1
|
Factor VII Activating Protease (FSAP) and Its Importance in Hemostasis—Part I: FSAP Structure, Synthesis and Activity Regulation: A Narrative Review. Int J Mol Sci 2023; 24:ijms24065473. [PMID: 36982544 PMCID: PMC10052181 DOI: 10.3390/ijms24065473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 03/11/2023] [Accepted: 03/12/2023] [Indexed: 03/15/2023] Open
Abstract
Factor VII activating protease (FSAP) was first isolated from human plasma less than 30 years ago. Since then, many research groups have described the biological properties of this protease and its role in hemostasis and other processes in humans and other animals. With the progress of knowledge about the structure of FSAP, several of its relationships with other proteins or chemical compounds that may modulate its activity have been explained. These mutual axes are described in the present narrative review. The first part of our series of manuscripts on FSAP describes the structure of this protein and the processes leading to the enhancement and inhibition of its activities. The following parts, II and III, concern the role of FSAP in hemostasis and in the pathophysiology of human diseases, with particular emphasis on cardiovascular diseases.
Collapse
|
2
|
Cheng L, Wang D, Wang Z, Li H, Wang G, Wu Z, Xu M, Yan S, Zhan H, Wang H, Zhang X, Liang T, Wei C, Zhang F, Zheng W, Yu X, Li Y. Proteomics Landscape Mapping of Organ-Resolved Behçet's Disease Using In-Depth Plasma Proteomics for Identifying Hyaluronic Binding Protein 2 Expression Associated With Vascular Involvement. Arthritis Rheumatol 2023; 75:424-437. [PMID: 36122191 DOI: 10.1002/art.42348] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 08/19/2022] [Accepted: 09/07/2022] [Indexed: 01/14/2023]
Abstract
OBJECTIVE This study was undertaken to elucidate the pathogenesis and heterogeneity of Behçet's disease (BD) involving different organs using in-depth proteomics to identify the biomarkers for clinical assessment and treatment of patients with BD. METHODS We measured the expression levels of proteins in plasma samples from 98 patients with BD and from 31 healthy controls using our in-depth proteomics platform with a data-independent acquisition mass spectrometer and antibody microarray. We performed bioinformatics analyses of the biologic processes and signaling pathways that were changed in the BD group and constructed a proteomics landscape of organ-resolved BD pathogenesis. We then validated the biomarkers of disease severity and the vascular subset in an independent cohort of 108 BD patients and 29 healthy controls using an enzyme-linked immunosorbent assay. RESULTS The BD group had 220 differentially expressed proteins, which discriminated between BD patients (88.6%) and healthy controls (95.5%). The bioinformatics analyses revealed different biologic processes associated with BD pathogeneses, including complement activation, wound healing, angiogenesis, and leukocyte-mediated immunity. Furthermore, the constructed proteomics landscape of organ-resolved BD identified proteomics features of BD associated with different organs and protein targets that could be used for the development of therapeutic treatment. Hyaluronic binding protein 2, tenascin, and serpin A3 were validated as potential biomarkers for the clinical assessment of vascular BD and treatment targets. CONCLUSION Our results provide valuable insight into the pathogenesis of organ-resolved BD in terms of proteomics characteristics and potential biomarkers for clinical assessment and potential therapies for vascular BD.
Collapse
Affiliation(s)
- Linlin Cheng
- Department of Clinical Laboratory, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Dongxue Wang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Science-Beijing (PHOENIX Center), Beijing Institute of Lifeomics, Beijing, China
| | - Zhimian Wang
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, State Key Laboratory of Complex Severe and Rare Diseases, and Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Haolong Li
- Department of Clinical Laboratory, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Guibin Wang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Science-Beijing (PHOENIX Center), Beijing Institute of Lifeomics, Beijing, China
| | - Ziyan Wu
- Department of Clinical Laboratory, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Meng Xu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Science-Beijing (PHOENIX Center), Beijing Institute of Lifeomics, Beijing, China
| | - Songxin Yan
- Department of Clinical Laboratory, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Haoting Zhan
- Department of Clinical Laboratory, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Hongye Wang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Science-Beijing (PHOENIX Center), Beijing Institute of Lifeomics, Beijing, China
| | - Xiaomei Zhang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Science-Beijing (PHOENIX Center), Beijing Institute of Lifeomics, Beijing, China
| | - Te Liang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Science-Beijing (PHOENIX Center), Beijing Institute of Lifeomics, Beijing, China
| | - Chundi Wei
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Science-Beijing (PHOENIX Center), Beijing Institute of Lifeomics, Beijing, China
| | - Fengchun Zhang
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, State Key Laboratory of Complex Severe and Rare Diseases, and Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Wenjie Zheng
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, State Key Laboratory of Complex Severe and Rare Diseases, and Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Xiaobo Yu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Science-Beijing (PHOENIX Center), Beijing Institute of Lifeomics, Beijing, China
| | - Yongzhe Li
- Department of Clinical Laboratory, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| |
Collapse
|
3
|
Jiang Y, Li J, Sang C, Cao G, Wang S. Diagnostic and prognostic value of HABP2 as a novel biomarker for endometrial cancer. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:1164. [PMID: 33241013 PMCID: PMC7576057 DOI: 10.21037/atm-20-5744] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
BACKGROUND Endometrial cancer is the fifth most common malignant disorder in women, with its incidence increasing. A biomarker with diagnostic and prognostic value remains to be found. The HABP2 protein, or Factor VII-activating protease, encodes a hyaluronic acid-binding protein. METHODS Patient data including clinical characteristics and RNAseq information of HABP2 was obtained from The Cancer Genome Atlas (TCGA), and analyzed by R statistic packages. A total of 370 women with endometrial cancer were enrolled in the study. To study the diagnostic value of HABP2 in patients with endometrial cancer, receiver operating characteristic (ROC) curves were plotted by the pROC package. To study the prognostic value of HABP2 in patients with endometrial cancer, the survival package in R was used and the Cox model was established. RESULTS HABP2 expression was lower in endometrial cancer compared with normal endometrial tissues. HABP2 showed moderate diagnostic value for endometrial cancer, with HBP2 expression associated with vital status, histologic grade, and residual tumor. HABP2 was an independent prognostic factor, with low HABP2 expression indicating a better overall survival. CONCLUSIONS HABP2 has diagnostic and prognostic value and maybe a novel biomarker for endometrial cancer.
Collapse
Affiliation(s)
- Ying Jiang
- Department of Obstetrics and Gynecology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Jinfeng Li
- Department of Obstetrics and Gynecology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Cuiqin Sang
- Department of Obstetrics and Gynecology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Guangming Cao
- Department of Obstetrics and Gynecology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Shuzhen Wang
- Department of Obstetrics and Gynecology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
4
|
Byskov K, Le Gall SM, Thiede B, Camerer E, Kanse SM. Protease activated receptors (PAR)-1 and -2 mediate cellular effects of factor VII activating protease (FSAP). FASEB J 2019; 34:1079-1090. [PMID: 31914657 DOI: 10.1096/fj.201801986rr] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 11/13/2019] [Accepted: 11/14/2019] [Indexed: 01/30/2023]
Abstract
Factor VII activating protease (FSAP) is a circulating serine protease implicated in thrombosis, atherosclerosis, stroke, and cancer. Using an overexpression strategy, we have systematically investigated the role of protease activated receptors (PAR)-1, -2, -3, and -4 on FSAP-mediated signaling in HEK293T and A549 cells. Cleavage of PAR-reporter constructs and MAPK phosphorylation was used to monitor receptor activation. FSAP cleaved PAR-2 and to a lesser degree PAR-1, but not PAR-3 or PAR-4 in both cell types. Robust MAPK activation in response to FSAP was observed after PAR-2, but not PAR-1 overexpression in HEK293T. Recombinant serine protease domain of wild type FSAP, but not the Marburg I isoform of FSAP, could reproduce the effects of plasma purified FSAP. Canonical cleavage of both PARs was suggested by mass spectrometric analysis of synthetic peptide substrates from the N-terminus of PARs and site directed mutagenesis studies. Surprisingly, knockdown of endogenous PAR-1, but not PAR-2, prevented the apoptosis-inhibitory effect of FSAP, suggesting that PAR1 is nevertheless a direct or indirect target in some cell types. This molecular characterization of PAR-1 and -2 as cellular receptors of FSAP will help to define the actions of FSAP in the context of cancer and vascular biology.
