51
|
Abstract
Thrombin is a multifunctional serine protease generated in injured cells. The generation of thrombin in coagulation plays a central role in the functioning of haemostasis. The last enzyme in the coagulation cascade is thrombin, with the function of cleaving fibrinogen to fibrin, which forms the fibrin clot of a haemostatic plug. Although thrombin primarily converts fibrinogen to fibrin, it also has many other positive regulatory effects on coagulation. Thrombin has procoagulant, inflammatory, cellular proliferation and anticoagulant effects. In coagulation system, thrombin has two very distinct roles. Firstly, it acts as a procoagulant when it converts fibrinogen into an insoluble fibrin clot, activates factor (F) XIII, activates thrombin activatable fibrinolysis inhibitor (TAFI) and activates FV, FVIII and FXI. Thrombin also enhances platelet adhesion by inactivating a disintegrin and metalloprotease with thrombospondin type1 motif (ADAMTS13). However, when thrombin activates protein C, it acts as an anticoagulant. A natural anticoagulant pathway that supplies regulation of the blood coagulation system contains protein C, which is the key component. This is accomplished by the specific proteolytic inactivation of FV and FVIII. In this review, the multiple roles of thrombin in the haemostatic response to injury are studied in addition to the cofactors that determine thrombin activity and how thrombin activity is thought to be coordinated.
Collapse
|
52
|
Popescu NI, Lupu C, Lupu F. Disseminated intravascular coagulation and its immune mechanisms. Blood 2022; 139:1973-1986. [PMID: 34428280 PMCID: PMC8972096 DOI: 10.1182/blood.2020007208] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 06/02/2021] [Indexed: 11/26/2022] Open
Abstract
Disseminated intravascular coagulation (DIC) is a syndrome triggered by infectious and noninfectious pathologies characterized by excessive generation of thrombin within the vasculature and widespread proteolytic conversion of fibrinogen. Despite diverse clinical manifestations ranging from thrombo-occlusive damage to bleeding diathesis, DIC etiology commonly involves excessive activation of blood coagulation and overlapping dysregulation of anticoagulants and fibrinolysis. Initiation of blood coagulation follows intravascular expression of tissue factor or activation of the contact pathway in response to pathogen-associated or host-derived, damage-associated molecular patterns. The process is further amplified through inflammatory and immunothrombotic mechanisms. Consumption of anticoagulants and disruption of endothelial homeostasis lower the regulatory control and disseminate microvascular thrombosis. Clinical DIC development in patients is associated with worsening morbidities and increased mortality, regardless of the underlying pathology; therefore, timely recognition of DIC is critical for reducing the pathologic burden. Due to the diversity of triggers and pathogenic mechanisms leading to DIC, diagnosis is based on algorithms that quantify hemostatic imbalance, thrombocytopenia, and fibrinogen conversion. Because current diagnosis primarily assesses overt consumptive coagulopathies, there is a critical need for better recognition of nonovert DIC and/or pre-DIC states. Therapeutic strategies for patients with DIC involve resolution of the eliciting triggers and supportive care for the hemostatic imbalance. Despite medical care, mortality in patients with DIC remains high, and new strategies, tailored to the underlying pathologic mechanisms, are needed.
Collapse
Affiliation(s)
| | - Cristina Lupu
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK; and
| | - Florea Lupu
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK; and
- Department of Cell Biology
- Department of Pathology, and
- Department of Internal Medicine, Oklahoma University Health Sciences Center, Oklahoma City, OK
| |
Collapse
|
53
|
|
54
|
Antoniak S, Phungphong S, Cheng Z, Jensen BC. Novel Mechanisms of Anthracycline-Induced Cardiovascular Toxicity: A Focus on Thrombosis, Cardiac Atrophy, and Programmed Cell Death. Front Cardiovasc Med 2022; 8:817977. [PMID: 35111832 PMCID: PMC8801506 DOI: 10.3389/fcvm.2021.817977] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 12/23/2021] [Indexed: 01/13/2023] Open
Abstract
Anthracycline antineoplastic agents such as doxorubicin are widely used and highly effective component of adjuvant chemotherapy for breast cancer and curative regimens for lymphomas, leukemias, and sarcomas. The primary dose-limiting adverse effect of anthracyclines is cardiotoxicity that typically manifests as cardiomyopathy and can progress to the potentially fatal clinical syndrome of heart failure. Decades of pre-clinical research have explicated the complex and multifaceted mechanisms of anthracycline-induced cardiotoxicity. It is well-established that oxidative stress contributes to the pathobiology and recent work has elucidated important central roles for direct mitochondrial injury and iron overload. Here we focus instead on emerging aspects of anthracycline-induced cardiotoxicity that may have received less attention in other recent reviews: thrombosis, myocardial atrophy, and non-apoptotic programmed cell death.
Collapse
Affiliation(s)
- Silvio Antoniak
- Department of Pathology and Laboratory Medicine, University of North Carolina School of Medicine, Chapel Hill, NC, United States
- Blood Research Center, University of North Carolina School of Medicine, Chapel Hill, NC, United States
- *Correspondence: Silvio Antoniak
| | - Sukanya Phungphong
- Department of Pharmaceutical Sciences, Washington State University, Spokane, WA, United States
| | - Zhaokang Cheng
- Department of Pharmaceutical Sciences, Washington State University, Spokane, WA, United States
- Zhaokang Cheng
| | - Brian C. Jensen
- Cardiology Division, Department of Medicine, University of North Carolina School of Medicine, Chapel Hill, NC, United States
- Department of Pharmacology, University of North Carolina School of Medicine, Chapel Hill, NC, United States
- McAllister Heart Institute, University of North Carolina School of Medicine, Chapel Hill, NC, United States
| |
Collapse
|
55
|
Abstract
TAM receptors (Tyro3, Axl and MerTK) are a family of tyrosine kinase receptors that are expressed in a variety of cell populations, including liver parenchymal and non-parenchymal cells. These receptors are vital for immune homeostasis, as they regulate the innate immune response by suppressing inflammation via toll-like receptor inhibition and by promoting tissue resolution through efferocytosis. However, there is increasing evidence indicating that aberrant TAM receptor signaling may play a role in pathophysiological processes in the context of liver disease. This review will explore the roles of TAM receptors and their ligands in liver homeostasis as well as a variety of disease settings, including acute liver injury, steatosis, fibrosis, cirrhosis-associated immune dysfunction and hepatocellular carcinoma. A better understanding of our current knowledge of TAM receptors in liver disease may identify new opportunities for disease monitoring as well as novel therapeutic targets. Nonetheless, this review also aims to highlight areas where further research on TAM receptor biology in liver disease is required.
Collapse
|
56
|
Kamamoto T, Nakajima Y, Uchida Y, Nakagawa T, Tonegawa H, Tani Y, Nishimoto E, Takahashi Y, Nishikubo T, Nogami K. Protein C system in preterm babies with chronic lung disease: Prospective study. Pediatr Int 2022; 64:e15221. [PMID: 35912452 DOI: 10.1111/ped.15221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 04/09/2022] [Accepted: 04/15/2022] [Indexed: 11/28/2022]
Abstract
BACKGROUND Chronic lung disease (CLD) is a major neonatal pulmonary disorder associated with inflammation. Recent studies have shown that protein C anticoagulant pathways, such as those for protein C (PC), protein S (PS), and thrombomodulin (TM), could be useful indices for reflecting pulmonary injury. However, the involvement of these factors in preterm infants with very low birthweight (VLBW) who have developed CLD remains to be investigated. Here, we investigated whether PC pathway-related factors could predict the development of CLD in preterm infants with VLBW. METHODS We collected plasma samples from 26 preterm infants with VLBW (13 each from those with and without CLD) at the time of birth and measured TM, PC, and PS levels in their plasmas. We analyzed prospectively the relationship between these factors in infants with and without CLD. RESULTS There were significant differences in gestational age, birthweight, Apgar score (5 min), and duration of mechanical ventilation between the CLD and non-CLD groups. No significant differences in the PC and PS levels at birth were observed between the two groups, whereas the TM levels in the CLD group were significantly higher than those in the non-CLD group (P = 0.013). The TM levels correlated with gestational age and duration of mechanical ventilation. However, covariance analysis demonstrated that gestational age was significantly associated with TM levels, and consequently, development of CLD was not associated with TM level at birth. CONCLUSIONS Thrombomodulin, PC, and PS levels at birth could not predict the development of CLD in preterm infants with VLBW.
