1
|
Haynes LM, Holding ML, DiGiovanni HL, Siemieniak D, Ginsburg D. High-throughput amino acid-level characterization of the interactions of plasminogen activator inhibitor-1 with variably divergent proteases. Protein Sci 2025; 34:e70088. [PMID: 40100143 PMCID: PMC11917113 DOI: 10.1002/pro.70088] [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: 09/26/2024] [Revised: 01/09/2025] [Accepted: 02/18/2025] [Indexed: 03/20/2025]
Abstract
While members of large paralogous protein families share structural features, their functional niches often diverge significantly. Serine protease inhibitors (SERPINs), whose members typically function as covalent inhibitors of serine proteases, are one such family. Plasminogen activator inhibitor-1 (PAI-1) is a prototypic SERPIN, which canonically inhibits tissue- and urokinase-type plasminogen activators (tPA and uPA) to regulate fibrinolysis. PAI-1 has been shown to also inhibit other serine proteases, including coagulation factor XIIa (FXIIa) and transmembrane serine protease 2 (TMPRSS2). The structural determinants of PAI-1 inhibitory function toward these non-canonical protease targets, and the biological significance of these functions, are unknown. We applied deep mutational scanning (DMS) to assess the effects of ~80% of all possible single-amino acid substitutions in PAI-1 on its ability to inhibit three putative serine protease targets (uPA, FXIIa, and TMPRSS2). Selection with each target protease generated a unique PAI-1 mutational landscape, with the determinants of protease specificity distributed throughout PAI-1's primary sequence. Next, we conducted a comparative analysis of extant orthologous sequences, demonstrating that key residues modulating PAI-1 inhibition of uPA and FXIIa, but not TMPRSS2, are maintained by purifying selection (also referred to as "negative selection"). PAI-1's activity toward FXIIa may reflect how protease evolutionary relationships predict SERPIN functional divergence, which we support via a cophylogenetic analysis of secreted SERPINs and their cognate serine proteases. This work provides insight into the functional diversification of SERPINs and lays the framework for extending these studies to other proteases and their regulators.
Collapse
Affiliation(s)
- Laura M. Haynes
- Life Sciences InstituteUniversity of MichiganAnn ArborMichiganUSA
| | - Matthew L. Holding
- Life Sciences InstituteUniversity of MichiganAnn ArborMichiganUSA
- Department of Ecology and Evolutionary BiologyUniversity of MichiganAnn ArborMichiganUSA
| | | | - David Siemieniak
- Life Sciences InstituteUniversity of MichiganAnn ArborMichiganUSA
| | - David Ginsburg
- Life Sciences InstituteUniversity of MichiganAnn ArborMichiganUSA
- Department of Internal MedicineUniversity of MichiganAnn ArborMichiganUSA
- Department of Human GeneticsUniversity of MichiganAnn ArborMichiganUSA
- Department of PediatricsUniversity of MichiganAnn ArborMichiganUSA
| |
Collapse
|
2
|
Yang C, Lei C, Jing G, Xia Y, Zhou H, Wu D, Zuo J, Gong H, Wang X, Dong Y, Aidebaike D, Wu X, Song X. Erbin Regulates Tissue Factors Through Ras/Raf Pathway in Coagulation Disorders in Sepsis. J Inflamm Res 2025; 18:1739-1754. [PMID: 39931168 PMCID: PMC11808216 DOI: 10.2147/jir.s493093] [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: 09/16/2024] [Accepted: 01/06/2025] [Indexed: 02/13/2025] Open
Abstract
Background Sepsis, as a clinically critical disease, usually induces coagulation disorders. It has been reported that ERBB2 Interacting Protein (Erbin) is involved in the development of various inflammatory diseases, and macrophages are involved in the regulation of coagulation disorders in sepsis. However, the role of Erbin in coagulation disorders in sepsis and the relationship between Erbin and macrophage regulation of coagulation function are still unclear. Methods At the cellular level, macrophages were treated with lipopolysaccharide (LPS) or MEK inhibitor (PD98059), protein expression levels were detected by Western blot, co-immunoprecipitation (Co-IP), and immunofluorescence, mRNA expression levels were detected by quantitative real-time polymerase chain reaction (qPCR), and the concentration of tissue factor (TF) in cell supernatant was detected by enzyme linked immunosorbent assay (ELISA). At the animal level, the cecal ligation and perforation (CLP) model was constructed in mice, and the inflammatory response and coagulation disorder of mice were observed by hematoxylin-eosin (HE) staining, immunohistochemistry, ELISA, and automatic hemagglutination analyzer. The protein and mRNA expression level were detected by Western blot and qPCR. Pearson linear correlation analysis was used to analyze the correlation between the inflammation index and the coagulation function index. Results We confirmed that the Erbin is involved in the regulation of coagulation function by macrophages and plays a role in the coagulation disorder of sepsis. In vivo studies have shown that mice with Erbin deletion have more obvious enhanced coagulation function, and in vitro studies have shown that Erbin knockout mediated macrophage secretion of TF by activating the Ras/Raf pathway. Conclusion Erbin reduces the coagulation activation by inhibiting TF release from macrophages.
Collapse
Affiliation(s)
- Cheng Yang
- Research Centre of Anesthesiology and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei Province, 430062, People’s Republic of China
| | - Chuntian Lei
- Research Centre of Anesthesiology and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei Province, 430062, People’s Republic of China
| | - Guoqing Jing
- Research Centre of Anesthesiology and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei Province, 430062, People’s Republic of China
| | - Yun Xia
- Research Centre of Anesthesiology and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei Province, 430062, People’s Republic of China
| | - Huimin Zhou
- Research Centre of Anesthesiology and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei Province, 430062, People’s Republic of China
| | - Die Wu
- Research Centre of Anesthesiology and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei Province, 430062, People’s Republic of China
| | - Jing Zuo
- Research Centre of Anesthesiology and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei Province, 430062, People’s Republic of China
| | - Hailong Gong
- Research Centre of Anesthesiology and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei Province, 430062, People’s Republic of China
| | - Xing Wang
- Research Centre of Anesthesiology and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei Province, 430062, People’s Republic of China
| | - Yingyue Dong
- Research Centre of Anesthesiology and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei Province, 430062, People’s Republic of China
| | - Delida Aidebaike
- Research Centre of Anesthesiology and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei Province, 430062, People’s Republic of China
| | - Xiaojing Wu
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, 430060, People’s Republic of China
| | - Xuemin Song
- Research Centre of Anesthesiology and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei Province, 430062, People’s Republic of China
| |
Collapse
|
3
|
Puy C, Moellmer SA, Pang J, Vu HH, Melrose AR, Lorentz CU, Tucker EI, Shatzel JJ, Keshari RS, Lupu F, Gailani D, McCarty OJT. Coagulation factor XI regulates endothelial cell permeability and barrier function in vitro and in vivo. Blood 2024; 144:1821-1833. [PMID: 39158072 PMCID: PMC11830974 DOI: 10.1182/blood.2023022257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 06/26/2024] [Accepted: 07/21/2024] [Indexed: 08/20/2024] Open
Abstract
ABSTRACT Loss of endothelial barrier function contributes to the pathophysiology of many inflammatory diseases. Coagulation factor XI (FXI) plays a regulatory role in inflammation. Although activation of FXI increases vascular permeability in vivo, the mechanism by which FXI or its activated form FXIa disrupts endothelial barrier function is unknown. We investigated the role of FXIa in human umbilical vein endothelial cell (HUVEC) or human aortic endothelial cell (HAEC) permeability. The expression patterns of vascular endothelial (VE)-cadherin and other proteins of interest were examined by western blot or immunofluorescence. Endothelial cell permeability was analyzed by Transwell assay. We demonstrate that FXIa increases endothelial cell permeability by inducing cleavage of the VE-cadherin extracellular domain, releasing a soluble fragment. The activation of a disintegrin and metalloproteinase 10 (ADAM10) mediates the FXIa-dependent cleavage of VE-cadherin, because adding an ADAM10 inhibitor prevented the cleavage of VE-cadherin induced by FXIa. The binding of FXIa with plasminogen activator inhibitor 1 and very low-density lipoprotein receptor on HUVEC or HAEC surfaces activates vascular endothelial growth receptor factor 2 (VEGFR2). The activation of VEGFR2 triggers the mitogen-activated protein kinase (MAPK) signaling pathway and promotes the expression of active ADAM10 on the cell surface. In a pilot experiment using an established baboon model of sepsis, the inhibition of FXI activation significantly decreased the levels of soluble VE-cadherin to preserve barrier function. This study reveals a novel pathway by which FXIa regulates vascular permeability. The effect of FXIa on barrier function may be another way by which FXIa contributes to the development of inflammatory diseases.
