1
|
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
|
2
|
Sedzro JC, Smith SA, Scott A, Wang Y, Travers RJ, Hemp R, Morse CN, Morrissey JH. Antipolyphosphate monoclonal antibodies derived from autoimmune mice. Res Pract Thromb Haemost 2024; 8:102550. [PMID: 39309228 PMCID: PMC11414566 DOI: 10.1016/j.rpth.2024.102550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Revised: 07/29/2024] [Accepted: 08/09/2024] [Indexed: 09/25/2024] Open
Abstract
Background Inorganic polyphosphates (polyPs) are linear chains of phosphates that accelerate blood clotting. Targeting polyP in vivo has been shown to reduce thrombosis. Objectives To identify and characterize anti-polyP monoclonal antibodies that could be used as analytical tools and as antithrombotic agents. Methods Hybridomas were prepared from spleen cells from autoimmune NZBWF1/J female mice and screened for anti-polyP antibodies. Antibodies that bound polyP using enzyme-linked immunosorbent assay and pull-down assays were further characterized with plate binding, surface plasmon resonance, and plasma-based clotting assays. Antithrombotic potential was evaluated in a murine ferric chloride-induced carotid artery thrombosis model. Results Of 4 antibodies that bound polyP in our pull-down assay, 2 (PP2069 and PP2099) were available for further characterization. While analyzing these anti-polyP antibodies, we found secretory leukocyte peptidase inhibitor (SLPI) to be a common contaminant of these antibodies and that SLPI binds polyP. We removed SLPI quantitatively from our purified immunoglobulin G. Both PP2069 and PP2099 immunoglobulin G displayed high affinity for polyP but also bound to other polyanions such as DNA, heparin, and certain other glycosaminoglycans, indicating limited specificity. Both antibodies inhibited polyP-initiated plasma clotting in vitro. When tested in vivo in a mouse thrombosis model, however, neither PP2069 nor PP2099 exhibited a significant antithrombotic effect. Conclusion Autoimmune mice spontaneously produce antibodies against polyP. The 2 examples of anti-polyP monoclonal antibodies studied here not only bound to polyP with high affinity but also cross-reacted with DNA and heparin. Neither antibody protected against thrombosis in a mouse model, but they might have some utility for in vitro studies of polyP.
Collapse
Affiliation(s)
- Josepha C. Sedzro
- Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Stephanie A. Smith
- Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Alexander Scott
- Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Yuqi Wang
- Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Richard J. Travers
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Rachel Hemp
- Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Chase N. Morse
- Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - James H. Morrissey
- Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, Michigan, USA
| |
Collapse
|
3
|
Hambardikar V, Akosah YA, Scoma ER, Guitart-Mampel M, Urquiza P, Da Costa RT, Perez MM, Riggs LM, Patel R, Solesio ME. Toolkit for cellular studies of mammalian mitochondrial inorganic polyphosphate. Front Cell Dev Biol 2023; 11:1302585. [PMID: 38161329 PMCID: PMC10755588 DOI: 10.3389/fcell.2023.1302585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 11/28/2023] [Indexed: 01/03/2024] Open
Abstract
Introduction: Inorganic polyphosphate (polyP) is an ancient polymer which is extremely well-conserved throughout evolution, and found in every studied organism. PolyP is composed of orthophosphates linked together by high-energy bonds, similar to those found in ATP. The metabolism and the functions of polyP in prokaryotes and simple eukaryotes are well understood. However, little is known about its physiological roles in mammalian cells, mostly due to its unknown metabolism and lack of systematic methods and effective models for the study of polyP in these organisms. Methods: Here, we present a comprehensive set of genetically modified cellular models to study mammalian polyP. Specifically, we focus our studies on mitochondrial polyP, as previous studies have shown the potent regulatory role of mammalian polyP in the organelle, including bioenergetics, via mechanisms that are not yet fully understood. Results: Using SH-SY5Y cells, our results show that the enzymatic depletion of mitochondrial polyP affects the expression of genes involved in the maintenance of mitochondrial physiology, as well as the structure of the organelle. Furthermore, this depletion has deleterious effects on mitochondrial respiration, an effect that is dependent on the length of polyP. Our results also show that the depletion of mammalian polyP in other subcellular locations induces significant changes in gene expression and bioenergetics; as well as that SH-SY5Y cells are not viable when the amount and/or the length of polyP are increased in mitochondria. Discussion: Our findings expand on the crucial role of polyP in mammalian mitochondrial physiology and place our cell lines as a valid model to increase our knowledge of both mammalian polyP and mitochondrial physiology.
Collapse
Affiliation(s)
- Vedangi Hambardikar
- Department of Biology, and Center for Computational and Integrative Biology (CCIB), Rutgers University, Camden, NJ, United States
| | - Yaw A. Akosah
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York City, NY, United States
| | - Ernest R. Scoma
- Department of Biology, and Center for Computational and Integrative Biology (CCIB), Rutgers University, Camden, NJ, United States
| | - Mariona Guitart-Mampel
- Department of Biology, and Center for Computational and Integrative Biology (CCIB), Rutgers University, Camden, NJ, United States
| | - Pedro Urquiza
- Department of Biology, and Center for Computational and Integrative Biology (CCIB), Rutgers University, Camden, NJ, United States
| | - Renata T. Da Costa
- Department of Biology, and Center for Computational and Integrative Biology (CCIB), Rutgers University, Camden, NJ, United States
| | - Matheus M. Perez
- Department of Biology, and Center for Computational and Integrative Biology (CCIB), Rutgers University, Camden, NJ, United States
| | - Lindsey M. Riggs
- Department of Biology, and Center for Computational and Integrative Biology (CCIB), Rutgers University, Camden, NJ, United States
| | - Rajesh Patel
- Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ, United States
| | - Maria E. Solesio
- Department of Biology, and Center for Computational and Integrative Biology (CCIB), Rutgers University, Camden, NJ, United States
| |
Collapse
|
4
|
Borghi F, Saiardi A. Evolutionary perspective on mammalian inorganic polyphosphate (polyP) biology. Biochem Soc Trans 2023; 51:1947-1956. [PMID: 37844192 PMCID: PMC10657179 DOI: 10.1042/bst20230483] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 10/06/2023] [Accepted: 10/10/2023] [Indexed: 10/18/2023]
Abstract
Inorganic polyphosphate (polyP), the polymeric form of phosphate, is attracting ever-growing attention due to the many functions it appears to perform within mammalian cells. This essay does not aim to systematically review the copious mammalian polyP literature. Instead, we examined polyP synthesis and functions in various microorganisms and used an evolutionary perspective to theorise key issues of this field and propose solutions. By highlighting the presence of VTC4 in distinct species of very divergent eucaryote clades (Opisthokonta, Viridiplantae, Discoba, and the SAR), we propose that whilst polyP synthesising machinery was present in the ancestral eukaryote, most lineages subsequently lost it during evolution. The analysis of the bacteria-acquired amoeba PPK1 and its unique polyP physiology suggests that eukaryote cells must have developed mechanisms to limit cytosolic polyP accumulation. We reviewed the literature on polyP in the mitochondria from the perspective of its endosymbiotic origin from bacteria, highlighting how mitochondria could possess a polyP physiology reminiscent of their 'bacterial' beginning that is not yet investigated. Finally, we emphasised the similarities that the anionic polyP shares with the better-understood negatively charged polymers DNA and RNA, postulating that the nucleus offers an ideal environment where polyP physiology might thrive.
Collapse
Affiliation(s)
- Filipy Borghi
- Laboratory for Molecular Cell Biology, University College London, London WC1E 6BT, U.K
| | - Adolfo Saiardi
- Laboratory for Molecular Cell Biology, University College London, London WC1E 6BT, U.K
| |
Collapse
|
5
|
Liu W, Wang J, Comte‐Miserez V, Zhang M, Yu X, Chen Q, Jessen HJ, Mayer A, Wu S, Ye S. Cryo-EM structure of the polyphosphate polymerase VTC reveals coupling of polymer synthesis to membrane transit. EMBO J 2023; 42:e113320. [PMID: 37066886 PMCID: PMC10183816 DOI: 10.15252/embj.2022113320] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Revised: 03/18/2023] [Accepted: 03/27/2023] [Indexed: 04/18/2023] Open
Abstract
The eukaryotic vacuolar transporter chaperone (VTC) complex acts as a polyphosphate (polyP) polymerase that synthesizes polyP from adenosine triphosphate (ATP) and translocates polyP across the vacuolar membrane to maintain an intracellular phosphate (Pi ) homeostasis. To discover how the VTC complex performs its function, we determined a cryo-electron microscopy structure of an endogenous VTC complex (Vtc4/Vtc3/Vtc1) purified from Saccharomyces cerevisiae at 3.1 Å resolution. The structure reveals a heteropentameric architecture of one Vtc4, one Vtc3, and three Vtc1 subunits. The transmembrane region forms a polyP-selective channel, likely adopting a resting state conformation, in which a latch-like, horizontal helix of Vtc4 limits the entrance. The catalytic Vtc4 central domain is located on top of the pseudo-symmetric polyP channel, creating a strongly electropositive pathway for nascent polyP that can couple synthesis to translocation. The SPX domain of the catalytic Vtc4 subunit positively regulates polyP synthesis by the VTC complex. The noncatalytic Vtc3 regulates VTC through a phosphorylatable loop. Our findings, along with the functional data, allow us to propose a mechanism of polyP channel gating and VTC complex activation.