Collapse
Affiliation(s)
- Kristina Byskov
- Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Sylvain M Le Gall
- INSERM U970, Paris Cardiovascular Research Centre, Université de Paris, Paris, France
| | - Bernd Thiede
- Department of Biosciences, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway
| | - Eric Camerer
- INSERM U970, Paris Cardiovascular Research Centre, Université de Paris, Paris, France
| | - Sandip M Kanse
- Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway.,Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
5
|
VEGF-A-Cleavage by FSAP and Inhibition of Neo-Vascularization. Cells 2019; 8:cells8111396. [PMID: 31698750 PMCID: PMC6912458 DOI: 10.3390/cells8111396] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 10/28/2019] [Accepted: 11/04/2019] [Indexed: 12/23/2022] Open
Abstract
Alternative splicing leads to the secretion of multiple forms of vascular endothelial growth factor-A (VEGF-A) that differ in their activity profiles with respect to neovascularization. FSAP (factor VII activating protease) is the zymogen form of a plasma protease that is activated (FSAPa) upon tissue injury via the release of histones. The purpose of the study was to determine if FSAPa regulates VEGF-A activity in vitro and in vivo. FSAP bound to VEGF165, but not VEGF121, and VEGF165 was cleaved in its neuropilin/proteoglycan binding domain. VEGF165 cleavage did not alter its binding to VEGF receptors but diminished its binding to neuropilin. The stimulatory effects of VEGF165 on endothelial cell proliferation, migration, and signal transduction were not altered by FSAP. Similarly, proliferation of VEGF receptor-expressing BAF3 cells, in response to VEGF165, was not modulated by FSAP. In the mouse matrigel model of angiogenesis, FSAP decreased the ability of VEGF165, basic fibroblast growth factor (bFGF), and their combination, to induce neovascularization. Lack of endogenous FSAP in mice did not influence neovascularization. Thus, FSAP inhibited VEGF165-mediated angiogenesis in the matrigel model in vivo, where VEGF’s interaction with the matrix and its diffusion are important.
Collapse
|
6
|
Vadgama N, Lamont D, Hardy J, Nasir J, Lovering RC. Distinct proteomic profiles in monozygotic twins discordant for ischaemic stroke. Mol Cell Biochem 2019; 456:157-165. [PMID: 30694515 DOI: 10.1007/s11010-019-03501-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 01/19/2019] [Indexed: 01/13/2023]
Abstract
Stroke is a common disorder with significant morbidity and mortality, and complex aetiology involving both environmental and genetic risk factors. Although some of the major risk factors for stoke, such as smoking and hypertension, are well-documented, the underlying genetic and detailed molecular mechanisms remain elusive. Exploring the relevant biochemical pathways may contribute to the clinical diagnosis of stroke and shed light on its aetiology. A comparative proteomic analysis of blood serum of a pair of monozygotic (MZ) twins discordant for ischaemic stroke (IS) was performed using a label-free quantitative proteomics approach. To overcome the limit of reproducibility in the serum preparation, two separate runs were performed, each consisting of three technical replicates per sample. Biological processes associated with proteins differentially expressed between the twins were explored with gene ontology (GO) classification using the functional analysis tool g:Profiler. ANOVA test performed in Progenesis LC-MS identified 179 (run 1) and 209 (run 2) proteins as differentially expressed between the affected and unaffected twin (p < 0.05). Furthermore, the level of serum fibulin 1, an extracellular matrix protein associated with arterial stiffness, was on average 13.37-fold higher in the affected twin. Each dataset was then analysed independently, and the proteins were classified according to GO terms. The categories overrepresented in the affected twin predominantly corresponded to stroke-relevant processes, including wound healing, blood coagulation and haemostasis, with a high proportion of the proteins overexpressed in the affected twin associated with these terms. By contrast, in the unaffected twin diagnosed with atopic dermatitis, there were increased levels of keratin proteins and GO terms associated with skin development. The identification of cellular pathways enriched in IS as well as the upregulation of fibulin 1 sheds new light on the underlying disease-causing mechanisms at the molecular level. Our findings of distinct proteomic signatures associated with IS and atopic dermatitis suggest proteomic profiling could be used as a general approach for improved diagnostic, prognostic and therapeutic strategies.
Collapse
Affiliation(s)
- Nirmal Vadgama
- Institute of Neurology, University College London, London, UK
- Cell Biology and Genetics Research Centre, St. George's University of London, London, UK
| | - Douglas Lamont
- College of Life Sciences, University of Dundee, Dundee, UK
| | - John Hardy
- Institute of Neurology, University College London, London, UK
| | - Jamal Nasir
- Cell Biology and Genetics Research Centre, St. George's University of London, London, UK.
- Molecular Biosciences Research Group, Faculty of Health & Society, University of Northampton, Northampton, UK.
| | - Ruth C Lovering
- Centre for Cardiovascular Genetics, Institute of Cardiovascular Science, University College London, London, UK
| |
Collapse
|
7
|
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: 1.5] [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
|
8
|
Etscheid M, Kanse SM. Factor VII activating protease. Hamostaseologie 2017; 31:174-178. [DOI: 10.5482/ha-1159] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2011] [Accepted: 05/26/2011] [Indexed: 02/06/2023] Open
Abstract
SummaryFactor VII activating protease (FSAP) is a circulating serine protease with high homology to fibrinolytic enzymes. A role in the regulation of coagulation and fibrinolysis is suspected based on in vitro studies demonstrating activation of FVII or pro-urokinase plasminogen activator (uPA). However, considering the paucity of any studies in animal models or any correlative studies in humans the role of FSAP in haemostasis remains unclear. In relation to vascular remodeling processes or inflammation it has been convincingly shown that FSAP interacts with growth factors as well as protease activated receptors (PAR). Against this sparse background there are a plethora of studies which have investigated the linkage of single nucleotide polymorphisms (SNP) in the FSAP gene (HABP2) to various diseases. The G534E SNP of FSAP is associated with a low proteolytic activity due to an amino acid exchange in the protease domain. This and other SNPs have been linked to carotid stenosis, stroke as well as thrombosis in the elderly and plaque calcification. These SNP analyses indicate an important role for FSAP in the regulation of the haemostasis system as well as fibroproliferative inflammatory processes.
Collapse
|
9
|
Stephan F, Aarden LA, Zeerleder S. FSAP, a new player in inflammation? Hamostaseologie 2017; 32:51-5. [DOI: 10.5482/ha-1187] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2011] [Accepted: 11/04/2011] [Indexed: 12/11/2022] Open
Abstract
SummaryFactor VII-activating protease (FSAP) is a serine protease in plasma that has a role in coagulation and fibrinolysis. FVII could be activated by purified FSAP in a tissue factor independent manner and pro-urokinase has been demonstrated to be a substrate for purified FSAP in-vitro. However, the physiological role of FSAP in haemostasis remains unclear. More recently FSAP is suggested to be involved in inflammation. It modulates vascular permeability directly and indirectly by the generation of bradykinin. Furthermore, FSAP is activated by dead cells induced by the inflammatory response and subsequently removes nucleosomes from apoptotic cells. FSAP activation can be detected in sepsis patients as well. However, whether FSAP activation upon inflammation is beneficial or detrimental remains an open question.In this review the structure, activation mechanisms and the possible role of FSAP in inflammation are discussed.