Collapse
Affiliation(s)
- Cristina Puy
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR
| | - Samantha A. Moellmer
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR
| | - Jiaqing Pang
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR
| | - Helen H. Vu
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR
| | - Alexander R. Melrose
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR
| | - Christina U. Lorentz
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR
- Aronora, Inc, Portland, OR
| | - Erik I. Tucker
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR
- Aronora, Inc, Portland, OR
| | - Joseph J. Shatzel
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR
- Division of Hematology and Medical Oncology, Oregon Health and Science University, Portland, OR
| | - Ravi S. Keshari
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
| | - Florea Lupu
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
| | - David Gailani
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN
| | - Owen J. T. McCarty
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR
- Division of Hematology and Medical Oncology, Oregon Health and Science University, Portland, OR
| |
Collapse
|
4
|
Lira AL, Kohs TC, Moellmer SA, Shatzel JJ, McCarty OJ, Puy C. Substrates, Cofactors, and Cellular Targets of Coagulation Factor XIa. Semin Thromb Hemost 2024; 50:962-969. [PMID: 36940715 PMCID: PMC11069399 DOI: 10.1055/s-0043-1764469] [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] [Indexed: 03/22/2023]
Abstract
Coagulation factor XI (FXI) has increasingly been shown to play an integral role in several physiologic and pathological processes. FXI is among several zymogens within the blood coagulation cascade that are activated by proteolytic cleavage, with FXI converting to the active serine protease form (FXIa). The evolutionary origins of FXI trace back to duplication of the gene that transcribes plasma prekallikrein, a key factor in the plasma kallikrein-kinin system, before further genetic divergence led to FXI playing a unique role in blood coagulation. While FXIa is canonically known for activating the intrinsic pathway of coagulation by catalyzing the conversion of FIX into FIXa, it is promiscuous in nature and has been shown to contribute to thrombin generation independent of FIX. In addition to its role in the intrinsic pathway of coagulation, FXI also interacts with platelets, endothelial cells, and mediates the inflammatory response through activation of FXII and cleavage of high-molecular-weight kininogen to generate bradykinin. In this manuscript, we critically review the current body of knowledge surrounding how FXI navigates the interplay of hemostasis, inflammatory processes, and the immune response and highlight future avenues for research. As FXI continues to be clinically explored as a druggable therapeutic target, understanding how this coagulation factor fits into physiological and disease mechanisms becomes increasingly important.
Collapse
Affiliation(s)
- André L. Lira
- Department of Biomedical Engineering, School of Medicine, Oregon Health & Science University, Portland, Oregon
| | - Tia C.L. Kohs
- Department of Biomedical Engineering, School of Medicine, Oregon Health & Science University, Portland, Oregon
| | - Samantha A. Moellmer
- Department of Biomedical Engineering, School of Medicine, Oregon Health & Science University, Portland, Oregon
| | - Joseph J. Shatzel
- Department of Biomedical Engineering, School of Medicine, Oregon Health & Science University, Portland, Oregon
- Divison of Hematology and Medical Oncology, Department of Medicine, Oregon Health & Science University, Portland, Oregon
| | - Owen J.T. McCarty
- Department of Biomedical Engineering, School of Medicine, Oregon Health & Science University, Portland, Oregon
- Divison of Hematology and Medical Oncology, Department of Medicine, Oregon Health & Science University, Portland, Oregon
| | - Cristina Puy
- Department of Biomedical Engineering, School of Medicine, Oregon Health & Science University, Portland, Oregon
- Divison of Hematology and Medical Oncology, Department of Medicine, Oregon Health & Science University, Portland, Oregon
| |
Collapse
|
5
|
Haynes LM, Holding ML, DiGiovanni H, Siemieniak D, Ginsburg D. High-throughput amino acid-level characterization of the interactions of plasminogen activator inhibitor-1 with variably divergent proteases. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.16.612699. [PMID: 39345533 PMCID: PMC11429915 DOI: 10.1101/2024.09.16.612699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
While members of large paralogous protein families share structural features, their functional niches often diverge significantly. Serine protease inhibitors (SERPINs), whose members typically function as covalent inhibitors of serine proteases, are one such family. Plasminogen activator inhibitor-1 (PAI-1) is a prototypic SERPIN, which canonically inhibits tissue-and urokinase-type plasminogen activators (tPA and uPA) to regulate fibrinolysis. PAI-1 has been shown to also inhibit other serine proteases, including coagulation factor XIIa (FXIIa) and transmembrane serine protease 2 (TMPRSS2). The structural determinants of PAI-1 inhibitory function toward these non-canonical protease targets, and the biological significance of these functions, are unknown. We applied deep mutational scanning (DMS) to assess the effects of ∼80% of all possible single amino acid substitutions in PAI-1 on its ability to inhibit three putative serine protease targets (uPA, FXIIa, and TMPRSS2). Selection with each target protease generated a unique PAI-1 mutational landscape, with the determinants of protease specificity distributed throughout PAI-1's primary sequence. Next, we conducted a comparative analysis of extant orthologous sequences, demonstrating that key residues modulating PAI-1 inhibition of uPA and FXIIa, but not TMPRSS2, are maintained by purifying selection. PAI-1's activity toward FXIIa may reflect how protease evolutionary relationships predict SERPIN functional divergence, which we support via a cophylogenetic analysis of all secreted SERPINs and their cognate serine proteases. This work provides insight into the functional diversification of SERPINs and lays the framework for extending these studies to other proteases and their regulators.