Collapse
Affiliation(s)
- Wei Liu
- Frontiers Science Center for Synthetic Biology (Ministry of Education), Tianjin Key Laboratory of Function and Application of Biological Macromolecular Structures, School of Life SciencesTianjin UniversityTianjinChina
| | - Jiening Wang
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Collaborative Innovation Center for Green Transformation of Bio‐Resources, Hubei Key Laboratory of Industrial Biotechnology, School of Life SciencesHubei UniversityWuhanChina
| | | | - Mengyu Zhang
- Frontiers Science Center for Synthetic Biology (Ministry of Education), Tianjin Key Laboratory of Function and Application of Biological Macromolecular Structures, School of Life SciencesTianjin UniversityTianjinChina
| | - Xuejing Yu
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Collaborative Innovation Center for Green Transformation of Bio‐Resources, Hubei Key Laboratory of Industrial Biotechnology, School of Life SciencesHubei UniversityWuhanChina
| | - Qingfeng Chen
- School of Life SciencesYunnan UniversityKunmingChina
| | - Henning Jacob Jessen
- Institute of Organic ChemistryUniversity of FreiburgFreiburgGermany
- CIBSS – Centre for Integrative Biological Signalling StudiesUniversity of FreiburgFreiburgGermany
| | - Andreas Mayer
- Département d'ImmunobiologieUniversité de LausanneEpalingesSwitzerland
| | - Shan Wu
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Collaborative Innovation Center for Green Transformation of Bio‐Resources, Hubei Key Laboratory of Industrial Biotechnology, School of Life SciencesHubei UniversityWuhanChina
| | - Sheng Ye
- Frontiers Science Center for Synthetic Biology (Ministry of Education), Tianjin Key Laboratory of Function and Application of Biological Macromolecular Structures, School of Life SciencesTianjin UniversityTianjinChina
- Life Sciences Institute, Zhejiang UniversityHangzhouChina
| |
Collapse
|
6
|
Ben S, Huang X, Shi Y, Xu Z, Xiao H. Change in cytokine profiles released by mast cells mediated by lung cancer-derived exosome activation may contribute to cancer-associated coagulation disorders. Cell Commun Signal 2023; 21:97. [PMID: 37143160 PMCID: PMC10161433 DOI: 10.1186/s12964-023-01110-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 03/22/2023] [Indexed: 05/06/2023] Open
Abstract
BACKGROUND Coagulation disorders are a significant cause of lung cancer mortality. Although mast cells are known to play a role in coagulation abnormalities, their specific role in this process has not yet been elucidated. METHOD We detected mast cells in the tumor microenvironment using single-cell sequencing data and examined their correlation with thrombosis-related genes, neutrophil-related genes, neutrophil extracellular trap-related signature genes, and immune infiltration levels in lung cancer patients through bioinformatics analysis. Bone marrow mast cell uptake of exosomes isolated from the lung adenocarcinoma cell line A549, which were labeled using PKH67, was observed using confocal microscopy. Mast cell degranulation was detected by measuring the β-hexosaminidase release rate. Additionally, cytokine array analysis was performed to identify altered mediators released by bone marrow mast cells after uptake of the exosomes. RESULTS In our study, we found a close correlation between the proportion of mast cells in lung cancer patients and the expression levels of thrombosis-related genes and neutrophil extracellular trap signature genes, both of which play a key role in thrombophilic disorder. Moreover, we discovered that lung cancer cell-derived exosomes can be taken up by mast cells, which in turn become activated to release procoagulant mediators. CONCLUSION Our study shows that exosomes derived from lung cancer cells can activate mast cells to release procoagulants that may contribute to abnormal blood clotting in lung cancer patients. Video Abstract.
Collapse
Affiliation(s)
- Suqin Ben
- Department of Respiratory and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiulin Huang
- Department of Respiratory and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yongxin Shi
- Department of Respiratory and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ziheng Xu
- Department of Respiratory and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hui Xiao
- Department of Respiratory and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
7
|
Konrath S, Mailer RK, Beerens M, Englert H, Frye M, Kuta P, Preston RJS, Maas C, Butler LM, Roest M, de Laat B, Renné T. Intrinsic coagulation pathway-mediated thrombin generation in mouse whole blood. Front Cardiovasc Med 2022; 9:1008410. [PMID: 36518684 PMCID: PMC9742269 DOI: 10.3389/fcvm.2022.1008410] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Accepted: 10/26/2022] [Indexed: 08/27/2023] Open
Abstract
Calibrated Automated Thrombography (CAT) is a versatile and sensitive method for analyzing coagulation reactions culminating in thrombin generation (TG). Here, we present a CAT method for analyzing TG in murine whole blood by adapting the CAT assay used for measuring TG in human plasma. The diagnostically used artificial and physiologic factor XII (FXII) contact activators kaolin, ellagic acid and polyphosphate (polyP) stimulated TG in murine blood in a dose-dependent manner resulting in a gradual increase in endogenous thrombin potential and peak thrombin, with shortened lag times and times to peak. The activated FXII inhibitor rHA-Infestin-4 and direct oral anticoagulants (DOACs) interfered with TG triggered by kaolin, ellagic acid and polyP and TG was completely attenuated in blood of FXII- (F12 -/-) and FXI-deficient (F11 -/-) mice. Moreover, reconstitution of blood from F12 -/- mice with human FXII restored impaired contact-stimulated TG. HEK293 cell-purified polyP also initiated FXII-driven TG in mouse whole blood and addition of the selective inhibitor PPX_Δ12 ablated natural polyP-stimulated TG. In conclusion, the data provide a method for analysis of contact activation-mediated TG in murine whole blood. As the FXII-driven intrinsic pathway of coagulation has emerged as novel target for antithrombotic agents that are validated in mouse thrombosis and bleeding models, our novel assay could expedite therapeutic drug development.
Collapse
Affiliation(s)
- Sandra Konrath
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Reiner K. Mailer
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Manu Beerens
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Hanna Englert
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Maike Frye
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Piotr Kuta
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Roger J. S. Preston
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Coen Maas
- Central Diagnostic Laboratory, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Lynn M. Butler
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of Clinical Medicine, The Arctic University of Norway, Tromsø, Norway
- Clinical Chemistry and Blood Coagulation Research, Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden
| | - Mark Roest
- Department of Platelet Pathophysiology, Synapse Research Institute, Maastricht, Netherlands
| | - Bas de Laat
- Department of Functional Coagulation, Synapse Research Institute, Maastricht, Netherlands
- Department of Data Analysis and Artificial Intelligence, Synapse Research Institute, Maastricht, Netherlands
| | - Thomas Renné
- Institute of Clinical Chemistry and Laboratory Medicine, 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
|
8
|
Montilla M, Liberato A, Ruiz-Ocaña P, Sáez-Benito A, Aguilar-Diosdado M, Lechuga-Sancho AM, Ruiz FA. Proinflammatory Polyphosphate Increases in Plasma of Obese Children with Insulin Resistance and Adults with Severe Type 2 Diabetes. Nutrients 2022; 14:nu14214601. [PMID: 36364861 PMCID: PMC9654964 DOI: 10.3390/nu14214601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 10/25/2022] [Accepted: 10/28/2022] [Indexed: 11/06/2022] Open
Abstract
Obesity increases the risk of insulin resistance and type 2 diabetes through increased inflammation at cellular and tissue levels. Therefore, study of the molecular elements involved in obesity-related inflammation may contribute to preventing and controlling it. Inorganic polyphosphate is a natural phosphate polymer that has recently been attracting more attention for its role in inflammation and hemostasis processes. Polyphosphates are one of the main constituents of human platelets, which are secreted after platelet activation. Among other roles, they interact with multiple proteins of the coagulation cascade, trigger bradykinin release, and inhibit the complement system. Despite its importance, determinations of polyphosphate levels in blood plasma had been elusive until recently, when we developed a method to detect these levels precisely. Here, we perform cross sectional studies to evaluate plasma polyphosphate in: 25 children, most of them with obesity and overweight, and 20 adults, half of them with severe type 2 diabetes. Our results show that polyphosphate increases, in a significant manner, in children with insulin resistance and in type 2 diabetes patients. As we demonstrated before that polyphosphate decreases in healthy overweight individuals, these results suggest that this polymer could be an inflammation biomarker in the metabolic disease onset before diabetes.
