1
|
Staubach P, Tachdjian R, Li HH, Hakl R, Aygören-Pürsün E, Wieman L, Lawo JP, Craig TJ. Timing of Onset of Garadacimab for Preventing Hereditary Angioedema Attacks. Clin Exp Allergy 2024. [PMID: 39353415 DOI: 10.1111/cea.14568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 08/13/2024] [Accepted: 08/22/2024] [Indexed: 10/04/2024]
Affiliation(s)
- Petra Staubach
- Department of Dermatology and Allergy, University Medical Center Mainz, Mainz, Germany
| | - Raffi Tachdjian
- Division of Allergy and Clinical Immunology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, USA
| | - H Henry Li
- Institute for Asthma and Allergy, Chevy Chase, Maryland, USA
| | - Roman Hakl
- Department of Clinical Immunology and Allergology, St. Anne's University Hospital and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Emel Aygören-Pürsün
- Department for Children and Adolescents, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt, Germany
| | | | | | - Timothy J Craig
- Allergy and Immunology, Tenured Professor of Medicine, Pediatrics and Biomedical Sciences, Hershey, Pennsylvania, USA
- Vinmec International Hospital, Times City, Hanoi, Vietnam
| |
Collapse
|
2
|
Cohn DM, Renné T. Targeting factor XIIa for therapeutic interference with hereditary angioedema. J Intern Med 2024; 296:311-326. [PMID: 39331688 DOI: 10.1111/joim.20008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/29/2024]
Abstract
Hereditary angioedema (HAE) is a rare, potentially life-threatening genetic disorder characterized by recurrent attacks of swelling. Local vasodilation and vascular leakage are stimulated by the vasoactive peptide bradykinin, which is excessively produced due to dysregulation of the activated factor XII (FXIIa)-driven kallikrein-kinin system. There is a need for novel treatments for HAE that provide greater efficacy, improved quality of life, minimal adverse effects, and reduced treatment burden over current first-line therapies. FXIIa is emerging as an attractive therapeutic target for interference with HAE attacks. In this review, we draw on preclinical, experimental animal, and in vitro studies, providing an overview on targeting FXIIa as the basis for pharmacologic interference in HAE. We highlight that there is a range of FXIIa inhibitors in development for different therapeutic areas. Of these, garadacimab, an FXIIa-targeted inhibitory monoclonal antibody, is the most advanced and has shown potential as a novel long-term prophylactic treatment for patients with HAE in clinical trials. The evidence from these trials is summarized and discussed, and we propose areas for future research where targeting FXIIa may have therapeutic potential beyond HAE.
Collapse
Affiliation(s)
- Danny M Cohn
- University of Amsterdam, Amsterdam Cardiovascular Sciences, Amsterdam UMC, Amsterdam, The Netherlands
| | - Thomas Renné
- University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
- Center for Thrombosis and Hemostasis (CTH), Johannes Gutenberg University Medical Center, Mainz, Germany
| |
Collapse
|
3
|
Schoeppe R, Waldmann M, Jessen HJ, Renné T. An Update on Polyphosphate In Vivo Activities. Biomolecules 2024; 14:937. [PMID: 39199325 PMCID: PMC11352482 DOI: 10.3390/biom14080937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 07/13/2024] [Accepted: 07/16/2024] [Indexed: 09/01/2024] Open
Abstract
Polyphosphate (polyP) is an evolutionary ancient inorganic molecule widespread in biology, exerting a broad range of biological activities. The intracellular polymer serves as an energy storage pool and phosphate/calcium ion reservoir with implications for basal cellular functions. Metabolisms of the polymer are well understood in procaryotes and unicellular eukaryotic cells. However, functions, regulation, and association with disease states of the polymer in higher eukaryotic species such as mammalians are just beginning to emerge. The review summarises our current understanding of polyP metabolism, the polymer's functions, and methods for polyP analysis. In-depth knowledge of the pathways that control polyP turnover will open future perspectives for selective targeting of the polymer.
Collapse
Affiliation(s)
- Robert Schoeppe
- Institute of Clinical Chemistry and Laboratory Medicine (O26), University Medical Center Hamburg-Eppendorf, D-20246 Hamburg, Germany
| | - Moritz Waldmann
- Institute of Clinical Chemistry and Laboratory Medicine (O26), University Medical Center Hamburg-Eppendorf, D-20246 Hamburg, Germany
| | - Henning J. Jessen
- Institute of Organic Chemistry, Albert-Ludwigs-University of Freiburg, D-79105 Freiburg, Germany;
| | - Thomas Renné
- Institute of Clinical Chemistry and Laboratory Medicine (O26), University Medical Center Hamburg-Eppendorf, D-20246 Hamburg, Germany
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, D02 YN77 Dublin, Ireland
- Center for Thrombosis and Haemostasis (CTH), Johannes Gutenberg University Medical Center, D-55131 Mainz, Germany
| |
Collapse
|
4
|
Coelho SVA, Augusto FM, de Arruda LB. Potential Pathways and Pathophysiological Implications of Viral Infection-Driven Activation of Kallikrein-Kinin System (KKS). Viruses 2024; 16:245. [PMID: 38400022 PMCID: PMC10892958 DOI: 10.3390/v16020245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Revised: 01/30/2024] [Accepted: 01/31/2024] [Indexed: 02/25/2024] Open
Abstract
Microcirculatory and coagulation disturbances commonly occur as pathological manifestations of systemic viral infections. Research exploring the role of the kallikrein-kinin system (KKS) in flavivirus infections has recently linked microvascular dysfunctions to bradykinin (BK)-induced signaling of B2R, a G protein-coupled receptor (GPCR) constitutively expressed by endothelial cells. The relevance of KKS activation as an innate response to viral infections has gained increasing attention, particularly after the reports regarding thrombogenic events during COVID-19. BK receptor (B2R and B1R) signal transduction results in vascular permeability, edema formation, angiogenesis, and pain. Recent findings unveiling the role of KKS in viral pathogenesis include evidence of increased activation of KKS with elevated levels of BK and its metabolites in both intravascular and tissue milieu, as well as reports demonstrating that virus replication stimulates BKR expression. In this review, we will discuss the mechanisms triggered by virus replication and by virus-induced inflammatory responses that may stimulate KKS. We also explore how KKS activation and BK signaling may impact virus pathogenesis and further discuss the potential therapeutic application of BKR antagonists in the treatment of hemorrhagic and respiratory diseases.
Collapse
Affiliation(s)
- Sharton Vinícius Antunes Coelho
- Departamento de Virologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil;
| | | | - Luciana Barros de Arruda
- Departamento de Virologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil;
| |
Collapse
|
5
|
Ren S, Cai D, Xiao L, Shen H, Ren C. Whole-exome sequencing reveals a novel frameshift mutation in a consanguineous family with a hereditary coagulation factor XII deficiency. Clin Biochem 2023:110602. [PMID: 37391120 DOI: 10.1016/j.clinbiochem.2023.110602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 06/15/2023] [Accepted: 06/17/2023] [Indexed: 07/02/2023]
Abstract
BACKGROUND AND AIMS We aimed to elucidate a hereditary mutation of coagulation factor XII (FXII) in a consanguineous Chinese family. METHODS Mutations were investigated using Sanger and whole-exome sequencing. FXII (FXII:C) activity and FXII antigen (FXII:Ag) were assessed using clotting assays and ELISA, respectively. Gene variants were annotated and the likelihood that amino acid mutations would affect protein function was predicted using bioinformatics. RESULTS Activated partial thromboplastin time was prolonged to >170 s (reference range, 22.3-32.5 s), and FXII:C and FXII:Ag were decreased to 0.3% and 1%, respectively, (normal range for both, 72%-150%) in the proband. Sequencing revealed a homozygous frameshift mutation c.150delC (p.Phe51Serfs*44) site in the F12 gene exon 3. This mutation results in premature termination of the encoded protein translation and the protein is truncated. Bioinformatic findings indicated a novel pathogenic frameshift mutation. CONCLUSION The c.150delC frameshift mutation p.Phe51Serfs*44 in the F12 gene likely explains the low FXII level and the molecular pathogenesis of an inherited FXII deficiency in a consanguineous family.
Collapse
Affiliation(s)
- Shuting Ren
- Department of Clinical Laboratory, The 904th Hospital of Joint Logistic Support Force of PLA, 101#, Xingyuan North Road, LiangXi District, Wuxi, Jiangsu 214044, PR China
| | - Dongping Cai
- Department of Clinical Laboratory, The 904th Hospital of Joint Logistic Support Force of PLA, 101#, Xingyuan North Road, LiangXi District, Wuxi, Jiangsu 214044, PR China
| | - Li Xiao
- Department of Clinical Laboratory, The 904th Hospital of Joint Logistic Support Force of PLA, 101#, Xingyuan North Road, LiangXi District, Wuxi, Jiangsu 214044, PR China
| | - Hongshi Shen
- Department of Hematology, The 904th Hospital of Joint Logistic Support Force of PLA, 101#, Xingyuan North Road, LiangXi District, Wuxi, Jiangsu 214044, PR China.
| | - Chuanlu Ren
- Department of Clinical Laboratory, The 904th Hospital of Joint Logistic Support Force of PLA, 101#, Xingyuan North Road, LiangXi District, Wuxi, Jiangsu 214044, PR China.
| |
Collapse
|
6
|
Umei N, Shin S, Lai A, Miller J, Roberts K, Strelkova D, Chaudhary N, Ichiba S, Sakamoto A, Whitehead K, Cook K. Factor XII Silencing Using siRNA Prevents Thrombus Formation in a Rat Model of Extracorporeal Life Support. ASAIO J 2023; 69:527-532. [PMID: 36728837 DOI: 10.1097/mat.0000000000001876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Heparin anticoagulation increases the bleeding risk during extracorporeal life support (ECLS). This study determined whether factor XII (FXII) silencing using short interfering RNA (siRNA) can provide ECLS circuit anticoagulation without bleeding. Adult male, Sprague-Dawley rats were randomized to four groups (n = 3 each) based on anticoagulant: (1) no anticoagulant, (2) heparin, (3) FXII siRNA, or (4) nontargeting siRNA. Heparin was administered intravenously before and during ECLS. FXII or nontargeting siRNA were administered intravenously 3 days before the initiation of ECLS via lipidoid nanoparticles. The rats were placed on pumped, arteriovenous ECLS for 8 hours or until the blood flow resistance reached three times its baseline resistance. Without anticoagulant, mock-oxygenator resistance tripled within 7 ± 2 minutes. The resistance in the FXII siRNA group did not increase for 8 hours. There were no significant differences in resistance or mock-oxygenator thrombus volume between the FXII siRNA and the heparin groups. However, the bleeding time in the FXII siRNA group (3.4 ± 0.6 minutes) was significantly shorter than that in the heparin group (5.5 ± 0.5 minutes, p < 0.05). FXII silencing using siRNA provided simpler anticoagulation of ECLS circuits with reduced bleeding time as compared to heparin. http://links.lww.com/ASAIO/A937.
Collapse
Affiliation(s)
- Nao Umei
- From the Departments of Anesthesiology
- Surgical Intensive Care Medicine, Nippon Medical School Hospital, Tokyo, Japan
- Departments of Biomedical Engineering
| | - Suji Shin
- Departments of Biomedical Engineering
| | | | | | | | - Daria Strelkova
- Chemical Engineering, Carnegie Mellon University, Pittsburgh, Pennsylvania
| | - Namit Chaudhary
- Chemical Engineering, Carnegie Mellon University, Pittsburgh, Pennsylvania
| | - Shingo Ichiba
- From the Departments of Anesthesiology
- Surgical Intensive Care Medicine, Nippon Medical School Hospital, Tokyo, Japan
| | - Atsuhiro Sakamoto
- From the Departments of Anesthesiology
- Surgical Intensive Care Medicine, Nippon Medical School Hospital, Tokyo, Japan
| | - Kathryn Whitehead
- Departments of Biomedical Engineering
- Chemical Engineering, Carnegie Mellon University, Pittsburgh, Pennsylvania
| | | |
Collapse
|
7
|
Chou SC, Lin CY, Lin HY, Pai CH, Yu CY, Kuo SF, Lin JS, Lin PT, Hung MH, Hsieh HN, Liu HC, Shen MC. Characterization of congenital factor XII deficiency in Taiwanese patients: identification of one novel and one common mutation. Int J Hematol 2022; 116:528-533. [PMID: 35675023 PMCID: PMC9174919 DOI: 10.1007/s12185-022-03390-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 05/13/2022] [Accepted: 05/16/2022] [Indexed: 11/29/2022]
Abstract
Background Factor XII (FXII) deficiency is an interesting condition that causes prolonged activated partial thromboplastin time without bleeding diathesis. FXII may be not important in hemostasis, but still plays roles in thrombosis and inflammation. In order to raise clinical awareness about this condition, we studied patients with severe FXII deficiency and their relatives. Methods Consecutive severely FXII deficient patients presenting from 1995 to 2020 were recruited from two medical centers in Taiwan. Index patients and their families were tested for FXII function, antigen and F12 gene. F12 variants were constructed into the pIRES-hrGFP vector and expressed on human embryonic kidney cells (HEK293T). FXII antigen and activity were analyzed. Results We found five severely FXII deficient patients, three women and two men, aged 44–71 years. FXII antigen results ranged from undetectable to 43.7%. Three different mutations were identified: c.1681C>A (p.Gly542Ser), c.1561G>A (p.Glu502Lys), and a novel mutation c.1556T>A (p.Leu500Gln). HEK293T cells expressed consistently low FXII activity with all mutations. FXII antigen expression was similar to the wild type in c.1681C>A (p.Gly542Ser), but reduced in c.1556T>A (p.Leu500Gln) and c.1561G>A (p.Glu502Lys). Conclusions We report five unrelated patients with severe FXII deficiency, one of whom carried a novel, cross-reacting material negative mutation c.1556T>A (p.Leu500Gln).
