA mutant (Lys158-->Glu) and protease inhibitors, that platelets increased activation to u-PA by a serine protease. Activation of scu-PA was platelet-dependent, even in plasma. It occurred in platelet-rich but not in platelet-poor plasma, as assessed by sodium dodecylsulfate polyacrylamide gel electrophoresis and zymography after addition of plasminogen activator inhibitor-1. Candidate proteases that are known to activate scu-PA and are present in platelet preparations were investigated. Factor VII activating protease was detected in platelet preparations by western blotting, but its inhibition by antibodies did not inhibit activation of scu-PA by platelets. Plasmin and plasma kallikrein both mimicked the platelet effect, but were distinguished by their responses to a range of inhibitors. Analysis of platelet-associated protease activity and the time course of scu-PA activation pointed towards plasminogen, and the data were consistent with a mechanism of reciprocal activation. The essential role of plasminogen was revealed using platelets from plasminogen-deficient mice, which could not activate scu-PA. Local plasminogen on platelet membranes was markedly more effective than solution-phase plasminogen in activation of scu-PA. CONCLUSIONS Platelets enhance fibrinolysis by scu-PA through reciprocal activation of scu-PA and platelet-associated plasminogen, a system that is potentially important in the lysis of platelet-rich thrombi.
Collapse
Affiliation(s)
- K M Baeten
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
| | | | | | | | | | | |
Collapse
|
22
|
Nishimura N, Takai M, Yamamoto E, Hasumi K. Purpurin as a specific inhibitor of spermidine-induced autoactivation of the protease plasma hyaluronan-binding protein. Biol Pharm Bull 2010; 33:1430-3. [PMID: 20686243 DOI: 10.1248/bpb.33.1430] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2023]
Abstract
Plasma hyaluronan-binding protein (PHBP), a serine protease that can activate coagulation factor VII and prourokinase, circulates as a single-chain form (pro-PHBP), and is autoproteolytically converted to an active two-chain form with the aid of an effector such as spermidine and heparin. In this study, we screened natural sources for inhibitors of spermidine-induced pro-PHBP autoactivation. As an active agent, we purified bikaverin from a culture of a fungus. Bikaverin inhibited spermidine-induced autoactivation with an IC(50) of 0.45 microM, while it also inhibited the active form of PHBP (IC(50)=0.8 microM). Additional screening of related compounds led to the identification of purpurin, a plant anthraquinone, as a specific inhibitor: IC(50)=6.6 microM for spermidine-induced autoactivation; no inhibition of heparin-induced autoactivation and active PHBP. Alizarin and emodin, which structurally differed from purpurin in the position or the number of the hydroxyl groups, were less active and nonspecific. Thus, the position and/or the number of the hydroxyl group affect both the potency and selectivity of the anthraquinone inhibitors.
Collapse
Affiliation(s)
- Naoko Nishimura
- Department of Applied Biological Science, Tokyo University of Agriculture and Technology, Fucho, Tokyo, Japan
| | | | | | | |
Collapse
|
23
|
Stephan S, Schwarz H, Borchert A, Bussfeld D, Quak E, Simshaeuser-Knaub B, Teigelkamp S, Behrens F, Vitzthum F. Tests for the measurement of factor VII-activating protease (FSAP) activity and antigen levels in citrated plasma, their correlation to PCR testing, and utility for the detection of the Marburg I-polymorphism of FSAP. Clin Chem Lab Med 2008; 46:1109-16. [PMID: 18605956 DOI: 10.1515/cclm.2008.218] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND The single nucleotide Marburg I (MRI) polymorphism of the factor VII-activating protease (FSAP) gene, the prourokinase-activating activity of FSAP, and antigen levels of FSAP in plasma have been associated with incidence and progression of carotid stenosis and venous thromboembolism. However, more information on the extent of