Collapse
Affiliation(s)
- Tomoyuki Kamamoto
- Division of Neonatal Intensive Care, Center of Perinatal Medicine, Nara Medical University Hospital, Kashihara, Nara, Japan
| | - Yuto Nakajima
- Department of Pediatrics, Nara Medical University, Kashihara, Nara, Japan.,Advanced Medical Science of Thrombosis and Hemostasis, Nara Medical University, Kashihara, Nara, Japan
| | - Yumiko Uchida
- Division of Neonatal Intensive Care, Center of Perinatal Medicine, Nara Medical University Hospital, Kashihara, Nara, Japan
| | - Takashi Nakagawa
- Division of Neonatal Intensive Care, Center of Perinatal Medicine, Nara Medical University Hospital, Kashihara, Nara, Japan
| | - Hitoshi Tonegawa
- Division of Neonatal Intensive Care, Center of Perinatal Medicine, Nara Medical University Hospital, Kashihara, Nara, Japan
| | - Yuki Tani
- Division of Neonatal Intensive Care, Center of Perinatal Medicine, Nara Medical University Hospital, Kashihara, Nara, Japan
| | - Eri Nishimoto
- Division of Neonatal Intensive Care, Center of Perinatal Medicine, Nara Medical University Hospital, Kashihara, Nara, Japan
| | | | - Toshiya Nishikubo
- Division of Neonatal Intensive Care, Center of Perinatal Medicine, Nara Medical University Hospital, Kashihara, Nara, Japan
| | - Keiji Nogami
- Department of Pediatrics, Nara Medical University, Kashihara, Nara, Japan
| |
Collapse
|
57
|
Rajagopal S, Gupta A, Parveen R, Shukla N, Bhattacharya S, Naravula J, Kumar S A, Mathur P, Simlot A, Mehta S, Bihari C, Mehta S, Mishra AK, Nair BG, Medicherla KM, Reddy GB, Sreenivasulu N, Kishor PK, Suravajhala P. Vitamin K in human health and metabolism: A nutri-genomics review. Trends Food Sci Technol 2022. [DOI: 10.1016/j.tifs.2021.12.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
58
|
Elieh Ali Komi D, Rahimi Y, Asghari R, Jafari R, Rasouli J, Mohebalizadeh M, Abbasi A, Nejadrahim R, Rezazadeh F, Shafiei-Irannejad V. Investigation of the Molecular Mechanism of Coagulopathy in Severe and Critical Patients With COVID-19. Front Immunol 2021; 12:762782. [PMID: 34975853 PMCID: PMC8716500 DOI: 10.3389/fimmu.2021.762782] [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: 08/22/2021] [Accepted: 11/26/2021] [Indexed: 01/22/2023] Open
Abstract
Coagulopathy is a frequently reported finding in the pathology of coronavirus disease 2019 (COVID-19); however, the molecular mechanism, the involved coagulation factors, and the role of regulatory proteins in homeostasis are not fully investigated. We explored the dynamic changes of nine coagulation tests in patients and controls to propose a molecular mechanism for COVID-19-associated coagulopathy. Coagulation tests including prothrombin time (PT), partial thromboplastin time (PTT), fibrinogen (FIB), lupus anticoagulant (LAC), proteins C and S, antithrombin III (ATIII), D-dimer, and fibrin degradation products (FDPs) were performed on plasma collected from 105 individuals (35 critical patients, 35 severe patients, and 35 healthy controls). There was a statically significant difference when the results of the critical (CRT) and/or severe (SVR) group for the following tests were compared to the control (CRL) group: PTCRT (15.014) and PTSVR (13.846) (PTCRL = 13.383, p < 0.001), PTTCRT (42.923) and PTTSVR (37.8) (PTTCRL = 36.494, p < 0.001), LACCRT (49.414) and LACSVR (47.046) (LACCRL = 40.763, p < 0.001), FIBCRT (537.66) and FIBSVR (480.29) (FIBCRL = 283.57, p < 0.001), ProCCRT (85.57%) and ProCSVR (99.34%) (ProCCRL = 94.31%, p = 0.04), ProSCRT (62.91%) and ProSSVR (65.06%) (ProSCRL = 75.03%, p < 0.001), D-dimer (p < 0.0001, χ2 = 34.812), and FDP (p < 0.002, χ2 = 15.205). No significant association was found in the ATIII results in groups (ATIIICRT = 95.71% and ATIIISVR = 99.63%; ATIIICRL = 98.74%, p = 0.321). D-dimer, FIB, PT, PTT, LAC, protein S, FDP, and protein C (ordered according to p-values) have significance in the prognosis of patients. Disruptions in homeostasis in protein C (and S), VIII/VIIIa and V/Va axes, probably play a role in COVID-19-associated coagulopathy.
Collapse
Affiliation(s)
- Daniel Elieh Ali Komi
- Cellular and Molecular Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia, Iran
| | - Yaghoub Rahimi
- Cellular and Molecular Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia, Iran
| | - Rahim Asghari
- Hematology, Immune Cell Therapy, and Stem Cells Transplantation Research Center, Clinical Research Institute, Urmia University of Medical Sciences, Urmia, Iran
| | - Reza Jafari
- Hematology, Immune Cell Therapy, and Stem Cells Transplantation Research Center, Clinical Research Institute, Urmia University of Medical Sciences, Urmia, Iran
- Nephrology and Kidney Transplant Research Center, Clinical Research Institute, Urmia University of Medical Sciences, Urmia, Iran
| | - Javad Rasouli
- Department of Epidemiology and Biostatistics, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Mehdi Mohebalizadeh
- Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran
| | - Ata Abbasi
- Department of Pathology, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Rahim Nejadrahim
- Department of Infectious Diseases, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Farzin Rezazadeh
- Department of Emergency Medicine, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Vahid Shafiei-Irannejad
- Cellular and Molecular Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia, Iran
| |
Collapse
|
59
|
Santler SC, Konstantiniuk P, Schramayer G, Prüller F, Siegl G, Demel U, Cohnert T. The impact of activated protein C resistance on the patency of arteriovenous grafts for hemodialysis access. Semin Dial 2021; 35:58-65. [PMID: 34882835 DOI: 10.1111/sdi.13042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 08/03/2021] [Accepted: 10/09/2021] [Indexed: 11/27/2022]
Abstract
INTRODUCTION Vascular access is required for hemodialysis treatment. An effect of activated protein C resistance on access thrombosis rates has not yet been investigated. The aim of this study is to determine whether an activated protein C resistance is correlated with the patency of polytetrafluoroethylene arteriovenous grafts. METHODS The primary endpoint was the impact of activated protein C resistance; secondary endpoints were the influence of Factor V Leiden thrombophilia, homocysteine, ß2-glycoprotein antibodies, and other laboratory values on the assisted primary patency. RESULTS Forty-three grafts in 43 patients were included. The overall mean assisted primary patency was 18.4 months (±3.16 SE). Activated protein C resistance (p = 0.01) and ß2-glycoprotein antibodies (p = 0.018) had a significant influence on the assisted primary patency. The assisted primary patency for patients with low (<4) activated protein C resistance was 9.3 months compared to 24.8 of those with a high (≥4) activated protein C resistance. Patients with low (≤2.6) ß2-glycoprotein antibodies presented an assisted primary patency of 31.8 months whereas those with high (>2.6) ß2-glycoprotein antibodies showed 9.3 months. In all patients with a pathologic activated protein C resistance, a heterozygous or homozygous Factor V Leiden thrombophilia was detected. CONCLUSIONS This study identified low activated protein C resistance and high ß2-glycoprotein antibodies as risk factors for thrombosis in polytetrafluoroethylene arteriovenous grafts. A prospective study is needed to clarify if oral anticoagulation should be administered to all patients with a pathologic activated protein C resistance blood value and/or factor V Leiden mutation.
Collapse
Affiliation(s)
| | - Peter Konstantiniuk
- Department of Surgery, Division of Vascular Surgery, Medical University of Graz, Graz, Austria
| | - Georg Schramayer
- Department of Surgery, Division of Vascular Surgery, Medical University of Graz, Graz, Austria
| | - Florian Prüller
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Gregor Siegl
- Department of Surgery, Division of Vascular Surgery, Medical University of Graz, Graz, Austria
| | - Ulike Demel
- Department of Internal Medicine, Division of Rheumatology and Immunology, Medical University of Graz, Graz, Austria
| | - Tina Cohnert
- Department of Surgery, Division of Vascular Surgery, Medical University of Graz, Graz, Austria
| |
Collapse
|
60
|
Zhang YY, Ning BT. Signaling pathways and intervention therapies in sepsis. Signal Transduct Target Ther 2021; 6:407. [PMID: 34824200 PMCID: PMC8613465 DOI: 10.1038/s41392-021-00816-9] [Citation(s) in RCA: 147] [Impact Index Per Article: 36.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 10/19/2021] [Accepted: 10/26/2021] [Indexed: 12/12/2022] Open
Abstract
Sepsis is defined as life-threatening organ dysfunction caused by dysregulated host systemic inflammatory and immune response to infection. Over decades, advanced understanding of host-microorganism interaction has gradually unmasked the genuine nature of sepsis, guiding toward new definition and novel therapeutic approaches. Diverse clinical manifestations and outcomes among infectious patients have suggested the heterogeneity of immunopathology, while systemic inflammatory responses and deteriorating organ function observed in critically ill patients imply the extensively hyperactivated cascades by the host defense system. From focusing on microorganism pathogenicity, research interests have turned toward the molecular basis of host responses. Though progress has been made regarding recognition and management of clinical sepsis, incidence and mortality rate remain high. Furthermore, clinical trials of therapeutics have failed to obtain promising results. As far as we know, there was no systematic review addressing sepsis-related molecular signaling pathways and intervention therapy in literature. Increasing studies have succeeded to confirm novel functions of involved signaling pathways and comment on efficacy of intervention therapies amid sepsis. However, few of these studies attempt to elucidate the underlining mechanism in progression of sepsis, while other failed to integrate preliminary findings and describe in a broader view. This review focuses on the important signaling pathways, potential molecular mechanism, and pathway-associated therapy in sepsis. Host-derived molecules interacting with activated cells possess pivotal role for sepsis pathogenesis by dynamic regulation of signaling pathways. Cross-talk and functions of these molecules are also discussed in detail. Lastly, potential novel therapeutic strategies precisely targeting on signaling pathways and molecules are mentioned.
Collapse
Affiliation(s)
- Yun-Yu Zhang
- Department of Pediatric Intensive Care Unit, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, 200127, Shanghai, China
| | - Bo-Tao Ning
- Department of Pediatric Intensive Care Unit, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, 200127, Shanghai, China.
| |
Collapse
|
61
|
Vitamin K Effects on Gas6 and Soluble Axl Receptors in Intensive Care Patients: An Observational Screening Study. Nutrients 2021; 13:nu13114101. [PMID: 34836355 PMCID: PMC8621311 DOI: 10.3390/nu13114101] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 10/29/2021] [Accepted: 11/11/2021] [Indexed: 12/14/2022] Open
Abstract
Growth arrest-specific gene 6 protein (Gas6) is avitamin K-dependent tissue bound protein. Gas6 has been shown to promote growth and therapy resistance among different types of cancer as well as thromboembolism. The aim of this prospective screening study: ClinicalTrials.gov; Identifier: NTC3782025, was to evaluate the effects of intravenously administered vitamin K1 on Gas6 and its soluble (s)Axl receptor plasma levels in intensive care patients. Vitamin K1 was intravenously injected in non-warfarin treated patients with prolonged Owren prothrombin time international normalized ratio (PT-INR) > 1.2 and blood samples were retrieved before and 20-28 h after injection. Citrate plasma samples from 52 intensive care patients were analysed for different vitamin K dependent proteins. There was a significant, but small increase in median Gas6. Only one patient had a large increase in sAxl, but overall, no significant changes in sAxl Gas6 did not correlate to PT-INR, thrombin generation assay, coagulation factors II, VII, IX and X, but to protein S and decarboxylated matrix Gla protein (dp-ucMGP). In conclusion, there was a small increase in Gas6 over 20-28 h. The pathophysiology and clinical importance of this remains to be investigated. To verify a true vitamin K effect, improvement of Gas6 carboxylation defects needs to be studied.