Collapse
Affiliation(s)
- Sheng-Chieh Chou
- Division of Hematology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Ching-Yeh Lin
- Department of Internal Medicine, Changhua Christian Hospital, 135 Nan-Hsiao Street, Changhua, 500, Taiwan
| | - Hsuan-Yu Lin
- Department of Internal Medicine, Changhua Christian Hospital, 135 Nan-Hsiao Street, Changhua, 500, Taiwan
| | - Chen-Hsueh Pai
- Department of Clinical Laboratory Sciences and Medical Biotechnology, National Taiwan University, Taipei, Taiwan
| | - Cheng-Ye Yu
- Department of Clinical Laboratory Sciences and Medical Biotechnology, National Taiwan University, Taipei, Taiwan
| | - Su-Feng Kuo
- Department of Laboratory Medicine, Changhua Christian Hospital, Changhua, Taiwan
| | - Jen-Shiou Lin
- Department of Laboratory Medicine, Changhua Christian Hospital, Changhua, Taiwan
| | - Po-Te Lin
- Department of Internal Medicine, Changhua Christian Hospital, 135 Nan-Hsiao Street, Changhua, 500, Taiwan
| | - Mei-Hua Hung
- Division of Hematology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Han-Ni Hsieh
- Department of Internal Medicine, Changhua Christian Hospital, 135 Nan-Hsiao Street, Changhua, 500, Taiwan
| | - Hsiang-Chun Liu
- Department of Medical Research, E-DA Hospital/I-Shou University, Kaohsiung, Taiwan
| | - Ming-Ching Shen
- Division of Hematology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan. .,Department of Internal Medicine, Changhua Christian Hospital, 135 Nan-Hsiao Street, Changhua, 500, Taiwan.
| |
Collapse
|
8
|
Nopp S, Kraemmer D, Ay C. Factor XI Inhibitors for Prevention and Treatment of Venous Thromboembolism: A Review on the Rationale and Update on Current Evidence. Front Cardiovasc Med 2022; 9:903029. [PMID: 35647061 PMCID: PMC9133368 DOI: 10.3389/fcvm.2022.903029] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 04/12/2022] [Indexed: 12/12/2022] Open
Abstract
Although anticoagulation therapy has evolved from non-specific drugs (i.e., heparins and vitamin K antagonists) to agents that directly target specific coagulation factors (i.e., direct oral anticoagulants, argatroban, fondaparinux), thrombosis remains a leading cause of death worldwide. Direct oral anticoagulants (i.e., factor IIa- and factor Xa-inhibitors) now dominate clinical practice because of their favorable pharmacological profile and ease of use, particularly in venous thromboembolism (VTE) treatment and stroke prevention in atrial fibrillation. However, despite having a better safety profile than vitamin K antagonists, their bleeding risk is not insignificant. This is true for all currently available anticoagulants, and a high bleeding risk is considered a contraindication to anticoagulation. As a result, ongoing research focuses on developing future anticoagulants with an improved safety profile. Several promising approaches to reduce the bleeding risk involve targeting the intrinsic (or contact activation) pathway of coagulation, with the ultimate goal of preventing thrombosis without impairing hemostasis. Based on epidemiological data on hereditary factor deficiencies and preclinical studies factor XI (FXI) emerged as the most promising candidate target. In this review, we highlight unmet clinical needs of anticoagulation therapy, outlay the rationale and evidence for inhibiting FXI, discuss FXI inhibitors in current clinical trials, conduct an exploratory meta-analysis on their efficacy and safety, and provide an outlook on the potential clinical application of these novel anticoagulants.
Collapse
Affiliation(s)
| | | | - Cihan Ay
- Clinical Division of Hematology and Hemostaseology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
9
|
Kılınç E, Can Timucin A, Selim Cinaroglu S, Timucin E. Modeling and dynamical analysis of the full-length structure of factor XII with zinc. J Mol Model 2022; 28:129. [PMID: 35469101 DOI: 10.1007/s00894-022-05113-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 04/05/2022] [Indexed: 11/24/2022]
Abstract
Zinc (II), the second most abundant transition metal in blood, binds to the initiator of the contact pathway, factor XII (FXII). This binding induces conformational changes in the structure of FXII eventually leading to its activation. Despite many in vitro and in vivo studies on zinc-mediated activation of FXII, its molecular mechanism remains elusive mainly due to absence of a full-length structural model of FXII. To this end, this study investigated the role of zinc in the structure and dynamics of the full-length structure FXII that was obtained through molecular modeling. We have used four structural templates covering more than 70% of the FXII sequence and the remaining interconnecting regions were built by loop modeling. The resulting full-length structure of FXII contained disordered regions, but in comparison to the AlphaFold (AF) prediction, our full-length model represented a more realistic structure because of the disordered regions which were modeled to yield a more compact full-length structure in our model than the AF structure. Other than the disordered regions, our model and AF prediction were highly similar. The resulting full-length FXII structure was used to generate different systems representing the zinc-bound form (holo). Further to assess the contribution of the disulfide bridges, we also analyzed the apo and holo FXII structures with oxidized or reduced cysteine side-chains. Simulations suggested zinc binding conferred rigidity to the structure, particularly to the light chain of FXII. Zinc binding alone was sufficient to limit the backbone flexibility while 15 disulfide bonds, which were scattered throughout the structure, made a less significant contribution to the backbone rigidity. Altogether our results provide insights into the first realistic full-length structure of FXII focusing on the impact of structural zinc and disulfide bridges in the dynamics of this structure.
Collapse
Affiliation(s)
- Evren Kılınç
- Department of Biophysics, School of Medicine, Acibadem Mehmet Ali Aydinlar University, Istanbul, 34752, Turkey
| | - Ahmet Can Timucin
- Department of Molecular Biology and Genetics, Faculty of Arts and Sciences, Acibadem Mehmet Ali Aydinlar University, Istanbul, 34752, Turkey
| | | | - Emel Timucin
- Department of Biostatistics and Medical Informatics, School of Medicine, Acibadem Mehmet Ali Aydinlar University, Istanbul, 34752, Turkey.
| |
Collapse
|
10
|
Zhang Z, Shen C, Fang M, Han Y, Long C, Liu W, Yang M, Liu M, Zhang D, Cao Q, Chen X, Fang Y, Lu Q, Hou Z, Li Y, Liu Z, Lei X, Ni H, Lai R. Novel contact-kinin inhibitor sylvestin targets thromboinflammation and ameliorates ischemic stroke. Cell Mol Life Sci 2022; 79:240. [PMID: 35416530 PMCID: PMC11071929 DOI: 10.1007/s00018-022-04257-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Revised: 03/15/2022] [Accepted: 03/17/2022] [Indexed: 11/26/2022]
Abstract
Ischemic stroke is a leading cause of death and disability worldwide. Increasing evidence indicates that ischemic stroke is a thromboinflammatory disease in which the contact-kinin pathway has a central role by activating pro-coagulant and pro-inflammatory processes. The blocking of distinct members of the contact-kinin pathway is a promising strategy to control ischemic stroke. Here, a plasma kallikrein and active FXII (FXIIa) inhibitor (sylvestin, contained 43 amino acids, with a molecular weight of 4790.4 Da) was first identified from forest leeches (Haemadipsa sylvestris). Testing revealed that sylvestin prolonged activated partial thromboplastin time without affecting prothrombin time. Thromboelastography and clot retraction assays further showed that it extended clotting time in whole blood and inhibited clot retraction in platelet-rich plasma. In addition, sylvestin prevented thrombosis in vivo in FeCl3-induced arterial and carrageenan-induced tail thrombosis models. The potential role of sylvestin in ischemic stroke was evaluated by transient and permanent middle cerebral artery occlusion models. Sylvestin administration profoundly protected mice from ischemic stroke by counteracting intracerebral thrombosis and inflammation. Importantly, sylvestin showed no signs of bleeding tendency. The present study identifies sylvestin is a promising contact-kinin pathway inhibitor that can proffer profound protection from ischemic stroke without increased risk of bleeding.
Collapse
Affiliation(s)
- Zhiye Zhang
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Kunming, Yunnan, 650107, China
| | - Chuanbin Shen
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, M5S 1A1, Canada
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, Senior Scientist of Canadian Blood Services Centre for Innovation, Platform Director for Hematology, Cancer and Immunological Diseases, St. Michael's Hospital, Room 421, LKSKI - Keenan Research Centre, Li Ka Shing Knowledge Institute, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, 209 Victoria Street, Toronto, ON, M5B 1W8, Canada
| | - Mingqian Fang
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Kunming, Yunnan, 650107, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, 650204, China
| | - Yajun Han
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Kunming, Yunnan, 650107, China
| | - Chengbo Long
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Kunming, Yunnan, 650107, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, 650204, China
| | - Weihui Liu
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Kunming, Yunnan, 650107, China
| | - Min Yang
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Kunming, Yunnan, 650107, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, 650204, China
| | - Ming Liu
- Department of Molecular and Cell Biology, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, 230027, China
| | - Dengdeng Zhang
- Department of Pharmaceutical Sciences, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Qiqi Cao
- Department of Zoology, College of Life Sciences, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, China
| | - Xue Chen
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Kunming, Yunnan, 650107, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, 650204, China
| | - Yaqun Fang
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Kunming, Yunnan, 650107, China
| | - Qiumin Lu
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Kunming, Yunnan, 650107, China
| | - Zongliu Hou
- Central Laboratory of Yan'an Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650000, China
| | - Yaxiong Li
- Department of Cardiovascular Surgery, Yan'an Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650000, China
| | - Zhenze Liu
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, Senior Scientist of Canadian Blood Services Centre for Innovation, Platform Director for Hematology, Cancer and Immunological Diseases, St. Michael's Hospital, Room 421, LKSKI - Keenan Research Centre, Li Ka Shing Knowledge Institute, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, 209 Victoria Street, Toronto, ON, M5B 1W8, Canada
| | - Xi Lei
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, Senior Scientist of Canadian Blood Services Centre for Innovation, Platform Director for Hematology, Cancer and Immunological Diseases, St. Michael's Hospital, Room 421, LKSKI - Keenan Research Centre, Li Ka Shing Knowledge Institute, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, 209 Victoria Street, Toronto, ON, M5B 1W8, Canada
| | - Heyu Ni
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, M5S 1A1, Canada.
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, Senior Scientist of Canadian Blood Services Centre for Innovation, Platform Director for Hematology, Cancer and Immunological Diseases, St. Michael's Hospital, Room 421, LKSKI - Keenan Research Centre, Li Ka Shing Knowledge Institute, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, 209 Victoria Street, Toronto, ON, M5B 1W8, Canada.
- Canadian Blood Services Centre for Innovation, Toronto, ON, M5G 2M1, Canada.
- Department of Physiology, University of Toronto, Toronto, ON, M5S 1A1, Canada.
- Department of Medicine, University of Toronto, Toronto, ON, M5S 1A1, Canada.
| | - Ren Lai
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Kunming, Yunnan, 650107, China.
- Sino-African Joint Research Center, Chinese Academy of Science, Wuhan, 430074, Hubei, China.
- Institutes for Drug Discovery and Development, Chinese Academy of Sciences, Shanghai, 201203, China.
- Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650223, China.
| |
Collapse
|
11
|
Truong TK, Malik RA, Yao X, Fredenburgh JC, Stafford AR, Madarati HM, Kretz CA, Weitz JI. Identification of the histidine-rich glycoprotein domains responsible for contact pathway inhibition. J Thromb Haemost 2022; 20:821-832. [PMID: 34967109 DOI: 10.1111/jth.15631] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 12/27/2021] [Indexed: 11/28/2022]
Abstract
BACKGROUND Previously, we showed that histidine-rich glycoprotein (HRG) binds factor (F) XIIa with high affinity, inhibits FXII autoactivation and FXIIa-mediated activation of FXI, and attenuates ferric chloride-induced arterial thrombosis in mice. Therefore, HRG downregulates the contact pathway in vitro and in vivo. OBJECTIVE To identify the domains on HRG responsible for contact pathway inhibition. METHODS Recombinant HRG domain constructs (N-terminal [N1, N2, and N1N2], proline-rich regions, histidine-rich region [HRR], and C-terminal) were expressed and purified. The affinities of plasma-derived HRG, HRG domain constructs, and synthetic HRR peptides for FXII, FXIIa, β-FXIIa, and polyphosphate (polyP) were determined using surface plasmon resonance, and their effects on polyP-induced FXII autoactivation, FXIIa-mediated activation of FXI and prekallikrein, the activated partial thromboplastin time (APTT), and thrombin generation were examined. RESULTS HRG and HRG domain constructs bind FXIIa, but not FXII or β-FXII. HRR, N1, and N1N2 bind FXIIa with affinities comparable with that of HRG, whereas the remaining domains bind with lower affinity. Synthetic HRR peptides bind FXIIa and polyP with high affinity. HRG and HRR significantly inhibit FXII autoactivation and prolong the APTT. Like HRG, synthetic HRR peptides inhibit FXII autoactivation, attenuate FXIIa-mediated activation of prekallikrein and FXI, prolong the APTT, and attenuate thrombin generation. CONCLUSION The interaction of HRG with FXIIa and polyP is predominantly mediated by the HRR domain. Like intact HRG, HRR downregulates the contact pathway and contributes to HRG-mediated down regulation of coagulation.