these associations, potential further ones, and respective clinical utilities remain to be determined. At present, testing is performed mainly by PCR assays based on probes or SYBR Green I. Some studies include testing for antigen levels of total FSAP and its ability to activate prourokinase. To test large cohorts, it is beneficial to rely on assays that are cost-effective, reliable, easy to use, rapid to perform, and that may eventually be automated. In addition, it appears advantageous to use functional tests or tests that determine antigen levels as they may relate more closely to the phenotype than the genotype does. METHODS Tests for the measurements of antigen levels of FSAP and its prourokinase-activating activity were improved and performance characteristics assessed. To determine the FSAP genotypes, an amplification created restriction site (ACRS) PCR test was developed. RESULTS Key performance characteristics of the FSAP activity and antigen tests were as follows: measuring range: 350-1400 mPEU/mL and 1.8-120 ng/mL, total coefficients of variation (CV): 5%-20% and 5%-14%, within-run CV: 4%-11% and 2.3%-12%, and run-to-run CV: 2%-17% and 4.3%-8.3%, respectively. The ratio of the activity and antigen level of FSAP correctly identified the FSAP genotypes of 126 samples tested. CONCLUSIONS The ACRS PCR test is useful for laboratories that do not have the equipment to perform probe or SYBR Green I based real-time PCR. Furthermore, the tests developed for the determination of FSAP activity and antigen levels are convenient for determining clinical correlations, even for large population studies. The ratio of activity and antigen level of FSAP appears to be a promising and efficient alternative to molecular diagnostic techniques to detect the MRI polymorphism of FSAP.
Collapse
Affiliation(s)
- Sina Stephan
- Dade Behring Marburg GmbH, A Siemens Company, Marburg, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Wygrecka M, Morty RE, Markart P, Kanse SM, Andreasen PA, Wind T, Guenther A, Preissner KT. Plasminogen activator inhibitor-1 is an inhibitor of factor VII-activating protease in patients with acute respiratory distress syndrome. J Biol Chem 2007; 282:21671-82. [PMID: 17540775 PMCID: PMC8025756 DOI: 10.1074/jbc.m610748200] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Factor VII-activating protease (FSAP) is a novel plasma-derived serine protease structurally homologous to tissue-type and urokinase-type plasminogen activators. We demonstrate that plasminogen activator inhibitor-1 (PAI-1), the predominant inhibitor of tissue-type and urokinase-type plasminogen activators in plasma and tissues, is an inhibitor of FSAP as well. We detected PAI-1·FSAP complexes in addition to high levels of extracellular RNA, an important FSAP cofactor, in bronchoalveolar lavage fluids from patients with acute respiratory distress syndrome. Hydrolytic activity of FSAP was inhibited by PAI-1 with a second-order inhibition rate constant (Ka) of 3.38 ± 1.12 × 105m–1·s–1. Residue Arg346 was a critical recognition element on PAI-1 for interaction with FSAP. RNA, but not DNA, fragments (>400 nucleotides in length) dramatically enhanced the reactivity of PAI-1 with FSAP, and 4 μg·ml–1 RNA increased the Ka to 1.61 ± 0.94 × 106m–1·s–1. RNA also stabilized the active conformation of PAI-1, increasing the half-life for spontaneous conversion of active to latent PAI-1 from 48.4 ± 8 min to 114.6 ± 5 min. In contrast, little effect of DNA on PAI-1 stability was apparent. Residues Arg76 and Lys80 in PAI-1 were key elements mediating binding of nucleic acids to PAI-1. FSAP-driven inhibition of vascular smooth muscle cell proliferation was antagonized by PAI-1, suggesting functional consequences for the FSAP-PAI-1 interaction. These data indicate that extracellular RNA and PAI-1 can regulate FSAP activity, thereby playing a potentially important role in hemostasis and cell functions under various pathophysiological conditions, such as acute respiratory distress syndrome.