Collapse
|
62
|
Reda S, Rühl H, Witkowski J, Müller J, Pavlova A, Oldenburg J, Pötzsch B. PC Deficiency Testing: Thrombin-Thrombomodulin as PC Activator and Aptamer-Based Enzyme Capturing Increase Diagnostic Accuracy. Front Cardiovasc Med 2021; 8:755281. [PMID: 34708097 PMCID: PMC8542722 DOI: 10.3389/fcvm.2021.755281] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Accepted: 09/12/2021] [Indexed: 11/30/2022] Open
Abstract
Protein C (PC) activity tests are routinely performed in a thrombophilia workup to screen for PC deficiency. Currently used tests combine conversion of PC to activated PC (APC) by the snake venom Protac with subsequent APC detection through hydrolysis of a chromogenic peptide substrate or prolongation of a clotting time. In this prospective cohort study, we analyzed how different modes of PC activation and subsequent APC determination influence the diagnostic accuracy of PC activity testing in a cohort of 31 patients with genetically confirmed PC deficiency. In addition to chromogenic and clot-based measurement, an oligonucleotide-based enzyme capture assay utilizing a basic exosite-targeting aptamer was used for APC detection. To study the influence of the PC activation step on diagnostic sensitivity, PC activation through Protac and through the thrombin-thrombomodulin (TM) complex were compared. Twenty-six (84%) and 24 (77%) PC deficient patients were identified as true-positive using the chromogenic and the clot-based PC activity assay, respectively. True-positive results increased to 27 (87%) when the basic exosite-targeting aptamer approach was used for APC measurement. Additional replacement of the PC activator Protac by thrombin-TM gave true-positive results in all patients. These data indicate that the mode of PC activation is crucial in determining the accuracy of PC activity testing and that diagnostic sensitivity can be significantly improved by replacing the PC activator Protac with thrombin-TM. APC detection using a basic exosite-targeting aptamer achieves high sensitivity toward mutations outside the active center while being less subject to interfering factors than clot-based PC activity assays.
Collapse
Affiliation(s)
- Sara Reda
- Institute of Experimental Hematology and Transfusion Medicine, University Hospital Bonn, Bonn, Germany
| | - Heiko Rühl
- Institute of Experimental Hematology and Transfusion Medicine, University Hospital Bonn, Bonn, Germany
| | - Jana Witkowski
- Institute of Experimental Hematology and Transfusion Medicine, University Hospital Bonn, Bonn, Germany
| | - Jens Müller
- Institute of Experimental Hematology and Transfusion Medicine, University Hospital Bonn, Bonn, Germany
| | - Anna Pavlova
- Institute of Experimental Hematology and Transfusion Medicine, University Hospital Bonn, Bonn, Germany
| | - Johannes Oldenburg
- Institute of Experimental Hematology and Transfusion Medicine, University Hospital Bonn, Bonn, Germany
| | - Bernd Pötzsch
- Institute of Experimental Hematology and Transfusion Medicine, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
63
|
Ziegler O, Sriram N, Gelev V, Radeva D, Todorov K, Feng J, Sellke FW, Robson SC, Hiromura M, Alexandrov BS, Usheva A. The cardiac molecular setting of metabolic syndrome in pigs reveals disease susceptibility and suggests mechanisms that exacerbate COVID-19 outcomes in patients. Sci Rep 2021; 11:19752. [PMID: 34611227 PMCID: PMC8492658 DOI: 10.1038/s41598-021-99143-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 07/21/2021] [Indexed: 12/02/2022] Open
Abstract
Although metabolic syndrome (MetS) is linked to an elevated risk of cardiovascular disease (CVD), the cardiac-specific risk mechanism is unknown. Obesity, hypertension, and diabetes (all MetS components) are the most common form of CVD and represent risk factors for worse COVID-19 outcomes compared to their non MetS peers. Here, we use obese Yorkshire pigs as a highly relevant animal model of human MetS, where pigs develop the hallmarks of human MetS and reproducibly mimics the myocardial pathophysiology in patients. Myocardium-specific mass spectroscopy-derived metabolomics, proteomics, and transcriptomics enabled the identity and quality of proteins and metabolites to be investigated in the myocardium to greater depth. Myocardium-specific deregulation of pro-inflammatory markers, propensity for arterial thrombosis, and platelet aggregation was revealed by computational analysis of differentially enriched pathways between MetS and control animals. While key components of the complement pathway and the immune response to viruses are under expressed, key N6-methyladenosin RNA methylation enzymes are largely overexpressed in MetS. Blood tests do not capture the entirety of metabolic changes that the myocardium undergoes, making this analysis of greater value than blood component analysis alone. Our findings create data associations to further characterize the MetS myocardium and disease vulnerability, emphasize the need for a multimodal therapeutic approach, and suggests a mechanism for observed worse outcomes in MetS patients with COVID-19 comorbidity.
Collapse
Affiliation(s)
- Olivia Ziegler
- Division of Cardiothoracic Surgery, Department of Surgery and, The Warren Alpert Medical School, Brown University, Providence, RI, 02903, USA
| | - Nivedita Sriram
- Division of Cardiothoracic Surgery, Department of Surgery and, The Warren Alpert Medical School, Brown University, Providence, RI, 02903, USA
| | - Vladimir Gelev
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02115, USA
- Department of Chemistry, Sofia University, Sofia, Bulgaria
| | - Denitsa Radeva
- Department of Chemistry, Sofia University, Sofia, Bulgaria
| | - Kostadin Todorov
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02115, USA
- Medical University, Sofia, Bulgaria
| | - Jun Feng
- Division of Cardiothoracic Surgery, Department of Surgery and, The Warren Alpert Medical School, Brown University, Providence, RI, 02903, USA
| | - Frank W Sellke
- Division of Cardiothoracic Surgery, Department of Surgery and, The Warren Alpert Medical School, Brown University, Providence, RI, 02903, USA
| | - Simon C Robson
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02115, USA
| | - Makoto Hiromura
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02115, USA
- Daiichi University of Pharmacy, Fukuoka, 815-8511, Japan
| | | | - Anny Usheva
- Division of Cardiothoracic Surgery, Department of Surgery and, The Warren Alpert Medical School, Brown University, Providence, RI, 02903, USA.
| |
Collapse
|
64
|
Gruzdeva OV, Bychkova EE, Penskaya TY, Kuzmina AA, Antonova LV, Barbarash LS. Comparative Analysis of the Hemostasiological Profile in Sheep and Patients with Cardiovascular Pathology as the Basis for Predicting Thrombotic Risks During Preclinical Tests of Vascular Prostheses. Sovrem Tekhnologii Med 2021; 13:52-56. [PMID: 34513066 PMCID: PMC8353693 DOI: 10.17691/stm2021.13.1.06] [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: 10/01/2020] [Indexed: 11/29/2022] Open
Abstract
The aim of the investigation was to study the details of hemostasiological profile in sheep and patients with coronary heart disease (CHD) and to find the possibility of predicting thrombotic risks during preclinical tests of vascular prostheses on a large laboratory animal model.
Collapse
Affiliation(s)
- O V Gruzdeva
- Head of the Laboratory of Homeostasis Research, Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Blvd, Kemerovo, 650002, Russia
| | - E E Bychkova
- Research Assistant, Laboratory of Homeostasis Research, Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Blvd, Kemerovo, 650002, Russia
| | - T Yu Penskaya
- Junior Researcher, Laboratory of Homeostasis Research, Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Blvd, Kemerovo, 650002, Russia
| | - A A Kuzmina
- Junior Researcher, Laboratory of Homeostasis Research, Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Blvd, Kemerovo, 650002, Russia
| | - L V Antonova
- Head of the Laboratory of Cell Technologies, Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Blvd, Kemerovo, 650002, Russia
| | - L S Barbarash
- Professor, Academician of the Russian Academy of Sciences, Chief Researcher, Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Blvd, Kemerovo, 650002, Russia
| |
Collapse
|
65
|
Rezus E, Tamba BI, Badescu MC, Popescu D, Bratoiu I, Rezus C. Osteonecrosis of the Femoral Head in Patients with Hypercoagulability-From Pathophysiology to Therapeutic Implications. Int J Mol Sci 2021; 22:ijms22136801. [PMID: 34202897 PMCID: PMC8268880 DOI: 10.3390/ijms22136801] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/19/2021] [Accepted: 06/21/2021] [Indexed: 12/24/2022] Open
Abstract
Osteonecrosis of the femoral head (ONFH) is a debilitating disease with major social and economic impacts. It frequently affects relatively young adults and has a predilection for rapid progression to femoral head collapse and end-stage hip arthritis. If not diagnosed and treated properly in the early stages, ONFH has devastating consequences and leads to mandatory total hip arthroplasty. The pathophysiology of non-traumatic ONFH is very complex and not fully understood. While multiple risk factors have been associated with secondary ONFH, there are still many cases in which a clear etiology cannot be established. Recognition of the prothrombotic state as part of the etiopathogeny of primary ONFH provides an opportunity for early medical intervention, with implications for both prophylaxis and therapy aimed at slowing or stopping the progression of the disease. Hereditary thrombophilia and hypofibrinolysis are associated with thrombotic occlusion of bone vessels. Anticoagulant treatment can change the natural course of the disease and improve patients' quality of life. The present work focused on highlighting the association between hereditary thrombophilia/hypofibrinolysis states and ONFH, emphasizing the importance of identifying this condition. We have also provided strong arguments to support the efficiency and safety of anticoagulant treatment in the early stages of the disease, encouraging etiological diagnosis and prompt therapeutic intervention. In the era of direct oral anticoagulants, new therapeutic options have become available, enabling better long-term compliance.
Collapse
Affiliation(s)
- Elena Rezus
- Department of Rheumatology and Physiotherapy, “Grigore T. Popa” University of Medicine and Pharmacy Iași, 16 University Street, 700115 Iasi, Romania; (E.R.); (I.B.)
- I Rheumatology Clinic, Clinical Rehabilitation Hospital, 14 Pantelimon Halipa Street, 700661 Iasi, Romania
| | - Bogdan Ionel Tamba
- Advanced Center for Research and Development in Experimental Medicine (CEMEX), “Grigore T. Popa” University of Medicine and Pharmacy Iasi, 9-13 Mihail Kogălniceanu Street, 700454 Iasi, Romania;
| | - Minerva Codruta Badescu
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania; (D.P.); (C.R.)
- III Internal Medicine Clinic, “St. Spiridon” County Emergency Clinical Hospital, 1 Independence Boulevard, 700111 Iasi, Romania
- Correspondence:
| | - Diana Popescu
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania; (D.P.); (C.R.)
- III Internal Medicine Clinic, “St. Spiridon” County Emergency Clinical Hospital, 1 Independence Boulevard, 700111 Iasi, Romania
| | - Ioana Bratoiu
- Department of Rheumatology and Physiotherapy, “Grigore T. Popa” University of Medicine and Pharmacy Iași, 16 University Street, 700115 Iasi, Romania; (E.R.); (I.B.)