Collapse
Affiliation(s)
- Tammy K Truong
- Thrombosis and Atherosclerosis Research Institute, Hamilton, Ontario, Canada
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
- Department of Medical Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Rida A Malik
- Thrombosis and Atherosclerosis Research Institute, Hamilton, Ontario, Canada
- Department of Medical Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Xintong Yao
- Thrombosis and Atherosclerosis Research Institute, Hamilton, Ontario, Canada
- Department of Medical Sciences, McMaster University, Hamilton, Ontario, Canada
| | - James C Fredenburgh
- Thrombosis and Atherosclerosis Research Institute, Hamilton, Ontario, Canada
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Alan R Stafford
- Thrombosis and Atherosclerosis Research Institute, Hamilton, Ontario, Canada
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Hasam M Madarati
- Thrombosis and Atherosclerosis Research Institute, Hamilton, Ontario, Canada
- Department of Medical Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Colin A Kretz
- Thrombosis and Atherosclerosis Research Institute, Hamilton, Ontario, Canada
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Jeffrey I Weitz
- Thrombosis and Atherosclerosis Research Institute, Hamilton, Ontario, Canada
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
- Department of Medical Sciences, McMaster University, Hamilton, Ontario, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
12
|
Abstract
Blood coagulation is essential to maintain the integrity of a closed circulatory system (hemostasis), but also contributes to thromboembolic occlusion of vessels (thrombosis). Thrombosis may cause deep vein thrombosis, pulmonary embolism, myocardial infarction, peripheral artery disease, and ischemic stroke, collectively the most common causes of death and disability in the developed world. Treatment for the prevention of thromboembolic diseases using anticoagulants such as heparin, coumarins, thrombin inhibitors, or antiplatelet drugs increase the risk of bleeding and are associated with an increase in potentially life-threatening hemorrhage, partially offsetting the benefits of reduced coagulation. Thus, drug development aiming at novel targets is needed to provide efficient and safe anticoagulation. Within the last decade, experimental and preclinical data have shown that some coagulation mechanisms principally differ in thrombosis and hemostasis. The plasma contact system protein factors XII and XI, high-molecular-weight kininogen, and plasma kallikrein specifically contribute to thrombosis, however, have minor, if any, role in hemostatic coagulation mechanisms. Inherited deficiency in contact system proteins is not associated with increased bleeding in humans and animal models. Therefore, targeting contact system proteins provides the exciting opportunity to interfere specifically with thromboembolic diseases without increasing the bleeding risk. Recent studies that investigated pharmacologic inhibition of contact system proteins have shown that this approach provides efficient and safe thrombo-protection that in contrast to classical anticoagulants is not associated with increased bleeding risk. This review summarizes therapeutic and conceptual developments for selective interference with pathological thrombus formation, while sparing physiologic hemostasis, that enables safe anticoagulation treatment.
Collapse
Affiliation(s)
- Reiner K Mailer
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Piotr Kuta
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Thomas Renné
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany.,Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland.,Center for Thrombosis and Hemostasis (CTH), Johannes Gutenberg University Medical Center, Mainz, Germany
| |
Collapse
|
13
|
Pawaskar D, Chen X, Glassman F, May F, Roberts A, Biondo M, McKenzie A, Nolte MW, Jusko WJ, Tortorici M. Pharmacokinetic/pharmacodynamic modeling for dose selection for the first-in-human trial of the activated Factor XII inhibitor garadacimab (CSL312). Clin Transl Sci 2021; 15:709-720. [PMID: 34811931 PMCID: PMC8932701 DOI: 10.1111/cts.13192] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 09/17/2021] [Accepted: 10/11/2021] [Indexed: 12/19/2022] Open
Abstract
Factor XII (FXII) is a serine protease involved in multiple cascades, including the kallikrein–kinin system. It may play a role in diseases in which the downstream cascades are dysregulated, such as hereditary angioedema. Garadacimab (CSL312) is a first‐in‐class, fully human, monoclonal antibody targeting activated FXII (FXIIa). We describe how translational pharmacokinetic (PK) and pharmacodynamic (PD) modeling enabled dose selection for the phase I, first‐in‐human trial of garadacimab. The PK/PD data used for modeling were derived from preclinical PK/PD and safety studies. Garadacimab plasma concentrations rose with increasing dose, and clear dose‐related PD effects were observed (e.g., a mechanism‐based prolongation of activated partial thromboplastin time). The PK/PD profile from cynomolgus monkeys was used to generate minimal physiologically‐based pharmacokinetic (mPBPK) models with target‐mediated drug disposition (TMDD) for data prediction in cynomolgus monkeys. These models were later adapted for prediction of human data to establish dose selection. Based on the final mPBPK model with TMDD and assuming a weight of 70 kg for an adult human, a minimal inhibition (<10%) of FXIIa with a starting dose of 0.1 mg/kg garadacimab and a near maximal inhibition (>95%) at 10 mg/kg garadacimab were predicted. The phase I study is complete, and data on exposure profiles and inhibition of FXIIa‐mediated kallikrein activity observed in the trial support and validate these simulations. This emphasizes the utility and relevance of translational modeling and simulation in drug development.
Collapse
Affiliation(s)
| | - Xi Chen
- Genentech, San Francisco, California, USA
| | | | - Frauke May
- CSL Behring Innovation GmbH, Marburg, Germany
| | | | | | | | | | - William J Jusko
- School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, Buffalo, New York, USA
| | | |
Collapse
|
14
|
Prevalence and Clinical Impact of Reduced Coagulation Factor XII Activity in Patients Receiving Extracorporeal Membrane Oxygenation. Crit Care Med 2021; 49:e1206-e1211. [PMID: 34259662 DOI: 10.1097/ccm.0000000000005179] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVES Extracorporeal membrane oxygenation provides large surface exposure to human blood leading to coagulation activation. Only limited clinical data are available on contact activation and coagulation factor XII activity in extracorporeal membrane oxygenation patients. DESIGN Prospective cohort study. SETTING Three medical ICUs at the Medical University of Vienna. PATIENTS Adult patients receiving venovenous or venoarterial extracorporeal membrane oxygenation. INTERVENTIONS None. MEASUREMENTS AND MAIN RESULTS The primary outcome was the change in coagulation factor XII activity in response to extracorporeal membrane oxygenation. Secondary outcomes included the prevalence of reduced coagulation factor XII activity (< 60%) among patients receiving extracorporeal membrane oxygenation and association of coagulation factor XII activity with thromboembolic and bleeding complications. An exploratory endpoint was the association of coagulation factor XII activity and activated partial thromboplastin time in heparinase-treated samples in vitro. Fifty-one patients with a total of 117 samples were included in the study between July 2018 and February 2020. Fifty patients (98%) had reduced coagulation factor XII activity at any timepoint during extracorporeal membrane oxygenation. Median coagulation factor XII activity during extracorporeal membrane oxygenation treatment was 30% (interquartile range, 21.5-41%) and increased after discontinuation (p = 0.047). Patients with thromboembolic complications had higher median coagulation factor XII activity during extracorporeal membrane oxygenation (34% vs 23%; p = 0.023). The odds of a thromboembolic event increased by 200% per tertile of median coagulation factor XII activity (crude odds ratio, 3.034; 95% CI, 1.21-7.63). No association with bleeding was observed. In heparinase-treated samples, coagulation factor XII activity correlated well with activated partial thromboplastin time (r = -0.789; p = 0.007). CONCLUSIONS We observed a high prevalence of reduced coagulation factor XII activity in adult patients on extracorporeal membrane oxygenation, which may confound activated partial thromboplastin time measurements and limit its clinical usefulness for monitoring and titrating anticoagulation with unfractionated heparin. Lower coagulation factor XII activity was associated with less thromboembolic complications, which may highlight the potential of coagulation factor XII to serve as a target for anticoagulation in extracorporeal membrane oxygenation.
Collapse
|
15
|
Obstals F, Witzdam L, Garay-Sarmiento M, Kostina NY, Quandt J, Rossaint R, Singh S, Grottke O, Rodriguez-Emmenegger C. Improving Hemocompatibility: How Can Smart Surfaces Direct Blood To Fight against Thrombi. ACS APPLIED MATERIALS & INTERFACES 2021; 13:11696-11707. [PMID: 33656864 DOI: 10.1021/acsami.1c01079] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Nature utilizes endothelium as a blood interface that perfectly controls hemostasis, preventing the uncontrolled formation of thrombi. The management of positive and negative feedback that finely tunes thrombosis and fibrinolysis is essential for human life, especially for patients who undergo extracorporeal circulation (ECC) after a severe respiratory or cardiac failure. The exposure of blood to a surface different from healthy endothelium inevitably initiates coagulation, drastically increasing the mortality rate by thromboembolic complications. In the present study, an ultrathin antifouling fibrinolytic coating capable of disintegrating thrombi in a self-regulated manner is reported. The coating system is composed of a polymer brush layer that can prevent any unspecific interaction with blood. The brushes are functionalized with a tissue plasminogen activator (tPA) to establish localized fibrinolysis that solely and exclusively is active when it is required. This interactive switching between the dormant and active state is realized through an amplification mechanism that increases (positive feedback) or restores (negative feedback) the activity of tPA depending on whether a thrombus is detected and captured or not. Thus, only a low surface density of tPA is necessary to lyse real thrombi. Our work demonstrates the first report of a coating that self-regulates its fibrinolytic activity depending on the conditions of blood.
Collapse
Affiliation(s)
- Fabian Obstals
- DWI - Leibniz-Institute for Interactive Materials e.V., Forckenbeckstraße 50, Aachen D-52074, Germany
- Institute of Technical and Macromolecular Chemistry, RWTH Aachen University, Forckenbeckstraße 50, Aachen D-52074, Germany
| | - Lena Witzdam
- DWI - Leibniz-Institute for Interactive Materials e.V., Forckenbeckstraße 50, Aachen D-52074, Germany
- Institute of Technical and Macromolecular Chemistry, RWTH Aachen University, Forckenbeckstraße 50, Aachen D-52074, Germany
| | - Manuela Garay-Sarmiento
- DWI - Leibniz-Institute for Interactive Materials e.V., Forckenbeckstraße 50, Aachen D-52074, Germany
- Institute of Technical and Macromolecular Chemistry, RWTH Aachen University, Forckenbeckstraße 50, Aachen D-52074, Germany
| | - Nina Yu Kostina
- DWI - Leibniz-Institute for Interactive Materials e.V., Forckenbeckstraße 50, Aachen D-52074, Germany
- Institute of Technical and Macromolecular Chemistry, RWTH Aachen University, Forckenbeckstraße 50, Aachen D-52074, Germany
| | - Jonas Quandt
- DWI - Leibniz-Institute for Interactive Materials e.V., Forckenbeckstraße 50, Aachen D-52074, Germany
- Institute of Technical and Macromolecular Chemistry, RWTH Aachen University, Forckenbeckstraße 50, Aachen D-52074, Germany
| | - Rolf Rossaint
- University Hospital Aachen, Pauwelsstraße 30, Aachen D-52074, Germany
| | - Smriti Singh
- DWI - Leibniz-Institute for Interactive Materials e.V., Forckenbeckstraße 50, Aachen D-52074, Germany
- Institute of Technical and Macromolecular Chemistry, RWTH Aachen University, Forckenbeckstraße 50, Aachen D-52074, Germany
| | - Oliver Grottke
- University Hospital Aachen, Pauwelsstraße 30, Aachen D-52074, Germany
| | - Cesar Rodriguez-Emmenegger
- DWI - Leibniz-Institute for Interactive Materials e.V., Forckenbeckstraße 50, Aachen D-52074, Germany
- Institute of Technical and Macromolecular Chemistry, RWTH Aachen University, Forckenbeckstraße 50, Aachen D-52074, Germany
| |
Collapse
|
16
|
Preissner KT, Fischer S, Deindl E. Extracellular RNA as a Versatile DAMP and Alarm Signal That Influences Leukocyte Recruitment in Inflammation and Infection. Front Cell Dev Biol 2020; 8:619221. [PMID: 33392206 PMCID: PMC7775424 DOI: 10.3389/fcell.2020.619221] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 11/30/2020] [Indexed: 12/11/2022] Open
Abstract
Upon vascular injury, tissue damage, ischemia, or microbial infection, intracellular material such as nucleic acids and histones is liberated and comes into contact with the vessel wall and circulating blood cells. Such "Danger-associated molecular patterns" (DAMPs) may thus have an enduring influence on the inflammatory defense process that involves leukocyte recruitment and wound healing reactions. While different species of extracellular RNA (exRNA), including microRNAs and long non-coding RNAs, have been implicated to influence inflammatory processes at different levels, recent in vitro and in vivo work has demonstrated a major impact of ribosomal exRNA as a prominent DAMP on various steps of leukocyte recruitment within the innate immune response. This includes the induction of vascular hyper-permeability and vasogenic edema by exRNA via the activation of the "vascular endothelial growth factor" (VEGF) receptor-2 system, as well as the recruitment of leukocytes to the inflamed endothelium, the M1-type polarization of inflammatory macrophages, or the role of exRNA as a pro-thrombotic cofactor to promote thrombosis. Beyond sterile inflammation, exRNA also augments the docking of bacteria to host cells and the subsequent microbial invasion. Moreover, upon vessel occlusion and ischemia, the shear stress-induced release of exRNA initiates arteriogenesis (i.e., formation of natural vessel bypasses) in a multistep process that resembles leukocyte recruitment. Although exRNA can be counteracted for by natural circulating RNase1, under the conditions mentioned, only the administration of exogenous, thermostable, non-toxic RNase1 provides an effective and safe therapeutic regimen for treating the damaging activities of exRNA. It remains to be investigated whether exRNA may also influence viral infections (including COVID-19), e.g., by supporting the interaction of host cells with viral particles and their subsequent invasion. In fact, as a consequence of the viral infection cycle, massive amounts of exRNA are liberated, which can provoke further tissue damage and enhance virus dissemination. Whether the application of RNase1 in this scenario may help to limit the extent of viral infections like COVID-19 and impact on leukocyte recruitment and emigration steps in immune defense in order to limit the extent of associated cardiovascular diseases remains to be studied.