Collapse
|
10
|
Factor VII activating protease (FSAP) regulates the expression of inflammatory genes in vascular smooth muscle and endothelial cells. Atherosclerosis 2017; 265:133-139. [DOI: 10.1016/j.atherosclerosis.2017.08.029] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 08/22/2017] [Accepted: 08/23/2017] [Indexed: 02/04/2023]
|
11
|
Ramanathan R, Gram JB, Sand NPR, Nørgaard BL, Diederichsen AC, Vitzthum F, Schwarz H, Sidelmann JJ. Factor VII-activating protease. Blood Coagul Fibrinolysis 2017; 28:558-563. [DOI: 10.1097/mbc.0000000000000640] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
12
|
Daniel JM, Reichel CA, Schmidt-Woell T, Dutzmann J, Zuchtriegel G, Krombach F, Herold J, Bauersachs J, Sedding DG, Kanse SM. Factor VII-activating protease deficiency promotes neointima formation by enhancing leukocyte accumulation. J Thromb Haemost 2016; 14:2058-2067. [PMID: 27431088 DOI: 10.1111/jth.13417] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 07/01/2016] [Indexed: 01/08/2023]
Abstract
Essentials Factor VII-activating protease (FSAP) is a plasma protease involved in vascular processes. Neointima formation was investigated after vascular injury in FSAP-/- mice. The neointimal lesion size and the accumulation of macrophages were increased in FSAP-/- mice. This was due to an increased activity of the chemokine (C-C motif) ligand 2 (CCL2). SUMMARY Background Factor VII-activating protease (FSAP) is a multifunctional circulating plasma serine protease involved in thrombosis and vascular remodeling processes. The Marburg I single-nucleotide polymorphism (MI-SNP) in the FSAP-coding gene is characterized by low proteolytic activity, and is associated with increased rates of stroke and carotid stenosis in humans. Objectives To determine whether neointima formation after vascular injury is increased in FSAP-/- mice. Methods and Results The neointimal lesion size and the proliferation of vascular smooth muscle cells (VSMCs) were significantly enhanced in FSAP-/- mice as compared with C57BL/6 control mice after wire-induced injury of the femoral artery. Accumulation of leukocytes and macrophages was increased within the lesions of FSAP-/- mice at day 3 and day 14. Quantitative zymography demonstrated enhanced activity of gelatinases/matrix metalloproteinase (MMP)-2 and MMP-9 within the neointimal lesions of FSAP-/- mice, and immunohistochemistry showed particular costaining of MMP-9 with accumulating leukocytes. Using intravital microscopy, we observed that FSAP deficiency promoted the intravascular adherence and the subsequent transmigration of leukocytes in vivo in response to chemokine ligand 2 (CCL2). CCL2 expression was increased in FSAP-/- monocytes but not in the vessel wall. There was no difference in the expression of platelet-derived growth factor (PDGF-BB). Conclusions FSAP deficiency causes an increase in CCL2 expression and CCL2-mediated infiltration of leukocytes into the injured vessel, thereby promoting SMC proliferation and migration by the activation of leukocyte-derived gelatinases. These results provide a possible explanation for the observed association of the loss-of-function MI-SNP with vascular proliferative diseases.
Collapse
Affiliation(s)
- J-M Daniel
- Department of Biochemistry, Justus-Liebig-Universität Giessen, Giessen, Germany
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - C A Reichel
- Department of Otorhinolaryngology, Head and Neck Surgery, Ludwig-Maximilians-Universität München, Munich, Germany
- Walter Brendel Center of Experimental Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
| | - T Schmidt-Woell
- Department of Biochemistry, Justus-Liebig-Universität Giessen, Giessen, Germany
| | - J Dutzmann
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - G Zuchtriegel
- Department of Otorhinolaryngology, Head and Neck Surgery, Ludwig-Maximilians-Universität München, Munich, Germany
- Walter Brendel Center of Experimental Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
| | - F Krombach
- Walter Brendel Center of Experimental Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
| | - J Herold
- Department of Cardiology, Angiology and Pneumology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - J Bauersachs
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - D G Sedding
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - S M Kanse
- Department of Biochemistry, Justus-Liebig-Universität Giessen, Giessen, Germany.
- Oslo University Hospital and University of Oslo, Oslo, Norway.
| |
Collapse
|
13
|
Coagulation Factors in the Interstitial Space. Protein Sci 2016. [DOI: 10.1201/9781315374307-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
14
|
Bustamante A, Díaz-Fernández B, Giralt D, Boned S, Pagola J, Molina CA, García-Berrocoso T, Kanse SM, Montaner J. Factor seven activating protease (FSAP) predicts response to intravenous thrombolysis in acute ischemic stroke. Int J Stroke 2016; 11:646-55. [PMID: 27073188 DOI: 10.1177/1747493016641949] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Accepted: 01/17/2016] [Indexed: 11/15/2022]
Abstract
BACKGROUND Prediction of recanalization after intravenous thrombolysis could be important to direct secondary reperfusion techniques. Factor seven activating protease (FSAP) has been described to have a relevant pathophysiological role in stroke. AIM The aim is to determine whether plasma FSAP levels are associated with recanalization after tissue plasminogen activator in acute stroke. METHODS FSAP antigen, activity, and FSAP-inhibitor complexes were measured in 120 acute stroke patients admitted to Hospital Vall d'Hebron with arterial occlusions, before intravenous thrombolysis. Recanalization was assessed by transcranial Doppler 2 h after thrombolysis. Predictors of recanalization were determined by logistic regression analysis and the additional predictive value of FSAP over them was determined by integrated discrimination improvement index. RESULTS Complete recanalization was achieved in 31 patients. FSAP antigen levels were lower in patients achieving recanalization (8.2 (6.3-11.7) µg/mL vs. 9.8 (7.6-12.8) µg/mL; p = 0.046). After adjustment by age, sex, and National Institutes of Health Stroke Scale, Oxfordshire Community Stroke Project (odds ratio = 0.33 (0.13-0.82), p = 0.017) and FSAP antigen (odds ratio = 3.22 (1.22-8.47), p = 0.018) were independently associated with recanalization, and the addition of FSAP improved the model discrimination (integrated discrimination improvement = 5.5% (1.4-9.7), p = 0.009). CONCLUSIONS Our study showed that lower FSAP antigen plasma levels were associated with a higher chance of arterial recanalization after tissue plasminogen activator treatment, suggesting an involvement of FSAP in tissue plasminogen activator-induced clot lysis. FSAP antigen determination might be useful in predicting tissue plasminogen activator response in stroke patients.