Collapse
Affiliation(s)
- Marcela Montilla
- Research Unit, Hospital Universitario Puerta del Mar, 11009 Cadiz, Spain
- Instituto de Investigación e Innovación Biomédica de Cádiz (INiBICA), 11009 Cadiz, Spain
- Medical School, Universidad Cooperativa de Colombia, Villavicencio 500003, Colombia
| | - Andrea Liberato
- Research Unit, Hospital Universitario Puerta del Mar, 11009 Cadiz, Spain
- Instituto de Investigación e Innovación Biomédica de Cádiz (INiBICA), 11009 Cadiz, Spain
| | - Pablo Ruiz-Ocaña
- Pediatric Endocrinology and Diabetes, Department of Pediatrics, Hospital Universitario Puerta del Mar, 11009 Cadiz, Spain
| | - Ana Sáez-Benito
- Instituto de Investigación e Innovación Biomédica de Cádiz (INiBICA), 11009 Cadiz, Spain
- Clinical Analysis Department, Hospital Universitario Puerta del Mar, 11009 Cadiz, Spain
| | - Manuel Aguilar-Diosdado
- Instituto de Investigación e Innovación Biomédica de Cádiz (INiBICA), 11009 Cadiz, Spain
- Endocrinology and Metabolism Department, Hospital Universitario Puerta del Mar, and Universidad de Cádiz, 11009 Cadiz, Spain
| | - Alfonso Maria Lechuga-Sancho
- Research Unit, Hospital Universitario Puerta del Mar, 11009 Cadiz, Spain
- Instituto de Investigación e Innovación Biomédica de Cádiz (INiBICA), 11009 Cadiz, Spain
- Pediatric Endocrinology and Diabetes, Department of Pediatrics, Hospital Universitario Puerta del Mar, 11009 Cadiz, Spain
- Area of Pediatrics, Medical School, Universidad de Cádiz, 11003 Cadiz, Spain
| | - Felix A. Ruiz
- Research Unit, Hospital Universitario Puerta del Mar, 11009 Cadiz, Spain
- Instituto de Investigación e Innovación Biomédica de Cádiz (INiBICA), 11009 Cadiz, Spain
- Area of Nutrition and Bromatology, Medical School, Universidad de Cádiz, 11003 Cadiz, Spain
- Correspondence: ; Tel.: +34-690395217
| |
Collapse
|
9
|
Keppler M, Moser S, Jessen HJ, Held C, Andexer JN. Make or break: the thermodynamic equilibrium of polyphosphate kinase-catalysed reactions. Beilstein J Org Chem 2022; 18:1278-1288. [PMID: 36225726 PMCID: PMC9520863 DOI: 10.3762/bjoc.18.134] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 08/30/2022] [Indexed: 11/23/2022] Open
Abstract
Polyphosphate kinases (PPKs) have become popular biocatalysts for nucleotide 5'-triphosphate (NTP) synthesis and regeneration. Two unrelated families are described: PPK1 and PPK2. They are structurally unrelated and use different catalytic mechanisms. PPK1 enzymes prefer the usage of adenosine 5'-triphosphate (ATP) for polyphosphate (polyP) synthesis while PPK2 enzymes favour the reverse reaction. With the emerging use of PPK enzymes in biosynthesis, a deeper understanding of the enzymes and their thermodynamic reaction course is of need, especially in comparison to other kinases. Here, we tested four PPKs from different organisms under the same conditions without any coupling reactions. In comparison to other kinases using phosphate donors with comparably higher phosphate transfer potentials that are characterised by reaction yields close to full conversion, the PPK-catalysed reaction reaches an equilibrium in which about 30% ADP is left. These results were obtained for PPK1 and PPK2 enzymes, and are supported by theoretical data on the basic reaction. At high concentrations of substrate, the different kinetic preferences of PPK1 and PPK2 can be observed. The implications of these results for the application of PPKs in chemical synthesis and as enzymes for ATP regeneration systems are discussed.
Collapse
Affiliation(s)
- Michael Keppler
- Institute of Pharmaceutical Sciences, University of Freiburg, Albertstr. 25, 79104 Freiburg, Germany
| | - Sandra Moser
- Institute of Organic Chemistry, University of Freiburg, Albertstr. 21, 79104 Freiburg, Germany
| | - Henning J Jessen
- Institute of Organic Chemistry, University of Freiburg, Albertstr. 21, 79104 Freiburg, Germany
| | - Christoph Held
- Department of Biochemical and Chemical Engineering, TU Dortmund University, Emil-Figge-Str. 70, 44227 Dortmund, Germany
| | - Jennifer N Andexer
- Institute of Pharmaceutical Sciences, University of Freiburg, Albertstr. 25, 79104 Freiburg, Germany
| |
Collapse
|
10
|
Abbina S, La CC, Vappala S, Kalathottukaren MT, Abbasi U, Gill A, Smith SA, Haynes CA, Morrissey JH, Kizhakkedathu JN. Influence of Steric Shield on Biocompatibility and Antithrombotic Activity of Dendritic Polyphosphate Inhibitor. Mol Pharm 2022; 19:1853-1865. [PMID: 35500201 DOI: 10.1021/acs.molpharmaceut.1c00934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The polyanion, inorganic polyphosphate (polyP), is a procoagulant molecule which has become a promising therapeutic target in the development of antithrombotics. Neutralizing polyP's prothrombotic activity using polycationic inhibitors is one of the viable strategies to design new polyP inhibitors. However, in this approach, a fine balance between the electrostatic interaction of polyP and the inhibitor is needed. Any unprotected polycations are known to interact with negatively charged blood components, potentially resulting in platelet activation, cellular toxicity, and bleeding. Thus, designing potent polycationic polyP inhibitors with good biocompatibility is a major challenge. Building on our previous research on universal heparin reversal agent (UHRA), we report polyP inhibitors with a modified steric shield design. The molecular weight, number of cationic binding groups, and the length of the polyethylene glycol (PEG) chains were varied to arrive at the desired inhibitor. We studied two different PEG lengths (mPEG-750 versus mPEG-350) on the polyglycerol scaffold and investigated their influence on biocompatibility and polyP neutralization activity. The polyP inhibitor with mPEG-750 brush layer, mPEG750 UHRA-10, showed superior biocompatibility compared to its mPEG-350 analogs by a number of measured parameters without losing its neutralization activity. An increase in cationic binding groups (25 groups in mPEG750 UHRA-8 and 32 in mPEG750 UHRA-10 [HC]) did not alter the neutralization activity, which suggested that the mPEG-750 shield layer provides significant protection of cationic binding groups and thus helps to minimize unwanted nonspecific interactions. Furthermore, these modified polyP inhibitors are highly biocompatible compared to conventional polycations that have been previously used as polyP inhibitors (e.g., PAMAM dendrimers and polyethylenimine). Through this study, we demonstrated the importance of the design of steric shield toward highly biocompatible polyP inhibitors. This approach can be exploited in the design of highly biocompatible macromolecular inhibitors.