Collapse
Affiliation(s)
- Klaus T. Preissner
- Department of Biochemistry, Medical School, Justus Liebig University Giessen, Giessen, Germany
- Kerckhoff-Heart-Research-Institute, Department of Cardiology, Medical School, Justus Liebig University Giessen, Giessen, Germany
| | - Silvia Fischer
- Department of Biochemistry, Medical School, Justus Liebig University Giessen, Giessen, Germany
| | - Elisabeth Deindl
- Walter-Brendel-Centre of Experimental Medicine, University Hospital, LMU Munich, Munich, Germany
- Biomedical Center, Institute of Cardiovascular Physiology and Pathophysiology, LMU Munich, Munich, Germany
| |
Collapse
|
17
|
Factor XII blockade inhibits aortic dilatation in angiotensin II-infused apolipoprotein E-deficient mice. Clin Sci (Lond) 2020; 134:1049-1061. [PMID: 32309850 DOI: 10.1042/cs20191020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 04/05/2020] [Accepted: 04/20/2020] [Indexed: 12/23/2022]
Abstract
Abdominal aortic aneurysm (AAA) is an important cause of mortality in older adults. Chronic inflammation and excessive matrix remodelling are considered important in AAA pathogenesis. Kinins are bioactive peptides important in regulating inflammation. Stimulation of the kinin B2 receptor has been previously reported to promote AAA development and rupture in a mouse model. The endogenous B2 receptor agonist, bradykinin, is generated from the kallikrein-kinin system following activation of plasma kallikrein by Factor XII (FXII). In the current study whole-body FXII deletion, or neutralisation of activated FXII (FXIIa), inhibited expansion of the suprarenal aorta (SRA) of apolipoprotein E-deficient mice in response to angiotensin II (AngII) infusion. FXII deficiency or FXIIa neutralisation led to decreased aortic tumor necrosis factor-α-converting enzyme (TACE/a disintegrin and metalloproteinase-17 (aka tumor necrosis factor-α-converting enzyme) (ADAM-17)) activity, plasma kallikrein concentration, and epithelial growth factor receptor (EGFR) phosphorylation compared with controls. FXII deficiency or neutralisation also reduced Akt1 and Erk1/2 phosphorylation and decreased expression and levels of active matrix metalloproteinase (Mmp)-2 and Mmp-9. The findings suggest that FXII, kallikrein, ADAM-17, and EGFR are important molecular mediators by which AngII induces aneurysm in apolipoprotein E-deficient mice. This could be a novel pathway to target in the design of drugs to limit AAA progression.
Collapse
|
18
|
Gerling K, Ölschläger S, Avci-Adali M, Neumann B, Schweizer E, Schlensak C, Wendel HP, Stoppelkamp S. A Novel C1-Esterase Inhibitor Oxygenator Coating Prevents FXII Activation in Human Blood. Biomolecules 2020; 10:biom10071042. [PMID: 32668719 PMCID: PMC7407883 DOI: 10.3390/biom10071042] [Citation(s) in RCA: 6] [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: 06/07/2020] [Revised: 07/04/2020] [Accepted: 07/09/2020] [Indexed: 01/06/2023] Open
Abstract
The limited hemocompatibility of currently used oxygenator membranes prevents long-term use of artificial lungs in patients with lung failure. To improve hemocompatibility, we developed a novel covalent C1-esterase inhibitor (C1-INH) coating. Besides complement inhibition, C1-INH also prevents FXII activation, a very early event of contact phase activation at the crossroads of coagulation and inflammation. Covalently coated heparin, as the current anticoagulation gold standard, served as control. Additionally, a combination of both coatings (C1-INH/heparin) was established. The coatings were tested for their hemocompatibility by dynamic incubation with freshly drawn human whole blood. The analysis of various blood and plasma parameters revealed that C1-INH-containing coatings were able to markedly reduce FXIIa activity compared to heparin coating. Combined C1-INH/heparin coatings yielded similarly low levels of thrombin-antithrombin III complex formation as heparin coating. In particular, adhesion of monocytes and platelets as well as the diminished formation of fibrin networks were observed for combined coatings. We could show for the first time that a covalent coating with complement inhibitor C1-INH was able to ameliorate hemocompatibility. Thus, the early inhibition of the coagulation cascade is likely to have far-reaching consequences for the other cross-reacting plasma protein pathways.
Collapse
Affiliation(s)
- Katharina Gerling
- University Hospital Tuebingen, Clinic for Thoracic and Cardiovascular Surgery, Calwerstr, 7/1, 72076 Tuebingen, Germany; (K.G.); (S.Ö.); (M.A.-A.); (B.N.); (C.S.); (H.-P.W.)
| | - Sabrina Ölschläger
- University Hospital Tuebingen, Clinic for Thoracic and Cardiovascular Surgery, Calwerstr, 7/1, 72076 Tuebingen, Germany; (K.G.); (S.Ö.); (M.A.-A.); (B.N.); (C.S.); (H.-P.W.)
| | - Meltem Avci-Adali
- University Hospital Tuebingen, Clinic for Thoracic and Cardiovascular Surgery, Calwerstr, 7/1, 72076 Tuebingen, Germany; (K.G.); (S.Ö.); (M.A.-A.); (B.N.); (C.S.); (H.-P.W.)
| | - Bernd Neumann
- University Hospital Tuebingen, Clinic for Thoracic and Cardiovascular Surgery, Calwerstr, 7/1, 72076 Tuebingen, Germany; (K.G.); (S.Ö.); (M.A.-A.); (B.N.); (C.S.); (H.-P.W.)
| | - Ernst Schweizer
- University Hospital Tuebingen, Section Medical Materials and Technology, Osianderstr, 2-8, 72076 Tuebingen, Germany;
| | - Christian Schlensak
- University Hospital Tuebingen, Clinic for Thoracic and Cardiovascular Surgery, Calwerstr, 7/1, 72076 Tuebingen, Germany; (K.G.); (S.Ö.); (M.A.-A.); (B.N.); (C.S.); (H.-P.W.)
| | - Hans-Peter Wendel
- University Hospital Tuebingen, Clinic for Thoracic and Cardiovascular Surgery, Calwerstr, 7/1, 72076 Tuebingen, Germany; (K.G.); (S.Ö.); (M.A.-A.); (B.N.); (C.S.); (H.-P.W.)
| | - Sandra Stoppelkamp
- University Hospital Tuebingen, Clinic for Thoracic and Cardiovascular Surgery, Calwerstr, 7/1, 72076 Tuebingen, Germany; (K.G.); (S.Ö.); (M.A.-A.); (B.N.); (C.S.); (H.-P.W.)
- Correspondence: ; Tel.: +49-7071-29-83340
| |
Collapse
|
19
|
From multi-target anticoagulants to DOACs, and intrinsic coagulation factor inhibitors. Blood Rev 2020; 39:100615. [DOI: 10.1016/j.blre.2019.100615] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2019] [Revised: 08/08/2019] [Accepted: 08/27/2019] [Indexed: 01/10/2023]
|
20
|
Winter WE, Greene DN, Beal SG, Isom JA, Manning H, Wilkerson G, Harris N. Clotting factors: Clinical biochemistry and their roles as plasma enzymes. Adv Clin Chem 2019; 94:31-84. [PMID: 31952574 DOI: 10.1016/bs.acc.2019.07.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The purpose of this review is to describe structure and function of the multiple proteins of the coagulation system and their subcomponent domains. Coagulation is the process by which flowing liquid blood plasma is converted to a soft, viscous gel entrapping the cellular components of blood including red cells and platelets and thereby preventing extravasation of blood. This process is triggered by the minimal proteolysis of plasma fibrinogen. This transforms the latter to sticky fibrin monomers which polymerize into a network. The proteolysis of fibrinogen is a function of the trypsin-like enzyme termed thrombin. Thrombin in turn is activated by a cascade of trypsin-like enzymes that we term coagulation factors. In this review we examine the mechanics of the coagulation cascade with a view to the structure-function relationships of the proteins. We also note that two of the factors have no trypsin like protease domain but are essential cofactors or catalysts for the proteases. This review does not discuss the major role of platelets except to highlight their membrane function with respect to the factors. Coagulation testing is a major part of routine diagnostic clinical pathology. Testing is performed on specimens from individuals either with bleeding or with thrombotic disorders and those on anticoagulant medications. We examine the basic in-vitro laboratory coagulation tests and review the literature comparing the in vitro and in vivo processes. In vitro clinical testing typically utilizes plasma specimens and non-physiological or supraphysiological activators. Because the review focuses on coagulation factor structure, a brief overview of the evolutionary origins of the coagulation system is included.
Collapse
Affiliation(s)
- William E Winter
- University of Florida, Department of Pathology, Immunology & Laboratory Medicine, Gainesville, FL, United States
| | - Dina N Greene
- Laboratory Services, Kaiser Permanente, Renton, WA, United States
| | - Stacy G Beal
- University of Florida, Department of Pathology, Immunology & Laboratory Medicine, Gainesville, FL, United States
| | - James A Isom
- University of Florida, Department of Pathology, Immunology & Laboratory Medicine, Gainesville, FL, United States
| | | | | | - Neil Harris
- University of Florida, Department of Pathology, Immunology & Laboratory Medicine, Gainesville, FL, United States.
| |
Collapse
|
21
|
Mailer RKW, Hänel L, Allende M, Renné T. Polyphosphate as a Target for Interference With Inflammation and Thrombosis. Front Med (Lausanne) 2019; 6:76. [PMID: 31106204 PMCID: PMC6499166 DOI: 10.3389/fmed.2019.00076] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 03/28/2019] [Indexed: 12/19/2022] Open
Abstract
Activated platelets and mast cells expose the inorganic polymer, polyphosphate (polyP) on their surfaces. PolyP initiates procoagulant and proinflammatory reactions and the polymer has been recognized as a therapeutic target for interference with blood coagulation and vascular hyperpermeability. PolyP content and chain length depend on the specific cell type and energy status, which may affect cellular functions. PolyP metabolism has mainly been studied in bacteria and yeast, but its roles in eukaryotic cells and mammalian systems have remained enigmatic. In this review, we will present an overview of polyP functions, focusing on intra- and extracellular roles of the polymer and discuss open questions that emerge from the current knowledge on polyP regulation.
Collapse
Affiliation(s)
- Reiner K W Mailer
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Lorena Hänel
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Mikel Allende
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Thomas Renné
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
22
|
Zhang H, Liu S, Lin C, Luo S, Yang L, Jin Y, Zhu L, Wang M. Compound heterozygous mutations Glu502Lys and Met527Thr of the FXII gene in a patient with factor XII deficiency. ACTA ACUST UNITED AC 2019; 24:420-425. [PMID: 30929639 DOI: 10.1080/16078454.2019.1598679] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
OBJECTIVE To study the gene mutation of human coagulation factor XII (FXII) in a Chinese family with FXII deficiency and it will help us to understand the pathogenesis of this type of disease. CLINICAL PRESENTATION The proband was a 50-year-old male who had a fracture of the right humerus. The routine presurgical coagulation test showed a significant prolonged activated partial thromboplastin time (APTT) at 59.1s (reference range, 29.0-43.0s). TECHNIQUES FXII activity (FXII:C) and FXII antigen (FXII:Ag) were detected by the one-stage clotting method and ELISA, respectively. To identify mutations, the FXII whole exon and flanking sequences were carried out. Suspected mutations were confirmed by reverse sequencing. The conservatism and possible impact of the amino acid substitution were analyzed by ClustalX-2.1-win and four online bioinformatics tools. RESULTS Phenotypic analysis revealed the FXII:C and FXII:Ag of the proband were 4% and 5%, respectively (normal range, 72-113%). Gene sequencing detected compound heterozygous mutations c.1561G > A (Glu502Lys) and c.1637T > C (Met527Thr) in exon 13. Bioinformatics and model analysis indicated that mutations probably had disrupted the function and structure of the FXII protein. CONCLUSION We detected two missense mutations Glu502Lys and Met527Thr in the catalytic domain of the proband, of which Met527Thr was first reported in the world. Our findings suggest that the double mutations in the FXII gene were the causing reasons for the decreased FXII:C and FXII:Ag. These results not only enriched the F12 mutation database in this condition, but also helped to identify the genetic defects of FXII in China.