Collapse
|
6
|
Ballard-Kordeliski A, Lee RH, O’Shaughnessy EC, Kim PY, Jones SR, Pawlinski R, Flick MJ, Paul DS, Mackman N, Adalsteinsson DA, Bergmeier W. 4D intravital imaging studies identify platelets as the predominant cellular procoagulant surface in a mouse hemostasis model. Blood 2024; 144:1116-1126. [PMID: 38820498 PMCID: PMC11406176 DOI: 10.1182/blood.2023022608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 05/12/2024] [Accepted: 05/13/2024] [Indexed: 06/02/2024] Open
Abstract
ABSTRACT Interplay between platelets, coagulation factors, endothelial cells (ECs), and fibrinolytic factors is necessary for effective hemostatic plug formation. This study describes a 4-dimensional (4D) imaging platform to visualize and quantify hemostatic plug components in mice with high spatiotemporal resolution. Fibrin accumulation after laser-induced vascular injury was observed at the platelet plug-EC interface, controlled by the antagonistic balance between fibrin generation and breakdown. We observed less fibrin accumulation in mice expressing low levels of tissue factor or F12-/-mice compared with controls, whereas increased fibrin accumulation, including on the vasculature adjacent to the platelet plug, was observed in plasminogen-deficient mice or wild-type mice treated with tranexamic acid. Phosphatidylserine (PS), a membrane lipid critical for the assembly of coagulation factors, was first detected at the platelet plug-EC interface, followed by exposure across the endothelium. Impaired PS exposure resulted in a significant reduction in fibrin accumulation in cyclophilin D-/-mice. Adoptive transfer studies demonstrated a key role for PS exposure on platelets, and to a lesser degree on ECs, in fibrin accumulation during hemostatic plug formation. Together, these studies suggest that (1) platelets are the functionally dominant procoagulant cellular surface, and (2) plasmin is critical for limiting fibrin accumulation at the site of a forming hemostatic plug.
Collapse
Affiliation(s)
- Abigail Ballard-Kordeliski
- Department of Biochemistry and Biophysics, The University of North Carolina at Chapel Hill, Chapel Hill, NC
- UNC Blood Research Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Robert H. Lee
- Department of Biochemistry and Biophysics, The University of North Carolina at Chapel Hill, Chapel Hill, NC
- UNC Blood Research Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Ellen C. O’Shaughnessy
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Paul Y. Kim
- Department of Medicine, McMaster University, Hamilton, ON, Canada
- Thrombosis and Atherosclerosis Research Institute, Hamilton, ON, Canada
| | - Summer R. Jones
- Department of Biochemistry and Biophysics, The University of North Carolina at Chapel Hill, Chapel Hill, NC
- UNC Blood Research Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Rafal Pawlinski
- UNC Blood Research Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC
- Department of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Matthew J. Flick
- UNC Blood Research Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC
- Department of Pathology and Laboratory Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - David S. Paul
- Department of Biochemistry and Biophysics, The University of North Carolina at Chapel Hill, Chapel Hill, NC
- UNC Blood Research Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Nigel Mackman
- UNC Blood Research Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC
- Department of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - David A. Adalsteinsson
- Department of Mathematics, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Wolfgang Bergmeier
- Department of Biochemistry and Biophysics, The University of North Carolina at Chapel Hill, Chapel Hill, NC
- UNC Blood Research Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| |
Collapse
|
7
|
Taskin B, Kohs TC, Shatzel JJ, Puy C, McCarty OJ. Factor XI as a therapeutic target in neuroinflammatory disease. Curr Opin Hematol 2024; 31:32-38. [PMID: 37694771 PMCID: PMC10843631 DOI: 10.1097/moh.0000000000000787] [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] [Indexed: 09/12/2023]
Abstract
PURPOSE OF REVIEW This review summarizes the pathophysiology and potential therapeutic options for treatment of multiple sclerosis, a common neuronal demyelinating disorder affecting 2.2 million people worldwide. As an autoimmune disorder, multiple sclerosis is associated with neuroinflammation and increased permeability of the blood-brain barrier (BBB), although the cause linking multiple sclerosis with compromised barrier function remains ill-defined. It has been previously shown that coagulation factors, including thrombin and fibrin, exacerbate the inflammatory processes and permeability of the BBB. RECENT FINDINGS Increased levels of the coagulation factor (F) XII have been found in patients presenting with relapsing-remitting multiple sclerosis, with a deleterious role for FXII being validated in murine model of multiple sclerosis, experimental autoimmune encephalitis (EAE). Recent work has uncovered a role for the major substrate activated by FXII and thrombin, FXI, in the disorder of EAE. The study found that pharmacological targeting of FXI decreased clinical symptoms, lymphocyte invasion, and white matter destruction in a multiple sclerosis model. SUMMARY This review emphasizes the role of FXII and FXI in regulating barrier function and the immune response in neuroinflammation. These new findings broaden the potential for therapeutic utility of FXI inhibitors beyond thrombosis to include neuroinflammatory diseases associated with compromised BBB function, including multiple sclerosis.
Collapse
Affiliation(s)
- Berk Taskin
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, US
| | - Tia C.L. Kohs
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, US
| | - Joseph J. Shatzel
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, US
- Division of Hematology and Medical Oncology, Oregon Health and Science University, Portland, OR
| | - Cristina Puy
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, US
| | - Owen J.T. McCarty
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, US
- Division of Hematology and Medical Oncology, Oregon Health and Science University, Portland, OR
| |
Collapse
|
8
|
Kohs TC, Vu HH, Jordan KR, Parra-Izquierdo I, Hinds MT, Shatzel JJ, Kievit P, Morgan TK, Yunga ST, Ngo TT, Aslan JE, Wallisch M, Lorentz CU, Tucker EI, Gailani D, Lindner JR, Puy C, McCarty OJ. Activation of coagulation FXI promotes endothelial inflammation and amplifies platelet activation in a nonhuman primate model of hyperlipidemia. Res Pract Thromb Haemost 2024; 8:102276. [PMID: 38226339 PMCID: PMC10788631 DOI: 10.1016/j.rpth.2023.102276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 09/16/2023] [Accepted: 09/19/2023] [Indexed: 01/17/2024] Open
Abstract
Background Hyperlipidemia is associated with chronic inflammation and thromboinflammation. This is an underlying cause of several cardiovascular diseases, including atherosclerosis. In diseased blood vessels, rampant thrombin generation results in the initiation of the coagulation cascade, activation of platelets, and endothelial cell dysfunction. Coagulation factor (F) XI represents a promising therapeutic target to reduce thromboinflammation, as it is uniquely positioned at an intersection between inflammation and thrombin generation. Objectives This study aimed to investigate the role of FXI in promoting platelet and endothelial cell activation in a model of hyperlipidemia. Methods Nonhuman primates (NHPs) were fed a standard chow diet (lean, n = 6) or a high-fat diet (obese, n = 8) to establish a model of hyperlipidemia. Obese NHPs were intravenously administered a FXI blocking antibody (2 mg/kg) and studied at baseline and at 1, 7, 14, 21, and 28 days after drug administration. Platelet activation and inflammatory markers were measured using fluorescence-activated cell sorting or enzyme-linked immunosorbent assay. Molecular imaging was used to quantify vascular cell adhesion molecule 1 (VCAM-1) expression at the carotid bifurcation. Results Obese NHPs demonstrated increased sensitivity for platelet P-selectin expression and phosphatidylserine exposure in response to platelet GPVI or PAR agonists compared with lean NHPs. Obese NHPs exhibited elevated levels of C-reactive protein, cathepsin D, and myeloperoxidase compared with lean NHPs. Following pharmacological inhibition of FIX activation by FXIa, platelet priming for activation by GPVI or PAR agonists, C-reactive protein levels, and endothelial VCAM-1 levels were reduced in obese NHPs. Conclusion FXI activation promotes the proinflammatory phenotype of hyperlipidemia by priming platelet activation and inciting endothelial cell dysfunction.