Collapse
Affiliation(s)
- Srinivas Abbina
- Department of Pathology and Laboratory Medicine, The University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada.,Centre for Blood Research, Life Science Institute, The University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - Chanel C La
- Centre for Blood Research, Life Science Institute, The University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada.,Department of Chemistry, The University of British Columbia, Vancouver, British Columbia V6T 1Z3 Canada
| | - Sreeparna Vappala
- Department of Pathology and Laboratory Medicine, The University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada.,Centre for Blood Research, Life Science Institute, The University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - Manu Thomas Kalathottukaren
- Department of Pathology and Laboratory Medicine, The University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada.,Centre for Blood Research, Life Science Institute, The University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - Usama Abbasi
- Department of Pathology and Laboratory Medicine, The University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada.,Centre for Blood Research, Life Science Institute, The University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - Arshdeep Gill
- Centre for Blood Research, Life Science Institute, The University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada.,Department of Chemistry, The University of British Columbia, Vancouver, British Columbia V6T 1Z3 Canada
| | - Stephanie A Smith
- Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, Michigan 48109, United States
| | - Charles A Haynes
- Centre for Blood Research, Life Science Institute, The University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada.,Michael Smith Laboratories, Department of Chemical and Biological Engineering, The University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada
| | - James H Morrissey
- Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, Michigan 48109, United States.,Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan 48109, United States
| | - Jayachandran N Kizhakkedathu
- Department of Pathology and Laboratory Medicine, The University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada.,Centre for Blood Research, Life Science Institute, The University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada.,Department of Chemistry, The University of British Columbia, Vancouver, British Columbia V6T 1Z3 Canada.,School of Biomedical Engineering, The University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| |
Collapse
|
11
|
Kazmer ST, Hartel G, Robinson H, Richards RS, Yan K, van Hal SJ, Chan R, Hind A, Bradley D, Zieschang F, Rawle DJ, Le TT, Reid DW, Suhrbier A, Hill MM. Pathophysiological Response to SARS-CoV-2 Infection Detected by Infrared Spectroscopy Enables Rapid and Robust Saliva Screening for COVID-19. Biomedicines 2022; 10:biomedicines10020351. [PMID: 35203562 PMCID: PMC8962262 DOI: 10.3390/biomedicines10020351] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/10/2022] [Accepted: 01/26/2022] [Indexed: 02/04/2023] Open
Abstract
Fourier transform infrared (FTIR) spectroscopy provides a (bio)chemical snapshot of the sample, and was recently used in proof-of-concept cohort studies for COVID-19 saliva screening. However, the biological basis of the proposed technology has not been established. To investigate underlying pathophysiology, we conducted controlled infection experiments on Vero E6 cells in vitro and K18-hACE2 mice in vivo. Potentially infectious culture supernatant or mouse oral lavage samples were treated with ethanol or 75% (v/v) Trizol for attenuated total reflectance (ATR)-FTIR spectroscopy and proteomics, or RT-PCR, respectively. Controlled infection with UV-inactivated SARS-CoV-2 elicited strong biochemical changes in culture supernatant/oral lavage despite a lack of viral replication, determined by RT-PCR or a cell culture infectious dose 50% assay. Nevertheless, SARS-CoV-2 infection induced additional FTIR signals over UV-inactivated SARS-CoV-2 infection in both cell and mouse models, which correspond to aggregated proteins and RNA. Proteomics of mouse oral lavage revealed increased secretion of kallikreins and immune modulatory proteins. Next, we collected saliva from a cohort of human participants (n = 104) and developed a predictive model for COVID-19 using partial least squares discriminant analysis. While high sensitivity of 93.48% was achieved through leave-one-out cross-validation, COVID-19 patients testing negative on follow-up on the day of saliva sampling using RT-PCR was poorly predicted in this model. Importantly, COVID-19 vaccination did not lead to the misclassification of COVID-19 negatives. Finally, meta-analysis revealed that SARS-CoV-2 induced increases in the amide II band in all arms of this study and in recently published cohort studies, indicative of altered β-sheet structures in secreted proteins. In conclusion, this study reveals a consistent secretory pathophysiological response to SARS-CoV-2, as well as a simple, robust method for COVID-19 saliva screening using ATR-FTIR.
Collapse
Affiliation(s)
- Seth T. Kazmer
- Precision & Systems Biomedicine Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia; (S.T.K.); (H.R.); (R.S.R.)
| | - Gunter Hartel
- Biostatistics Unit, QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia;
| | - Harley Robinson
- Precision & Systems Biomedicine Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia; (S.T.K.); (H.R.); (R.S.R.)
| | - Renee S. Richards
- Precision & Systems Biomedicine Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia; (S.T.K.); (H.R.); (R.S.R.)
| | - Kexin Yan
- Inflammation Biology Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia; (K.Y.); (D.J.R.); (T.T.L.); (A.S.)
| | - Sebastiaan J. van Hal
- New South Wales Health Pathology-Royal Prince Alfred Hospital, Camperdown, NSW 2050, Australia; (S.J.v.H.); (R.C.)
| | - Raymond Chan
- New South Wales Health Pathology-Royal Prince Alfred Hospital, Camperdown, NSW 2050, Australia; (S.J.v.H.); (R.C.)
| | - Andrew Hind
- Agilent Technologies Australia, Mulgrave, VIC 3170, Australia; (A.H.); (D.B.); (F.Z.)
| | - David Bradley
- Agilent Technologies Australia, Mulgrave, VIC 3170, Australia; (A.H.); (D.B.); (F.Z.)
| | - Fabian Zieschang
- Agilent Technologies Australia, Mulgrave, VIC 3170, Australia; (A.H.); (D.B.); (F.Z.)
| | - Daniel J. Rawle
- Inflammation Biology Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia; (K.Y.); (D.J.R.); (T.T.L.); (A.S.)
| | - Thuy T. Le
- Inflammation Biology Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia; (K.Y.); (D.J.R.); (T.T.L.); (A.S.)
| | - David W. Reid
- Lung Inflammation & Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia;
- The Prince Charles Hospital, Chermside, QLD 4032, Australia
| | - Andreas Suhrbier
- Inflammation Biology Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia; (K.Y.); (D.J.R.); (T.T.L.); (A.S.)
- Australian Infectious Disease Research Centre, GVN Centre of Excellence, Brisbane, QLD 4029, Australia
| | - Michelle M. Hill
- Precision & Systems Biomedicine Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia; (S.T.K.); (H.R.); (R.S.R.)
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Herston, QLD 4006, Australia
- Correspondence:
| |
Collapse
|
12
|
Polyphosphate expression by cancer cell extracellular vesicles mediates binding of factor XII and contact activation. Blood Adv 2021; 5:4741-4751. [PMID: 34597365 PMCID: PMC8759128 DOI: 10.1182/bloodadvances.2021005116] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 08/13/2021] [Indexed: 01/04/2023] Open
Abstract
Cleaved HK is observed in many patients with cancer, suggesting activation of the contact system. EVs from cancer cell lines or patients with cancer express polyphosphate, bind and activate FXII, and are prothrombotic.
Extracellular vesicles (EV) have been implicated in diverse biological processes, including intracellular communication, transport of nucleic acids, and regulation of vascular function. Levels of EVs are elevated in cancer, and studies suggest that EV may stimulate thrombosis in patients with cancer through expression of tissue factor. However, limited data also implicate EV in the activation of the contact pathway of coagulation through activation of factor XII (FXII) to FXIIa. To better define the ability of EV to initiate contact activation, we compared the ability of EV derived from different cancer cell lines to activate FXII. EV from all cell lines activated FXII, with those derived from pancreatic and lung cancer cell lines demonstrating the most potent activity. Concordant with the activation of FXII, EV induced the cleavage of high molecular weight kininogen (HK) to cleaved kininogen. We also observed that EVs from patients with cancer stimulated FXII activation and HK cleavage. To define the mechanisms of FXII activation by EV, EV were treated with calf intestinal alkaline phosphatase or Escherichia coli exopolyphosphatase to degrade polyphosphate; this treatment blocked binding of FXII to EVs and the ability of EV to mediate FXII activation. In vivo, EV induced pulmonary thrombosis in wild-type mice, with protection conferred by a deficiency in FXII, HK, or prekallikrein. Moreover, pretreatment of EVs with calf intestinal alkaline phosphatase inhibited their prothrombotic effect. These results indicate that polyphosphate mediates the binding of contact factors to EV and that EV-associated polyphosphate may contribute to the prothrombotic effects of EV in cancer.
Collapse
|
13
|
Mailer RK, Rangaswamy C, Konrath S, Emsley J, Renné T. An update on factor XII-driven vascular inflammation. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2021; 1869:119166. [PMID: 34699874 DOI: 10.1016/j.bbamcr.2021.119166] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 10/01/2021] [Accepted: 10/11/2021] [Indexed: 12/11/2022]
Abstract
The plasma protein factor XII (FXII) is the liver-derived zymogen of the serine protease FXIIa that initiates an array of proteolytic cascades. Zymogen activation, enzymatic FXIIa activity and functions are regulated by interactions with cell receptors, negatively charged surfaces, other serine proteases, and serpin inhibitors, which bind to distinct protein domains and regions in FXII(a). FXII exerts mitogenic activity, while FXIIa initiates the pro-inflammatory kallikrein-kinin pathway and the pro-thrombotic intrinsic coagulation pathway, respectively. Growing evidence indicates that FXIIa-mediated thrombo-inflammation plays a crucial role in various pathological states besides classical thrombosis, such as endothelial dysfunction. Consistently, increased FXIIa levels are associated with hypercholesterolemia and hypertriglyceridemia. In contrast, FXII deficiency protects from thrombosis but is otherwise not associated with prolonged bleeding or other adverse clinical manifestations. Here, we review current concepts for FXII(a)-driven vascular inflammation focusing on endothelial hyperpermeability, receptor signaling, atherosclerosis and immune cell activation.