Collapse
Affiliation(s)
- Haiyue Zhang
- a Department of Clinical Laboratory , The First Affiliated Hospital of Wenzhou Medical University , Wenzhou , People's Republic of China
| | - Siqi Liu
- a Department of Clinical Laboratory , The First Affiliated Hospital of Wenzhou Medical University , Wenzhou , People's Republic of China
| | - Chanchan Lin
- a Department of Clinical Laboratory , The First Affiliated Hospital of Wenzhou Medical University , Wenzhou , People's Republic of China
| | - Shasha Luo
- a Department of Clinical Laboratory , The First Affiliated Hospital of Wenzhou Medical University , Wenzhou , People's Republic of China
| | - Lihong Yang
- a Department of Clinical Laboratory , The First Affiliated Hospital of Wenzhou Medical University , Wenzhou , People's Republic of China
| | - Yanhui Jin
- a Department of Clinical Laboratory , The First Affiliated Hospital of Wenzhou Medical University , Wenzhou , People's Republic of China
| | - Liqing Zhu
- a Department of Clinical Laboratory , The First Affiliated Hospital of Wenzhou Medical University , Wenzhou , People's Republic of China
| | - Mingshan Wang
- a Department of Clinical Laboratory , The First Affiliated Hospital of Wenzhou Medical University , Wenzhou , People's Republic of China
| |
Collapse
|
23
|
Liu J, Qin J, Borodovsky A, Racie T, Castoreno A, Schlegel M, Maier MA, Zimmerman T, Fitzgerald K, Butler J, Akinc A. An investigational RNAi therapeutic targeting Factor XII (ALN-F12) for the treatment of hereditary angioedema. RNA (NEW YORK, N.Y.) 2019; 25:255-263. [PMID: 30463937 PMCID: PMC6348991 DOI: 10.1261/rna.068916.118] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 11/20/2018] [Indexed: 06/09/2023]
Abstract
Hereditary angioedema (HAE) is a genetic disorder mostly caused by mutations in the C1 esterase inhibitor gene (C1INH) that results in poor control of contact pathway activation and excess bradykinin generation. Bradykinin increases vascular permeability and is ultimately responsible for the episodes of swelling characteristic of HAE. We hypothesized that the use of RNA interference (RNAi) to reduce plasma Factor XII (FXII), which initiates the contact pathway signaling cascade, would reduce contact pathway activation and prevent excessive bradykinin generation. A subcutaneously administered GalNAc-conjugated small-interfering RNA (siRNA) targeting F12 mRNA (ALN-F12) was developed, and potency was evaluated in mice, rats, and cynomolgus monkeys. The effect of FXII reduction by ALN-F12 administration was evaluated in two different vascular leakage mouse models. An ex vivo assay was developed to evaluate the correlation between human plasma FXII levels and high-molecular weight kininogen (HK) cleavage. A single subcutaneous dose of ALN-F12 led to potent, dose-dependent reduction of plasma FXII in mice, rats, and NHP. In cynomolgus monkeys, a single subcutaneous dose of ALN-F12 at 3 mg/kg resulted in >85% reduction of plasma FXII. Administration of ALN-F12 resulted in dose-dependent reduction of vascular permeability in two different mouse models of bradykinin-driven vascular leakage, demonstrating that RNAi-mediated reduction of FXII can potentially mitigate excess bradykinin stimulation. Lastly, ex vivo human plasma HK cleavage assay indicated FXII-dependent bradykinin generation. Together, these data suggest that RNAi-mediated knockdown of FXII by ALN-F12 is a potentially promising approach for the prophylactic treatment of HAE.
Collapse
Affiliation(s)
- Jingxuan Liu
- Alnylam Pharmaceuticals, Cambridge, Massachusetts 02142, USA
| | - June Qin
- Alnylam Pharmaceuticals, Cambridge, Massachusetts 02142, USA
| | - Anna Borodovsky
- Alnylam Pharmaceuticals, Cambridge, Massachusetts 02142, USA
| | - Timothy Racie
- Alnylam Pharmaceuticals, Cambridge, Massachusetts 02142, USA
| | - Adam Castoreno
- Alnylam Pharmaceuticals, Cambridge, Massachusetts 02142, USA
| | - Mark Schlegel
- Alnylam Pharmaceuticals, Cambridge, Massachusetts 02142, USA
| | - Martin A Maier
- Alnylam Pharmaceuticals, Cambridge, Massachusetts 02142, USA
| | - Tracy Zimmerman
- Alnylam Pharmaceuticals, Cambridge, Massachusetts 02142, USA
| | | | - James Butler
- Alnylam Pharmaceuticals, Cambridge, Massachusetts 02142, USA
| | - Akin Akinc
- Alnylam Pharmaceuticals, Cambridge, Massachusetts 02142, USA
| |
Collapse
|
24
|
Hogwood J, Naggi A, Torri G, Page C, Rigsby P, Mulloy B, Gray E. The effect of increasing the sulfation level of chondroitin sulfate on anticoagulant specific activity and activation of the kinin system. PLoS One 2018; 13:e0193482. [PMID: 29494632 PMCID: PMC5832253 DOI: 10.1371/journal.pone.0193482] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Accepted: 02/12/2018] [Indexed: 11/29/2022] Open
Abstract
Oversulfated chondroitin sulfate (OSCS) was identified as a contaminant in certain heparin preparations as the cause of adverse reactions in patients. OSCS was found to possess both plasma anticoagulant activity and the ability to activate prekallikrein to kallikrein. Differentially sulfated chondroitin sulfates were prepared by synthetic modification of chondroitin sulfate and were compared to the activity of OSCS purified from contaminated heparin. Whilst chondroitin sulfate was found to have minimal anticoagulant activity, increasing sulfation levels produced an anticoagulant response which we directly show for the first time is mediated through heparin cofactor II. However, the tetra-sulfated preparations did not possess any higher anticoagulant activity than several tri-sulfated variants, and also had lower heparin cofactor II mediated activity. Activation of prekallikrein was concentration dependent for all samples, and broadly increased with the degree of sulfation, though the di-sulfated preparation was able to form more kallikrein than some of the tri-sulfated preparations. The ability of the samples to activate the kinin system, as measured by bradykinin, was observed to be through kallikrein generation. These results show that whilst an increase in sulfation of chondroitin sulfate did cause an increase in anticoagulant activity and activation of the kinin system, there may be subtler structural interactions other than sulfation at play given the different responses observed.
Collapse
Affiliation(s)
- J. Hogwood
- National Institute for Biological Standards and Control, Blanche Lane, Herts, United Kingdom
- Sacker Institute of Pulmonary Pharmacology, Institute of Pharmaceutical Science, King’s College London, United Kingdom
- * E-mail:
| | - A. Naggi
- Institute for Chemical and Biochemical Research ‘‘G. Ronzoni”, Milan, Italy
| | - G. Torri
- Institute for Chemical and Biochemical Research ‘‘G. Ronzoni”, Milan, Italy
| | - C. Page
- Sacker Institute of Pulmonary Pharmacology, Institute of Pharmaceutical Science, King’s College London, United Kingdom
| | - P. Rigsby
- National Institute for Biological Standards and Control, Blanche Lane, Herts, United Kingdom
| | - B. Mulloy
- Sacker Institute of Pulmonary Pharmacology, Institute of Pharmaceutical Science, King’s College London, United Kingdom
| | - E. Gray
- National Institute for Biological Standards and Control, Blanche Lane, Herts, United Kingdom
- Sacker Institute of Pulmonary Pharmacology, Institute of Pharmaceutical Science, King’s College London, United Kingdom
| |
Collapse
|
25
|
Lowe G, Cate HT. Coagulation proteases and cardiovascular disease. Thromb Haemost 2017; 112:858-9. [DOI: 10.1160/th14-09-0781] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Accepted: 09/19/2014] [Indexed: 11/05/2022]
|
26
|
Ding C, van 't Veer C, Roelofs JJTH, Shukla M, McCrae KR, Revenko AS, Crosby J, van der Poll T. Limited role of kininogen in the host response during gram-negative pneumonia-derived sepsis. Am J Physiol Lung Cell Mol Physiol 2017; 314:L397-L405. [PMID: 29122754 DOI: 10.1152/ajplung.00288.2017] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
High-molecular-weight kininogen (HK), together with factor XI, factor XII and prekallikrein, is part of the contact system that has proinflammatory, prothrombotic, and vasoactive properties. We hypothesized that HK plays a role in the host response during pneumonia-derived sepsis. To this end mice were depleted of kininogen (KNG) to plasma HK levels of 28% of normal by repeated treatment with a specific antisense oligonucleotide (KNG ASO) for 3 wk before infection with the common human sepsis pathogen Klebsiella pneumoniae via the airways. Whereas plasma HK levels increased during infection in mice treated with a scrambled control ASO (Ctrl ASO), HK level in the KNG ASO-treated group remained reduced to 25-30% of that in the corresponding Ctrl ASO group both before and after infection. KNG depletion did not influence bacterial growth in lungs or dissemination to distant body sites. KNG depletion was associated with lower lung CXC chemokine and myeloperoxidase levels but did not impact neutrophil influx, lung pathology, activation of the vascular endothelium, activation of the coagulation system, or the extent of distant organ injury. These results were corroborated by studies in mice with a genetic deficiency of KNG, which were indistinguishable from wild-type mice during Klebsiella-induced sepsis. Both KNG depletion and KNG deficiency were associated with strongly reduced plasma prekallikrein levels, indicating the carrier function of HK for this zymogen. This study suggests that KNG does not significantly contribute to the host defense during gram-negative pneumonia-derived sepsis.
Collapse
Affiliation(s)
- Chao Ding
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University , Nanjing , China.,Center of Experimental and Molecular Medicine, Academic Medical Center, University of Amsterdam , Amsterdam , The Netherlands
| | - Cornelis van 't Veer
- Center of Experimental and Molecular Medicine, Academic Medical Center, University of Amsterdam , Amsterdam , The Netherlands
| | - Joris J T H Roelofs
- Department of Pathology, Academic Medical Center, University of Amsterdam, Amsterdam , The Netherlands
| | - Meenal Shukla
- Departments of Hematology-Oncology and Cellular and Molecular Medicine, Cleveland Clinic, Cleveland, Ohio
| | - Keith R McCrae
- Departments of Hematology-Oncology and Cellular and Molecular Medicine, Cleveland Clinic, Cleveland, Ohio
| | - Alexey S Revenko
- Antisense Drug Discovery, IONIS Pharmaceuticals, Carlsbad, California
| | - Jeff Crosby
- Antisense Drug Discovery, IONIS Pharmaceuticals, Carlsbad, California
| | - Tom van der Poll
- Center of Experimental and Molecular Medicine, Academic Medical Center, University of Amsterdam , Amsterdam , The Netherlands.,Division of Infectious Diseases, Academic Medical Center, University of Amsterdam, Amsterdam , The Netherlands
| |
Collapse
|
27
|
Yang A, Chen F, He C, Zhou J, Lu Y, Dai J, Birge RB, Wu Y. The Procoagulant Activity of Apoptotic Cells Is Mediated by Interaction with Factor XII. Front Immunol 2017; 8:1188. [PMID: 28993777 PMCID: PMC5622377 DOI: 10.3389/fimmu.2017.01188] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Accepted: 09/07/2017] [Indexed: 11/13/2022] Open
Abstract
Apoptotic cells, by externalizing phosphatidylserine (PS) as a hallmark feature, are procoagulant. However, the mechanism by which apoptotic cells activate coagulation system remains unknown. Intrinsic coagulation pathway is initiated by coagulation factor XII (FXII) of contact activation system. The purpose of this study was to determine whether FXII is involved in procoagulant activity of apoptotic cells. Using western blotting and chromogenic substrate assay, we found that incubation with apoptotic cells, but not with viable cells, resulted in rapid cleavage and activation of FXII in the presence of prekallikrein and high molecular weight kininogen (HK), other two components of contact activation system. As detected by flow cytometry, FXII bound to apoptotic cells in a concentration-dependent manner, which was inhibited by annexin V and PS liposome. Direct association of FXII with PS was confirmed in a surface plasmon resonance assay. Clotting time of FXII-deficient plasma induced by apoptotic cells was significantly prolonged, which was fully reversed by replenishment with FXII. Corn trypsin inhibitor, a FXII inhibitor, completely prevented apoptotic cells-induced intrinsic tenase complex formation. Consistently, apoptotic cells significantly increased thrombin production in normal plasma, which was not affected by an inhibitory anti-tissue factor antibody. However, blocking of PS by annexin V, inhibition of FXII, or the deficiency of FXII suppressed apoptotic cells-induced thrombin generation. Addition of purified FXII to FXII-deficient plasma recovered thrombin generation to the normal plasma level. In conclusion, FXII binds to apoptotic cells via PS and becomes activated, thereby constituting a novel mechanism mediating the procoagulant activity of apoptotic cells.
Collapse
Affiliation(s)
- Aizhen Yang
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Fengwu Chen
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Chao He
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Junsong Zhou
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China.,The Sol Sherry Thrombosis Research Center, Temple University School of Medicine, Philadelphia, PA, United States
| | - Yi Lu
- Wuhan Thalys Medical Technology Inc., Wuhan, China
| | - Jihong Dai
- The Sol Sherry Thrombosis Research Center, Temple University School of Medicine, Philadelphia, PA, United States.,Department of Pathology and Laboratory Medicine, Rutgers University-New Jersey Medical School, Newark, NJ, United States
| | - Raymond B Birge
- Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers University-New Jersey Medical School, Newark, NJ, United States
| | - Yi Wu
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China.,The Sol Sherry Thrombosis Research Center, Temple University School of Medicine, Philadelphia, PA, United States
| |
Collapse
|
28
|
The plasma contact system, a protease cascade at the nexus of inflammation, coagulation and immunity. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2017; 1864:2118-2127. [PMID: 28743596 DOI: 10.1016/j.bbamcr.2017.07.009] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Revised: 07/14/2017] [Accepted: 07/19/2017] [Indexed: 01/11/2023]
Abstract
The contact system is a potent procoagulant and proinflammatory plasma protease cascade that is initiated by binding ("contact")-induced, auto-activation of factor XII zymogen. Formed active serine protease FXIIa then cleaves plasma prekallikrein to kallikrein that in turn liberates the mediator bradykinin from its precursor high molecular weight kininogen. Bradykinin induces inflammation with implications for host defense and innate immunity. FXIIa also triggers the intrinsic pathway of coagulation that has been shown to critically contribute to thrombosis. Vice versa, FXII deficiency impairs thrombosis in animal models without inducing abnormal excessive bleeding. Recent work has established the FXIIa-driven contact system as promising target for anticoagulant and anti-inflammatory drugs. This review focuses on the biochemistry of the contact system, its regulation by endogenous and exogenous inhibitors, and roles in disease states. This article is part of a Special Issue entitled: Proteolysis as a Regulatory Event in Pathophysiology edited by Stefan Rose-John.