Collapse
Affiliation(s)
- Alejandro Bustamante
- Neurovascular Research Laboratory, Institut de Recerca, Universitat Autònoma de Barcelona, Barcelona, Spain
| | | | - Dolors Giralt
- Neurovascular Research Laboratory, Institut de Recerca, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Sandra Boned
- Stroke Unit, Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | - Jorge Pagola
- Stroke Unit, Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | - Carlos A Molina
- Stroke Unit, Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | - Teresa García-Berrocoso
- Neurovascular Research Laboratory, Institut de Recerca, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Sandip M Kanse
- Oslo University Hospital, University of Olso, Oslo, Norway
| | - Joan Montaner
- Neurovascular Research Laboratory, Institut de Recerca, Universitat Autònoma de Barcelona, Barcelona, Spain Stroke Unit, Hospital Universitari Vall d'Hebron, Barcelona, Spain
| |
Collapse
|
15
|
Mudadu MA, Porto-Neto LR, Mokry FB, Tizioto PC, Oliveira PSN, Tullio RR, Nassu RT, Niciura SCM, Tholon P, Alencar MM, Higa RH, Rosa AN, Feijó GLD, Ferraz ALJ, Silva LOC, Medeiros SR, Lanna DP, Nascimento ML, Chaves AS, Souza ARDL, Packer IU, Torres RAA, Siqueira F, Mourão GB, Coutinho LL, Reverter A, Regitano LCA. Genomic structure and marker-derived gene networks for growth and meat quality traits of Brazilian Nelore beef cattle. BMC Genomics 2016; 17:235. [PMID: 26979536 PMCID: PMC4791965 DOI: 10.1186/s12864-016-2535-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 02/25/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Nelore is the major beef cattle breed in Brazil with more than 130 million heads. Genome-wide association studies (GWAS) are often used to associate markers and genomic regions to growth and meat quality traits that can be used to assist selection programs. An alternative methodology to traditional GWAS that involves the construction of gene network interactions, derived from results of several GWAS is the AWM (Association Weight Matrices)/PCIT (Partial Correlation and Information Theory). With the aim of evaluating the genetic architecture of Brazilian Nelore cattle, we used high-density SNP genotyping data (~770,000 SNP) from 780 Nelore animals comprising 34 half-sibling families derived from highly disseminated and unrelated sires from across Brazil. The AWM/PCIT methodology was employed to evaluate the genes that participate in a series of eight phenotypes related to growth and meat quality obtained from this Nelore sample. RESULTS Our results indicate a lack of structuring between the individuals studied since principal component analyses were not able to differentiate families by its sires or by its ancestral lineages. The application of the AWM/PCIT methodology revealed a trio of transcription factors (comprising VDR, LHX9 and ZEB1) which in combination connected 66 genes through 359 edges and whose biological functions were inspected, some revealing to participate in biological growth processes in literature searches. CONCLUSIONS The diversity of the Nelore sample studied is not high enough to differentiate among families neither by sires nor by using the available ancestral lineage information. The gene networks constructed from the AWM/PCIT methodology were a useful alternative in characterizing genes and gene networks that were allegedly influential in growth and meat quality traits in Nelore cattle.
Collapse
Affiliation(s)
- Maurício A Mudadu
- Embrapa Agricultural Informatics, Av. André Tosello, 209, Campinas, SP, Brazil. .,Embrapa Southeast Livestock, Rodovia Washington Luiz, Km 234, São Carlos, SP, Brazil.
| | - Laercio R Porto-Neto
- Commonwealth Scientific and Industrial Research Organization - Agriculture, 306 Carmody Road, Brisbane, QLD, Australia
| | - Fabiana B Mokry
- Department of Genetics and Evolution, Federal University of São Carlos, Rodovia Washington Luiz, Km 235, São Carlos, SP, Brazil
| | - Polyana C Tizioto
- Department of Genetics and Evolution, Federal University of São Carlos, Rodovia Washington Luiz, Km 235, São Carlos, SP, Brazil
| | - Priscila S N Oliveira
- Department of Genetics and Evolution, Federal University of São Carlos, Rodovia Washington Luiz, Km 235, São Carlos, SP, Brazil
| | - Rymer R Tullio
- Embrapa Southeast Livestock, Rodovia Washington Luiz, Km 234, São Carlos, SP, Brazil
| | - Renata T Nassu
- Embrapa Southeast Livestock, Rodovia Washington Luiz, Km 234, São Carlos, SP, Brazil
| | - Simone C M Niciura
- Embrapa Southeast Livestock, Rodovia Washington Luiz, Km 234, São Carlos, SP, Brazil
| | - Patrícia Tholon
- Embrapa Southeast Livestock, Rodovia Washington Luiz, Km 234, São Carlos, SP, Brazil
| | - Maurício M Alencar
- Embrapa Southeast Livestock, Rodovia Washington Luiz, Km 234, São Carlos, SP, Brazil
| | - Roberto H Higa
- Embrapa Agricultural Informatics, Av. André Tosello, 209, Campinas, SP, Brazil
| | - Antônio N Rosa
- Embrapa Beef Cattle, Av. Rádio Maia, 830, Campo Grande, MS, Brazil
| | - Gélson L D Feijó
- Embrapa Beef Cattle, Av. Rádio Maia, 830, Campo Grande, MS, Brazil
| | - André L J Ferraz
- State University of Mato Grosso do Sul, Rodovia Uems-Aquidauana km 12, Aquidauana, MS, Brazil
| | - Luiz O C Silva
- Embrapa Beef Cattle, Av. Rádio Maia, 830, Campo Grande, MS, Brazil
| | | | - Dante P Lanna
- Department of Animal Science, University of São Paulo, Av. Padua Dias, 11306, Piracicaba, SP, Brazil
| | - Michele L Nascimento
- Department of Animal Science, University of São Paulo, Av. Padua Dias, 11306, Piracicaba, SP, Brazil
| | - Amália S Chaves
- Department of Animal Science, University of São Paulo, Av. Padua Dias, 11306, Piracicaba, SP, Brazil
| | - Andrea R D L Souza
- Faculdade de Medicina Veterinaria e Zootecnia, Federal University of Mato Grosso do Sul, Av. Senador Filinto Müller, 2443, Campo Grande, MS, Brazil
| | - Irineu U Packer
- Department of Animal Science, University of São Paulo, Av. Padua Dias, 11306, Piracicaba, SP, Brazil
| | | | - Fabiane Siqueira
- Embrapa Beef Cattle, Av. Rádio Maia, 830, Campo Grande, MS, Brazil
| | - Gerson B Mourão
- Department of Animal Science, University of São Paulo, Av. Padua Dias, 11306, Piracicaba, SP, Brazil
| | - Luiz L Coutinho
- Department of Animal Science, University of São Paulo, Av. Padua Dias, 11306, Piracicaba, SP, Brazil
| | - Antonio Reverter
- Commonwealth Scientific and Industrial Research Organization - Agriculture, 306 Carmody Road, Brisbane, QLD, Australia
| | - Luciana C A Regitano
- Embrapa Southeast Livestock, Rodovia Washington Luiz, Km 234, São Carlos, SP, Brazil
| |
Collapse
|
16
|
Induction of a heparin-stimulated serine proteinase in sex accessory gland tumors of the Lobund-Wistar rat. Exp Mol Pathol 2015; 99:39-43. [DOI: 10.1016/j.yexmp.2015.04.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Accepted: 04/18/2015] [Indexed: 11/22/2022]
|
17
|
Martinez-Palacian A, Kanse SM, Weiskirchen R. Factor VII activating protease (FSAP): A novel protective factor in liver fibrosis. Proteomics Clin Appl 2014; 8:438-46. [DOI: 10.1002/prca.201300078] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2013] [Revised: 10/25/2013] [Accepted: 11/27/2013] [Indexed: 02/06/2023]
Affiliation(s)
| | - Sandip M. Kanse
- Department of Biochemistry; University of Oslo; Oslo Norway
- Institute for Biochemistry; Justus-Liebig-University; Giessen Germany
| | - Ralf Weiskirchen
- Institute of Clinical Chemistry and Pathobiochemistry; RWTH University Hospital; Aachen Germany
| |
Collapse
|
18
|
Roedel EK, Schwarz E, Kanse SM. The factor VII-activating protease (FSAP) enhances the activity of bone morphogenetic protein-2 (BMP-2). J Biol Chem 2013; 288:7193-203. [PMID: 23341458 DOI: 10.1074/jbc.m112.433029] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Factor VII-activating protease (FSAP) is a circulating protease involved in the pathogenesis of atherosclerosis, calcification, and fibrotic processes. To understand how FSAP controls the balance of local growth factors, we have investigated its effect on the regulation of bone morphogenetic proteins (BMPs). BMP-2 is produced as a large pro-form and secreted as a mature heparin-binding growth factor after intracellular processing by pro-protein convertases (PCs). In this study, we discovered that FSAP enhances the biological activity of mature BMP-2 as well as its pro-form, as shown by osteogenic differentiation of C2C12 myoblasts. These findings were complemented by knockdown of FSAP in hepatocytes, which revealed BMP-2 processing by endogenous FSAP. N-terminal sequencing indicated that pro-BMP-2 was cleaved by FSAP at the canonical PC cleavage site, giving rise to mature BMP-2 (Arg(282)↓Gln(283)), as well as in the N-terminal heparin binding region of mature BMP-2, generating a truncated mature BMP-2 peptide (Arg(289)↓Lys(290)). Similarly, mature BMP-2 was also cleaved to a truncated peptide within its N-terminal region (Arg(289)↓Lys(290)). Plasmin exhibited a similar activity, but it was weaker compared with FSAP. Thrombin, Factor VIIa, Factor Xa, and activated protein C were not effective. These results were further supported by the observation that the mutation of the heparin binding region of BMP-2 inhibited the processing by FSAP but not by PC. Thus, the proteolysis and activation of pro-BMP-2 and mature BMP-2 by FSAP can regulate cell differentiation and calcification in vasculature and may explain why polymorphisms in the gene encoding for FSAP are related to vascular diseases.