Collapse
Affiliation(s)
- Reiner K Mailer
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Chandini Rangaswamy
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sandra Konrath
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jonas Emsley
- Centre for Biomolecular Sciences, School of Pharmacy, University of Nottingham, Nottingham, UK
| | - Thomas Renné
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Center for Thrombosis and Hemostasis (CTH), Johannes Gutenberg University Medical Center, Mainz, Germany.
| |
Collapse
|
14
|
Konrath S, Mailer RK, Renné T. Mechanism, Functions, and Diagnostic Relevance of FXII Activation by Foreign Surfaces. Hamostaseologie 2021; 41:489-501. [PMID: 34592776 DOI: 10.1055/a-1528-0499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
Factor XII (FXII) is a serine protease zymogen produced by hepatocytes and secreted into plasma. The highly glycosylated coagulation protein consists of six domains and a proline-rich region that regulate activation and function. Activation of FXII results from a conformational change induced by binding ("contact") with negatively charged surfaces. The activated serine protease FXIIa drives both the proinflammatory kallikrein-kinin pathway and the procoagulant intrinsic coagulation cascade, respectively. Deficiency in FXII is associated with a prolonged activated partial thromboplastin time (aPTT) but not with an increased bleeding tendency. However, genetic or pharmacological deficiency impairs both arterial and venous thrombosis in experimental models. This review summarizes current knowledge of FXII structure, mechanisms of FXII contact activation, and the importance of FXII for diagnostic coagulation testing and thrombosis.
Collapse
Affiliation(s)
- Sandra Konrath
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Reiner K Mailer
- 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
|
15
|
Polyphosphate-induced thrombosis in mice is factor XII dependent and is attenuated by histidine-rich glycoprotein. Blood Adv 2021; 5:3540-3551. [PMID: 34474475 DOI: 10.1182/bloodadvances.2021004567] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 05/20/2021] [Indexed: 12/24/2022] Open
Abstract
Histidine-rich glycoprotein (HRG) is an abundant plasma protein that binds factor XIIa (FXIIa) and inhibits factor XII (FXII) autoactivation and FXIIa-mediated activation of FXI. Polyphosphate (polyP), a potent procoagulant released from activated platelets, may serve as a physiological activator of the contact system. Previously, we showed that HRG binds DNA and neutralizes its procoagulant activity. Consequently, our goal was to determine whether the capacity of HRG to bind polyanions enables it to regulate polyP-induced thrombosis. In a plate-based assay, immobilized polyP bound HRG, FXII, and FXIIa in a zinc-dependent manner. Basal and polyP-induced thrombin generation was greater in plasma from HRG-deficient mice than in plasma from wild-type mice. Intraperitoneal injection of polyP shortened the activated partial thromboplastin time, enhanced thrombin generation, increased thrombin-antithrombin levels, reduced lung perfusion, and promoted pulmonary fibrin deposition to a greater extent in HRG-deficient mice than in wild-type mice, effects that were abrogated with FXII knockdown. HRG thus attenuates the procoagulant and prothrombotic effects of polyP in an FXII-dependent manner by modulating the contact system.
Collapse
|
16
|
Seidel H, Hertfelder HJ, Oldenburg J, Kruppenbacher JP, Afrin LB, Molderings GJ. Effects of Primary Mast Cell Disease on Hemostasis and Erythropoiesis. Int J Mol Sci 2021; 22:ijms22168960. [PMID: 34445665 PMCID: PMC8396658 DOI: 10.3390/ijms22168960] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 08/10/2021] [Accepted: 08/17/2021] [Indexed: 01/21/2023] Open
Abstract
Mast cell disease is an epigenetically and genetically determined disease entity with very diverse clinical manifestations in potentially every system and tissue due to inap pro priate release of variable subsets of mast cell mediators together with accumulation of either morphologically normal or altered mast cells. Easy bruising, excessive bleeding, and aberrancies of erythropoiesis can frequently be observed in patients with mast cell disease. A thorough history, including a family history, will guide the appropriate work-up, and laboratory evaluations may provide clues to diagnosis. In recent years, our understanding of the involvement of coagulation and anticoagulant pathways, the fibrinolytic system, and erythropoiesis in the pathophysiology of mast cell disease has increased considerably. This review summarizes current knowledge of the impact of the disturbed hemostatic and erythropoietic balance in patients with mast cell disease and describes options of treatment.
Collapse
Affiliation(s)
- Holger Seidel
- Center for Bleeding Disorders and Transfusion Medicine (CBT), Am Propsthof 3, D-53121 Bonn, Germany; (H.S.); (H.-J.H.); (J.P.K.)
| | - Hans-Jörg Hertfelder
- Center for Bleeding Disorders and Transfusion Medicine (CBT), Am Propsthof 3, D-53121 Bonn, Germany; (H.S.); (H.-J.H.); (J.P.K.)
- Institute of Experimental Haematology and Transfusion Medicine, University Hospital Bonn, Venusberg-Campus 1, D-53127 Bonn, Germany;
| | - Johannes Oldenburg
- Institute of Experimental Haematology and Transfusion Medicine, University Hospital Bonn, Venusberg-Campus 1, D-53127 Bonn, Germany;
| | - Johannes P. Kruppenbacher
- Center for Bleeding Disorders and Transfusion Medicine (CBT), Am Propsthof 3, D-53121 Bonn, Germany; (H.S.); (H.-J.H.); (J.P.K.)
| | - Lawrence B. Afrin
- Department of Mast Cell Studies, AIM Center for Personalized Medicine, 3010 Westchester Ave Suite 404, Purchase, NY 10577, USA;
| | - Gerhard J. Molderings
- Institute of Human Genetics, University Hospital of Bonn, Venusberg-Campus 1, D-53127 Bonn, Germany
- Correspondence: ; Tel.: +49-228-287-51000
| |
Collapse
|
17
|
Xenotropic and polytropic retrovirus receptor 1 regulates procoagulant platelet polyphosphate. Blood 2021; 137:1392-1405. [PMID: 32932519 DOI: 10.1182/blood.2019004617] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 08/25/2020] [Indexed: 12/15/2022] Open
Abstract
Polyphosphate is a procoagulant inorganic polymer of linear-linked orthophosphate residues. Multiple investigations have established the importance of platelet polyphosphate in blood coagulation; however, the mechanistic details of polyphosphate homeostasis in mammalian species remain largely undefined. In this study, xenotropic and polytropic retrovirus receptor 1 (XPR1) regulated polyphosphate in platelets and was implicated in thrombosis in vivo. We used bioinformatic analyses of omics data to identify XPR1 as a major phosphate transporter in platelets. XPR1 messenger RNA and protein expression inversely correlated with intracellular polyphosphate content and release. Pharmacological interference with XPR1 activity increased polyphosphate stores, led to enhanced platelet-driven coagulation, and amplified thrombus formation under flow via the polyphosphate/factor XII pathway. Conditional gene deletion of Xpr1 in platelets resulted in polyphosphate accumulation, accelerated arterial thrombosis, and augmented activated platelet-driven pulmonary embolism without increasing bleeding in mice. These data identify platelet XPR1 as an integral regulator of platelet polyphosphate metabolism and reveal a fundamental role for phosphate homeostasis in thrombosis.
Collapse
|
18
|
Fluorometric Quantification of Human Platelet Polyphosphate Using 4',6-Diamidine-2-phenylindole Dihydrochloride: Applications in the Japanese Population. Int J Mol Sci 2021; 22:ijms22147257. [PMID: 34298874 PMCID: PMC8307652 DOI: 10.3390/ijms22147257] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 06/29/2021] [Accepted: 07/01/2021] [Indexed: 02/07/2023] Open
Abstract
Polyphosphate (polyP), a biopolymer of inorganic phosphate, is widely distributed in living organisms. In platelets, polyP is released upon activation and plays important roles in coagulation and tissue regeneration. However, the lack of a specific quantification method has delayed the in-depth study of polyP. The fluorescent dye 4′,6-diamidine-2-phenylindole dihydrochloride (DAPI) has recently received attention as a promising probe for the visualization and quantification of cellular polyP levels. In this study, we further optimized quantification conditions and applied this protocol in quantification of platelet polyP levels in a Japanese population. Blood samples were collected from non-smoking, healthy Japanese subjects (23 males, 23 females). Washed platelets were fixed and probed with DAPI for fluorometric determination. PolyP levels per platelet count were significantly higher in women than that in men. A moderate negative correlation between age and polyP levels was found in women. Responsiveness to CaCl2 stimulation was also significantly higher in women than that in men. Overall, our optimized protocol requires neither purification nor degradation steps, reducing both the time and bias for reproducible quantification. Thus, we suggest that despite its low specificity, this DAPI-based protocol would be useful in routine laboratory testing to quantify platelet polyP levels efficiently and economically.