Collapse
|
29
|
Bauer JW, Xu LC, Vogler EA, Siedlecki CA. Surface dependent contact activation of factor XII and blood plasma coagulation induced by mixed thiol surfaces. Biointerphases 2017; 12:02D410. [PMID: 28514863 PMCID: PMC5435513 DOI: 10.1116/1.4983634] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 04/27/2017] [Accepted: 05/04/2017] [Indexed: 12/20/2022] Open
Abstract
Studies of the activation of FXII in both platelet poor plasma and in neat buffer solutions were undertaken for a series of mixed thiol self-assembled monolayers spanning a broad range of water wettability. A wide spectrum of carboxyl/methyl-, hydroxyl/methyl-, and amine/methyl-thiol modified surfaces were prepared, characterized, and then utilized as the procoagulant materials in a series of FXII activation studies. X-ray photoelectron spectroscopy was utilized to verify the sample surface's thiol composition and contact angles measured to determine the sample surface's wettability. These samples were then used in in vitro coagulation assays using a 50% mixture of recalcified plasma in phosphate buffered saline. Alternatively, the samples were placed into purified FXII solutions for 30 min to assess FXII activation in neat buffer solution. Plasma coagulation studies supported a strong role for anionic surfaces in contact activation, in line with the traditional models of coagulation, while the activation results in neat buffer solution demonstrated that FXIIa production is related to surface wettability with minimum levels of enzyme activation observed at midrange wettabilities, and no statistically distinguishable differences in FXII activation seen between highly wettable and highly nonwettable surfaces. Results demonstrated that the composition of the solution and the surface properties of the material all contribute to the observation of contact activation, and the activation of FXII is not specific to anionic surfaces as has been long believed.
Collapse
Affiliation(s)
- James W Bauer
- Department of Bioengineering, Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033
| | - Li-Chong Xu
- Department of Surgery, Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033
| | - Erwin A Vogler
- Department of Bioengineering, Pennsylvania State University, University Park, Pennsylvania 16802 and Department of Materials Science and Engineering, Pennsylvania State University, University Park, Pennsylvania 16802
| | - Christopher A Siedlecki
- Department of Bioengineering, Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033 and Department of Surgery, Pennsylvania State University College of Medicine, 500 University Drive, H151, Hershey, Pennsylvania 17033
| |
Collapse
|
30
|
Hopp S, Nolte MW, Stetter C, Kleinschnitz C, Sirén AL, Albert-Weissenberger C. Alleviation of secondary brain injury, posttraumatic inflammation, and brain edema formation by inhibition of factor XIIa. J Neuroinflammation 2017; 14:39. [PMID: 28219400 PMCID: PMC5319055 DOI: 10.1186/s12974-017-0815-8] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Accepted: 02/08/2017] [Indexed: 11/15/2022] Open
Abstract
Background Traumatic brain injury (TBI) is a devastating neurological condition and a frequent cause of permanent disability. Posttraumatic inflammation and brain edema formation, two pathological key events contributing to secondary brain injury, are mediated by the contact-kinin system. Activation of this pathway in the plasma is triggered by activated factor XII. Hence, we set out to study in detail the influence of activated factor XII on the abovementioned pathophysiological features of TBI. Methods Using a cortical cryogenic lesion model in mice, we investigated the impact of genetic deficiency of factor XII and inhibition of activated factor XII with a single bolus injection of recombinant human albumin-fused Infestin-4 on the release of bradykinin, the brain lesion size, and contact-kinin system-dependent pathological events. We determined protein levels of bradykinin, intracellular adhesion molecule-1, CC-chemokine ligand 2, and interleukin-1β by enzyme-linked immunosorbent assays and mRNA levels of genes related to inflammation by quantitative real-time PCR. Brain lesion size was determined by tetrazolium chloride staining. Furthermore, protein levels of the tight junction protein occludin, integrity of the blood-brain barrier, and brain water content were assessed by Western blot analysis, extravasated Evans Blue dye, and the wet weight-dry weight method, respectively. Infiltration of neutrophils and microglia/activated macrophages into the injured brain lesions was quantified by immunohistological stainings. Results We show that both genetic deficiency of factor XII and inhibition of activated factor XII in mice diminish brain injury-induced bradykinin release by the contact-kinin system and minimize brain lesion size, blood-brain barrier leakage, brain edema formation, and inflammation in our brain injury model. Conclusions Stimulation of bradykinin release by activated factor XII probably plays a prominent role in expanding secondary brain damage by promoting brain edema formation and inflammation. Pharmacological blocking of activated factor XII could be a useful therapeutic principle in the treatment of TBI-associated pathologic processes by alleviating posttraumatic inflammation and brain edema formation.
Collapse
Affiliation(s)
- Sarah Hopp
- Department of Neurology, University Hospital Würzburg, Würzburg, Germany.,Department of Neurosurgery, University Hospital Würzburg, Josef-Schneider-Strasse 11, Würzburg, Germany
| | | | - Christian Stetter
- Department of Neurosurgery, University Hospital Würzburg, Josef-Schneider-Strasse 11, Würzburg, Germany
| | - Christoph Kleinschnitz
- Department of Neurology, University Hospital Würzburg, Würzburg, Germany.,Department of Neurology, University Duisburg-Essen, Essen, Germany
| | - Anna-Leena Sirén
- Department of Neurosurgery, University Hospital Würzburg, Josef-Schneider-Strasse 11, Würzburg, Germany
| | | |
Collapse
|
31
|
Middendorp SJ, Wilbs J, Quarroz C, Calzavarini S, Angelillo-Scherrer A, Heinis C. Peptide Macrocycle Inhibitor of Coagulation Factor XII with Subnanomolar Affinity and High Target Selectivity. J Med Chem 2017; 60:1151-1158. [DOI: 10.1021/acs.jmedchem.6b01548] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Affiliation(s)
- Simon J. Middendorp
- Institute
of Chemical Sciences and Engineering, Ecole Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| | - Jonas Wilbs
- Institute
of Chemical Sciences and Engineering, Ecole Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| | - Claudia Quarroz
- University Clinic
of Hematology and Central Hematology Laboratory, Bern University Hospital,
University of Bern, Inselspital, CH-3010 Bern, Switzerland
- Department
of Clinical Research, University of Bern, CH-3010 Bern, Switzerland
| | - Sara Calzavarini
- University Clinic
of Hematology and Central Hematology Laboratory, Bern University Hospital,
University of Bern, Inselspital, CH-3010 Bern, Switzerland
- Department
of Clinical Research, University of Bern, CH-3010 Bern, Switzerland
| | - Anne Angelillo-Scherrer
- University Clinic
of Hematology and Central Hematology Laboratory, Bern University Hospital,
University of Bern, Inselspital, CH-3010 Bern, Switzerland
- Department
of Clinical Research, University of Bern, CH-3010 Bern, Switzerland
| | - Christian Heinis
- Institute
of Chemical Sciences and Engineering, Ecole Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| |
Collapse
|
32
|
Barbieri CM, Wang X, Wu W, Zhou X, Ogawa AM, O'Neill K, Chu D, Castriota G, Seiffert DA, Gutstein DE, Chen Z. Factor XIIa as a Novel Target for Thrombosis: Target Engagement Requirement and Efficacy in a Rabbit Model of Microembolic Signals. J Pharmacol Exp Ther 2016; 360:466-475. [PMID: 28035006 DOI: 10.1124/jpet.116.238493] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2016] [Accepted: 12/22/2016] [Indexed: 11/22/2022] Open
Abstract
Coagulation Factor XII (FXII) plays a critical role in thrombosis. What is unclear is the level of enzyme occupancy of FXIIa that is needed for efficacy and the impact of FXIIa inhibition on cerebral embolism. A selective activated FXII (FXIIa) inhibitor, recombinant human albumin-tagged mutant Infestin-4 (rHA-Mut-inf), was generated to address these questions. rHA-Mut-inf displayed potency comparable to the original wild-type HA-Infestin-4 (human FXIIa inhibition constant = 0.07 and 0.12 nM, respectively), with markedly improved selectivity against Factor Xa (FXa) and plasmin. rHA-Mut-inf binds FXIIa, but not FXII zymogen, and competitively inhibits FXIIa protease activity. Its mode of action is hence akin to typical small-molecule inhibitors. Plasma shift and aPTT studies with rHA-Mut-inf demonstrated that calculated enzyme occupancy for FXIIa in achieving a putative aPTT doubling target in human, nonhuman primate, and rabbit is more than 99.0%. The effects of rHA-Mut-inf in carotid arterial thrombosis and microembolic signal (MES) in middle cerebral artery were assessed simultaneously in rabbits. Dose-dependent inhibition was observed for both arterial thrombosis and MES. The ED50 of thrombus formation was 0.17 mg/kg i.v. rHA-Mut-inf for the integrated blood flow and 0.16 mg/kg for thrombus weight; the ED50 for MES was 0.06 mg/kg. Ex vivo aPTT tracked with efficacy. In summary, our findings demonstrated that very high enzyme occupancy will be required for FXIIa active site inhibitors, highlighting the high potency and exquisite selectivity necessary for achieving efficacy in humans. Our MES studies suggest that targeting FXIIa may offer a promising strategy for stroke prevention associated with thromboembolic events.
Collapse
Affiliation(s)
- Christopher M Barbieri
- In Vitro Pharmacology (C.M.B., A.M.O., K.O., D.C.) and Cardiometabolic Diseases (X.W., W.W., X.Z., G.C., D.A.S., D.E.G., Z.C.), Merck & Co., Inc., Kenilworth, New Jersey
| | - Xinkang Wang
- In Vitro Pharmacology (C.M.B., A.M.O., K.O., D.C.) and Cardiometabolic Diseases (X.W., W.W., X.Z., G.C., D.A.S., D.E.G., Z.C.), Merck & Co., Inc., Kenilworth, New Jersey
| | - Weizhen Wu
- In Vitro Pharmacology (C.M.B., A.M.O., K.O., D.C.) and Cardiometabolic Diseases (X.W., W.W., X.Z., G.C., D.A.S., D.E.G., Z.C.), Merck & Co., Inc., Kenilworth, New Jersey
| | - Xueping Zhou
- In Vitro Pharmacology (C.M.B., A.M.O., K.O., D.C.) and Cardiometabolic Diseases (X.W., W.W., X.Z., G.C., D.A.S., D.E.G., Z.C.), Merck & Co., Inc., Kenilworth, New Jersey
| | - Aimie M Ogawa
- In Vitro Pharmacology (C.M.B., A.M.O., K.O., D.C.) and Cardiometabolic Diseases (X.W., W.W., X.Z., G.C., D.A.S., D.E.G., Z.C.), Merck & Co., Inc., Kenilworth, New Jersey
| | - Kim O'Neill
- In Vitro Pharmacology (C.M.B., A.M.O., K.O., D.C.) and Cardiometabolic Diseases (X.W., W.W., X.Z., G.C., D.A.S., D.E.G., Z.C.), Merck & Co., Inc., Kenilworth, New Jersey
| | - Donald Chu
- In Vitro Pharmacology (C.M.B., A.M.O., K.O., D.C.) and Cardiometabolic Diseases (X.W., W.W., X.Z., G.C., D.A.S., D.E.G., Z.C.), Merck & Co., Inc., Kenilworth, New Jersey
| | - Gino Castriota
- In Vitro Pharmacology (C.M.B., A.M.O., K.O., D.C.) and Cardiometabolic Diseases (X.W., W.W., X.Z., G.C., D.A.S., D.E.G., Z.C.), Merck & Co., Inc., Kenilworth, New Jersey
| | - Dietmar A Seiffert
- In Vitro Pharmacology (C.M.B., A.M.O., K.O., D.C.) and Cardiometabolic Diseases (X.W., W.W., X.Z., G.C., D.A.S., D.E.G., Z.C.), Merck & Co., Inc., Kenilworth, New Jersey
| | - David E Gutstein
- In Vitro Pharmacology (C.M.B., A.M.O., K.O., D.C.) and Cardiometabolic Diseases (X.W., W.W., X.Z., G.C., D.A.S., D.E.G., Z.C.), Merck & Co., Inc., Kenilworth, New Jersey
| | - Zhu Chen
- In Vitro Pharmacology (C.M.B., A.M.O., K.O., D.C.) and Cardiometabolic Diseases (X.W., W.W., X.Z., G.C., D.A.S., D.E.G., Z.C.), Merck & Co., Inc., Kenilworth, New Jersey
| |
Collapse
|
33
|
The initiation and effects of plasma contact activation: an overview. Int J Hematol 2016; 105:235-243. [PMID: 27848184 DOI: 10.1007/s12185-016-2132-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 10/31/2016] [Accepted: 11/02/2016] [Indexed: 10/20/2022]
Abstract
The plasma contact system sits atop the intrinsic coagulation cascade and plasma kallikrein-kinin pathway, and in vivo its activation contributes, respectively, to coagulation and inflammation mainly via two downstream pathways. This system has been widely investigated, its activation mechanisms by negatively charged surfaces and the interactions within its components, factor XII, prekallikrein and high molecular weight kininogen are well understood at the biochemical level. However, as most of the activators that have been discovered by in vitro experiments are exogenous, the physiological activators and roles of the contact system have remained unclear and controversial. In the last two decades, several physiological activators have been identified, and a better understanding of its roles and its connection with other signaling pathways has been obtained from in vivo studies. In this article, we present an overview of the contact pathway with a focus on the activation mechanisms, natural stimuli, possible physiological roles, potential risks of its excessive activation, remaining questions and future prospects.