Collapse
Affiliation(s)
- Elfie Kathrin Roedel
- Institute for Biochemistry, Justus-Liebig-University Giessen, 35392 Giessen, Germany
| | | | | |
Collapse
|
19
|
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
|
20
|
Affiliation(s)
- J H McVey
- Medical Research Council Centre for Transplantation, King's College London, King's Health Partners, Guy's Hospital, London, UK.
| |
Collapse
|
21
|
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: 1.6] [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
|
22
|
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
|
23
|
Yamamichi S, Fujiwara Y, Kikuchi T, Nishitani M, Matsushita Y, Hasumi K. Extracellular histone induces plasma hyaluronan-binding protein (factor VII activating protease) activation in vivo. Biochem Biophys Res Commun 2011; 409:483-8. [DOI: 10.1016/j.bbrc.2011.05.030] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2011] [Accepted: 05/05/2011] [Indexed: 10/18/2022]
|
24
|
Altmäe S, Kallak TK, Fridén B, Stavreus-Evers A. Variation in hyaluronan-binding protein 2 (HABP2) promoter region is associated with unexplained female infertility. Reprod Sci 2010; 18:485-92. [PMID: 21098215 DOI: 10.1177/1933719110388849] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
We set up to analyze polymorphisms in hyaluronan-binding protein 2 (HABP2) gene in healthy fertile women (n = 158) and in women with unexplained infertility (n = 116) and to investigate the potential role of HABP2 in receptive endometrium. Minor rs1157916 A and the major rs2240879 A alleles together with AA genotypes were significantly less frequent in infertile women than in controls. Immunohistochemistry analysis of endometrial HABP2 expression at the time of implantation identified significantly lower HABP2 protein level in infertile women in stroma and vessels than in fertile women. Migration assay analysis of cultured trophoblast and endothelial cells toward HABP2 protein referred to the function of HABP2 in endometrial endothelial cells. In conclusion, our results indicate that polymorphisms in the regulatory region of HABP2 gene could influence gene expression levels in the receptive endometrium and could thereby be one reason for infertility complications in women with unexplained infertility. Additionally, HABP2 protein involvement in endometrial angiogenesis is proposed.
Collapse
Affiliation(s)
- Signe Altmäe
- Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden.
| | | | | | | |
Collapse
|
25
|
Schwarz H, Stephan S, Sidelmann JJ, Fischer B, Althaus H, Hahn M, Kappel A, Ehm M, Vitzthum F. Qualitative detection of the Marburg I alloenzyme of factor VII-activating protease by an immunoassay and its comparison to PCR testing. Clin Chem Lab Med 2010; 48:1745-9. [PMID: 20828365 DOI: 10.1515/cclm.2010.334] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [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, thromboembolism, atherosclerosis, and the incidence and progression of carotid stenosis. At present, MRI SNP testing is mainly performed using costly nucleic acid analysis. The ratio between FSAP activity and antigen concentrations in citrated plasma has been used to assess the FSAP genotype. METHODS This article describes the development of a prototype ELISA for the detection of the MRI FSAP alloenzyme, and its correlation to FSAP genotypes to assess whether a positive MRI FSAP ELISA result may be used as a surrogate marker for the presence of the MRI SNP. RESULTS ELISA results were correlated with FSAP genotypes from 523 blood donors measured using PCR. Diagnostic sensitivity and specificity of the assay for determination of the genotype were 100% (95% confidence interval [CI]: 93.36-100) and 99.79% (95% CI: 98.80-99.96), respectively. Maximum run-to-run, within-run, and total coefficients of variation were 7.8%, 7.9%, and 9.9%, respectively. No cross-reactivities with homologues of the MRI FSAP alloenzyme were observed. Test performance was not affected by typical interfering compounds. CONCLUSIONS The data demonstrate that an immunoassay applying antibodies specific to the MRI FSAP alloenzyme can provide sufficiently accurate detection of the MRI SNP. This will significantly simplify MRI FSAP testing, particularly in large cohorts.
Collapse
Affiliation(s)
- Herbert Schwarz
- Siemens Healthcare Diagnostics Products GmbH, Marburg, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Sidelmann JJ, Skouby SO, Vitzthum F, Schwarz H, Jespersen J. Hormone therapy affects plasma measures of factor VII-activating protease in younger postmenopausal women. Climacteric 2010; 13:340-6. [DOI: 10.3109/13697131003597027] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
27
|
Muhl L, Galuska SP, Öörni K, Hernández-Ruiz L, Andrei-Selmer LC, Geyer R, Preissner KT, Ruiz FA, Kovanen PT, Kanse SM. High negative charge-to-size ratio in polyphosphates and heparin regulates factor VII-activating protease. FEBS J 2009; 276:4828-39. [DOI: 10.1111/j.1742-4658.2009.07183.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
28
|
Muhl L, Hersemeyer K, Preissner KT, Weimer T, Kanse SM. Structure - function analysis of factor VII activating protease (FSAP): Sequence determinants for heparin binding and cellular functions. FEBS Lett 2009; 583:1994-8. [DOI: 10.1016/j.febslet.2009.05.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2009] [Revised: 04/20/2009] [Accepted: 05/08/2009] [Indexed: 02/07/2023]
|
29
|
Roderfeld M, Weiskirchen R, Atanasova S, Gressner AM, Preissner KT, Roeb E, Kanse SM. Altered factor VII activating protease expression in murine hepatic fibrosis and its influence on hepatic stellate cells. Liver Int 2009; 29:686-91. [PMID: 19018983 DOI: 10.1111/j.1478-3231.2008.01897.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/13/2023]
Abstract
BACKGROUND Platelet-derived growth factor-BB (PDGF-BB) is a profibrotic factor in liver fibrosis through its ability to stimulate hepatic stellate cells (HSC). The liver-derived serine protease factor VII activating protease (FSAP) regulates the activities of PDGF-BB in a cell-specific manner. AIMS Our aim was to determine the influence of FSAP on the activation of HSC and to analyse the regulation of FSAP in hepatic fibrogenesis. METHODS The effect of FSAP on PDGF-stimulated p42/p44 mitogen-activated protein kinase (MAPK) activation in primary rat HSC was determined by Western blotting. Migration and proliferation of HSC was evaluated in Boyden chamber experiments and (3)H-thymidine incorporation assays respectively. Expression of FSAP was analysed in a CCl(4) mouse model of liver fibrosis by Western blot, quantitative real-time polymerase chain reaction and immunohistochemistry. RESULTS FSAP inhibited PDGF-BB-stimulated p42/p44 MAPK phosphorylation, proliferation and migration of HSC. FSAP mRNA expression level was increased 3 h after CCl(4) application and decreased after 18 h and, in established fibrosis, after chronic CCl(4) administration. In parallel, there was a decrease in the circulating FSAP protein in chronic fibrosis. Concurrently, the homogenous hepatic expression pattern of FSAP was disturbed. Immunohistochemistry revealed a decrease of FSAP in hepatocytes in inflammatory and fibrotic lesions. CONCLUSIONS Our results demonstrate an inhibitory effect of FSAP on PDGF-mediated activation of HSC. In addition, FSAP expression is transiently increased in acute-phase reaction but decreased during chronic fibrogenesis, which in turn may influence PDGF-BB availability and myofibroblast activity.