Collapse
|
19
|
Ohkubo YZ, Madsen JJ. Uncovering Membrane-Bound Models of Coagulation Factors by Combined Experimental and Computational Approaches. Thromb Haemost 2021; 121:1122-1137. [PMID: 34214998 PMCID: PMC8432591 DOI: 10.1055/s-0040-1722187] [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] [Indexed: 11/19/2022]
Abstract
In the life sciences, including hemostasis and thrombosis, methods of structural biology have become indispensable tools for shedding light on underlying mechanisms that govern complex biological processes. Advancements of the relatively young field of computational biology have matured to a point where it is increasingly recognized as trustworthy and useful, in part due to their high space–time resolution that is unparalleled by most experimental techniques to date. In concert with biochemical and biophysical approaches, computational studies have therefore proven time and again in recent years to be key assets in building or suggesting structural models for membrane-bound forms of coagulation factors and their supramolecular complexes on membrane surfaces where they are activated. Such endeavors and the proposed models arising from them are of fundamental importance in describing and understanding the molecular basis of hemostasis under both health and disease conditions. We summarize the body of work done in this important area of research to drive forward both experimental and computational studies toward new discoveries and potential future therapeutic strategies.
Collapse
Affiliation(s)
- Y Zenmei Ohkubo
- Department of Bioinformatics, School of Life and Natural Sciences, Abdullah Gül University, Kayseri, Turkey
| | - Jesper J Madsen
- Global and Planetary Health, College of Public Health, University of South Florida, Tampa, Florida, United States
| |
Collapse
|
20
|
Douxfils J, Favresse J, Dogné JM, Lecompte T, Susen S, Cordonnier C, Lebreton A, Gosselin R, Sié P, Pernod G, Gruel Y, Nguyen P, Vayne C, Mullier F. Hypotheses behind the very rare cases of thrombosis with thrombocytopenia syndrome after SARS-CoV-2 vaccination. Thromb Res 2021; 203:163-171. [PMID: 34029848 PMCID: PMC8123522 DOI: 10.1016/j.thromres.2021.05.010] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 05/07/2021] [Accepted: 05/08/2021] [Indexed: 01/17/2023]
Abstract
As of 4 April 2021, a total of 169 cases of cerebral venous sinus thrombosis (CVST) and 53 cases of splanchnic vein thrombosis were reported to EudraVigilance among around 34 million people vaccinated in the European Economic Area and United Kingdom with COVID-19 Vaccine AstraZeneca, a chimpanzee adenoviral vector (ChAdOx1) encoding the spike protein antigen of the SARS-CoV-2 virus. The first report of the European Medicines Agency gathering data on 20 million people vaccinated with Vaxzevria® in the UK and the EEA concluded that the number of post-vaccination cases with thromboembolic events as a whole reported to EudraVigilance in relation to the number of people vaccinated was lower than the estimated rate of such events in the general population. However, the EMA's Pharmacovigilance Risk Assessment Committee concluded that unusual thromboses with low blood platelets should be listed as very rare side effects of Vaxzevria®, pointing to a possible link. The same issue was identified with the COVID-19 Vaccine Janssen (Ad26.COV2.S). Currently, there is still a sharp contrast between the clinical or experimental data reported in the literature on COVID-19 and the scarcity of data on the unusual thrombotic events observed after the vaccination with these vaccines. Different hypotheses might support these observations and should trigger further in vitro and ex vivo investigations. Specialized studies were needed to fully understand the potential relationship between vaccination and possible risk factors in order to implement risk minimization strategies.
Collapse
Affiliation(s)
- Jonathan Douxfils
- University of Namur, Department of Pharmacy, Namur Research for Life Sciences, Namur Thrombosis and Hemostasis Center, Namur, Belgium; QUALIblood s.a., Namur, Belgium.
| | - Julien Favresse
- University of Namur, Department of Pharmacy, Namur Research for Life Sciences, Namur Thrombosis and Hemostasis Center, Namur, Belgium; Clinique Saint-Luc Bouge, Department of Laboratory Medicine, Bouge, Belgium
| | - Jean-Michel Dogné
- University of Namur, Department of Pharmacy, Namur Research for Life Sciences, Namur Thrombosis and Hemostasis Center, Namur, Belgium
| | - Thomas Lecompte
- Départements de Médecine, Hôpitaux Universitaires de Genève, service d'angiologie et d'hémostase et Faculté de Médecine, Geneva Platelet Group (GpG), Université de Genève, Geneva, Switzerland
| | - Sophie Susen
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011- EGID, F-59000 Lille, France
| | - Charlotte Cordonnier
- Univ Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience & Cognition, F-59000 Lille, France
| | - Aurélien Lebreton
- Service d'hématologie biologique, CHU Clermont-Ferrand, Clermont-Ferrand, France
| | - Robert Gosselin
- University of California, Davis Health System, Thrombosis and Hemostasis Center, Sacramento, United States
| | - Pierre Sié
- University Paul Sabatier, CHU of Toulouse, Laboratory of Hematology, F-31069 Toulouse, France
| | - Gilles Pernod
- CHU Grenoble Alpes, Department of Vascular Medicine, CNRS/TIMC-IMAG UMR 5525/Themas, Grenoble, France
| | - Yves Gruel
- University of Tours, EA7501 GICC, CHRU de Tours, Department of Haemostasis, Tours, France
| | | | - Caroline Vayne
- University of Tours, EA7501 GICC, CHRU de Tours, Department of Haemostasis, Tours, France
| | - François Mullier
- CHU UCL Namur, Université catholique de Louvain, Hematology Laboratory, Namur Research for Life Sciences, Namur Thrombosis and Hemostasis Center, Yvoir, Belgium
| |
Collapse
|
21
|
Abstract
The association between inflammation, infection, and venous thrombosis has long been recognized; yet, only in the last decades have we begun to understand the mechanisms through which the immune and coagulation systems interact and reciprocally regulate one another. These interconnected networks mount an effective response to injury and pathogen invasion, but if unregulated can result in pathological thrombosis and organ damage. Neutrophils, monocytes, and platelets interact with each other and the endothelium in host defense and also play critical roles in the formation of venous thromboembolism. This knowledge has advanced our understanding of both human physiology and pathophysiology, as well as identified mechanisms of anticoagulant resistance and novel therapeutic targets for the prevention and treatment of thrombosis. In this review, we discuss the contributions of inflammation and infection to venous thromboembolism.
Collapse
Affiliation(s)
- Meaghan E. Colling
- Hematology Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
- Clinical Division of Haematology and Haemostaseology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Benjamin E. Tourdot
- Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Yogendra Kanthi
- Laboratory of Vascular Thrombosis and Inflammation, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
22
|
Baijal K, Downey M. The promises of lysine polyphosphorylation as a regulatory modification in mammals are tempered by conceptual and technical challenges. Bioessays 2021; 43:e2100058. [PMID: 33998006 DOI: 10.1002/bies.202100058] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 04/13/2021] [Accepted: 04/16/2021] [Indexed: 12/17/2022]
Abstract
Polyphosphate (polyP) is a ubiquitous biomolecule thought to be present in all cells on Earth. PolyP is deceivingly simple, consisting of repeated units of inorganic phosphates polymerized in long energy-rich chains. PolyP is involved in diverse functions in mammalian systems-from cell signaling to blood clotting. One exciting avenue of research is a new nonenzymatic post-translational modification, termed lysine polyphosphorylation, wherein polyP chains are covalently attached to lysine residues of target proteins. While the modification was first characterized in budding yeast, recent work has now identified the first human targets. There is significant promise in this area of biomedical research, but a number of technical issues and knowledge gaps present challenges to rapid progress. In this review, the current state of the field is summarized and existing roadblocks related to the study of lysine polyphosphorylation in higher eukaryotes are introduced. It is discussed how limited methods to identify targets of polyphosphorylation are further impacted by low concentration, unknown regulatory enzymes, and sequestration of polyP into compartments in mammalian systems. Furthermore, suggestions on how these obstacles could be addressed or what their physiological relevance may be within mammalian cells are presented.