Collapse
Affiliation(s)
- Tia C.L. Kohs
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon, USA
| | - Helen H. Vu
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon, USA
| | - Kelley R. Jordan
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon, USA
| | - Iván Parra-Izquierdo
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon, USA
| | - Monica T. Hinds
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon, USA
| | - Joseph J. Shatzel
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon, USA
- Division of Hematology and Oncology, Oregon Health & Science University, Portland, Oregon, USA
| | - Paul Kievit
- Division of Cardiometabolic Health, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Terry K. Morgan
- Department of Pathology and Laboratory Medicine, Oregon Health & Science University, Portland, Oregon, USA
| | - Samuel Tassi Yunga
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon, USA
- Cancer Early Detection Advanced Research Center, Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon, USA
| | - Thuy T.M. Ngo
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon, USA
- Cancer Early Detection Advanced Research Center, Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon, USA
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, Oregon, USA
| | - Joseph E. Aslan
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon, USA
- Knight Cardiovascular Institute, School of Medicine, Oregon Health & Science University, Portland, Oregon, USA
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, Oregon, USA
| | - Michael Wallisch
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon, USA
- Aronora, Inc, Portland, Oregon, USA
| | - Christina U. Lorentz
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon, USA
- Aronora, Inc, Portland, Oregon, USA
| | - Erik I. Tucker
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon, USA
- Aronora, Inc, Portland, Oregon, USA
| | - David Gailani
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Jonathan R. Lindner
- Division of Cardiovascular Medicine and Robert M. Berne Cardiovascular Research Institute, University of Virginia, Charlottesville, Virginia, USA
| | - Cristina Puy
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon, USA
| | - Owen J.T. McCarty
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon, USA
- Division of Hematology and Oncology, Oregon Health & Science University, Portland, Oregon, USA
| |
Collapse
|
9
|
Joubert J, Meiring SM, Janse van Rensburg WJ. The thrombin generation capability of the Chacma baboon (Papio ursinus): implications for haemostatic disease models. Sci Rep 2023; 13:22968. [PMID: 38151511 PMCID: PMC10752904 DOI: 10.1038/s41598-023-50341-8] [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: 10/20/2023] [Accepted: 12/19/2023] [Indexed: 12/29/2023] Open
Abstract
Baboon models are often used to investigate haemostatic diseases, such as acquired thrombotic thrombocytopenic purpura or bacterial sepsis-induced disseminated intravascular coagulation, and their potential treatment with novel drugs. Thrombin generation is vital for these models, and an important potential therapeutic target. We investigated the thrombin generation profile of the Chacma baboon (Papio ursinus - a common pre-clinical model) including the effects of sex and ABO blood group. Thrombin generation curves, lag times, peak heights, times-to-peak, velocity indexes and Endogenous Thrombin Potentials (ETPs) of 40 adult Chacma baboons were assessed and compared with normal human plasma, using a low concentration of tissue factor (1 pM) and phospholipids. Reference intervals were calculated, and results compared between O and non-O ABO blood groups, and between males and females. Lag times of all baboons fell within the human reference interval. Most animals (n = 32; 80%) had times-to-peak above, and velocity indexes and peak heights markedly below (n = 27; 68%) the human range. However, 97.5% of baboons had an ETP above the human reference interval, indicating greater overall thrombin generation. ABO blood group had no effect, but males (n = 14; 35%) had less potent thrombin generation than females (n = 26; 65%), with significantly longer lag times (p = 0.0475), lower peak thrombin concentrations (p = 0.0203), and lower ETPs (p = 0.0238). Chacma baboons have greater overall endogenous thrombin generation potentials than humans, which is even more prominent in females. This should be considered when designing future baboon model experiments involving the haemostatic system, or when evaluating novel therapies in these animals.
Collapse
Affiliation(s)
- J Joubert
- Department of Haematology and Cell Biology, School of Pathology, Faculty of Health Sciences, University of the Free State, 205 Nelson Mandela Drive, PO Box 339 (G2), Bloemfontein, 9300, South Africa.
- National Health Laboratory Service, Universitas Academic Laboratories, Haematology, Bloemfontein, South Africa.
| | - S M Meiring
- Department of Haematology and Cell Biology, School of Pathology, Faculty of Health Sciences, University of the Free State, 205 Nelson Mandela Drive, PO Box 339 (G2), Bloemfontein, 9300, South Africa
- National Health Laboratory Service, Universitas Academic Laboratories, Haematology, Bloemfontein, South Africa
| | - W J Janse van Rensburg
- Human Molecular Biology Unit, School of Biomedical Sciences, Faculty of Health Sciences, University of the Free State, Bloemfontein, South Africa
| |
Collapse
|
10
|
Ballard-Kordeliski A, Lee RH, O'Shaughnessy EC, Kim PY, Jones S, Mackman N, Flick MJ, Paul DS, Adalsteinsson D, Bergmeier W. 4D intravital imaging studies identify platelets as the predominant cellular procoagulant surface in a mouse model of hemostasis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.25.554449. [PMID: 37662350 PMCID: PMC10473702 DOI: 10.1101/2023.08.25.554449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
Interplay between platelets, coagulation/fibrinolytic factors, and endothelial cells (ECs) is necessary for effective hemostatic plug formation. This study describes a novel four-dimensional (4D) imaging platform to visualize and quantify hemostatic plug components with high spatiotemporal resolution. Fibrin accumulation following laser-induced endothelial ablation was observed at the EC-platelet plug interface, controlled by the antagonistic balance between fibrin generation and breakdown. Phosphatidylserine (PS) was first detected in close physical proximity to the fibrin ring, followed by exposure across the endothelium. Impaired PS exposure in cyclophilinD -/- mice resulted in a significant reduction in fibrin accumulation. Adoptive transfer and inhibitor studies demonstrated a key role for platelets, but not ECs, in fibrin generation during hemostatic plug formation. Inhibition of fibrinolysis with tranexamic acid (TXA) led to increased fibrin accumulation in WT mice, but not in cyclophilinD -/- mice or WT mice treated with antiplatelet drugs. These studies implicate platelets as the functionally dominant procoagulant surface during hemostatic plug formation. In addition, they suggest that impaired fibrin formation due to reduced platelet procoagulant activity is not reversed by TXA treatment.