Collapse
Affiliation(s)
- Martin Roderfeld
- Department of Medicine II, Gastroenterology, Justus-Liebig-University Giessen, Giessen, Germany
| | | | | | | | | | | | | |
Collapse
|
30
|
Wasmuth HE, Tag CG, Van de Leur E, Hellerbrand C, Mueller T, Berg T, Puhl G, Neuhaus P, Samuel D, Trautwein C, Kanse SM, Weiskirchen R. The Marburg I variant (G534E) of the factor VII-activating protease determines liver fibrosis in hepatitis C infection by reduced proteolysis of platelet-derived growth factor BB. Hepatology 2009; 49:775-80. [PMID: 19105210 DOI: 10.1002/hep.22707] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
UNLABELLED Genetic risk factors play an important role for the progression of liver fibrosis in chronic hepatitis C virus (HCV) infection, but functional data on specific alleles and their related proteins are limited. Platelet-derived growth factor BB (PDGF-BB) is one of the strongest mitogens for hepatic stellate cells and is considered as a critical soluble mediator of liver fibrosis in vitro and in vivo. The biological activity of PDGF-BB is dependent on its degradation by the factor VII-activating protease (FSAP). Here, we demonstrate that a coding polymorphism (G534E) in the gene for FSAP is significantly associated with severe HCV-induced liver fibrosis (odds ratio, 2.59; P = 0.017), which is independent of age, gender, and presence of diabetes in multivariate analysis. These genetic findings were replicated in a cohort of patients with liver transplantation due to HCV-induced cirrhosis (OR, 2.56; P = 0.011). Functional dissection of the association demonstrates that the single amino acid change encoded by G534E in the FSAP protein does not influence PDGFbeta receptor or alpha-smooth muscle actin expression but completely abrogates FSAP-mediated inhibition of PDGF-BB-induced proliferation of primary stellate cells in vitro. CONCLUSION The G534E variant of FSAP is a risk locus for HCV-induced liver fibrosis and cirrhosis by determining PDGF-BB-mediated hepatic stellate cell proliferation through a single amino acid substitution in FSAP. FSAP G534E might be useful for risk stratification in patients with HCV infection.
Collapse
Affiliation(s)
- Hermann E Wasmuth
- Department of Internal Medicine III, University Hospital Aachen, Aachen, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
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: 11] [Impact Index Per Article: 0.7] [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
|
32
|
Zeerleder S, Zwart B, te Velthuis H, Stephan F, Manoe R, Rensink I, Aarden LA. Nucleosome-releasing factor: a new role for factor VII-activating protease (FSAP). FASEB J 2008; 22:4077-84. [PMID: 18753248 DOI: 10.1096/fj.08-110429] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Plasma proteins such as early complement components and IgM are involved in the removal of late apoptotic or secondary necrotic (sn) cells. We have recently described how a plasma protease that could be inhibited by the protease inhibitor aprotinin was essential to remove nucleosomes from sn cells. An obvious candidate, plasmin, did indeed have nucleosome-releasing factor (NRF) activity. However, recalcified plasma (r-plasma) retained its NRF activity after plasminogen depletion, which suggests the existence of another protease responsible for NRF activity in plasma. In this study we have used size-exclusion and anion-exchange chromatography to purify the protease responsible for NRF activity in plasma. SDS-PAGE analysis of chromatography fractions containing NRF activity revealed a protein band corresponding with NRF activity. Sequence analysis showed this band to be factor VII-activating protease (FSAP). We developed monoclonal antibodies to FSAP and were able to completely inhibit NRF activity in plasma with monoclonal antibodies to FSAP. Using affinity chromatography we were able to purify single-chain (sc) FSAP from r-plasma. Purified scFSAP efficiently removes nucleosomes from sn cells. We report that factor VII-activating protease may function in cellular homeostasis by catalyzing the release of nucleosomes from secondary necrotic cells.
Collapse
Affiliation(s)
- Sacha Zeerleder
- Department of Immunopathology, Sanquin Research at CLB, Plesmanlaan 125, 1066 CX Amsterdam, The Netherlands.
| | | | | | | | | | | | | |
Collapse
|
33
|
Factor VII-activating protease in patients with acute deep venous thrombosis. Thromb Res 2008; 122:848-53. [PMID: 18394684 DOI: 10.1016/j.thromres.2008.02.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2007] [Revised: 01/22/2008] [Accepted: 02/03/2008] [Indexed: 11/24/2022]
Abstract
Factor VII-activating protease (FSAP) is involved in haemostasis and inflammation. FSAP cleaves single chain urokinase-type plasminogen activator (scu-PA). The 1601GA genotype of the 1601G/A polymorphism in the FSAP gene leads to the expression of a FSAP variant with reduced ability to activate scu-PA, without affecting the ability to activate coagulation Factor VII (FVII). Previous studies have investigated the association of the 1601GA genotype with incidence and progression of carotid stenosis and deep venous thrombosis (DVT). The present study is the first to evaluate the potential association between the FSAP phenotype and DVT. We studied the association between the 1601G/A polymorphism, FSAP activity, FSAP antigen, Factor VIIa (FVIIa), prothrombin fragment 1+2 (F1+2), and C-reactive protein (CRP) in plasmas of 170 patients suspected for DVT. FSAP genotypes were equally distributed in patients with (n=64) and without DVT (n=106), (P=0.94). The 1601GA genotype was associated with significant reduction of FSAP activity (P<0.001) and FSAP antigen levels (P=0.04). Patients with DVT showed significantly higher FSAP activity (P=0.008), FSAP antigen (P=0.003), and F1+2 levels (P<0.001) than patients without DVT. The association between the FSAP measures and DVT disappeared when adjusted for CRP levels. F1+2 correlated positively to FSAP antigen (P=0.01), while FVIIa-levels were comparable in patients with and without DVT. We conclude that even though FSAP measures are significantly increased in patients with acute DVT, alterations in the scu-PA activating properties of FSAP are presumably not markedly involved in the development of acute DVT, and that the association between FSAP and DVT disappears after adjustment for CRP.
Collapse
|
34
|
Parahuleva MS, Kanse SM, Parviz B, Barth A, Tillmanns H, Bohle RM, Sedding DG, Hölschermann H. Factor Seven Activating Protease (FSAP) expression in human monocytes and accumulation in unstable coronary atherosclerotic plaques. Atherosclerosis 2008; 196:164-171. [PMID: 17482622 DOI: 10.1016/j.atherosclerosis.2007.03.042] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2006] [Revised: 03/16/2007] [Accepted: 03/22/2007] [Indexed: 11/29/2022]
Abstract
OBJECTIVE The Factor Seven Activating Protease (FSAP) is known to influence fibrinolysis and to play a critical role in the inhibition of vascular smooth muscle cell (VSMC) proliferation and migration as well as neointima formation. In order to define the role of FSAP in vascular pathophysiology we have investigated the expression of FSAP protein and mRNA in human vascular cells and coronary atherosclerotic plaques with defined clinical features. METHODS AND RESULTS Directional coronary atherectomy (DCA) specimens from 40 lesions were analyzed for FSAP antigen and mRNA expression. Higher level of FSAP mRNA (p<0.001) as well as FSAP immunostaining (p<0.005) was observed in patients with acute coronary syndromes compared to patients with stable angina pectoris. FSAP antigen was found to be focally accumulated in hypocellular and lipid-rich areas within the necrotic core of atherosclerotic plaques. FSAP was also co-localized with CD11b/CD68 expressing cells in macrophage-rich shoulder regions of the plaques. Monocyte-derived macrophages expressed FSAP in vitro and this was further induced by pro-inflammatory mediators. CONCLUSIONS FSAP accumulation in coronary atherosclerotic lesions is due to either local synthesis by monocytes/macrophages, or uptake from the plasma due to plaque hemorrhage. The higher expression of FSAP in unstable plaques suggests that it may destabilize plaque through reducing VSMC proliferation/migration and altering the hemostatic balance.