- I Rheumatology Clinic, Clinical Rehabilitation Hospital, 14 Pantelimon Halipa Street, 700661 Iasi, Romania
| | - Ciprian Rezus
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania; (D.P.); (C.R.)
- III Internal Medicine Clinic, “St. Spiridon” County Emergency Clinical Hospital, 1 Independence Boulevard, 700111 Iasi, Romania
| |
Collapse
|
66
|
Abstract
A receptor for phospholipid antibodies drives clotting and inflammation
Collapse
Affiliation(s)
- Mariana J Kaplan
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
67
|
Thrombomodulin is essential for maintaining quiescence in vascular endothelial cells. Proc Natl Acad Sci U S A 2021; 118:2022248118. [PMID: 33836597 DOI: 10.1073/pnas.2022248118] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Thrombomodulin (TM) is a thrombin receptor on endothelial cells that is involved in promoting activation of the anticoagulant protein C pathway during blood coagulation. TM also exerts protective anti-inflammatory properties through a poorly understood mechanism. In this study, we investigated the importance of TM signaling to cellular functions by deleting it from endothelial cells by CRISPR-Cas9 technology and analyzed the resultant phenotype of TM-deficient (TM -/- ) cells. Deficiency of TM in endothelial cells resulted in increased basal permeability and hyperpermeability when stimulated by thrombin and TNF-α. The loss of the basal barrier permeability function was accompanied by increased tyrosine phosphorylation of VE-cadherin and reduced polymerization of F-actin filaments at cellular junctions. A significant increase in basal NF-κB signaling and expression of inflammatory cell adhesion molecules was observed in TM -/- cells that resulted in enhanced adhesion of leukocytes to TM -/- cells in flow chamber experiments. There was also a marked increase in expression, storage, and release of the von Willebrand factor (VWF) and decreased storage and release of angiopoietin-2 in TM -/- cells. In a flow chamber assay, isolated platelets adhered to TM -/- cells, forming characteristic VWF-platelet strings. Increased VWF levels and inflammatory foci were also observed in the lungs of tamoxifen-treated ERcre-TMf/f mice. Reexpression of the TM construct in TM -/- cells, but not treatment with soluble TM, normalized the cellular phenotype. Based on these results, we postulate cell-bound TM endows a quiescent cellular phenotype by tightly regulating expression of procoagulant, proinflammatory, and angiogenic molecules in vascular endothelial cells.
Collapse
|
68
|
Rehill AM, McCluskey S, O'Donnell JS, Dockal M, Preston RJS. Heterogeneity in Bleeding Tendency and Arthropathy Development in Individuals with Hemophilia. Semin Thromb Hemost 2021; 47:183-191. [PMID: 33636749 DOI: 10.1055/s-0041-1723769] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
People with hemophilia (PWH) have an increased tendency to bleed, often into their joints, causing debilitating joint disease if left untreated. To reduce the incidence of bleeding events, PWH receive prophylactic replacement therapy with recombinant factor VIII (FVIII) or FIX. Bleeding events in PWH are typically proportional to their plasma FVIII or IX levels; however, in many PWH, bleeding tendency and the likelihood of developing arthropathy often varies independently of endogenous factor levels. Consequently, many PWH suffer repeated bleeding events before correct dosing of replacement factor can be established. Diagnostic approaches to define an individual's bleeding tendency remain limited. Multiple modulators of bleeding phenotype in PWH have been proposed, including the type of disease-causing variant, age of onset of bleeding episodes, plasma modifiers of blood coagulation or clot fibrinolysis pathway activity, interindividual differences in platelet reactivity, and endothelial anticoagulant activity. In this review, we summarize current knowledge of established factors modulating bleeding tendency and discuss emerging concepts of additional biological elements that may contribute to variable bleeding tendency in PWH. Finally, we consider how variance in responses to new gene therapies may also necessitate consideration of patient-specific tailoring of treatment. Cumulatively, these studies highlight the need to reconsider the current "one size fits all" approach to treatment regimens for PWH and consider therapies guided by the bleeding phenotype of each individual PWH at the onset of therapy. Further characterization of the biological bases of bleeding heterogeneity in PWH, combined with the development of novel diagnostic assays to identify those factors that modulate bleeding risk in PWH, will be required to meet these aspirations.
Collapse
Affiliation(s)
- Aisling M Rehill
- Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Seán McCluskey
- Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, Dublin, Ireland.,National Children's Research Centre, Our Lady's Children's Hospital, Crumlin, Dublin, Ireland
| | - James S O'Donnell
- Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, Dublin, Ireland.,National Children's Research Centre, Our Lady's Children's Hospital, Crumlin, Dublin, Ireland.,St James' Hospital, Dublin, Ireland
| | - Michael Dockal
- Baxalta Innovations GmbH, A Member of the Takeda Group of Companies, Vienna, Austria
| | - Roger J S Preston
- Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, Dublin, Ireland.,National Children's Research Centre, Our Lady's Children's Hospital, Crumlin, Dublin, Ireland
| | | |
Collapse
|
69
|
Portal Vein Thrombosis following Total Colectomy due to Colonic Inertia: A Case Report and Evaluation of Risk Factors. Case Rep Hematol 2021; 2021:8895206. [PMID: 33532102 PMCID: PMC7840229 DOI: 10.1155/2021/8895206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Revised: 11/04/2020] [Accepted: 01/12/2021] [Indexed: 11/17/2022] Open
Abstract
The portal vein could be occluded by blood clots partially or completely causing portal vein thrombosis (PVT). The acute episode may be asymptomatic or manifested by abdominal pain, increasing body temperature, and unspecific dyspeptic symptoms. The main causes of PVT are categorized into local, acquired, and genetic thrombophilic factors. To our knowledge, this is the 2nd recognized case of PVT following colectomy for colonic inertia successfully treated with an effective anticoagulation therapy. The patient received unfractionated heparin as soon the diagnosis was implemented. The patient was a 34-year-old lady with chief complaint of severe abdominal pain, nausea, vomiting, and anorexia 10 days after the first hospital admission for subtotal colectomy due to colonic inertia. Spiral abdominal CT scan with intravenous (IV) contrast showed thrombosis in main portal vein with its extension to right and left intrahepatic branches. Our case showed that we should keep in mind PVT in patients who present with upper gastrointestinal symptoms several days after a major surgery (open colectomy) as a risk factor and oral contraceptive pills (OCP) usage, postpregnancy, and immobility as other risk factors, that the protein C, S, and FVL deficiencies were secondary, and that the PVT can be managed by low molecular weight heparin plus oral warfarin therapy in the continue.
Collapse
|
70
|
Marlar RA, Gausman JN, Tsuda H, Rollins-Raval MA, Brinkman HJM. Recommendations for clinical laboratory testing for protein S deficiency: Communication from the SSC committee plasma coagulation inhibitors of the ISTH. J Thromb Haemost 2021; 19:68-74. [PMID: 33405382 DOI: 10.1111/jth.15109] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Revised: 09/01/2020] [Accepted: 09/08/2020] [Indexed: 11/26/2022]
Abstract
Hereditary deficiencies of protein S (PS) increase the risk of venous thrombosis; however, assessing the plasma levels of PS can be difficult because of its complex physiological interactions in plasma, sample-related preanalytical variables, and numerous acquired disease processes. Reliable laboratory assays are essential for accurate evaluation of PS when diagnosing a congenital deficiency based on the plasma phenotype alone. This report presents the current evidence-based recommendations for clinical PS assays as well as when to test for PS abnormalities.
Collapse
Affiliation(s)
- Richard A Marlar
- Department of Pathology, TriCore Reference Laboratories, University of New Mexico, Albuquerque, NM, USA
| | - Jana N Gausman
- OU Medicine, Inc, Laboratory, University of Oklahoma Medical Center, Oklahoma City, OK, USA
| | - Hiroko Tsuda
- Department of Nutritional Sciences, Nakamura Gakuen University, Fukuoka, Japan
| | - Marian A Rollins-Raval
- Department of Pathology, TriCore Reference Laboratories, University of New Mexico, Albuquerque, NM, USA
| | - Herm Jan M Brinkman
- Department of Molecular and Cellular Hemostasis, Sanquin Research, Amsterdam, The Netherlands
| |
Collapse
|
71
|
Clinical and Molecular Study of Common Thrombophilia Mutation Prothrombin G20210A. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1339:331-336. [DOI: 10.1007/978-3-030-78787-5_40] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
72
|
Eliwan H, Omer M, McKenna E, Kelly LA, Nolan B, Regan I, Molloy EJ. Protein C Pathway in Paediatric and Neonatal Sepsis. Front Pediatr 2021; 9:562495. [PMID: 35186813 PMCID: PMC8849213 DOI: 10.3389/fped.2021.562495] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 12/20/2021] [Indexed: 11/24/2022] Open
Abstract
Protein C plays a major role in the physiological regulation of coagulation pathways through inactivation of factor Va, factor VIIIa, and plasminogen activator inhibitor. Protein C is involved in the control of inflammation during sepsis, by inhibiting release of pro-inflammatory cytokines, thereby controlling neutrophil, and monocyte effects on injured tissue. Recombinant human activated protein C (rhAPC) reduced mortality in adult sepsis in earlier studies but had no significant benefit in more recent trials. Protein C levels are reduced during paediatric and neonatal sepsis, which may play a major role in the development of disseminated intravascular thrombosis, purpura fulminans, and multiorgan dysfunction. The role of protein C in paediatric sepsis requires further clinical and immunological evaluation to define the patient subgroups who may benefit from this therapy. Newer versions of rhAPC are under development with less risk of haemorrhage potentially broadening the scope of this intervention.