Collapse
Affiliation(s)
- Malgorzata Wygrecka
- Department of Biochemistry, Faculty of Medicine, Justus-Liebig-University Giessen, Giessen, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Shibamiya A, Muhl L, Tannert-Otto S, Preissner K, Kanse S. Nucleic acids potentiate Factor VII-activating protease (FSAP)-mediated cleavage of platelet-derived growth factor-BB and inhibition of vascular smooth muscle cell proliferation. Biochem J 2007; 404:45-50. [PMID: 17300216 PMCID: PMC1868837 DOI: 10.1042/bj20070166] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
FSAP (Factor VII-activating protease) can cleave and inactivate PDGF-BB (platelet-derived growth factor-BB) and thereby inhibits VSMC (vascular smooth-muscle cell) proliferation. The auto-activation of FSAP is facilitated by negatively charged polyanions such as heparin, dextransulfate or extracellular ribonucleic acids. Since auto-activation is essential for the anti-proliferative function of FSAP, the influence of nucleic acids as cofactors for the FSAP-mediated inhibition of PDGF-BB was investigated. Natural or artificial RNA was an effective cofactor for FSAP mediated PDGF-BB degradation, whereas the effect of DNA was weak. RNA-induced cleavage of PDGF-BB was inhibited by serine protease inhibitors. The pattern of PDGF-BB cleavage was identical with either heparin or RNA as a cofactor. One of the cleavage sites in PDGF-BB was at the positions 160-162 (R160KK162), which is an important region for receptor binding and activation. In VSMCs, PDGF-BB-stimulated DNA synthesis was inhibited by FSAP in the presence of RNA. RNA was more effective than DNA and the cofactor activity of RNA was neutralized after pretreatment with RNase. FSAP binding to RNA protected the nucleic acid from degradation by RNase. These data are relevant to situations where extracellular nucleic acids released from necrotic or apoptotic cells could activate local FSAP, leading to inhibition of PDGF-BB.
Collapse
Affiliation(s)
- Aya Shibamiya
- *Institute for Biochemistry, Medical Faculty, Justus-Liebig-University, 35392 Giessen, Germany
- †Graduate School of Health Sciences, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo 113-8519, Japan
| | - Lars Muhl
- *Institute for Biochemistry, Medical Faculty, Justus-Liebig-University, 35392 Giessen, Germany
| | - Susanne Tannert-Otto
- *Institute for Biochemistry, Medical Faculty, Justus-Liebig-University, 35392 Giessen, Germany
| | - Klaus T. Preissner
- *Institute for Biochemistry, Medical Faculty, Justus-Liebig-University, 35392 Giessen, Germany
| | - Sandip M. Kanse
- *Institute for Biochemistry, Medical Faculty, Justus-Liebig-University, 35392 Giessen, Germany
- To whom correspondence should be addressed (email )
| |
Collapse
|
26
|
Wygrecka M, Markart P, Fink L, Guenther A, Preissner KT. Raised protein levels and altered cellular expression of factor VII activating protease (FSAP) in the lungs of patients with acute respiratory distress syndrome (ARDS). Thorax 2007; 62:880-8. [PMID: 17483138 PMCID: PMC2094251 DOI: 10.1136/thx.2006.069658] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
BACKGROUND The acute respiratory distress syndrome (ARDS) is characterised by inflammation of the lung parenchyma and changes in alveolar haemostasis with extravascular fibrin deposition. Factor VII activating protease (FSAP) is a recently described serine protease in plasma and tissues known to be involved in haemostasis, cell proliferation and migration. METHODS The level of FSAP protein expression was examined by western blotting/ELISA/immunohistochemistry and its activity was investigated by coagulation/fibrinolysis assays in plasma, bronchoalveolar lavage (BAL) fluid and lung tissue of mechanically ventilated patients with early ARDS and compared with patients with cardiogenic pulmonary oedema and healthy controls. Cell culture experiments were performed to assess the influence of different inflammatory stimuli on FSAP expression by various cell populations of the lung. RESULTS FSAP protein level and activity were markedly increased in the plasma and BAL fluid of patients with ARDS with a significant contribution to the increased alveolar procoagulant activity. Immunoreactivity for FSAP was observed in alveolar macrophages, bronchial epithelial and endothelial cells of lungs of patients with ARDS, while in controls the immunoreactivity for FSAP was restricted to alveolar macrophages. Only a low basal level of FSAP expression was detected in these cell populations. However, FSAP-specific mRNA expression was induced by lipopolysaccharide and interleukin-8 in human lung microvascular endothelial cells and in bronchial epithelial cells. FSAP was also found to be taken up by alveolar macrophages and degraded within the lysosomal compartment. CONCLUSIONS Increased levels of FSAP and an altered cellular expression pattern are found in the lungs of patients with ARDS. This may represent a novel pathological mechanism which contributes to pulmonary extravascular fibrin deposition and may also modulate inflammation in the acutely injured lung via haemostasis-independent cellular activities of FSAP.