Collapse
|
11
|
Barrett L, Curry N, Abu-Hanna J. Experimental Models of Traumatic Injuries: Do They Capture the Coagulopathy and Underlying Endotheliopathy Induced by Human Trauma? Int J Mol Sci 2023; 24:11174. [PMID: 37446351 PMCID: PMC10343021 DOI: 10.3390/ijms241311174] [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: 06/13/2023] [Revised: 07/03/2023] [Accepted: 07/05/2023] [Indexed: 07/15/2023] Open
Abstract
Trauma-induced coagulopathy (TIC) is a major cause of morbidity and mortality in patients with traumatic injury. It describes the spectrum of coagulation abnormalities that occur because of the trauma itself and the body's response to the trauma. These coagulation abnormalities range from hypocoagulability and hyperfibrinolysis, resulting in potentially fatal bleeding, in the early stages of trauma to hypercoagulability, leading to widespread clot formation, in the later stages. Pathological changes in the vascular endothelium and its regulation of haemostasis, a phenomenon known as the endotheliopathy of trauma (EoT), are thought to underlie TIC. Our understanding of EoT and its contribution to TIC remains in its infancy largely due to the scarcity of experimental research. This review discusses the mechanisms employed by the vascular endothelium to regulate haemostasis and their dysregulation following traumatic injury before providing an overview of the available experimental in vitro and in vivo models of trauma and their applicability for the study of the EoT and its contribution to TIC.
Collapse
Affiliation(s)
- Liam Barrett
- Division of Anaesthesia, Department of Medicine, University of Cambridge, Cambridge CB2 1TN, UK;
- Emergency Department, Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, UK
| | - Nicola Curry
- Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK;
- Oxford Haemophilia and Thrombosis Centre, Oxford University Hospitals NHS Foundation Trust, Oxford OX3 7LD, UK
| | - Jeries Abu-Hanna
- Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK;
| |
Collapse
|
12
|
Association of FXI activity with thrombo-inflammation, extracellular matrix, lipid metabolism and apoptosis in venous thrombosis. Sci Rep 2022; 12:9761. [PMID: 35697739 PMCID: PMC9192691 DOI: 10.1038/s41598-022-13174-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 02/17/2022] [Indexed: 12/31/2022] Open
Abstract
Animal experiments and early phase human trials suggest that inhibition of factor XIa (FXIa) safely prevents venous thromboembolism (VTE), and specific murine models of sepsis have shown potential efficacy in alleviating cytokine storm. These latter findings support the role of FXI beyond coagulation. Here, we combine targeted proteomics, machine learning and bioinformatics, to discover associations between FXI activity (FXI:C) and the plasma protein profile of patients with VTE. FXI:C was measured with a modified activated partial prothrombin time (APTT) clotting time assay. Proximity extension assay-based protein profiling was performed on plasma collected from subjects from the Genotyping and Molecular Phenotyping of Venous Thromboembolism (GMP-VTE) Project, collected during an acute VTE event (n = 549) and 12-months after (n = 187). Among 444 proteins investigated, N = 21 and N = 66 were associated with FXI:C during the acute VTE event and at 12 months follow-up, respectively. Seven proteins were identified as FXI:C-associated at both time points. These FXI-related proteins were enriched in immune pathways related to causes of thrombo-inflammation, extracellular matrix interaction, lipid metabolism, and apoptosis. The results of this study offer important new avenues for future research into the multiple properties of FXI, which are of high clinical interest given the current development of FXI inhibitors.
Collapse
|
13
|
Lakshmanan HHS, Estonilo A, Reitsma SE, Melrose AR, Subramanian J, Zheng TJ, Maddala J, Tucker EI, Gailani D, McCarty OJT, Jurney PL, Puy C. Revised model of the tissue factor pathway of thrombin generation: Role of the feedback activation of FXI. J Thromb Haemost 2022; 20:1350-1363. [PMID: 35352494 PMCID: PMC9590754 DOI: 10.1111/jth.15716] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 02/26/2022] [Accepted: 03/16/2022] [Indexed: 11/27/2022]
Abstract
BACKGROUND Biochemical reaction networks are self-regulated in part due to feedback activation mechanisms. The tissue factor (TF) pathway of blood coagulation is a complex reaction network controlled by multiple feedback loops that coalesce around the serine protease thrombin. OBJECTIVES Our goal was to evaluate the relative contribution of the feedback activation of coagulation factor XI (FXI) in TF-mediated thrombin generation using a comprehensive systems-based analysis. MATERIALS AND METHODS We developed a systems biology model that improves the existing Hockin-Mann (HM) model through an integrative approach of mathematical modeling and in vitro experiments. Thrombin generation measured using in vitro assays revealed that the feedback activation of FXI contributes to the propagation of thrombin generation based on the initial concentrations of TF or activated coagulation factor X (FXa). We utilized experimental data to improve the robustness of the HM model to capture thrombin generation kinetics without a role for FXI before including the feedback activation of FXI by thrombin to construct the extended (ext.) HM model. RESULTS AND CONCLUSIONS Using the ext.HM model, we predicted that the contribution of positive feedback of FXI activation by thrombin can be abolished by selectively eliminating the inhibitory function of tissue factor pathway inhibitor (TFPI), a serine protease inhibitor of FXa and TF-activated factor VII (FVIIa) complex. This prediction from the ext.HM model was experimentally validated using thrombin generation assays with function blocking antibodies against TFPI and plasmas depleted of FXI. Together, our results demonstrate the applications of combining experimental and modeling techniques in predicting complex biochemical reaction systems.