Collapse
Affiliation(s)
- Hassan Eliwan
- National Children's Research Centre, Dublin, Ireland.,Department of Paediatrics, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland
| | - Murwan Omer
- Department of Paediatrics, Children's Health Ireland at Tallaght, Dublin, Ireland
| | - Ellen McKenna
- Department of Paediatrics, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland
| | - Lynne A Kelly
- National Children's Research Centre, Dublin, Ireland.,Department of Paediatrics, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland.,Trinity Research in Childhood Centre, Dublin, Ireland
| | - Beatrice Nolan
- Department of Haematology, Children's Health Ireland at Crumlin, Dublin, Ireland
| | - Irene Regan
- National Children's Research Centre, Dublin, Ireland.,Department of Haematology, Children's Health Ireland at Crumlin, Dublin, Ireland
| | - Eleanor J Molloy
- National Children's Research Centre, Dublin, Ireland.,Department of Paediatrics, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland.,Department of Paediatrics, Children's Health Ireland at Tallaght, Dublin, Ireland.,Trinity Research in Childhood Centre, Dublin, Ireland.,Department of Neonatology, Children's Health Ireland at Crumlin, Dublin, Ireland.,Department of Paediatrics, Coombe Women's and Infant's University Hospital, Dublin, Ireland
| |
Collapse
|
73
|
Sriwastva MK, Kunjunni R, Andrabi M, Prasad K, Saxena R, Subbiah V. Neuroprotective Effects of Activated Protein C Involve the PARP/AIF Pathway against Oxygen-Glucose Deprivation in SH-SY5Y Cells. Brain Sci 2020; 10:brainsci10120959. [PMID: 33321687 PMCID: PMC7764138 DOI: 10.3390/brainsci10120959] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 12/03/2020] [Accepted: 12/07/2020] [Indexed: 12/20/2022] Open
Abstract
Protein C, a member of the zymogen family of serine proteases in plasma, is one of the several vitamin K dependent glycoproteins known to induce anti-apoptotic activity. However, the target molecule involved in the mechanism needs to be investigated. We sought to investigate the pathways involved in the anti-apoptotic role of activated protein C (APC) on oxygen-glucose deprivation (OGD) induced ischemic conditions in in-vitro SH-SY5Y cells. SH-SY5Y cells were exposed to OGD in an airtight chamber containing 95% N2 and 5% CO2 and media deprived of glucose for 4 h following 24 h of reoxygenation. The cell toxicity, viability, expression of receptors such as endothelial cell protein C receptor (EPCR), protease-activated receptor (PAR)1, PAR3, and apoptosis-related proteins B-cell lymphoma 2 (BCL-2), BCL-2-like protein 4 (Bax), Poly [ADP-ribose] polymerase-1 (PARP-1) were assessed. Administration of APC decreased the cellular injury when compared to the OGD exposed group in a dose-dependent manner and displayed increased expression of PAR-1, PAR-3, and EPCR. The APC treatment leads to a reduction in PARP-1 expression and cleavage and apoptosis-inducing factor (AIF) expression. The reduction of caspase-3 activity and PARP-1 and AIF expression following APC administration results in restoring mitochondrial function with decreased cellular injury and apoptosis. Our results suggested that APC has potent protective effects against in-vitro ischemia in SH-SY5Y cells by modulating mitochondrial function.
Collapse
Affiliation(s)
- Mukesh Kumar Sriwastva
- Department of Neurobiochemistry, All India Institute of Medical Sciences, New Delhi 110029, India; (R.K.); (M.A.); (V.S.)
- Correspondence: ; Tel.: +91-112659-4488
| | - Remesh Kunjunni
- Department of Neurobiochemistry, All India Institute of Medical Sciences, New Delhi 110029, India; (R.K.); (M.A.); (V.S.)
| | - Mutahar Andrabi
- Department of Neurobiochemistry, All India Institute of Medical Sciences, New Delhi 110029, India; (R.K.); (M.A.); (V.S.)
| | - Kameshwar Prasad
- Department of Neurology, All India Institute of Medical Sciences, New Delhi 110029, India;
| | - Renu Saxena
- Department of Hematology, All India Institute of Medical Sciences, New Delhi 110029, India;
| | - Vivekanandhan Subbiah
- Department of Neurobiochemistry, All India Institute of Medical Sciences, New Delhi 110029, India; (R.K.); (M.A.); (V.S.)
| |
Collapse
|
74
|
Shrimp J, Kales SC, Sanderson PE, Simeonov A, Shen M, Hall MD. An Enzymatic TMPRSS2 Assay for Assessment of Clinical Candidates and Discovery of Inhibitors as Potential Treatment of COVID-19. ACS Pharmacol Transl Sci 2020; 3:997-1007. [PMID: 33062952 PMCID: PMC7507803 DOI: 10.1021/acsptsci.0c00106] [Citation(s) in RCA: 91] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Indexed: 12/12/2022]
Abstract
SARS-CoV-2 is the viral pathogen causing the COVID19 global pandemic. Consequently, much research has gone into the development of preclinical assays for the discovery of new or repurposing of FDA-approved therapies. Preventing viral entry into a host cell would be an effective antiviral strategy. One mechanism for SARS-CoV-2 entry occurs when the spike protein on the surface of SARS-CoV-2 binds to an ACE2 receptor followed by cleavage at two cut sites ("priming") that causes a conformational change allowing for viral and host membrane fusion. TMPRSS2 has an extracellular protease domain capable of cleaving the spike protein to initiate membrane fusion. A validated inhibitor of TMPRSS2 protease activity would be a valuable tool for studying the impact TMPRSS2 has in viral entry and potentially be an effective antiviral therapeutic. To enable inhibitor discovery and profiling of FDA-approved therapeutics, we describe an assay for the biochemical screening of recombinant TMPRSS2 suitable for high throughput application. We demonstrate effectiveness to quantify inhibition down to subnanomolar concentrations by assessing the inhibition of camostat, nafamostat, and gabexate, clinically approved agents in Japan. Also, we profiled a camostat metabolite, FOY-251, and bromhexine hydrochloride, an FDA-approved mucolytic cough suppressant. The rank order potency for the compounds tested are nafamostat (IC50 = 0.27 nM), camostat (IC50 = 6.2 nM), FOY-251 (IC50 = 33.3 nM), and gabexate (IC50 = 130 nM). Bromhexine hydrochloride showed no inhibition of TMPRSS2. Further profiling of camostat, nafamostat, and gabexate against a panel of recombinant proteases provides insight into selectivity and potency.
Collapse
Affiliation(s)
- Jonathan
H. Shrimp
- National
Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850, United States
| | - Stephen C. Kales
- National
Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850, United States
| | - Philip E. Sanderson
- National
Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850, United States
| | - Anton Simeonov
- National
Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850, United States
| | - Min Shen
- National
Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850, United States
| | - Matthew D. Hall
- National
Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850, United States
| |
Collapse
|
75
|
Zhang F, Pu J, Gu Z, Mao H. DIA proteomics reveals hypotensive and immune-enhancing constituents in buffalo whey from different altitudes. Int J Biol Macromol 2020; 164:4146-4154. [PMID: 32882282 DOI: 10.1016/j.ijbiomac.2020.08.213] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Revised: 08/27/2020] [Accepted: 08/27/2020] [Indexed: 11/29/2022]
Abstract
The selection of raw milk with high levels of functional components that have health-promoting activities is very important for the exploitation and production of functional milk, but the differences in the functional components of whey from buffalo raised at different altitudes have not been thoroughly investigated. Here, we detected the effects of altitudes on the functional components in whey from dairy buffalo farms situated at low altitude (LA), medium altitude (MA), and high altitude (HA) sites with data-independent acquisition proteomic approaches. In 33 samples, 9331 peptides corresponding to 1008 high-confidence proteins were detected. HA-whey had a lower level of angiotensinogen than that of the LA- and MA-whey, and conversely contained higher levels of immune-enhancing components than for the latter two groups. Differential proteins were involved in vascular smooth muscle contraction, complement and coagulation cascades, and the secretion, production and regulation pathways in immune components. LA-whey showed higher levels of lymphocyte antigen and selenoprotein F than that of the HA-whey. Owing to the biological functions of their most abundant components, HA- and LA-whey are suitable for the processing of functional milk for lowering blood pressure, and the production of immune milk, respectively.
Collapse
Affiliation(s)
- Fulan Zhang
- Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China
| | - Jinhui Pu
- Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China
| | - Zhaobing Gu
- Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China.
| | - Huaming Mao
- Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China
| |
Collapse
|
76
|
Zöller B, Svensson PJ, Dahlbäck B, Lind-Hallden C, Hallden C, Elf J. Genetic risk factors for venous thromboembolism. Expert Rev Hematol 2020; 13:971-981. [DOI: 10.1080/17474086.2020.1804354] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Bengt Zöller
- Center for Primary Health Care Research, Lund University, Malmö, Sweden
| | - Peter J. Svensson
- Center for Thrombosis and Haemostasis, Lund University, Skåne University Hospital, Malmö, Sweden
| | - Björn Dahlbäck
- Department of Translational Medicine, Lund University, Skåne University Hospital, Malmö, Sweden
| | - Christina Lind-Hallden
- Department of Environmental Science and Bioscience, Kristianstad University, Kristianstad, Sweden
| | - Christer Hallden
- Department of Environmental Science and Bioscience, Kristianstad University, Kristianstad, Sweden
| | - Johan Elf
- Center for Thrombosis and Haemostasis, Lund University, Skåne University Hospital, Malmö, Sweden
| |
Collapse
|
77
|
Shrimp JH, Kales SC, Sanderson PE, Simeonov A, Shen M, Hall MD. An Enzymatic TMPRSS2 Assay for Assessment of Clinical Candidates and Discovery of Inhibitors as Potential Treatment of COVID-19. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2020. [PMID: 32596694 DOI: 10.1101/2020.06.23.167544] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
SARS-CoV-2 is the viral pathogen causing the COVID19 global pandemic. Consequently, much research has gone into the development of pre-clinical assays for the discovery of new or repurposing of FDA-approved therapies. Preventing viral entry into a host cell would be an effective antiviral strategy. One mechanism for SARS-CoV-2 entry occurs when the spike protein on the surface of SARS-CoV-2 binds to an ACE2 receptor followed by cleavage at two cut sites ("priming") that causes a conformational change allowing for viral and host membrane fusion. TMPRSS2 has an extracellular protease domain capable of cleaving the spike protein to initiate membrane fusion. A validated inhibitor of TMPRSS2 protease activity would be a valuable tool for studying the impact TMPRSS2 has in viral entry and potentially be an effective antiviral therapeutic. To enable inhibitor discovery and profiling of FDA-approved therapeutics, we describe an assay for the biochemical screening of recombinant TMPRSS2 suitable for high throughput application. We demonstrate effectiveness to quantify inhibition down to subnanomolar concentrations by assessing the inhibition of camostat, nafamostat and gabexate, clinically approved agents in Japan. Also, we profiled a camostat metabolite, FOY-251, and bromhexine hydrochloride, an FDA-approved mucolytic cough suppressant. The rank order potency for the compounds tested are: nafamostat (IC 50 = 0.27 nM), camostat (IC 50 = 6.2 nM), FOY-251 (IC 50 = 33.3 nM) and gabexate (IC 50 = 130 nM). Bromhexine hydrochloride showed no inhibition of TMPRSS2. Further profiling of camostat, nafamostat and gabexate against a panel of recombinant proteases provides insight into selectivity and potency.