Collapse
Affiliation(s)
- Mariana S Parahuleva
- Internal Medicine I/Cardiology and Angiology, Justus-Liebig-University, Giessen, Germany
| | - Sandip M Kanse
- Institute for Biochemistry, Justus-Liebig-University, Giessen, Germany
| | - Behnoush Parviz
- Internal Medicine I/Cardiology and Angiology, Justus-Liebig-University, Giessen, Germany
| | - Andreas Barth
- Internal Medicine I/Cardiology and Angiology, Justus-Liebig-University, Giessen, Germany
| | - Harald Tillmanns
- Internal Medicine I/Cardiology and Angiology, Justus-Liebig-University, Giessen, Germany
| | - Rainer M Bohle
- Institute of General and Specialist Pathology, Homburg, Germany
| | - Daniel G Sedding
- Internal Medicine I/Cardiology and Angiology, Justus-Liebig-University, Giessen, Germany
| | - Hans Hölschermann
- Hochtaunusklinik Internal Medicine I/Cardiology and Angiology, Urseler Straße 33, 61348 Bad Homburg, Germany.
| |
Collapse
|
35
|
The hyaluronic acid-binding protease: a novel vascular and inflammatory mediator? Int Immunopharmacol 2007; 8:166-70. [PMID: 18182221 DOI: 10.1016/j.intimp.2007.10.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2007] [Revised: 10/10/2007] [Accepted: 10/11/2007] [Indexed: 10/22/2022]
Abstract
A serine protease in human plasma termed hyaluronan-binding protease HABP is structurally related to plasminogen-activators, coagulation FXII and hepathocyte growth factor activator. This protease has coagulation and fibrinolysis-related activities, although a physiologic role in haemostasis still requires confirmation. In more recent years accumulating information became available supporting the hypothesis that HABP plays also a significant role in the regulation of cells in the vasculature and in the perivascular environment. On the one hand HABP generates bradykinin or bFGF on the surface of human umbilical vein endothelial cells (HUVECs), triggering intracellular signalling via the bradykinin receptor 2 and FGFR-1. Other data indicate that beside endothelial cells also vascular smooth muscle cells are a target for HABP. As major mechanism of cell regulation a high affinity of HABP to growth factors with the subsequent proteolytic cleavage and inactivation has been identified. The current knowledge of the physiologic and clinical relevance of HABP as a vascular and possibly inflammatory mediator is summarized in this review.
Collapse
|
36
|
Muhl L, Nykjaer A, Wygrecka M, Monard D, Preissner K, Kanse S. Inhibition of PDGF-BB by Factor VII-activating protease (FSAP) is neutralized by protease nexin-1, and the FSAP-inhibitor complexes are internalized via LRP. Biochem J 2007; 404:191-6. [PMID: 17298300 PMCID: PMC1868796 DOI: 10.1042/bj20061630] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
FSAP (Factor VII-activating protease) can inhibit neointima formation and VSMC (vascular smooth-muscle cell) proliferation by cleavage of PDGF-BB (platelet-derived growth factor-BB). Negatively charged polyanions lead to autoactivation of the FSAP, but no information is available concerning the potential regulation of FSAP activity and its metabolism in the vessel wall. In the present study, we demonstrate that the enzymatic activity of FSAP can be inhibited by the serine protease inhibitor, PN-1 (protease nexin-1), that is found in the vasculature. This leads to the loss of the inhibitory effect of FSAP on PDGF-BB-mediated DNA synthesis and mitogen-activated protein kinase phosphorylation in VSMCs. The FSAP-PN-1 complexes bind to the LRP (low-density lipoprotein receptor-related protein) and are subsequently internalized. This binding is inhibited by receptor-associated protein, an antagonist of LRP, as well as heparin. While PDGFbetaR (PDGFbeta receptor) is internalized by an LRP-dependent mechanism after stimulation of cells by PDGF-BB, the FSAP-PN-1 complex neither influenced PDGF-BB-mediated phosphorylation of PDGFbetaR nor its internalization via LRP. Hence, PN-1 inhibits the enzymatic activity of FSAP and neutralizes its effect on PDGF-BB-mediated VSMC proliferation. The FSAP-inhibitor complexes are internalized via LRP without influencing the PDGF-BB signal transduction pathway.
Collapse
Affiliation(s)
- Lars Muhl
- *Institute for Biochemistry, Justus-Liebig-University, 35392 Giessen, Germany
| | - Anders Nykjaer
- †Institute of Medical Biochemistry, University of Aarhus, Aarhus, Denmark
| | - Malgorzata Wygrecka
- *Institute for Biochemistry, Justus-Liebig-University, 35392 Giessen, Germany
| | - Denis Monard
- ‡Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | - Klaus T. Preissner
- *Institute for Biochemistry, Justus-Liebig-University, 35392 Giessen, Germany
| | - Sandip M. Kanse
- *Institute for Biochemistry, Justus-Liebig-University, 35392 Giessen, Germany
- To whom correspondence should be addressed (email )
| |
Collapse
|
37
|
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: 2.1] [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
|
38
|
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: 0.9] [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
|
39
|
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: 1.4] [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
|
40
|
Sedding D, Daniel JM, Muhl L, Hersemeyer K, Brunsch H, Kemkes-Matthes B, Braun-Dullaeus RC, Tillmanns H, Weimer T, Preissner KT, Kanse SM. The G534E polymorphism of the gene encoding the factor VII-activating protease is associated with cardiovascular risk due to increased neointima formation. ACTA ACUST UNITED AC 2006; 203:2801-7. [PMID: 17145954 PMCID: PMC2118185 DOI: 10.1084/jem.20052546] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The G534E polymorphism (Marburg I [MI]) of factor VII–activating protease (FSAP) is associated with carotid stenosis and cardiovascular disease. We have previously demonstrated that FSAP is present in atherosclerotic plaques and it is a potent inhibitor of vascular smooth muscle proliferation and migration in vitro. The effect of wild-type (WT)- and MI-FSAP on neointima formation in the mouse femoral artery after wire-induced injury was investigated. Local application of WT-FSAP led to a 70% reduction in the neointima formation, and this effect was dependent on the protease activity of FSAP. MI-FSAP did not inhibit neointima formation in vivo. This is due to a reduced proteolytic activity of MI-FSAP, compared to WT-FSAP, toward platelet-derived growth factor BB, a key mediator of neointima development. The inability of MI-FSAP to inhibit vascular smooth muscle accumulation explains the observed linkage between the MI-polymorphism and increased cardiovascular risk. Hence, FSAP has a protective function in the vasculature, and analysis of MI polymorphism is likely to be clinically relevant in restenosis.
Collapse
Affiliation(s)
- Daniel Sedding
- Institute for Biochemistry, Justus-Liebig-University, 35392 Giessen, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Altincicek B, Shibamiya A, Trusheim H, Tzima E, Niepmann M, Linder D, Preissner K, Kanse S. A positively charged cluster in the epidermal growth factor-like domain of Factor VII-activating protease (FSAP) is essential for polyanion binding. Biochem J 2006; 394:687-92. [PMID: 16332249 PMCID: PMC1383718 DOI: 10.1042/bj20051563] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [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 novel plasma-derived serine protease that regulates haemostasis as well as vascular cell proliferation. FSAP undergoes autoactivation in the presence of polyanionic macromolecules such as heparin and RNA. Competition experiments suggest that RNA and heparin bind to the same or overlapping interaction sites. A proteolysis approach, where FSAP was hydrolysed into smaller fragments, was used to identify the polyanion-binding site. The EGF (epidermal growth factor)-like domains EGF2 and EGF3 of FSAP are the major interaction domains for RNA. The amino acids Arg170, Arg171, Ser172 and Lys173 within the EGF3 domain were essential for this binding. This is also the region with the highest positive net charge in the protein and is most probably located in an exposed loop. It is also highly conserved across five species. Disruption of disulphide bridges led to the loss of RNA and heparin binding, indicating that the three-dimensional structure of the EGF3 domain is essential for binding to negatively charged heparin or RNA. The identification of polyanion-binding sites will help to define the role of FSAP in the vasculature.