Collapse
Affiliation(s)
| | - Aldrich Estonilo
- Department of Biomedical Engineering, San Jose State University, San Jose, California, USA
| | - Stéphanie E. Reitsma
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon, USA
| | - Alexander R. Melrose
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon, USA
| | | | - Tony J. Zheng
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon, USA
| | - Jeevan Maddala
- Department of Chemical and Biomedical Engineering, West Virginia University, Morgantown, West Virginia, USA
| | - Erik I. Tucker
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon, USA
- Aronora, Inc., Portland, Oregon, USA
| | - David Gailani
- Department of Pathology, Microbiology and Immunology, Vanderbilt University, Nashville, Tennessee, USA
| | - Owen J. T. McCarty
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon, USA
| | - Patrick L. Jurney
- Department of Biomedical Engineering, San Jose State University, San Jose, California, USA
| | - Cristina Puy
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon, USA
| |
Collapse
|
14
|
Bai Q, Lu Y, Chen Y, Zhang H, Zhang W, Wu H, Wen A. Endothelial METTL3 (Methyltransferase-Like 3) Inhibits Fibrinolysis by Promoting PAI-1 (Plasminogen Activator Inhibitor-1) Expression Through Enhancing Jun Proto-Oncogene N6-Methyladenosine Modification. Arterioscler Thromb Vasc Biol 2021; 41:2877-2889. [PMID: 34645279 PMCID: PMC8608005 DOI: 10.1161/atvbaha.121.316414] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 09/23/2021] [Indexed: 11/16/2022]
Abstract
OBJECTIVE METTL3 (methyltransferase-like protein 3)-mediated N6-methyladenosine modification is the most abundant RNA modification on eukaryote mRNAs and plays a crucial role in diverse physiological and pathological processes. However, whether N6-methyladenosine modification has function in thrombosis is unknown. This study aims to determine the role of METTL3 in the endothelial cells-mediated thrombosis. Approach and Results: RNA-sequencing and real-time quantitative PCR revealed that the expression of PAI-1 (plasminogen activator inhibitor-1) was downregulated in METTL3 knockdown human umbilical vein endothelial cells. In vitro experiments showed that METTL3 suppressed fibrinolysis. Mechanically, RNA methylation sequencing and meRIP-quantitative real-time PCR showed that METTL3 catalyzed N6-methyladenosine modification on 3' UTR of JUN mRNA. Western blotting analysis showed that METTL3 promoted JUN protein expression. Chromatin immunoprecipitation analysis demonstrated that JUN bound to the PAI-1 promoter in human umbilical vein endothelial cells. Furthermore, mice challenged with lipopolysaccharide resulted in higher METTL3 expression in vessels. Endothelial-specific knockdown of Mettl3 decreased expression of active PAI-1 in plasma and attenuated fibrin deposition in livers and lungs during endotoxemia. CONCLUSIONS Our study reveals that METTL3-mediated N6-methyladenosine modification plays a crucial role in fibrinolysis and is an underlying target for the therapy of thrombotic disorders.
Collapse
Affiliation(s)
- Qin Bai
- Department of Blood Transfusion, Daping Hospital, Army Medical University, Chongqing, China
| | - Yao Lu
- Department of Blood Transfusion, Daping Hospital, Army Medical University, Chongqing, China
| | - Yanhua Chen
- Department of Blood Transfusion, Daping Hospital, Army Medical University, Chongqing, China
| | - Han Zhang
- Department of Blood Transfusion, Daping Hospital, Army Medical University, Chongqing, China
| | - Weiwei Zhang
- Department of Blood Transfusion, Daping Hospital, Army Medical University, Chongqing, China
| | - Huang Wu
- Department of Blood Transfusion, Daping Hospital, Army Medical University, Chongqing, China
| | - Aiqing Wen
- Department of Blood Transfusion, Daping Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
15
|
Hendley SA, Dimov A, Bhargava A, Snoddy E, Mansour D, Afifi RO, Wool GD, Zha Y, Sammet S, Lu ZF, Ahmed O, Paul JD, Bader KB. Assessment of histological characteristics, imaging markers, and rt-PA susceptibility of ex vivo venous thrombi. Sci Rep 2021; 11:22805. [PMID: 34815441 PMCID: PMC8610976 DOI: 10.1038/s41598-021-02030-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 11/08/2021] [Indexed: 12/27/2022] Open
Abstract
Venous thromboembolism is a significant source of morbidity and mortality worldwide. Catheter-directed thrombolytics is the primary treatment used to relieve critical obstructions, though its efficacy varies based on the thrombus composition. Non-responsive portions of the specimen often remain in situ, which prohibits mechanistic investigation of lytic resistance or the development of diagnostic indicators for treatment outcomes. In this study, thrombus samples extracted from venous thromboembolism patients were analyzed ex vivo to determine their histological properties, susceptibility to lytic therapy, and imaging characteristics. A wide range of thrombus morphologies were observed, with a dependence on age and etymology of the specimen. Fibrinolytic inhibitors including PAI-1, alpha 2-antiplasmin, and TAFI were present in samples, which may contribute to the response venous thrombi to catheter-directed thrombolytics. Finally, a weak but significant correlation was observed between the response of the sample to lytic drug and its magnetic microstructure assessed with a quantitative MRI sequence. These findings highlight the myriad of changes in venous thrombi that may promote lytic resistance, and imaging metrics that correlate with treatment outcomes.
Collapse
Affiliation(s)
- Samuel A Hendley
- Committee on Medical Physics, University of Chicago, Chicago, IL, 60637, USA
| | - Alexey Dimov
- Department of Radiology, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Aarushi Bhargava
- Department of Radiology, University of Chicago, Chicago, IL, 60637, USA
| | - Erin Snoddy
- Department of Radiology, University of Chicago, Chicago, IL, 60637, USA
| | - Daniel Mansour
- Department of Radiology, University of Chicago, Chicago, IL, 60637, USA
| | - Rana O Afifi
- Department of Cardiothoracic and Vascular Surgery, University of Texas at Houston, Houston, TX, 77030, USA
| | - Geoffrey D Wool
- Department of Pathology, University of Chicago, Chicago, IL, 60637, USA
| | - Yuanyuan Zha
- The Human Immunological Monitoring Facility, University of Chicago, Chicago, IL, 60637, USA
| | - Steffen Sammet
- Committee on Medical Physics, University of Chicago, Chicago, IL, 60637, USA
- Department of Radiology, University of Chicago, Chicago, IL, 60637, USA
| | - Zheng Feng Lu
- Committee on Medical Physics, University of Chicago, Chicago, IL, 60637, USA
- Department of Radiology, University of Chicago, Chicago, IL, 60637, USA
| | - Osman Ahmed
- Department of Radiology, University of Chicago, Chicago, IL, 60637, USA
| | - Jonathan D Paul
- Department of Medicine, University of Chicago, Chicago, IL, 60637, USA
| | - Kenneth B Bader
- Committee on Medical Physics, University of Chicago, Chicago, IL, 60637, USA.
- Department of Radiology, University of Chicago, Chicago, IL, 60637, USA.
| |
Collapse
|
16
|
Lagrange J, Lacolley P, Wahl D, Peyrin-Biroulet L, Regnault V. Shedding Light on Hemostasis in Patients With Inflammatory Bowel Diseases. Clin Gastroenterol Hepatol 2021; 19:1088-1097.e6. [PMID: 31972287 DOI: 10.1016/j.cgh.2019.12.043] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Revised: 12/19/2019] [Accepted: 12/31/2019] [Indexed: 02/07/2023]
Abstract
Patients with inflammatory bowel diseases (IBD) have an increased risk of thrombosis, possibly due to changes in blood cells and molecules involved in hemostasis. They have increased platelet counts and reactivity as well as increased platelet-derived large extracellular vesicles. Coagulation is continuously activated in patients with IBD, based on measured markers of thrombin generation, and the anticoagulant functions of endothelial cells are damaged. Furthermore, fibrinogen is increased and fibrin clots are denser. However, pathogenesis of thrombosis in patients with IBD appears to differ from that of patients without IBD. Patients with IBD also take drugs that might contribute to risk of thrombosis, complicating the picture. We review the features of homeostasis that are altered in patients with IBD and possible mechanisms of this relationship.