Collapse
Affiliation(s)
- Kanchi Baijal
- Department of Cellular & Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada.,Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, Ontario, Canada
| | - Michael Downey
- Department of Cellular & Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada.,Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
23
|
Komorowicz E, Balázs N, Tanka-Salamon A, Varga Z, Szabó L, Bóta A, Longstaff C, Kolev K. Size- and charge-dependent modulation of the lytic susceptibility and mechanical stability of fibrin-histone clots by heparin and polyphosphate variants. J Thromb Haemost 2021; 19:1307-1318. [PMID: 33609065 DOI: 10.1111/jth.15258] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 01/21/2021] [Accepted: 01/22/2021] [Indexed: 12/16/2022]
Abstract
BACKGROUND Neutrophil extracellular traps (NETs) containing DNA and histones are expelled from neutrophils in infection and thrombosis. Heparins, anticoagulant polyanions, can neutralize histones with a potential therapeutic advantage in sepsis. Polyphosphates, procoagulant polyanions, are released by platelets and microorganisms. OBJECTIVES To characterize the combined effects of NET components and polyanions on clot structure, mechanical properties and lytic susceptibility. METHODS Scanning electron microscopy, pressure-driven permeation, turbidimetry, and oscillation rheometry were used for the characterization of the structure, viscoelasticity, and kinetics of formation and lysis of fibrin and plasma clots containing histones+/-DNA in combination with unfractionated heparin, its desulfated derivatives, low molecular weight heparin (LMWH), pentasaccharide, and polyphosphates of different sizes. RESULTS Histones and DNA inhibited fibrin lysis by plasmin, but this behavior was not neutralized by negatively charged heparins or short polyphosphates. Rather, fibrin lysis was further inhibited by added polyanions. Histones inhibited plasma clot lysis by tissue plasminogen activator and the response to added heparin was size dependent. Unfractionated heparin, LMWH, and pentasaccharide had no effect, exacerbated, or reversed histone inhibition, respectively. Histones increased the mechanical strength of fibrin, which was exacerbated by smaller heparin and polyphosphate molecules. Histones increased fibrin diameter and pore size of fibrin clots and this effect was neutralized by all heparin variants but enhanced by polyphosphates. CONCLUSIONS Despite their common polyanionic character, heparins and polyphosphates exert distinct effects on fibrin mechanical and fibrinolytic stability. Anti-fibrinolytic effects of histones were more often enhanced by polyanions not counteracted. Careful selection of anti-histone strategies is required if they are to be combined with thrombolytic therapy.
Collapse
Affiliation(s)
- Erzsébet Komorowicz
- Department of Biochemistry, Institute of Biochemistry and Molecular Biology, Semmelweis University, Budapest, Hungary
| | - Nóra Balázs
- Department of Biochemistry, Institute of Biochemistry and Molecular Biology, Semmelweis University, Budapest, Hungary
| | - Anna Tanka-Salamon
- Department of Biochemistry, Institute of Biochemistry and Molecular Biology, Semmelweis University, Budapest, Hungary
| | - Zoltán Varga
- Biological Nanochemistry Research Group, Institute of Materials and Environmental Chemistry, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
| | - László Szabó
- Department of Biochemistry, Institute of Biochemistry and Molecular Biology, Semmelweis University, Budapest, Hungary
| | - Attila Bóta
- Biological Nanochemistry Research Group, Institute of Materials and Environmental Chemistry, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
| | - Colin Longstaff
- Biotherapeutics, Haemostasis Section, National Institute for Biological Standards and Control, South Mimms, UK
| | - Krasimir Kolev
- Department of Biochemistry, Institute of Biochemistry and Molecular Biology, Semmelweis University, Budapest, Hungary
| |
Collapse
|
24
|
Nakamura M, Aizawa H, Kawabata H, Sato A, Watanabe T, Isobe K, Kitamura Y, Tanaka T, Kawase T. Platelet adhesion on commercially pure titanium plates in vitro III: effects of calcium phosphate-blasting on titanium plate biocompatibility. Int J Implant Dent 2020; 6:74. [PMID: 33215329 PMCID: PMC7677422 DOI: 10.1186/s40729-020-00270-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 10/25/2020] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Platelet-rich plasma (PRP) is often used to improve surface biocompatibility. We previously found that platelets rapidly adhere to plain commercially pure titanium (cp-Ti) plates in the absence, but not in the presence, of plasma proteins. To further expand on these findings, in the present study, we switched titanium plates from a plain surface to a rough surface that is blasted with calcium phosphate (CaP) powder and then examined platelet adhesion and activation. METHODS Elemental distribution in CaP-blasted cp-Ti plates was analyzed using energy-dispersive X-ray spectroscopy. PRP samples prepared from anticoagulated blood samples of six healthy, non-smoking adult male donors were loaded on CaP-blasted cp-Ti plates for 1 h and fixed for examination of platelet morphology and visualization of PDGF-B and platelet surface markers (CD62P, CD63) using scanning electron microscopy and fluorescence microscopy. Plain SUS316L stainless steel plates used in injection needles were also examined for comparison. RESULTS Significant amounts of calcium and phosphate were detected on the CaP-blasted cp-Ti surface. Platelets rapidly adhered to this surface, leading to higher activation. Platelets also adhered to the plain stainless surface; however, the levels of adhesion and activation were much lower than those observed on the CaP-blasted cp-Ti plate. CONCLUSIONS The CaP-blasted cp-Ti surface efficiently entraps and activates platelets. Biomolecules released from the activated platelets could be retained by the fibrin matrix on the surface to facilitate regeneration of the surrounding tissues. Thus, PRP immersion could not only eliminate surface air bubbles but also improve the biocompatibility of the implant surface.
Collapse
Affiliation(s)
| | | | | | - Atsushi Sato
- Tokyo Plastic Dental Society, Kita-ku, Tokyo, Japan
| | | | | | | | - Takaaki Tanaka
- Department of Materials Science and Technology, Niigata University, Niigata, Japan
| | - Tomoyuki Kawase
- Division of Oral Bioengineering, Institute of Medicine and Dentistry, Niigata University, Niigata, Japan.
| |
Collapse
|
25
|
Fang C, Schmaier AH. Novel anti-thrombotic mechanisms mediated by Mas receptor as result of balanced activities between the kallikrein/kinin and the renin-angiotensin systems. Pharmacol Res 2020; 160:105096. [PMID: 32712319 PMCID: PMC7378497 DOI: 10.1016/j.phrs.2020.105096] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 07/16/2020] [Accepted: 07/19/2020] [Indexed: 12/17/2022]
Abstract
The risk of thrombosis, a globally growing challenge and a major cause of death, is influenced by various factors in the intravascular coagulation, vessel wall, and cellular systems. Among the contributors to thrombosis, the contact activation system and the kallikrein/kinin system, two overlapping plasma proteolytic systems that are often considered as synonymous, regulate thrombosis from different aspects. On one hand, components of the contact activation system such as factor XII initiates activation of the coagulation proteins promoting thrombus formation on artificial surfaces through factor XI- and possibly prekallikrein-mediated intrinsic coagulation. On the other hand, physiological activation of plasma prekallikrein in the kallikrein/kinin system on endothelial cells liberates bradykinin from associated high-molecular-weight kininogen to stimulate the constitutive bradykinin B2 receptor to generate nitric oxide and prostacyclin to induce vasodilation and counterbalance angiotensin II signaling from the renin-angiotensin system which stimulates vasoconstriction. In addition to vascular tone regulation, this interaction between the kallikrein/kinin and renin-angiotensin systems has a thrombo-regulatory role independent of the contact pathway. At the level of the G-protein coupled receptors of these systems, defective bradykinin signaling due to attenuated bradykinin formation and/or decreased B2 receptor expression, as seen in murine prekallikrein and B2 receptor null mice, respectively, leads to compensatory overexpressed Mas, the receptor for angiotensin-(1-7) of the renin-angiotensin system. Mas stimulation and/or its increased expression contributes to maintaining a healthy vascular homeostasis by generating graded elevation of plasma prostacyclin which reduces thrombosis through two independent pathways: (1) increasing the vasoprotective transcription factor Sirtuin 1 to suppress tissue factor expression, and (2) inhibiting platelet activation. This review will summarize the recent advances in this field that support these understandings. Appreciating these subtle mechanisms help to develop novel anti-thrombotic strategies by targeting the vascular receptors in the renin-angiotensin and the kallikrein/kinin systems to maintain healthy vascular homeostasis.
Collapse
Affiliation(s)
- Chao Fang
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology and the Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan, Hubei, 430030, China.
| | - Alvin H. Schmaier
- Division of Hematology and Oncology, Department of Medicine, University Hospitals Cleveland Medical Center, Case Western Reserve University, Cleveland, OH, 44106, USA
| |
Collapse
|
26
|
Campos J, Brill A. The role of bone marrow-derived cells in venous thromboembolism. Int J Biochem Cell Biol 2020; 128:105850. [PMID: 32950686 PMCID: PMC7607213 DOI: 10.1016/j.biocel.2020.105850] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 09/03/2020] [Accepted: 09/14/2020] [Indexed: 12/22/2022]
Abstract
Venous thrombosis is a life-threatening condition with high morbidity and mortality. Abnormal functioning of different cells in the blood is an integral part of its pathogenesis. In this review, we describe the contribution of bone marrow-derived cells to the development of this debilitating disease. We present both epidemiological and clinical data demonstrating involvement of various cell types in venous thrombosis, and discuss potential mechanisms underlying these effects. Modern concepts including recently discovered new paradigms in thrombosis, such as neutrophil extracellular traps, mast cells, and polyphosphate, are summarized.