Collapse
|
78
|
De Fenza M, Eremeeva E, Troisi R, Yang H, Esposito A, Sica F, Herdewijn P, D'Alonzo D, Guaragna A. Structure-Activity Relationship Study of a Potent α-Thrombin Binding Aptamer Incorporating Hexitol Nucleotides. Chemistry 2020; 26:9589-9597. [PMID: 32363791 DOI: 10.1002/chem.202001504] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 04/29/2020] [Indexed: 12/13/2022]
Abstract
The replacement of one or more nucleotide residues in the potent α-thrombin-binding aptamer NU172 with hexitol-based nucleotides has been devised to study the effect of these substitutions on the physicochemical and functional properties of the anticoagulant agent. The incorporation of single hexitol nucleotides at the T9 and G18 positions of NU172 substantially retained the physicochemical features of the parent oligonucleotide, as a result of the biomimetic properties of the hexitol backbone. Importantly, the NU172-TH 9 mutant exhibited a higher binding affinity toward human α-thrombin than the native aptamer and an improved stability even after 24 h in 90 % human serum, with a significant increase in the estimated half-life. The anticoagulant activity of the modified oligonucleotide was also found to be slightly preferable to NU172. Overall, these results confirm the potential of hexitol nucleotides as biomimetic agents, while laying the foundations for the development of NU172-inspired α-thrombin-binding aptamers.
Collapse
Affiliation(s)
- Maria De Fenza
- Department of Chemical Sciences, Università degli Studi di Napoli Federico II, via Cintia, 80126, Napoli, Italy
| | - Elena Eremeeva
- Rega Institute for Medical Research, Herestraat 49-box 1041, 3000, Leuven, Belgium
| | - Romualdo Troisi
- Department of Chemical Sciences, Università degli Studi di Napoli Federico II, via Cintia, 80126, Napoli, Italy
| | - Hui Yang
- Rega Institute for Medical Research, Herestraat 49-box 1041, 3000, Leuven, Belgium
| | - Anna Esposito
- Department of Chemical Sciences, Università degli Studi di Napoli Federico II, via Cintia, 80126, Napoli, Italy
| | - Filomena Sica
- Department of Chemical Sciences, Università degli Studi di Napoli Federico II, via Cintia, 80126, Napoli, Italy
| | - Piet Herdewijn
- Rega Institute for Medical Research, Herestraat 49-box 1041, 3000, Leuven, Belgium
| | - Daniele D'Alonzo
- Department of Chemical Sciences, Università degli Studi di Napoli Federico II, via Cintia, 80126, Napoli, Italy
| | - Annalisa Guaragna
- Department of Chemical Sciences, Università degli Studi di Napoli Federico II, via Cintia, 80126, Napoli, Italy
| |
Collapse
|
79
|
Hamedani NS, Müller J, Tolle F, Rühl H, Pezeshkpoor B, Liphardt K, Oldenburg J, Mayer G, Pötzsch B. Selective Modulation of the Protease Activated Protein C Using Exosite Inhibiting Aptamers. Nucleic Acid Ther 2020; 30:276-288. [PMID: 32486960 DOI: 10.1089/nat.2020.0844] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Activated protein C (APC) is a serine protease with anticoagulant and cytoprotective activities. Nonanticoagulant APC mutants show beneficial effects as cytoprotective agents. To study, if such biased APC signaling can be achieved by APC-binding ligands, the aptamer technology has been used. A G-quadruplex-containing aptamer, G-NB3, has been selected that binds to the basic exosite of APC with a KD of 0.2 nM and shows no binding to APC-related serine proteases or the zymogen protein C. G-NB3 inhibits the inactivation of activated cofactors V and VIII with IC50 values of 11.6 and 13.1 nM, respectively, without inhibiting the cytoprotective and anti-inflammatory functions of APC as tested using a staurosporine-induced apoptosis assay and a vascular barrier protection assay. In addition, G-NB3 prolongs the plasma half-life of APC through inhibition of APC-serine protease inhibitor complex formation. These physicochemical and functional characteristics qualify G-NB3 as a promising therapeutic agent usable to enhance the cytoprotective functions of APC without increasing the risk of APC-related hemorrhage.
Collapse
Affiliation(s)
- Nasim Shahidi Hamedani
- Institute of Experimental Hematology and Transfusion Medicine, University of Bonn Medical Center, Bonn, Germany
| | - Jens Müller
- Institute of Experimental Hematology and Transfusion Medicine, University of Bonn Medical Center, Bonn, Germany
| | - Fabian Tolle
- Life and Medical Sciences Institute, University of Bonn, Bonn, Germany
| | - Heiko Rühl
- Institute of Experimental Hematology and Transfusion Medicine, University of Bonn Medical Center, Bonn, Germany
| | - Behnaz Pezeshkpoor
- Institute of Experimental Hematology and Transfusion Medicine, University of Bonn Medical Center, Bonn, Germany
| | - Kerstin Liphardt
- Institute of Experimental Hematology and Transfusion Medicine, University of Bonn Medical Center, Bonn, Germany
| | - Johannes Oldenburg
- Institute of Experimental Hematology and Transfusion Medicine, University of Bonn Medical Center, Bonn, Germany
| | - Günter Mayer
- Life and Medical Sciences Institute, University of Bonn, Bonn, Germany
| | - Bernd Pötzsch
- Institute of Experimental Hematology and Transfusion Medicine, University of Bonn Medical Center, Bonn, Germany
| |
Collapse
|
80
|
Lee J, Park A, Mun S, Kim HJ, Son H, Choi H, Kim D, Lee SJ, Kim JG, Kang HG. Proteomics-Based Identification of Diagnostic Biomarkers Related to Risk Factors and Pathogenesis of Ischemic Stroke. Diagnostics (Basel) 2020; 10:diagnostics10050340. [PMID: 32466277 PMCID: PMC7278009 DOI: 10.3390/diagnostics10050340] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 05/20/2020] [Accepted: 05/22/2020] [Indexed: 12/04/2022] Open
Abstract
Ischemic stroke is caused by blood clot formation and consequent vessel blockage. Proteomic approaches provide a cost-effective alternative to current diagnostic methods, including computerized tomography (CT) scans and magnetic resonance imaging (MRI). To identify diagnostic biomarkers associated with ischemic stroke risk factors, we performed individual proteomic analysis of serum taken from 20 healthy controls and 20 ischemic stroke patients. We then performed SWATH analysis, a data-independent method, to assess quantitative changes in protein expression between the two experimental conditions. Our analysis identified several candidate protein biomarkers, 11 of which were validated by multiple reaction monitoring (MRM) analysis as novel diagnostic biomarkers associated with ischemic stroke risk factors. Our study identifies new biomarkers associated with the risk factors and pathogenesis of ischemic stroke which, to the best of our knowledge, were previously unknown. These markers may be effective in not only the diagnosis but also the prevention and management of ischemic stroke.
Collapse
Affiliation(s)
- Jiyeong Lee
- Department of Biomedical Laboratory Science, School of Medicine, Eulji University, Daejeon 34824, Korea;
| | - Arum Park
- Department of Senior Healthcare, BK21 Plus Program, Graduate School, Eulji University, Seongnam 13135, Korea; (A.P.); (S.M.); (H.-J.K.); (H.S.); (H.C.)
| | - Sora Mun
- Department of Senior Healthcare, BK21 Plus Program, Graduate School, Eulji University, Seongnam 13135, Korea; (A.P.); (S.M.); (H.-J.K.); (H.S.); (H.C.)
| | - Hyo-Jin Kim
- Department of Senior Healthcare, BK21 Plus Program, Graduate School, Eulji University, Seongnam 13135, Korea; (A.P.); (S.M.); (H.-J.K.); (H.S.); (H.C.)
| | - Hyunsong Son
- Department of Senior Healthcare, BK21 Plus Program, Graduate School, Eulji University, Seongnam 13135, Korea; (A.P.); (S.M.); (H.-J.K.); (H.S.); (H.C.)
| | - Hyebin Choi
- Department of Senior Healthcare, BK21 Plus Program, Graduate School, Eulji University, Seongnam 13135, Korea; (A.P.); (S.M.); (H.-J.K.); (H.S.); (H.C.)
| | - Doojin Kim
- Department of Laboratory Medicine, Seongnam Central Hospital, Seongnam 13161, Korea;
| | - Soo Joo Lee
- Department of Neurology, Eulji University Hospital, School of Medicine, Eulji University, Daejeon 35233, Korea; (S.J.L.); (J.G.K.)
| | - Jae Guk Kim
- Department of Neurology, Eulji University Hospital, School of Medicine, Eulji University, Daejeon 35233, Korea; (S.J.L.); (J.G.K.)
| | - Hee-Gyoo Kang
- Department of Senior Healthcare, BK21 Plus Program, Graduate School, Eulji University, Seongnam 13135, Korea; (A.P.); (S.M.); (H.-J.K.); (H.S.); (H.C.)
- Department of Biomedical Laboratory Science, School of Medicine, Eulji University, Seongnam 13135, Korea
- Seongnam Senior Industry Innovation Center, Eulji University, Seongnam 13503, Korea
- Correspondence: ; Tel.: +82-31-740-7315; Fax: +82-31-740-7448
| |
Collapse
|
81
|
Yamashita A, Zhang Y, Sanner MF, Griffin JH, Mosnier LO. C-terminal residues of activated protein C light chain contribute to its anticoagulant and cytoprotective activities. J Thromb Haemost 2020; 18:1027-1038. [PMID: 32017367 PMCID: PMC7380734 DOI: 10.1111/jth.14756] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 01/23/2020] [Accepted: 01/28/2020] [Indexed: 12/18/2022]
Abstract
BACKGROUND Activated protein C (APC) is an important homeostatic blood coagulation protease that conveys anticoagulant and cytoprotective activities. Proteolytic inactivation of factors Va and VIIIa facilitated by cofactor protein S is responsible for APC's anticoagulant effects, whereas cytoprotective effects of APC involve primarily the endothelial protein C receptor (EPCR), protease activated receptor (PAR)1 and PAR3. OBJECTIVE To date, several binding exosites in the protease domain of APC have been identified that contribute to APC's interaction with its substrates but potential contributions of the C-terminus of the light chain have not been studied in detail. METHODS Site-directed Ala-scanning mutagenesis of six positively charged residues within G142-L155 was used to characterize their contributions to APC's anticoagulant and cytoprotective activities. RESULTS AND CONCLUSIONS K151 was involved in protein S dependent-anticoagulant activity of APC with some contribution of K150. 3D structural analysis supported that these two residues were exposed in an extended protein S binding site on one face of APC. Both K150 and K151 were important for PAR1 and PAR3 cleavage by APC, suggesting that this region may also mediate interactions with PARs. Accordingly, APC's cytoprotective activity as determined by endothelial barrier protection was impaired by Ala substitutions of these residues. Thus, both K150 and K151 are involved in APC's anticoagulant and cytoprotective activities. The differential contribution of K150 relative to K151 for protein S-dependent anticoagulant activity and PAR cleavage highlights that binding exosites for protein S binding and for PAR cleavage in the C-terminal region of APC's light chain overlap.