Collapse
Affiliation(s)
- Jeremy Lagrange
- INSERM U1116, Faculté de Médecine, Université de Lorraine, Vandœuvre-lès-Nancy, France; Université de Lorraine, Nancy, France; Center for Thrombosis and Hemostasis, University Medical Center Mainz, Mainz, Germany.
| | - Patrick Lacolley
- INSERM U1116, Faculté de Médecine, Université de Lorraine, Vandœuvre-lès-Nancy, France; Université de Lorraine, Nancy, France; Centre Hospitalier Régionale Universitaire de Nancy, Vandœuvre-lès-Nancy, France
| | - Denis Wahl
- INSERM U1116, Faculté de Médecine, Université de Lorraine, Vandœuvre-lès-Nancy, France; Université de Lorraine, Nancy, France; Division of Vascular Medicine, Centre Hospitalier Régionale Universitaire de Nancy, Vandœuvre-lès-Nancy, France
| | - Laurent Peyrin-Biroulet
- Université de Lorraine, Nancy, France; INSERM U1256, Faculté de Médecine, Université de Lorraine, Vandœuvre-lès-Nancy, France; Department of Gastroenterology, Centre Hospitalier Régionale Universitaire de Nancy, Vandœuvre-lès-Nancy, France
| | - Véronique Regnault
- INSERM U1116, Faculté de Médecine, Université de Lorraine, Vandœuvre-lès-Nancy, France; Université de Lorraine, Nancy, France; Centre Hospitalier Régionale Universitaire de Nancy, Vandœuvre-lès-Nancy, France
| |
Collapse
|
17
|
Kohs TCL, Lorentz CU, Johnson J, Puy C, Olson SR, Shatzel JJ, Gailani D, Hinds MT, Tucker EI, Gruber A, McCarty OJT, Wallisch M. Development of Coagulation Factor XII Antibodies for Inhibiting Vascular Device-Related Thrombosis. Cell Mol Bioeng 2020; 14:161-175. [PMID: 33868498 DOI: 10.1007/s12195-020-00657-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Accepted: 09/26/2020] [Indexed: 11/26/2022] Open
Abstract
Introduction Vascular devices such as stents, hemodialyzers, and membrane oxygenators can activate blood coagulation and often require the use of systemic anticoagulants to selectively prevent intravascular thrombotic/embolic events or extracorporeal device failure. Coagulation factor (F)XII of the contact activation system has been shown to play an important role in initiating vascular device surface-initiated thrombus formation. As FXII is dispensable for hemostasis, targeting the contact activation system holds promise as a significantly safer strategy than traditional antithrombotics for preventing vascular device-associated thrombosis. Objective Generate and characterize anti-FXII monoclonal antibodies that inhibit FXII activation or activity. Methods Monoclonal antibodies against FXII were generated in FXII-deficient mice and evaluated for their binding and anticoagulant properties in purified and plasma systems, in whole blood flow-based assays, and in an in vivo non-human primate model of vascular device-initiated thrombus formation. Results A FXII antibody screen identified over 400 candidates, which were evaluated in binding studies and clotting assays. One non-inhibitor and six inhibitor antibodies were selected for characterization in functional assays. The most potent inhibitory antibody, 1B2, was found to prolong clotting times, inhibit fibrin generation on collagen under shear, and inhibit platelet deposition and fibrin formation in an extracorporeal membrane oxygenator deployed in a non-human primate. Conclusion Selective contact activation inhibitors hold potential as useful tools for research applications as well as safe and effective inhibitors of vascular device-related thrombosis.
Collapse
Affiliation(s)
- T C L Kohs
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR 97239 USA
| | - C U Lorentz
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR 97239 USA
- Aronora Inc., Portland, OR USA
| | - J Johnson
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR 97239 USA
| | - C Puy
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR 97239 USA
| | - S R Olson
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR 97239 USA
- Division of Hematology& Medical Oncology, Department of Medicine, Oregon Health & Science University, Portland, OR USA
| | - J J Shatzel
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR 97239 USA
- Division of Hematology& Medical Oncology, Department of Medicine, Oregon Health & Science University, Portland, OR USA
| | - D Gailani
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, Nashville, TN USA
| | - M T Hinds
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR 97239 USA
| | - E I Tucker
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR 97239 USA
- Aronora Inc., Portland, OR USA
| | - A Gruber
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR 97239 USA
- Aronora Inc., Portland, OR USA
- Division of Hematology& Medical Oncology, Department of Medicine, Oregon Health & Science University, Portland, OR USA
| | - O J T McCarty
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR 97239 USA
- Division of Hematology& Medical Oncology, Department of Medicine, Oregon Health & Science University, Portland, OR USA
| | - M Wallisch
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR 97239 USA
- Aronora Inc., Portland, OR USA
| |
Collapse
|
18
|
Li X, Sim MMS, Wood JP. Recent Insights Into the Regulation of Coagulation and Thrombosis. Arterioscler Thromb Vasc Biol 2020; 40:e119-e125. [PMID: 32320291 DOI: 10.1161/atvbaha.120.312674] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Xian Li
- From the Saha Cardiovascular Research Center (X.L., J.P.W.), University of Kentucky, Lexington
| | - Martha M S Sim
- Department of Molecular and Cellular Biochemistry (M.M.S.S., J.P.W.), University of Kentucky, Lexington
| | - Jeremy P Wood
- From the Saha Cardiovascular Research Center (X.L., J.P.W.), University of Kentucky, Lexington.,Department of Molecular and Cellular Biochemistry (M.M.S.S., J.P.W.), University of Kentucky, Lexington.,Division of Cardiovascular Medicine (J.P.W.), University of Kentucky, Lexington
| |
Collapse
|
19
|
Abstract
Transcytosis of macromolecules through lung endothelial cells is the primary route of transport from the vascular compartment into the interstitial space. Endothelial transcytosis is mostly a caveolae-dependent process that combines receptor-mediated endocytosis, vesicle trafficking via actin-cytoskeletal remodeling, and SNARE protein directed vesicle fusion and exocytosis. Herein, we review the current literature on caveolae-mediated endocytosis, the role of actin cytoskeleton in caveolae stabilization at the plasma membrane, actin remodeling during vesicle trafficking, and exocytosis of caveolar vesicles. Next, we provide a concise summary of experimental methods employed to assess transcytosis. Finally, we review evidence that transcytosis contributes to the pathogenesis of acute lung injury. © 2020 American Physiological Society. Compr Physiol 10:491-508, 2020.