Collapse
|
34
|
Serum stimulation of CCR7 chemotaxis due to coagulation factor XIIa-dependent production of high-molecular-weight kininogen domain 5. Proc Natl Acad Sci U S A 2016; 113:E7059-E7068. [PMID: 27791187 DOI: 10.1073/pnas.1615671113] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Chemokines and their receptors play a critical role in immune function by directing cell-specific movement. C-C chemokine receptor 7 (CCR7) facilitates entry of T cells into lymph nodes. CCR7-dependent chemotaxis requires either of the cognate ligands C-C chemokine ligand 19 (CCL19) or CCL21. Although CCR7-dependent chemotaxis can be augmented through receptor up-regulation or by increased chemokine concentrations, we found that chemotaxis is also markedly enhanced by serum in vitro. Upon purification, the serum cofactor activity was ascribed to domain 5 of high-molecular-weight kininogen. This peptide was necessary and sufficient for accelerated chemotaxis. The cofactor activity in serum was dependent on coagulation factor XIIa, a serine protease known to induce cleavage of high-molecular-weight kininogen (HK) at sites of inflammation. Within domain 5, we synthesized a 24-amino acid peptide that could recapitulate the activity of intact serum through a mechanism distinct from up-regulating CCR7 expression or promoting chemokine binding to CCR7. This peptide interacts with the extracellular matrix protein thrombospondin 4 (TSP4), and antibodies to TSP4 neutralize its activity. In vivo, an HK domain 5 peptide stimulated homing of both T and B cells to lymph nodes. A circulating cofactor that is activated at inflammatory foci to enhance lymphocyte chemotaxis represents a powerful mechanism coupling inflammation to adaptive immunity.
Collapse
|
35
|
Cooley BC. The dirty side of the intrinsic pathway of coagulation. Thromb Res 2016; 145:159-60. [DOI: 10.1016/j.thromres.2016.06.028] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 06/24/2016] [Accepted: 06/25/2016] [Indexed: 10/21/2022]
|
36
|
Gulpen AJW, Ten Cate-Hoek AJ, Ten Cate H. Upstream versus downstream thrombin inhibition. Expert Rev Cardiovasc Ther 2016; 14:1273-1282. [DOI: 10.1080/14779072.2016.1224179] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|
37
|
Farkas H. Icatibant as acute treatment for hereditary angioedema in adults. Expert Rev Clin Pharmacol 2016; 9:779-88. [DOI: 10.1080/17512433.2016.1182425] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Affiliation(s)
- Henriette Farkas
- Hungarian Angioedema Center, 3rd Department of Internal Medicine, Semmelweis University, Budapest, Hungary
| |
Collapse
|
38
|
Bane CE, Ivanov I, Matafonov A, Boyd KL, Cheng Q, Sherwood ER, Tucker EI, Smiley ST, McCarty OJT, Gruber A, Gailani D. Factor XI Deficiency Alters the Cytokine Response and Activation of Contact Proteases during Polymicrobial Sepsis in Mice. PLoS One 2016; 11:e0152968. [PMID: 27046148 PMCID: PMC4821616 DOI: 10.1371/journal.pone.0152968] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Accepted: 03/22/2016] [Indexed: 11/18/2022] Open
Abstract
Sepsis, a systemic inflammatory response to infection, is often accompanied by abnormalities of blood coagulation. Prior work with a mouse model of sepsis induced by cecal ligation and puncture (CLP) suggested that the protease factor XIa contributed to disseminated intravascular coagulation (DIC) and to the cytokine response during sepsis. We investigated the importance of factor XI to cytokine and coagulation responses during the first 24 hours after CLP. Compared to wild type littermates, factor XI-deficient (FXI-/-) mice had a survival advantage after CLP, with smaller increases in plasma levels of TNF-α and IL-10 and delayed IL-1β and IL-6 responses. Plasma levels of serum amyloid P, an acute phase protein, were increased in wild type mice 24 hours post-CLP, but not in FXI-/- mice, supporting the impression of a reduced inflammatory response in the absence of factor XI. Surprisingly, there was little evidence of DIC in mice of either genotype. Plasma levels of the contact factors factor XII and prekallikrein were reduced in WT mice after CLP, consistent with induction of contact activation. However, factor XII and PK levels were not reduced in FXI-/- animals, indicating factor XI deficiency blunted contact activation. Intravenous infusion of polyphosphate into WT mice also induced changes in factor XII, but had much less effect in FXI deficient mice. In vitro analysis revealed that factor XIa activates factor XII, and that this reaction is enhanced by polyanions such polyphosphate and nucleic acids. These data suggest that factor XI deficiency confers a survival advantage in the CLP sepsis model by altering the cytokine response to infection and blunting activation of the contact (kallikrein-kinin) system. The findings support the hypothesis that factor XI functions as a bidirectional interface between contact activation and thrombin generation, allowing the two processes to influence each other.
Collapse
Affiliation(s)
- Charles E. Bane
- Department of Pathology, Microbiology and Immunology, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Ivan Ivanov
- Department of Pathology, Microbiology and Immunology, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Anton Matafonov
- Department of Pathology, Microbiology and Immunology, Vanderbilt University, Nashville, Tennessee, United States of America
- Department of Bioengineering and Organic Chemistry, Tomsk Polytechnic University, Tomsk, Russia
| | - Kelli L. Boyd
- Department of Pathology, Microbiology and Immunology, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Qiufang Cheng
- Department of Pathology, Microbiology and Immunology, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Edward R. Sherwood
- Department of Anesthesiology, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Erik I. Tucker
- Aronora, Inc., Portland, Oregon, United States of America
| | - Stephen T. Smiley
- National Institute of Allergy and Infectious Disease, Bethesda, Maryland, United States of America
| | - Owen J. T. McCarty
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Andras Gruber
- Aronora, Inc., Portland, Oregon, United States of America
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon, United States of America
| | - David Gailani
- Department of Pathology, Microbiology and Immunology, Vanderbilt University, Nashville, Tennessee, United States of America
- * E-mail:
| |
Collapse
|
39
|
Long AT, Kenne E, Jung R, Fuchs TA, Renné T. Contact system revisited: an interface between inflammation, coagulation, and innate immunity. J Thromb Haemost 2016; 14:427-37. [PMID: 26707513 DOI: 10.1111/jth.13235] [Citation(s) in RCA: 213] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Accepted: 11/22/2015] [Indexed: 12/12/2022]
Abstract
The contact system is a plasma protease cascade initiated by factor XII (FXII) that activates the proinflammatory kallikrein-kinin system and the procoagulant intrinsic coagulation pathway. Anionic surfaces induce FXII zymogen activation to form proteolytically active FXIIa. Bacterial surfaces also have the ability to activate contact system proteins, indicating an important role for host defense using the cooperation of the inflammatory and coagulation pathways. Recent research has shown that inorganic polyphosphate found in platelets activates FXII in vivo and can induce coagulation in pathological thrombus formation. Experimental studies have shown that interference with FXII provides thromboprotection without a therapy-associated increase in bleeding, renewing interest in the FXIIa-driven intrinsic pathway of coagulation as a therapeutic target. This review summarizes how the contact system acts as the cross-road of inflammation, coagulation, and innate immunity.
Collapse
Affiliation(s)
- A T Long
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - E Kenne
- Division of Clinical Chemistry, Department of Molecular Medicine and Surgery and Center for Molecular Medicine, Karolinska Institutet and University Hospital, Stockholm, Sweden
| | - R Jung
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - T A Fuchs
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Division of Clinical Chemistry, Department of Molecular Medicine and Surgery and Center for Molecular Medicine, Karolinska Institutet and University Hospital, Stockholm, Sweden
| | - T Renné
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Division of Clinical Chemistry, Department of Molecular Medicine and Surgery and Center for Molecular Medicine, Karolinska Institutet and University Hospital, Stockholm, Sweden
| |
Collapse
|
40
|
Kokoye Y, Ivanov I, Cheng Q, Matafonov A, Dickeson SK, Mason S, Sexton DJ, Renné T, McCrae K, Feener EP, Gailani D. A comparison of the effects of factor XII deficiency and prekallikrein deficiency on thrombus formation. Thromb Res 2016; 140:118-124. [PMID: 26950760 DOI: 10.1016/j.thromres.2016.02.020] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Revised: 01/14/2016] [Accepted: 02/16/2016] [Indexed: 11/29/2022]
Abstract
Studies with animal models implicate the plasma proteases factor XIIa (FXIIa) and α-kallikrein in arterial and venous thrombosis. As congenital deficiencies of factor XII (FXII) or prekallikrein (PK), the zymogens of FXIIa and α-kallikrein respectively, do not cause bleeding disorders, inhibition of these enzymes may have therapeutic benefit without compromising hemostasis. The relative contributions of FXIIa and α-kallikrein to thrombosis in animal models are not clear. We compared mice lacking FXII or PK to wild type mice in established models of arterial thrombosis. Wild type mice developed carotid artery occlusion when the vessel was exposed to a 3.5% solution of ferric chloride (FeCl3). FXII-deficient mice were resistant to occlusion at 5% FeCl3 and partially resistant at 10% FeCl3. PK-deficient mice were resistant at 3.5% FeCl3 and partially resistant at 5% FeCl3. Mice lacking high molecular weight kininogen, a cofactor for PK activation and activity, were also partially resistant to thrombosis at 5% FeCl3. Induction of carotid artery thrombosis with Rose Bengal was delayed in FXII-deficient mice compared to wild type or PK-deficient animals. In human plasma supplemented with silica, DNA or collagen to induce contact activation, an antibody to the FXIIa active site was more effective at preventing thrombin generation than an antibody to the α-kallikrein active site. Similarly, the FXIIa antibody was more effective at reducing fibrin formation in human blood flowing through collagen coated-tubes. The findings suggest that inhibitors of FXIIa will have more potent anti-thrombotic effects than inhibitors of α-kallikrein.
Collapse
Affiliation(s)
- Yasin Kokoye
- Department of Pathology, Microbiology and Immunology, Vanderbilt University, Nashville, TN, USA
| | - Ivan Ivanov
- Department of Pathology, Microbiology and Immunology, Vanderbilt University, Nashville, TN, USA
| | - Qiufang Cheng
- Department of Pathology, Microbiology and Immunology, Vanderbilt University, Nashville, TN, USA
| | - Anton Matafonov
- Department of Pathology, Microbiology and Immunology, Vanderbilt University, Nashville, TN, USA; Department of Bioengineering and Organic Chemistry, Tomsk Polytechnic University, Tomsk, Russia
| | - S Kent Dickeson
- Department of Pathology, Microbiology and Immunology, Vanderbilt University, Nashville, TN, USA
| | | | | | - Thomas Renné
- Department of Molecular Medicine and Surgery, Karolinska Institutet and University Hospital, Stockholm, Sweden; Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Keith McCrae
- Department of Hematology and Oncology, Cleveland Clinic, Cleveland, OH, USA
| | - Edward P Feener
- Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - David Gailani
- Department of Pathology, Microbiology and Immunology, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
41
|
De Meyer SF, Denorme F, Langhauser F, Geuss E, Fluri F, Kleinschnitz C. Thromboinflammation in Stroke Brain Damage. Stroke 2016; 47:1165-72. [PMID: 26786115 DOI: 10.1161/strokeaha.115.011238] [Citation(s) in RCA: 216] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 10/26/2015] [Indexed: 02/07/2023]
Affiliation(s)
- Simon F De Meyer
- From the Laboratory for Thrombosis Research, KU Leuven Campus Kulak Kortrijk, Kortrijk, Belgium (S.F.D.M., F.D.); and Department of Neurology, University Clinic of Würzburg, Würzburg, Germany (F.L., E.G., F.F., C.K.).
| | - Frederik Denorme
- From the Laboratory for Thrombosis Research, KU Leuven Campus Kulak Kortrijk, Kortrijk, Belgium (S.F.D.M., F.D.); and Department of Neurology, University Clinic of Würzburg, Würzburg, Germany (F.L., E.G., F.F., C.K.)
| | - Friederike Langhauser
- From the Laboratory for Thrombosis Research, KU Leuven Campus Kulak Kortrijk, Kortrijk, Belgium (S.F.D.M., F.D.); and Department of Neurology, University Clinic of Würzburg, Würzburg, Germany (F.L., E.G., F.F., C.K.)
| | - Eva Geuss
- From the Laboratory for Thrombosis Research, KU Leuven Campus Kulak Kortrijk, Kortrijk, Belgium (S.F.D.M., F.D.); and Department of Neurology, University Clinic of Würzburg, Würzburg, Germany (F.L., E.G., F.F., C.K.)
| | - Felix Fluri
- From the Laboratory for Thrombosis Research, KU Leuven Campus Kulak Kortrijk, Kortrijk, Belgium (S.F.D.M., F.D.); and Department of Neurology, University Clinic of Würzburg, Würzburg, Germany (F.L., E.G., F.F., C.K.)
| | - Christoph Kleinschnitz
- From the Laboratory for Thrombosis Research, KU Leuven Campus Kulak Kortrijk, Kortrijk, Belgium (S.F.D.M., F.D.); and Department of Neurology, University Clinic of Würzburg, Würzburg, Germany (F.L., E.G., F.F., C.K.).
| |
Collapse
|
42
|
Factor XII full and partial null in rat confers robust antithrombotic efficacy with no bleeding. Blood Coagul Fibrinolysis 2015; 26:893-902. [DOI: 10.1097/mbc.0000000000000337] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
43
|
Madhusudhan T, Kerlin BA, Isermann B. The emerging role of coagulation proteases in kidney disease. Nat Rev Nephrol 2015; 12:94-109. [PMID: 26592189 DOI: 10.1038/nrneph.2015.177] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
A role of coagulation proteases in kidney disease beyond their function in normal haemostasis and thrombosis has long been suspected, and studies performed in the past 15 years have provided novel insights into the mechanisms involved. The expression of protease-activated receptors (PARs) in renal cells provides a molecular link between coagulation proteases and renal cell function and revitalizes research evaluating the role of haemostasis regulators in renal disease. Renal cell-specific expression and activity of coagulation proteases, their regulators and their receptors are dynamically altered during disease processes. Furthermore, renal inflammation and tissue remodelling are not only associated, but are causally linked with altered coagulation activation and protease-dependent signalling. Intriguingly, coagulation proteases signal through more than one receptor or induce formation of receptor complexes in a cell-specific manner, emphasizing context specificity. Understanding these cell-specific signalosomes and their regulation in kidney disease is crucial to unravelling the pathophysiological relevance of coagulation regulators in renal disease. In addition, the clinical availability of small molecule targeted anticoagulants as well as the development of PAR antagonists increases the need for in-depth knowledge of the mechanisms through which coagulation proteases might regulate renal physiology.