Collapse
Affiliation(s)
- Malgorzata Wygrecka
- Department of Biochemistry, Faculty of Medicine, University of Giessen Lung Center, Friedrichstrasse 24, 35392 Giessen, Germany.
| | | | | | | | | |
Collapse
|
27
|
Nakazawa F, Kannemeier C, Shibamiya A, Song Y, Tzima E, Schubert U, Koyama T, Niepmann M, Trusheim H, Engelmann B, Preissner K. Extracellular RNA is a natural cofactor for the (auto-)activation of Factor VII-activating protease (FSAP). Biochem J 2005; 385:831-8. [PMID: 15654766 PMCID: PMC1134760 DOI: 10.1042/bj20041021] [Citation(s) in RCA: 87] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
FSAP (Factor VII-activating protease) is a new plasma-derived serine protease with putative dual functions in haemostasis, including activation of coagulation Factor VII and generation of urinary-type plasminogen activator (urokinase). The (auto-)activation of FSAP is facilitated by polyanionic glycosaminoglycans, such as heparin or dextran sulphate, whereas calcium ions stabilize the active form of FSAP. In the present study, extracellular RNA was identified and characterized as a novel FSAP cofactor. The conditioned medium derived from various cell types such as smooth muscle cells, endothelial cells, osteosarcoma cells or CHO (Chinese-hamster ovary) cells contained an acidic factor that initiated (auto-)activation of FSAP. RNase A, but not other hydrolytic enzymes (proteases, glycanases and DNase), abolished the FSAP cofactor activity, which was subsequently isolated by anion-exchange chromatography and unequivocally identified as RNA. In purified systems, as well as in plasma, different forms of natural RNA (rRNA, tRNA, viral RNA and artificial RNA) were able to (auto-)activate FSAP into the two-chain enzyme form. The specific binding of FSAP to RNA (but not to DNA) was shown by mobility-shift assays and UV crosslinking, thereby identifying FSAP as a new extracellular RNA-binding protein, the K(D) estimated to be 170-350 nM. Activation of FSAP occurred through an RNA-dependent template mechanism involving a nucleic acid size of at least 100 nt. In a purified system, natural RNA augmented the FSAP-dependent Factor VII activation several-fold (as shown by subsequent Factor Xa generation), as well as the FSAP-mediated generation of urokinase. Our results provide evidence for the first time that extracellular RNA, present at sites of cell damage or vascular injury, can serve an important as yet unrecognized cofactor function in haemostasis by inducing (auto-)activation of FSAP through a novel surface-dependent mechanism.