Collapse
Affiliation(s)
- Boran Altincicek
- Biochemisches Institut, Justus-Liebig-Universität Giessen, Friedrichstrasse 24, D-35392 Giessen, Germany
| | - Aya Shibamiya
- Biochemisches Institut, Justus-Liebig-Universität Giessen, Friedrichstrasse 24, D-35392 Giessen, Germany
| | - Heidi Trusheim
- Biochemisches Institut, Justus-Liebig-Universität Giessen, Friedrichstrasse 24, D-35392 Giessen, Germany
| | - Eleni Tzima
- Biochemisches Institut, Justus-Liebig-Universität Giessen, Friedrichstrasse 24, D-35392 Giessen, Germany
| | - Michael Niepmann
- Biochemisches Institut, Justus-Liebig-Universität Giessen, Friedrichstrasse 24, D-35392 Giessen, Germany
| | - Dietmar Linder
- Biochemisches Institut, Justus-Liebig-Universität Giessen, Friedrichstrasse 24, D-35392 Giessen, Germany
| | - Klaus T. Preissner
- Biochemisches Institut, Justus-Liebig-Universität Giessen, Friedrichstrasse 24, D-35392 Giessen, Germany
| | - Sandip M. Kanse
- Biochemisches Institut, Justus-Liebig-Universität Giessen, Friedrichstrasse 24, D-35392 Giessen, Germany
- To whom correspondence should be addressed (email )
| |
Collapse
|
42
|
Kress JA, Seitz R, Dodt J, Etscheid M. Induction of intracellular signalling in human endothelial cells by the hyaluronan-binding protease involves two distinct pathways. Biol Chem 2006; 387:1275-83. [PMID: 16972797 DOI: 10.1515/bc.2006.158] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Recently a novel plasma serine protease with high affinity to hyaluronic acid and glycosaminoglycans, such as heparin and heparan sulfate, has been described and termed hyaluronan-binding protease (HABP). HABP cleaves kininogen in vitro, releasing the vasoactive peptide bradykinin, and activates plasminogen activators, suggesting a vascular cell-directed physiological function of this novel plasma protease. Here we show that HABP stimulates human umbilical vein endothelial cells (HUVECs) by activating two distinct cell-surface receptors. On the one hand, HABP releases bradykinin from cell surface-bound or soluble kininogen and triggers a bradykinin B2-receptor-dependent mobilisation of intracellular Ca2+. On the other hand, HABP activates the p44/42-dependent MAPK (ERK1/2) signalling cascade independent of the B2-receptor, but involving the fibroblast growth factor receptor-1 and basic fibroblast growth factor. This signalling pathway leads to phosphorylation of the kinases Raf, MEK1/2 and ERK1/2. The extracellular activity of HABP also affects the gene expression level through phosphorylation of two transcription factors, the cAMP-responsive element binding protein CREB and the proto-oncogene c-Myc. Our results indicate a proangiogenic potential of HABP, which, in combination with a profibrinolytic activity, directs the physiological function of this plasma protease to processes in which clot lysis, cell motility and neovascularisation are pivotal processes, e.g., in wound healing, tissue repair and tumour progression.
Collapse
Affiliation(s)
- Julia Anne Kress
- Department of Haematology and Transfusion Medicine, Paul-Ehrlich-Institute, Federal Agency for Sera and Vaccines, D-63225 Langen, Germany
| | | | | | | |
Collapse
|
43
|
Etscheid M, Beer N, Dodt J. The hyaluronan-binding protease upregulates ERK1/2 and PI3K/Akt signalling pathways in fibroblasts and stimulates cell proliferation and migration. Cell Signal 2005; 17:1486-94. [PMID: 16153533 DOI: 10.1016/j.cellsig.2005.03.007] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2005] [Accepted: 03/04/2005] [Indexed: 11/21/2022]
Abstract
The hyaluronan-binding protease (HABP) is a serine protease in human plasma which is structurally related to plasminogen activators, coagulation factor XII and hepathocyte growth factor activator. It can in vitro activate the coagulation factor FVII, kininogen and plasminogen activators. The present study was initiated to gain a more complete picture of the cell-associated activities of this fibrinolysis-related protease. Treatment of lung fibroblasts with HABP lead to a rapid activation of signalling pathways, including the mitogen-activated protein kinase (MAPK) pathway with c-Raf, MEK and ERK1/2. Additionally the activation of the PI3K/Akt pathway and of several translation-related proteins was found. Proliferation assays confirmed the assumption of a strong growth-stimulating effect of HABP on human lung and skin fibroblasts. Intracellular signalling and growth stimulation were strongly dependent on the proteolytic activity of HABP. Stimulation of signalling and proliferation by HABP involved the fibroblast growth factor receptor 1 (FGFR-1). HABP-stimulated proliferation of lung fibroblasts MRC-5 was accompanied by a significant intracellular increase in basic fibroblast growth factor (bFGF), the major ligand of FGFR-1; bFGF could however not be identified in the supernatant of HABP-treated cells. Though, the conditioned medium from HABP-treated cells showed a strong growth-promoting activity on quiescent fibroblasts, indicating the release of a yet unknown growth factor amplifying the initial growth stimulus. In a two-dimensional wound model HABP stimulated the invasion of fibroblasts into a scratch area, adding a strong pro-migratory activity to this plasma protease. In summary, HABP exhibits a significant growth factor-like activity on quiescent human lung and dermal fibroblasts. Our findings suggest that this fibrinolysis-related plasma protease may participate in physiologic or pathologic processes where cell proliferation and migration are pivotal, like tissue repair, vascular remodelling, wound healing or tumor development.
Collapse
Affiliation(s)
- M Etscheid
- Department of Hematology/Transfusion Medicine, Paul Ehrlich Institute, Federal Agency for Sera and Vaccines, Paul-Ehrlich-Strasse 51-59, 63225 Langen, Germany.
| | | | | |
Collapse
|
44
|
Kanse SM, Liang O, Schubert U, Haas H, Preissner KT, Doenhoff MJ, Dennis RD. Characterisation and partial purification of Schistosoma mansoni egg-derived pro-angiogenic factor. Mol Biochem Parasitol 2005; 144:76-85. [PMID: 16169609 DOI: 10.1016/j.molbiopara.2005.08.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2005] [Revised: 07/29/2005] [Accepted: 08/03/2005] [Indexed: 12/11/2022]
Abstract
Pathogenesis of Schistosoma mansoni infection is due to the accumulation of eggs in the liver and intestine of the host followed by granuloma formation and fibrosis at the site of egg embolism. Nevertheless, total hepatic blood flow and hepatic function appear to be maintained by neovascularisation of the periportal fibrotic tissue. Here we demonstrate that intact live eggs, excretory/secretory products of eggs and the extracts of homogenised eggs stimulate the proliferation and migration of endothelial cells. Formation of endothelial capillary-like outgrowths, as well as, the activation of p42/p44 mitogen-activated protein kinase signal transduction pathway was stimulated by egg extracts, whereas other stages of the schistosome life-cycle did not exhibit such activity. We have characterised and partially purified the pro-angiogenic factor. The active pro-angiogenic component was fast-acting, heat-stable, protease-resistant, weakly heparin-binding and a non-lipid. It could be extracted with acidified ethanol or a hot acetic acid solution and could be partially purified by reverse-phase HPLC. The S. mansoni egg-derived pro-angiogenic factor, described here, may directly contribute to vascular remodelling in the liver and illustrates how helminth parasites may modulate angiogenesis.
Collapse
Affiliation(s)
- Sandip M Kanse
- Institut für Biochemie, Justus-Liebig-Universität, Friedrichstrasse 24, D-35392 Giessen, Germany
| | | | | | | | | | | | | |
Collapse
|