Collapse
Affiliation(s)
- Joana Campos
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Alexander Brill
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK; Department of Pathophysiology, Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia.
| |
Collapse
|
27
|
Abstract
Platelet-derived extracellular polyphosphate (PolyP) is a major mediator of thrombosis. PolyP is a linear chain of inorganic phosphate (Pi) and is stored in platelet dense granules. Pi enters cells from the extracellular fluid through phosphate transporters and may be stored as PolyP. Here we show that high extracellular Pi concentration in vitro increases platelet PolyP content, in a manner dependent on phosphate transporters, IP6K and V-type ATPases. The increased PolyP also enhanced PolyP-dependent coagulation in platelet-rich plasma. These data suggest a mechanistic link between hyperphosphatemia, PolyP and enhanced coagulation, which may be important in pathologies such as chronic kidney disease.
Collapse
Affiliation(s)
- Nima Abbasian
- Department of Pharmacology, University of Cambridge, Cambridge, UK
| | - Matthew T Harper
- Department of Pharmacology, University of Cambridge, Cambridge, UK
| |
Collapse
|
28
|
Meini S, Zanichelli A, Sbrojavacca R, Iuri F, Roberts AT, Suffritti C, Tascini C. Understanding the Pathophysiology of COVID-19: Could the Contact System Be the Key? Front Immunol 2020; 11:2014. [PMID: 32849666 PMCID: PMC7432138 DOI: 10.3389/fimmu.2020.02014] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 07/24/2020] [Indexed: 12/20/2022] Open
Abstract
To date the pathophysiology of COVID-19 remains unclear: this represents a factor determining the current lack of effective treatments. In this paper, we hypothesized a complex host response to SARS-CoV-2, with the Contact System (CS) playing a pivotal role in innate immune response. CS is linked with different proteolytic defense systems operating in human vasculature: the Kallikrein–Kinin (KKS), the Coagulation/Fibrinolysis and the Renin–Angiotensin (RAS) Systems. We investigated the role of the mediators involved. CS consists of Factor XII (FXII) and plasma prekallikrein (complexed to high-molecular-weight kininogen-HK). Autoactivation of FXII by contact with SARS-CoV-2 could lead to activation of intrinsic coagulation, with fibrin formation (microthrombosis), and fibrinolysis, resulting in increased D-dimer levels. Activation of kallikrein by activated FXII leads to production of bradykinin (BK) from HK. BK binds to B2-receptors, mediating vascular permeability, vasodilation and edema. B1-receptors, binding the metabolite [des-Arg9]-BK (DABK), are up-regulated during infections and mediate lung inflammatory responses. BK could play a relevant role in COVID-19 as already described for other viral models. Angiotensin-Converting-Enzyme (ACE) 2 displays lung protective effects: it inactivates DABK and converts Angiotensin II (Ang II) into Angiotensin-(1-7) and Angiotensin I into Angiotensin-(1-9). SARS-CoV-2 binds to ACE2 for cell entry, downregulating it: an impaired DABK inactivation could lead to an enhanced activity of B1-receptors, and the accumulation of Ang II, through a negative feedback loop, may result in decreased ACE activity, with consequent increase of BK. Therapies targeting the CS, the KKS and action of BK could be effective for the treatment of COVID-19.
Collapse
Affiliation(s)
- Simone Meini
- Internal Medicine Unit, Azienda USL Toscana Centro, Santa Maria Annunziata Hospital, Florence, Italy
| | - Andrea Zanichelli
- General Medicine Unit, ASST Fatebenefratelli Sacco, Ospedale Luigi Sacco-Università degli Studi di Milano, Milan, Italy
| | - Rodolfo Sbrojavacca
- Infectious Diseases Clinic, Santa Maria Misericordia Hospital, Università degli Studi di Udine, Udine, Italy
| | - Federico Iuri
- Department of Emergency, Santa Maria Misericordia Hospital, Università degli Studi di Udine, Udine, Italy
| | | | - Chiara Suffritti
- General Medicine Unit, ASST Fatebenefratelli Sacco, Ospedale Luigi Sacco-Università degli Studi di Milano, Milan, Italy
| | - Carlo Tascini
- Infectious Diseases Clinic, Santa Maria Misericordia Hospital, Università degli Studi di Udine, Udine, Italy
| |
Collapse
|
29
|
Dupuis A, Bordet JC, Eckly A, Gachet C. Platelet δ-Storage Pool Disease: An Update. J Clin Med 2020; 9:jcm9082508. [PMID: 32759727 PMCID: PMC7466064 DOI: 10.3390/jcm9082508] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 07/21/2020] [Accepted: 07/28/2020] [Indexed: 12/15/2022] Open
Abstract
Platelet dense-granules are small organelles specific to the platelet lineage that contain small molecules (calcium, adenyl nucleotides, serotonin) and are essential for the activation of blood platelets prior to their aggregation in the event of a vascular injury. Delta-storage pool diseases (δ-SPDs) are platelet pathologies leading to hemorrhagic syndromes of variable severity and related to a qualitative (content) or quantitative (numerical) deficiency in dense-granules. These pathologies appear in a syndromic or non-syndromic form. The syndromic forms (Chediak–Higashi disease, Hermansky–Pudlak syndromes), whose causative genes are known, associate immune deficiencies and/or oculocutaneous albinism with a platelet function disorder (PFD). The non-syndromic forms correspond to an isolated PFD, but the genes responsible for the pathology are not yet known. The diagnosis of these pathologies is complex and poorly standardized. It is based on orientation tests performed by light transmission aggregometry or flow cytometry, which are supplemented by complementary tests based on the quantification of platelet dense-granules by electron microscopy using the whole platelet mount technique and the direct determination of granule contents (ADP/ATP and serotonin). The objective of this review is to present the state of our knowledge concerning platelet dense-granules and the tools available for the diagnosis of different forms of δ-SPD.
Collapse
Affiliation(s)
- Arnaud Dupuis
- INSERM, EFS Grand Est, BPPS UMR-S 1255, FMTS, Université de Strasbourg, F-67000 Strasbourg, France; (A.E.); (C.G.)
- Correspondence: ; Tel.: +33-38-821-2506
| | - Jean-Claude Bordet
- Laboratoire D’hématologie, Hospices Civils de Lyon, 59 Bd Pinel, CEDEX, 69677 Bron, France;
| | - Anita Eckly
- INSERM, EFS Grand Est, BPPS UMR-S 1255, FMTS, Université de Strasbourg, F-67000 Strasbourg, France; (A.E.); (C.G.)
| | - Christian Gachet
- INSERM, EFS Grand Est, BPPS UMR-S 1255, FMTS, Université de Strasbourg, F-67000 Strasbourg, France; (A.E.); (C.G.)
| |
Collapse
|
30
|
Kusadasi N, Sikma M, Huisman A, Westerink J, Maas C, Schutgens R. A Pathophysiological Perspective on the SARS-CoV-2 Coagulopathy. Hemasphere 2020; 4:e457. [PMID: 32885147 PMCID: PMC7430228 DOI: 10.1097/hs9.0000000000000457] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 06/22/2020] [Indexed: 01/08/2023] Open
Abstract
Recent evidence is focusing on the presence of a hypercoagulable state with development of both venous and arterial thromboembolic complications in patients infected with SARS-CoV-2. The ongoing activation of coagulation related to the severity of the illness is further characterized by thrombotic microangiopathy and endotheliitis. These microangiopathic changes cannot be classified as classical disseminated intravascular coagulation (DIC). In this short review we describe the interaction between coagulation and inflammation with focus on the possible mechanisms that might be involved in SARS-CoV-2 infection associated coagulopathy in the critically ill.
Collapse
Affiliation(s)
- Nuray Kusadasi
- Department of Intensive Care Medicine, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Maaike Sikma
- Department of Intensive Care Medicine, University Medical Center Utrecht, Utrecht, The Netherlands
- Dutch Poisons Information Center, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Albert Huisman
- Central Diagnostic Laboratory, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Jan Westerink
- Department of Vascular Medicine, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Coen Maas
- Central Diagnostic Laboratory, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Roger Schutgens
- Van Creveldkliniek, Benign Hematology Center, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
31
|
Amiral J. Measurement of blood activation markers applied to the early diagnosis of cardiovascular alterations. Expert Rev Mol Diagn 2019; 20:85-98. [DOI: 10.1080/14737159.2020.1704258] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Jean Amiral
- Scientific-Hemostasis-Consulting, Scientific Director and Consultant in Thrombosis-Hemostasis, Andrésy, France
| |
Collapse
|