Collapse
Affiliation(s)
- Fumie Nakazawa
- *Graduate School of Allied Health Sciences, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo 113-8519, Japan
| | | | - Aya Shibamiya
- ‡Institute for Biochemistry, Medical Faculty, Justus-Liebig-Universität, D-35392 Giessen, Germany
| | - Yutong Song
- ‡Institute for Biochemistry, Medical Faculty, Justus-Liebig-Universität, D-35392 Giessen, Germany
| | - Eleni Tzima
- ‡Institute for Biochemistry, Medical Faculty, Justus-Liebig-Universität, D-35392 Giessen, Germany
| | - Uwe Schubert
- ‡Institute for Biochemistry, Medical Faculty, Justus-Liebig-Universität, D-35392 Giessen, Germany
| | - Takatoshi Koyama
- *Graduate School of Allied Health Sciences, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo 113-8519, Japan
| | - Michael Niepmann
- ‡Institute for Biochemistry, Medical Faculty, Justus-Liebig-Universität, D-35392 Giessen, Germany
| | - Heidi Trusheim
- ‡Institute for Biochemistry, Medical Faculty, Justus-Liebig-Universität, D-35392 Giessen, Germany
| | - Bernd Engelmann
- §Institute of Clinical Chemistry, Ludwig-Maximilians-Universität, D-81377 München, Germany
| | - Klaus T. Preissner
- ‡Institute for Biochemistry, Medical Faculty, Justus-Liebig-Universität, D-35392 Giessen, Germany
- To whom correspondence should addressed (email )
| |
Collapse
|
28
|
Abstract
The molecular mechanisms that finely co-ordinate fibrin formation and fibrinolysis are now well defined. The structure and function of all major fibrinolytic proteins, which include serine proteases, their inhibitors, activators and receptors, have been characterized. Measurements of real time, dynamic molecular interactions during fibrinolysis of whole blood clots can now be carried out in vitro. The development of gene-targeted mice deficient in one or more fibrinolytic protein(s) has demonstrated expected and unexpected roles for these proteins in both intravascular and extravascular settings. In addition, genetic analysis of human deficiency syndromes has revealed specific mutations that result in human disorders that are reflective of either fibrinolytic deficiency or excess. Elucidation of the fine control of fibrinolysis under different physiological and pathological haemostatic states will undoubtedly lead to novel therapeutic interventions. Here, we review the fundamental features of intravascular plasmin generation, and consider the major clinical syndromes resulting from abnormalities in fibrinolysis.
Collapse
Affiliation(s)
- Gabriela Cesarman-Maus
- Department of Cell and Developmental Biology, Weill Medical College of Cornell University, New York-Presbyterian Hospital, New York, NY 10021, USA
| | | |
Collapse
|
29
|
Anderson L. Candidate-based proteomics in the search for biomarkers of cardiovascular disease. J Physiol 2005; 563:23-60. [PMID: 15611012 PMCID: PMC1665562 DOI: 10.1113/jphysiol.2004.080473] [Citation(s) in RCA: 267] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2004] [Accepted: 12/16/2004] [Indexed: 11/08/2022] Open
Abstract
The key concept of proteomics (looking at many proteins at once) opens new avenues in the search for clinically useful biomarkers of disease, treatment response and ageing. As the number of proteins that can be detected in plasma or serum (the primary clinical diagnostic samples) increases towards 1000, a paradoxical decline has occurred in the number of new protein markers approved for diagnostic use in clinical laboratories. This review explores the limitations of current proteomics protein discovery platforms, and proposes an alternative approach, applicable to a range of biological/physiological problems, in which quantitative mass spectrometric methods developed for analytical chemistry are employed to measure limited sets of candidate markers in large sets of clinical samples. A set of 177 candidate biomarker proteins with reported associations to cardiovascular disease and stroke are presented as a starting point for such a 'directed proteomics' approach.
Collapse
|
30
|
Liu S, Aaronson H, Mitola DJ, Leppla SH, Bugge TH. Potent antitumor activity of a urokinase-activated engineered anthrax toxin. Proc Natl Acad Sci U S A 2003; 100:657-62. [PMID: 12525700 PMCID: PMC141052 DOI: 10.1073/pnas.0236849100] [Citation(s) in RCA: 91] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The acquisition of cell-surface urokinase plasminogen activator activity is a hallmark of malignancy. We generated an engineered anthrax toxin that is activated by cell-surface urokinase in vivo and displays limited toxicity to normal tissue but broad and potent tumoricidal activity. Native anthrax toxin protective antigen, when administered with a chimeric anthrax toxin lethal factor, Pseudomonas exotoxin fusion protein, was extremely toxic to mice, causing rapid and fatal organ damage. Replacing the furin activation sequence in anthrax toxin protective antigen with an artificial peptide sequence efficiently activated by urokinase greatly attenuated toxicity to mice. In addition, the mutation conferred cell-surface urokinase-dependent toxin activation in vivo, as determined by using a panel of plasminogen, plasminogen activator, plasminogen activator receptor, and plasminogen activator inhibitor-deficient mice. Surprisingly, toxin activation critically depended on both urokinase plasminogen activator receptor and plasminogen in vivo, showing that both proteins are essential cofactors for the generation of cell-surface urokinase. The engineered toxin displayed potent tumor cell cytotoxicity to a spectrum of transplanted tumors of diverse origin and could eradicate established solid tumors. This tumoricidal activity depended strictly on tumor cell-surface plasminogen activation. The data show that a simple change of protease activation specificity converts anthrax toxin from a highly lethal to a potent tumoricidal agent.