Collapse
Affiliation(s)
- Atsuki Yamashita
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA
| | - Yuqi Zhang
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla
| | - Michel F. Sanner
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla
| | - John H. Griffin
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA
| | - Laurent O. Mosnier
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA
| |
Collapse
|
82
|
Aungraheeta R, FitzGibbon L, Reilly-Stitt C, Mumford AD. Differential effects of direct factor IIa and factor Xa inhibitors in protein C-deficient plasma detected using thrombin generation and viscoelastometry assays. Int J Lab Hematol 2019; 42:126-133. [PMID: 31756037 DOI: 10.1111/ijlh.13126] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 10/27/2019] [Accepted: 10/28/2019] [Indexed: 11/28/2022]
Abstract
INTRODUCTION Protein C (PC) deficiency results in dysregulated thrombin generation and increases thrombosis risk. METHODS In order to investigate the potential effects of anticoagulant drugs in PC deficiency, we evaluated the pharmacodynamic effect of selective direct factor (F) IIa inhibitors (dabigatran and argatroban), selective direct FXa inhibitors (rivaroxaban and apixaban) and an indirect FXa/FIIa inhibitor (enoxaparin) in commercial PC-deficient plasma using thrombin generation and viscoelastometry assays modified to reflect PC anticoagulant activity. RESULTS Endogenous thrombin potential (ETP) and peak thrombin concentration (PTC) were increased in PC-deficient plasma but this corrected completely with PC concentrate. Inhibition of FIIa and FXa with the selective inhibitors also corrected the increased ETP and PTC but required high drug concentrations. There was sustained low-level thrombin generation in PC-deficient plasma with FXa inhibitors but not with FIIa inhibitors. Adding PC concentrate to PC-deficient plasma anticoagulated with dabigatran had little additional effect on ETP or PTC. In contrast, addition of even small quantities of PC concentrate to PC-deficient plasma anticoagulated with rivaroxaban further diminished ETP, primarily by abolishing sustained thrombin generation. In the viscoelastometry assay, the coagulation time was shortened and α-angle increased in PC-deficient plasma. These abnormalities reversed with both dabigatran and rivaroxaban. CONCLUSION The selective direct FXa and FIIa inhibitors at high concentrations both counteracted the abnormal thrombin generation and clot formation observed in PC-deficient plasma, but with qualitative differences in their effects.
Collapse
Affiliation(s)
- Riyaad Aungraheeta
- School of Cellular and Molecular Medicine, Faculty of Life Sciences, University of Bristol, Bristol, UK
| | - Lucy FitzGibbon
- Department of Haematology, University Hospitals Bristol NHS Foundation Trust, Bristol, UK
| | | | - Andrew D Mumford
- School of Cellular and Molecular Medicine, Faculty of Life Sciences, University of Bristol, Bristol, UK.,Department of Haematology, University Hospitals Bristol NHS Foundation Trust, Bristol, UK
| |
Collapse
|
83
|
Zhou J, Shen W, Gu Y, Li M, Shen W. Compound heterozygous mutations identified in severe type I protein S deficiency impaired the secretion of protein S. J Clin Pathol 2019; 73:7-13. [PMID: 31422373 DOI: 10.1136/jclinpath-2019-205956] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 08/06/2019] [Accepted: 08/07/2019] [Indexed: 11/04/2022]
Abstract
AIMS Hereditary protein S (PS) deficiency is one of the natural anticoagulant deficiencies causing thrombophilia. We herein described a young male with recurrent deep venous thrombosis, who was diagnosed as type I PS deficiency with compound heterozygous mutations of PROS1 gene. We aimed to analyse the relationship between the genotype and phenotype detection and investigate the pathological mechanisms of PROS1 mutations causing PS deficiency. METHODS Genetic analysis of PROS1 gene was carried out by direct sequencing. Thrombin generation potential and the inhibition function of thrombin generation by plasma PS were detected by thrombin generation test (TGT). The mRNA transcription level of mutant PS in vitro was measured by real-time PCR, while the protein level was evaluated by western blot and ELISA. Cellular distribution of the protein was further analysed by immunofluorescence. RESULTS Compound heterozygous mutations (PROS1 c.1551_1552delinsG, p.Thr518Argfs*39 and PROS1 c.1681C>T, p.Arg561Trp) were identified in the propositus, and the former one was a novel small indel mutation. TGT results showed impaired inhibition of thrombin generation with the addition of activated protein C in his parents with certain heterozygous mutations. In vitro expression study, p.Thr518Argfs*39 mutant produced truncated protein retained in the cytoplasm, while p.Arg561Trp mutant partially affected the secretion of PS. Both mutations are located in C-terminal sex hormone-binding globulin (SHBG)-like domain of PS. CONCLUSIONS Compound heterozygous mutations identified in the study have strong detrimental effect, causing severe type I PS deficiency in the propositus. SHBG-like domain of PS might play an important role in PS secretion system.
Collapse
Affiliation(s)
- Jingyi Zhou
- Department of Laboratory Medicine, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Wenyan Shen
- Department of Laboratory Medicine, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yi Gu
- Department of Laboratory Medicine, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Min Li
- Department of Laboratory Medicine, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Wei Shen
- Department of Laboratory Medicine, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
84
|
Lang TC, Zhao R, Kim A, Wijewardena A, Vandervord J, McGrath R, Fitzpatrick S, Fulcher G, Jackson CJ. Plasma protein C levels are directly associated with better outcomes in patients with severe burns. Burns 2019; 45:1659-1672. [PMID: 31221425 DOI: 10.1016/j.burns.2019.05.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 03/20/2019] [Accepted: 05/01/2019] [Indexed: 11/30/2022]
Abstract
Protein C circulates in human plasma to regulate inflammation and coagulation. It has shown a crucial role in wound healing in animals, and low plasma levels predict the presence of a wound in diabetic patients. However, no detailed study has measured protein C levels in patients with severe burns over the course of a hospital admission. A severe burn is associated with dysfunction of inflammation and coagulation as well as a significant risk of morbidity and mortality. The current methods of burn assessment have shortcomings in reliability and have limited prognostic value. The discovery of a biomarker that estimates burn severity and predicts clinical events with greater accuracy than current methods may improve management, resource allocation and patient counseling. This is the first study to assess the potential role of protein C as a biomarker of burn severity. We measured the plasma protein C levels of 86 patients immediately following a severe burn, then every three days over the first three weeks of a hospital admission. We also analysed the relationships between burn characteristics, blood test results including plasma protein C levels and clinical events. We used a primary composite outcome of increased support utilisation defined as: a mean intravenous fluid administration volume of five litres or more per day over the first 72 h of admission, a length of stay in the intensive care unit of more than four days, or greater than four surgical procedures during admission. The hypothesis was that low protein C levels would be negatively associated with increased support utilisation. At presentation to hospital after a severe burn, the mean plasma protein C level was 76 ± 20% with a range of 34-130% compared to the normal range of 70-180%. The initial low can be plausibly explained by impaired synthesis, increased degradation and excessive consumption of protein C following a burn. Levels increased gradually over six days then remained at a steady-state until the end of the inpatient study period, day 21. A multivariable regression model (Nagelkerke's R2 = 0.83) showed that the plasma protein C level on admission contributed the most to the ability of the model to predict increased support utilisation (OR = 0.825 (95% CI = 0.698-0.977), P = 0.025), followed by burn size (OR = 1.252 (95% CI = 1.025-1.530), P = 0.027), burn depth (partial thickness was used as the reference, full thickness OR = 80.499 (1.569-4129.248), P = 0.029), and neutrophil count on admission (OR = 1.532 (95% CI = 0.950-2.473), P = 0.08). Together, these four variables predicted increased support utilisation with 93.2% accuracy, 83.3% sensitivity and 97.6% specificity. However if protein C values were disregarded, only 49.5% of the variance was explained, with 82% accuracy, 63% sensitivity and 91.5% specificity. Thus, protein C may be a useful biomarker of burn severity and study replication will enable validation of these novel findings.
Collapse
Affiliation(s)
- Thomas Charles Lang
- Sutton Laboratories Level 10, The Kolling Institute, The University of Sydney, Northern Clinical School, Royal North Shore Hospital, Reserve Rd, St. Leonards, 2065, NSW, Australia; Department of Anaesthesia, Prince of Wales and Sydney Children's Hospitals, Barker St, Randwick, 2031, NSW, Australia.
| | - Ruilong Zhao
- Sutton Laboratories Level 10, The Kolling Institute, The University of Sydney, Northern Clinical School, Royal North Shore Hospital, Reserve Rd, St. Leonards, 2065, NSW, Australia
| | - Albert Kim
- Royal North Shore Hospital, Reserve Rd St., Leonards, 2065, NSW, Australia
| | - Aruna Wijewardena
- Royal North Shore Hospital, Reserve Rd St., Leonards, 2065, NSW, Australia
| | - John Vandervord
- Royal North Shore Hospital, Reserve Rd St., Leonards, 2065, NSW, Australia
| | - Rachel McGrath
- Royal North Shore Hospital, Reserve Rd St., Leonards, 2065, NSW, Australia
| | | | - Gregory Fulcher
- Royal North Shore Hospital, Reserve Rd St., Leonards, 2065, NSW, Australia
| | - Christopher John Jackson
- Sutton Laboratories Level 10, The Kolling Institute, The University of Sydney, Northern Clinical School, Royal North Shore Hospital, Reserve Rd, St. Leonards, 2065, NSW, Australia
| |
Collapse
|
85
|
Myers KV, Amend SR, Pienta KJ. Targeting Tyro3, Axl and MerTK (TAM receptors): implications for macrophages in the tumor microenvironment. Mol Cancer 2019; 18:94. [PMID: 31088471 PMCID: PMC6515593 DOI: 10.1186/s12943-019-1022-2] [Citation(s) in RCA: 294] [Impact Index Per Article: 49.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 05/02/2019] [Indexed: 12/14/2022] Open
Abstract
Tumor-associated macrophages are an abundant cell type in the tumor microenvironment. These macrophages serve as a promising target for treatment of cancer due to their roles in promoting cancer progression and simultaneous immunosuppression. The TAM receptors (Tyro3, Axl and MerTK) are promising therapeutic targets on tumor-associated macrophages. The TAM receptors are a family of receptor tyrosine kinases with shared ligands Gas6 and Protein S that skew macrophage polarization towards a pro-tumor M2-like phenotype. In macrophages, the TAM receptors also promote apoptotic cell clearance, a tumor-promoting process called efferocytosis. The TAM receptors bind the "eat-me" signal phosphatidylserine on apoptotic cell membranes using Gas6 and Protein S as bridging ligands. Post-efferocytosis, macrophages are further polarized to a pro-tumor M2-like phenotype and secrete increased levels of immunosuppressive cytokines. Since M2 polarization and efferocytosis are tumor-promoting processes, the TAM receptors on macrophages serve as exciting targets for cancer therapy. Current TAM receptor-directed therapies in preclinical development and clinical trials may have anti-cancer effects though impacting macrophage phenotype and function in addition to the cancer cells.