Collapse
Affiliation(s)
- Joshua H. Jones
- Department of Pharmacology, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Richard D. Minshall
- Department of Pharmacology, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA,Department of Anesthesiology, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA,Correspondence to
| |
Collapse
|
20
|
Xian M, Ji S, Chen C, Liang S, Wang S. Sparganin A alleviates blood stasis syndrome and its key targets by molecular docking. RSC Adv 2019; 9:37978-37985. [PMID: 35541782 PMCID: PMC9075786 DOI: 10.1039/c9ra06329c] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 10/24/2019] [Indexed: 01/01/2023] Open
Abstract
Blood stasis syndrome is implicated in the development of chronic conditions, including cardio- and cerebrovascular diseases. Cyclo-(Tyr-Leu), named Sparganin A (SA), is a compound isolated from the ethanol extract of Rhizoma Sparganii. Here, the successful extraction of SA from Rhizoma Sparganii was verified by extensive spectral analysis using 1H NMR and 13C NMR. To determine the biological effects of SA, a mouse model of acute blood stasis was established by subcutaneous injection of adrenaline hydrochloride and placing the animals in an ice water bath. In this model, the concentration of TXB2, PAI-1, FIB, ET-1 was measured by ELISA, and thymus index (TI), hepatic index (HI), and spleen index (SI) were calculated. Molecular docking by SYBYL and functional analysis of the putative targets by STRING and Cytoscape were employed to identify the key targets of SA. The accumulated results documented that SA exhibits anticoagulative activity, and its key targets are VEGFA and SERPINE1. SA may be involved in the pathological process of complement and coagulation cascades. This study demonstrates that SA may be a promising drug to control coagulation in blood stasis syndrome.
Collapse
Affiliation(s)
- Minghua Xian
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University Guangzhou 510006 China +86-20-39352177
- Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica of State Administration of TCM, Guangdong Pharmaceutical University Guangzhou 510006 China +86-20-39352177
- Engineering & Technology Research Center for Chinese Materia Medica Quality of the Universities of Guangdong Province, Guangdong Pharmaceutical University Guangzhou 510006 China
| | - Sulong Ji
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University Guangzhou 510006 China +86-20-39352177
- Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica of State Administration of TCM, Guangdong Pharmaceutical University Guangzhou 510006 China +86-20-39352177
- Engineering & Technology Research Center for Chinese Materia Medica Quality of the Universities of Guangdong Province, Guangdong Pharmaceutical University Guangzhou 510006 China
| | - Chen Chen
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University Guangzhou 510006 China +86-20-39352177
- Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica of State Administration of TCM, Guangdong Pharmaceutical University Guangzhou 510006 China +86-20-39352177
- Engineering & Technology Research Center for Chinese Materia Medica Quality of the Universities of Guangdong Province, Guangdong Pharmaceutical University Guangzhou 510006 China
| | - Shengwang Liang
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University Guangzhou 510006 China +86-20-39352177
- Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica of State Administration of TCM, Guangdong Pharmaceutical University Guangzhou 510006 China +86-20-39352177
- Engineering & Technology Research Center for Chinese Materia Medica Quality of the Universities of Guangdong Province, Guangdong Pharmaceutical University Guangzhou 510006 China
| | - Shumei Wang
- Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica of State Administration of TCM, Guangdong Pharmaceutical University Guangzhou 510006 China +86-20-39352177
- Engineering & Technology Research Center for Chinese Materia Medica Quality of the Universities of Guangdong Province, Guangdong Pharmaceutical University Guangzhou 510006 China
- School of Pharmacy, Guangdong Pharmaceutical University Guangzhou 510006 China
| |
Collapse
|
21
|
Xu Z, Cheng B, Fu S, Liu X, Xie G, Li Z, Ji Y, Fu Q, Xu Z, Fang X. Coagulative biomarkers on admission to the ICU predict acute kidney injury and mortality in patients with septic shock caused by intra-abdominal infection. Infect Drug Resist 2019; 12:2755-2764. [PMID: 31564926 PMCID: PMC6732509 DOI: 10.2147/idr.s218592] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 08/26/2019] [Indexed: 12/29/2022] Open
Abstract
Purpose Sepsis-associated coagulopathy (SAC) contributes to the development of multiple organ failure (MOF) and increasing mortality. The present study was conducted to investigate whether coagulative biomarkers on admission to the intensive care unit (ICU) can predict acute kidney injury (AKI) and mortality in patients with septic shock caused by intra-abdominal infection (IAI). Patients and methods An observational retrospective study was conducted in the surgical ICU. We studied patients who met the criteria of septic shock (Sepsis-3) caused by IAI between January 1, 2013, and December 31, 2016. By adjusting for baseline characteristics, multivariate regression analyses were employed to identify independent risk factors for predicting AKI and mortality. Results Of the 138 enrolled patients, 65 patients developed AKI. The patients who developed AKI exhibited a dramatically higher Sequential Organ Failure Assessment (SOFA) score (median, 12), Acute Physiology and Chronic Health Evaluation (APACHE) II score (median, 27.5) and mortality rate. In both models, we found that activated partial thromboplastin time (APTT) (odds ratio (OR)=1.074, 95% confidence interval (CI) 1.030-1.120, p=0.001), prothrombin time (PT) (OR=1.162, 95% CI 1.037-1.302, p=0.010) and D-dimer level (OR=1.098, 95% CI 1.002-1.202, p=0.045) on admission to the ICU were significant risk factors for AKI. Moreover, Cox regression analysis showed that prolonged APTT (OR=1.065, 95% CI 1.025-1.107, p=0.001) was independently associated with high mortality. Conclusion In patients with septic shock caused by IAI, APTT, PT and D-dimer level on admission to the ICU were significantly associated with AKI. Furthermore, APTT was an independent predictor of 30-day mortality.
Collapse
Affiliation(s)
- Zhipeng Xu
- Department of Anesthesiology and Intensive Care Medicine, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou City, Zhejiang Province, People's Republic of China
| | - Baoli Cheng
- Department of Anesthesiology and Intensive Care Medicine, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou City, Zhejiang Province, People's Republic of China
| | - Shuiqiao Fu
- Department of Surgical Intensive Care Unit, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou City, Zhejiang Province, People's Republic of China
| | - Xu Liu
- Department of Intensive Care Unit, The Affiliated Hospital of Guizhou Medical University, Guiyang City, Guizhou Province, People's Republic of China
| | - Guohao Xie
- Department of Anesthesiology and Intensive Care Medicine, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou City, Zhejiang Province, People's Republic of China
| | - Zhongwang Li
- Department of Anesthesiology and Intensive Care Medicine, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou City, Zhejiang Province, People's Republic of China
| | - Yun Ji
- Department of Surgical Intensive Care Unit, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou City, Zhejiang Province, People's Republic of China
| | - Qinghui Fu
- Department of Surgical Intensive Care Unit, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou City, Zhejiang Province, People's Republic of China
| | - Zhinan Xu
- Institute of Biological Engineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou City, Zhejiang Province, People's Republic of China
| | - Xiangming Fang
- Department of Anesthesiology and Intensive Care Medicine, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou City, Zhejiang Province, People's Republic of China
| |
Collapse
|