Collapse
Affiliation(s)
- Shihui Liu
- Oral Infection and Immunity Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | |
Collapse
|
31
|
Curino A, Mitola DJ, Aaronson H, McMahon GA, Raja K, Keegan AD, Lawrence DA, Bugge TH. Plasminogen promotes sarcoma growth and suppresses the accumulation of tumor-infiltrating macrophages. Oncogene 2002; 21:8830-42. [PMID: 12483535 DOI: 10.1038/sj.onc.1205951] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2002] [Revised: 07/31/2002] [Accepted: 08/07/2002] [Indexed: 11/09/2022]
Abstract
The specific functions of plasminogen, stromal plasminogen activator, stromal plasminogen activator receptor, and stromal plasminogen activator inhibitor in the progression of the murine soft tissue sarcoma, T241 were investigated. Negation of plasminogen to the tumor blunted the orthotopic growth of the sarcoma in syngeneic mice. The reduced tumor growth was associated with a dramatic increase in tumor-infiltrating F4/80-positive macrophages and a diminution of vessel density, but not with obvious differences in fibrin and collagen deposition, or invasiveness of the tumor. Ablation of plasminogen activation by the tumor stroma only modestly impaired the prolonged growth of the sarcoma, suggesting that tumor cell-produced plasminogen activator is sufficient to mediate productive plasminogen activation. Plasminogen facilitated sarcoma progression, angiogenesis, and suppression of macrophage infiltration in the absence of either stromal urokinase plasminogen activator receptor or stromal plasminogen activator inhibitor. These data demonstrate that tumor cell-produced plasminogen activator and host plasminogen cooperate to facilitate soft tissue sarcoma growth and suppress the accumulation of tumor-infiltrating macrophages.
Collapse
Affiliation(s)
- Alejandro Curino
- Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, 30 Convent Drive, Bethesda, Maryland, MD 20892, USA
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Choi-Miura NH, Otsuyama K, Sano Y, Saito K, Takahashi K, Tomita M. Hepatic injury-specific conversion of mouse plasma hyaluronan binding protein to the active hetero-dimer form. Biol Pharm Bull 2001; 24:892-6. [PMID: 11510480 DOI: 10.1248/bpb.24.892] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Plasma hyaluronan binding protein (PHBP) is produced only in liver and kidney in mouse. The induction of PHBP mRNA and the conversion of pro PHBP to the active hetero-dimer form were studied after CCl4, D-galactosamine, HgCl2 or turpentine administration and after partial hepatectomy. The results indicated that the administrations of CCl4 and D-galactosamine, which caused hepatic failure, and the partial hepatectomy enhanced the conversion of pro PHBP to the active two-chain form in the plasma. On the other hand, HgCl2 which injured kidney and turpentine which led to inflammation were not involved in the activation of PHBP. The weak induction and suppression of PHBP mRNA were observed in the liver at 3 h and 12 h, respectively, after the CCl4 administration. However, HgCl2 and turpentine did not influence the amount of PHBP mRNA. These results suggested the hepatic injury-specific activation of PHBP in plasma. PHBP may act as an early factor in the cascade for the tissue remodeling in liver following hepatic injury, i.e., PHBP activates urokinase, urokinase activates matrix metalloproteinases (MMPs) and MMPs degrade extracellular matrix for liver regeneration.
Collapse
Affiliation(s)
- N H Choi-Miura
- Department of Physiological Chemistry, School of Pharmaceutical Sciences, Showa University, Shinagawa, Tokyo, Japan.
| | | | | | | | | | | |
Collapse
|