Collapse
Affiliation(s)
- Thati Madhusudhan
- Institute of Clinical Chemistry and Pathobiochemistry, Medical Faculty, Otto-von-Guericke-University, Magdeburg, Leipziger Strasse 44, Magdeburg D-39120, Germany
| | - Bryce A Kerlin
- Center for Clinical and Translational Research, Nationwide Children's Hospital, 700 Children's Drive, W325 Columbus, Ohio 43205, USA
| | - Berend Isermann
- Institute of Clinical Chemistry and Pathobiochemistry, Medical Faculty, Otto-von-Guericke-University, Magdeburg, Leipziger Strasse 44, Magdeburg D-39120, Germany
| |
Collapse
|
44
|
Deguchi H, Wolfbauer G, Cheung MC, Banerjee Y, Elias DJ, Fernández JA, Albers JJ, Griffin JH. Inhibition of thrombin generation in human plasma by phospholipid transfer protein. Thromb J 2015; 13:24. [PMID: 26185485 PMCID: PMC4504036 DOI: 10.1186/s12959-015-0054-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Accepted: 05/20/2015] [Indexed: 01/10/2023] Open
Abstract
Background Plasma phospholipid transfer protein (PLTP) transfers lipids between donors and acceptors (e.g., from HDL to VLDL) and modulates lipoprotein composition, size, and levels. No study has reported an assessment of the effects of PLTP on blood clotting reactions, such as reflected in thrombin generation assays, or on the association of venous thrombosis (VTE) risk with PLTP activity. Methods The in vitro effects of PLTP on blood coagulation reactions and the correlations between plasma PLTP activity levels and VTE were studied. Results Recombinant (r) PLTP concentration-dependently inhibited plasma thrombin generation and factor XII-dependent kallikrein generation when sulfatide was used to stimulate factor XII autoactivation in plasma. However, rPLTP did not inhibit thrombin generation in plasma induced by factor XIa or tissue factor, implicating an effect of PLTP on contact activation reactions. In purified systems, rPLTP inhibited factor XII autoactivation stimulated by sulfatide in the presence of VLDL. In surface plasmon resonance studies, purified factor XII bound to immobilized rPLTP, implying that rPLTP inhibits factor XII-dependent contact activation by binding factor XII in the presence of lipoproteins. Analysis of plasmas from 40 male patients with unprovoked VTE and 40 matched controls indicated that low PLTP lipid transfer activity (≤25th percentile) was associated with an increased risk of VTE after adjustment for body mass index, plasma lipids, and two known thrombophilic genetic risk factors. Conclusion These data imply that PLTP may be an antithrombotic plasma protein by inhibiting generation of prothrombotic factor XIIa in the presence of VLDL. This newly discovered anticoagulant activity of PLTP merits further clinical and biochemical studies. Electronic supplementary material The online version of this article (doi:10.1186/s12959-015-0054-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Hiroshi Deguchi
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, MEM180, 10550 North Torrey Pines Rd., La Jolla, CA 92037 USA
| | - Gertrud Wolfbauer
- Division of Metabolism, Endocrinology, and Nutrition, Northwest Lipid Metabolism and Diabetes Research Laboratories, Department of Medicine, University of Washington, Seattle, WA 98109 USA
| | - Marian C Cheung
- Division of Metabolism, Endocrinology, and Nutrition, Northwest Lipid Metabolism and Diabetes Research Laboratories, Department of Medicine, University of Washington, Seattle, WA 98109 USA
| | - Yajnavalka Banerjee
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, MEM180, 10550 North Torrey Pines Rd., La Jolla, CA 92037 USA ; Current Address: Department of Biochemistry, College of Medicine and Health Sciences, SQ University, Muscat, Oman
| | - Darlene J Elias
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, MEM180, 10550 North Torrey Pines Rd., La Jolla, CA 92037 USA
| | - José A Fernández
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, MEM180, 10550 North Torrey Pines Rd., La Jolla, CA 92037 USA
| | - John J Albers
- Division of Metabolism, Endocrinology, and Nutrition, Northwest Lipid Metabolism and Diabetes Research Laboratories, Department of Medicine, University of Washington, Seattle, WA 98109 USA
| | - John H Griffin
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, MEM180, 10550 North Torrey Pines Rd., La Jolla, CA 92037 USA
| |
Collapse
|
45
|
FXIa and platelet polyphosphate as therapeutic targets during human blood clotting on collagen/tissue factor surfaces under flow. Blood 2015; 126:1494-502. [PMID: 26136249 DOI: 10.1182/blood-2015-04-641472] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Accepted: 06/18/2015] [Indexed: 11/20/2022] Open
Abstract
Factor XIIa (FXIIa) and factor XIa (FXIa) contribute to thrombosis in animal models, whereas platelet-derived polyphosphate (polyP) may potentiate contact or thrombin-feedback pathways. The significance of these mediators in human blood under thrombotic flow conditions on tissue factor (TF) -bearing surfaces remains inadequately resolved. Human blood (corn trypsin inhibitor treated [4 μg/mL]) was tested by microfluidic assay for clotting on collagen/TF at TF surface concentration ([TF]wall) from ∼0.1 to 2 molecules per μm(2). Anti-FXI antibodies (14E11 and O1A6) or polyP-binding protein (PPXbd) were used to block FXIIa-dependent FXI activation, FXIa-dependent factor IX (FIX) activation, or platelet-derived polyP, respectively. Fibrin formation was sensitive to 14E11 at 0 to 0.1 molecules per µm(2) and sensitive to O1A6 at 0 to 0.2 molecules per µm(2). However, neither antibody reduced fibrin generation at ∼2 molecules per µm(2) when the extrinsic pathway became dominant. Interestingly, PPXbd reduced fibrin generation at low [TF]wall (0.1 molecules per µm(2)) but not at zero or high [TF]wall, suggesting a role for polyP distinct from FXIIa activation and requiring low extrinsic pathway participation. Regardless of [TF]wall, PPXbd enhanced fibrin sensitivity to tissue plasminogen activator and promoted clot retraction during fibrinolysis concomitant with an observed PPXbd-mediated reduction of fibrin fiber diameter. This is the first detection of endogenous polyP function in human blood under thrombotic flow conditions. When triggered by low [TF]wall, thrombosis may be druggable by contact pathway inhibition, although thrombolytic susceptibility may benefit from polyP antagonism regardless of [TF]wall.
Collapse
|
46
|
Dorgalaleh A, Hosseini MS, Mobaraki RN, Alizadeh S, Hosseini S, Tabibian S, Shamsizadeh M, Moghaddam ES, Khosravi S, Rahimizadeh A. Inhibition of factor XIIa, a new approach in management of thrombosis. ANNALS OF TRANSLATIONAL MEDICINE 2015; 3:S20. [PMID: 26046066 DOI: 10.3978/j.issn.2305-5839.2015.02.14] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Accepted: 02/06/2015] [Indexed: 11/14/2022]
Affiliation(s)
- Akbar Dorgalaleh
- 1 Departments of Hematology, School of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran ; 2 Department of Hematology, School of Allied Medicine, Tehran University of Medical Sciences, Tehran, Iran ; 3 School of Nursing and Midwifery, Shahroud University of Medical Sciences, Shahroud, Iran ; 4 Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine and Zahedan Regional Educational Blood Transfusion Center; 5 Hormozgan Institute of Health, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Maryam Sadat Hosseini
- 1 Departments of Hematology, School of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran ; 2 Department of Hematology, School of Allied Medicine, Tehran University of Medical Sciences, Tehran, Iran ; 3 School of Nursing and Midwifery, Shahroud University of Medical Sciences, Shahroud, Iran ; 4 Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine and Zahedan Regional Educational Blood Transfusion Center; 5 Hormozgan Institute of Health, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Rashideh Naseri Mobaraki
- 1 Departments of Hematology, School of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran ; 2 Department of Hematology, School of Allied Medicine, Tehran University of Medical Sciences, Tehran, Iran ; 3 School of Nursing and Midwifery, Shahroud University of Medical Sciences, Shahroud, Iran ; 4 Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine and Zahedan Regional Educational Blood Transfusion Center; 5 Hormozgan Institute of Health, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Shaban Alizadeh
- 1 Departments of Hematology, School of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran ; 2 Department of Hematology, School of Allied Medicine, Tehran University of Medical Sciences, Tehran, Iran ; 3 School of Nursing and Midwifery, Shahroud University of Medical Sciences, Shahroud, Iran ; 4 Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine and Zahedan Regional Educational Blood Transfusion Center; 5 Hormozgan Institute of Health, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Soudabeh Hosseini
- 1 Departments of Hematology, School of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran ; 2 Department of Hematology, School of Allied Medicine, Tehran University of Medical Sciences, Tehran, Iran ; 3 School of Nursing and Midwifery, Shahroud University of Medical Sciences, Shahroud, Iran ; 4 Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine and Zahedan Regional Educational Blood Transfusion Center; 5 Hormozgan Institute of Health, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Shadi Tabibian
- 1 Departments of Hematology, School of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran ; 2 Department of Hematology, School of Allied Medicine, Tehran University of Medical Sciences, Tehran, Iran ; 3 School of Nursing and Midwifery, Shahroud University of Medical Sciences, Shahroud, Iran ; 4 Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine and Zahedan Regional Educational Blood Transfusion Center; 5 Hormozgan Institute of Health, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Morteza Shamsizadeh
- 1 Departments of Hematology, School of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran ; 2 Department of Hematology, School of Allied Medicine, Tehran University of Medical Sciences, Tehran, Iran ; 3 School of Nursing and Midwifery, Shahroud University of Medical Sciences, Shahroud, Iran ; 4 Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine and Zahedan Regional Educational Blood Transfusion Center; 5 Hormozgan Institute of Health, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Esmaeil Saneei Moghaddam
- 1 Departments of Hematology, School of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran ; 2 Department of Hematology, School of Allied Medicine, Tehran University of Medical Sciences, Tehran, Iran ; 3 School of Nursing and Midwifery, Shahroud University of Medical Sciences, Shahroud, Iran ; 4 Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine and Zahedan Regional Educational Blood Transfusion Center; 5 Hormozgan Institute of Health, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Sohila Khosravi
- 1 Departments of Hematology, School of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran ; 2 Department of Hematology, School of Allied Medicine, Tehran University of Medical Sciences, Tehran, Iran ; 3 School of Nursing and Midwifery, Shahroud University of Medical Sciences, Shahroud, Iran ; 4 Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine and Zahedan Regional Educational Blood Transfusion Center; 5 Hormozgan Institute of Health, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Aziz Rahimizadeh
- 1 Departments of Hematology, School of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran ; 2 Department of Hematology, School of Allied Medicine, Tehran University of Medical Sciences, Tehran, Iran ; 3 School of Nursing and Midwifery, Shahroud University of Medical Sciences, Shahroud, Iran ; 4 Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine and Zahedan Regional Educational Blood Transfusion Center; 5 Hormozgan Institute of Health, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| |
Collapse
|
47
|
Labberton L, Kenne E, Renné T. New agents for thromboprotection. A role for factor XII and XIIa inhibition. Hamostaseologie 2015; 35:338-50. [PMID: 25609114 DOI: 10.5482/hamo-14-11-0060] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2014] [Accepted: 01/13/2015] [Indexed: 11/05/2022] Open
Abstract
Blood coagulation is essential for hemostasis, however excessive coagulation can lead to thrombosis. Factor XII starts the intrinsic coagulation pathway and contact-induced factor XII activation provides the mechanistic basis for the diagnostic aPTT clotting assay. Despite its function for fibrin formation in test tubes, patients and animals lacking factor XII have a completely normal hemostasis. The lack of a bleeding tendency observed in factor XII deficiency states is in sharp contrast to deficiencies of other components of the coagulation cascade and factor XII has been considered to have no function for coagulation in vivo. Recently, experimental animal models showed that factor XII is activated by an inorganic polymer, polyphosphate, which is released from procoagulant platelets and that polyphosphate-driven factor XII activation has an essential role in pathologic thrombus formation. Cumulatively, the data suggest to target polyphosphate, factor XII, or its activated form factor XIIa for anticoagulation. As the factor XII pathway specifically contributes to thrombosis but not to hemostasis, interference with this pathway provides a unique opportunity for safe anticoagulation that is not associated with excess bleeding. The review summarizes current knowledge on factor XII functions, activators and inhibitors.
Collapse
Affiliation(s)
| | | | - T Renné
- Thomas Renné, M.D. Ph.D., Clinical Chemistry, Department of Molecular Medicine and Surgery, Karolinska Institutet, Karolinska University Hospital Solna (L2:05), 171 76 Stockholm, Sweden, Tel. +46/8/51 77 33 90, +49/(0)40/741 05 89 84, Fax +46/31 03 76, +49/(0)40/741 05 75 76, E-mail:
| |
Collapse
|