Collapse
Affiliation(s)
- Kayla V. Myers
- 0000 0001 2171 9311grid.21107.35Department of Pharmacology and Molecular Sciences, The Johns Hopkins School of Medicine, Baltimore, MD USA ,0000 0001 2171 9311grid.21107.35The James Buchanan Brady Urological Institute, Department of Urology, The Johns Hopkins School of Medicine, Baltimore, MD USA
| | - Sarah R. Amend
- 0000 0001 2171 9311grid.21107.35The James Buchanan Brady Urological Institute, Department of Urology, The Johns Hopkins School of Medicine, Baltimore, MD USA
| | - Kenneth J. Pienta
- 0000 0001 2171 9311grid.21107.35Department of Pharmacology and Molecular Sciences, The Johns Hopkins School of Medicine, Baltimore, MD USA ,0000 0001 2171 9311grid.21107.35The James Buchanan Brady Urological Institute, Department of Urology, The Johns Hopkins School of Medicine, Baltimore, MD USA ,0000 0001 2171 9311grid.21107.35Department of Oncology, The Johns Hopkins School of Medicine, Baltimore, MD USA ,0000 0001 2171 9311grid.21107.35Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD USA
| |
Collapse
|
86
|
Dahlbäck B, Guo LJ, Zöller B, Tran S. New functional test for the TFPIα cofactor activity of Protein S working in synergy with FV-Short. J Thromb Haemost 2019; 17:585-595. [PMID: 30740865 DOI: 10.1111/jth.14405] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 01/30/2019] [Indexed: 08/31/2023]
Abstract
Essentials Protein S and FV-Short are synergistic cofactors to Tissue Factor Pathway Inhibitor α (TFPIα). An assay for the TFPIα synergistic cofactor activity of protein S with FV-Short was developed. The assay was specific for the synergistic TFPIα-cofactor activity of free protein S. Protein S deficient individuals with known mutations were correctly distinguished from controls. SUMMARY: Background Protein S is an anticoagulant cofactor to both activated protein C and tissue factor pathway inhibitor (TFPIα). The TFPIα-cofactor activity of protein S is stimulated by a short isoform of factor V (FV-Short), the two proteins functioning in synergy. Objective Using the synergistic TFPIα-cofactor activity between protein S and FV-Short to develop a functional test for plasma protein S. Patients/Methods TFPIα-mediated inhibition of FXa in the presence of FV-Short, protein S and negatively charged phospholipid vesicles was monitored in time by synthetic substrate S2765. TFPIα, FXa and FV-Short were purified proteins, whereas diluted plasma from protein S deficient patients or controls were used as source for protein S. Results The assay was specific for free protein S demonstrating good correlation to free protein S plasma levels (r = 0.92) with a Y-axis intercept of -5%. Correlation to concentrations of total protein S (free and C4BPβ+-bound) was lower (r = 0.88) and the Y-axis intercept was +46%, which is consistent with the specificity for free protein S. The test distinguished protein S-deficient individuals from 6 families with known ProS1 mutations from family members having no mutation. Protein S levels of warfarin-treated protein S deficient cases were lower than protein S in cases treated with warfarin for other causes. Conclusions We describe a new assay measuring the TFPIα-cofactor activity of plasma protein S. The test identifies type I/III protein S deficiencies and will be a useful tool to detect type II protein S deficiency having defective TFPIα-cofactor activity.
Collapse
Affiliation(s)
- Björn Dahlbäck
- Department of Translational Medicine, Lund University, Skåne University Hospital, Malmö, Sweden
| | - Li Jun Guo
- Department of Translational Medicine, Lund University, Skåne University Hospital, Malmö, Sweden
| | - Bengt Zöller
- Department of Clinical Sciences, Lund University, Skåne University Hospital, Malmö, Sweden
| | - Sinh Tran
- Department of Translational Medicine, Lund University, Skåne University Hospital, Malmö, Sweden
| |
Collapse
|
87
|
O'Donnell JS, O'Sullivan JM, Preston RJS. Advances in understanding the molecular mechanisms that maintain normal haemostasis. Br J Haematol 2019; 186:24-36. [DOI: 10.1111/bjh.15872] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- James S. O'Donnell
- Haemostasis Research Group Department of Molecular and Cellular Therapeutics Irish Centre for Vascular Biology Royal College of Surgeons in Ireland Dublin Ireland
| | - Jamie M. O'Sullivan
- Haemostasis Research Group Department of Molecular and Cellular Therapeutics Irish Centre for Vascular Biology Royal College of Surgeons in Ireland Dublin Ireland
| | - Roger J. S. Preston
- Haemostasis Research Group Department of Molecular and Cellular Therapeutics Irish Centre for Vascular Biology Royal College of Surgeons in Ireland Dublin Ireland
| |
Collapse
|
88
|
Searching for potent and specific antibiotics against pathogenic Helicobacter and Campylobacter strains. ACTA ACUST UNITED AC 2019; 46:409-414. [DOI: 10.1007/s10295-018-2108-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2018] [Accepted: 11/08/2018] [Indexed: 12/14/2022]
Abstract
Abstract
Menaquinone is an obligatory component of the electron-transfer pathway in microorganisms. Its biosynthetic pathway was established by pioneering studies with Escherichia coli and it was revealed to be derived from chorismate by Men enzymes. However, we identified an alternative pathway, the futalosine pathway, operating in some microorganisms including Helicobacter pylori and Campylobacter jejuni, which cause gastric carcinoma and diarrhea, respectively. Because some useful intestinal bacteria, such as lactobacilli, use the canonical pathway, the futalosine pathway is an attractive target for development of chemotherapeutics for the abovementioned pathogens. In this mini-review, we summarize compounds that inhibit Mqn enzymes involved in the futalosine pathway discovered to date.
Collapse
|
89
|
Post-transcriptional, post-translational and pharmacological regulation of tissue factor pathway inhibitor. Blood Coagul Fibrinolysis 2018; 29:668-682. [PMID: 30439766 DOI: 10.1097/mbc.0000000000000775] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
: Tissue factor (TF) pathway inhibitor (TFPI) is an endogenous natural anticoagulant that readily inhibits the extrinsic coagulation initiation complex (TF-FVIIa-Xa) and prothrombinase (FXa, FVa and calcium ions). Alternatively, spliced TFPI isoforms (α, β and δ) are expressed by vascular and extravascular cells and regulate thrombosis and haemostasis, as well as cell signalling functions of TF complexes via protease-activated receptors (PARs). Proteolysis of TFPI plays an important role in regulating physiological roles of the TF pathway in host defense and possibly haemostasis. Elimination of TFPI inhibition has therefore been proposed as an approach to improve haemostasis in haemophilia patients. In this review, we focus on posttranscription and translational modification of TFPI and its function in thrombosis and how pharmacological inhibitors and endogenous proteases interfere with TFPI and alter haemostasis.
Collapse
|
90
|
Salvagno GL, Pavan C, Lippi G. Rare thrombophilic conditions. ANNALS OF TRANSLATIONAL MEDICINE 2018; 6:342. [PMID: 30306081 DOI: 10.21037/atm.2018.08.12] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Thrombophilia, either acquired or inherited, can be defined as a predisposition to developing thromboembolic complications. Since the discovery of antithrombin deficiency in the 1965, many other conditions have been described so far, which have then allowed to currently detect an inherited or acquired predisposition in approximately 60-70% of patients with thromboembolic disorders. These prothrombotic risk factors mainly include qualitative or quantitative defects of endogenous coagulation factor inhibitors, increased concentration or function of clotting proteins, defects in the fibrinolytic system, impaired platelet function, and hyperhomocysteinemia. In this review article, we aim to provide an overview on epidemiologic, clinic and laboratory aspects of both acquired and inherited rare thrombophilic risk factors, especially including dysfibrinogenemia, heparin cofactor II, thrombomodulin, lipoprotein(a), sticky platelet syndrome, plasminogen activator inhibitor-1 apolipoprotein E, tissue factor pathway inhibitor, paroxysmal nocturnal haemoglobinuria and heparin-induced thrombocytopenia.
Collapse
Affiliation(s)
| | - Chiara Pavan
- Division of Geriatric Medicine, Mater Salutis Hospital, Legnago, Verona, Italy
| | - Giuseppe Lippi
- Section of Clinical Biochemistry, University of Verona, Verona, Italy
| |
Collapse
|
91
|
Valentino LA, Turecek PL, Gritsch H, Butenas S, Mann KG. Issues complicating precision dosing for factor VIII prophylaxis. Transfus Apher Sci 2018; 57:472-479. [DOI: 10.1016/j.transci.2018.07.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
92
|
Law LA, Graham DK, Di Paola J, Branchford BR. GAS6/TAM Pathway Signaling in Hemostasis and Thrombosis. Front Med (Lausanne) 2018; 5:137. [PMID: 29868590 PMCID: PMC5954114 DOI: 10.3389/fmed.2018.00137] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Accepted: 04/23/2018] [Indexed: 01/01/2023] Open
Abstract
The GAS6/TYRO3-AXL-MERTK (TAM) signaling pathway is essential for full and sustained platelet activation, as well as thrombus stabilization. Inhibition of this pathway decreases platelet aggregation, shape change, clot retraction, aggregate formation under flow conditions, and surface expression of activation markers. Transgenic mice deficient in GAS6, or any of the TAM family of receptors that engage this ligand, exhibit in vivo protection against arterial and venous thrombosis but do not demonstrate either spontaneous or prolonged bleeding compared to their wild-type counterparts. Comparable results are observed in wild-type mice treated with pharmacological inhibitors of the GAS6-TAM pathway. Thus, GAS6/TAM inhibition offers an attractive novel therapeutic option that may allow for a moderate reduction in platelet activation and decreased thrombosis while still permitting the primary hemostatic function of platelet plug formation.
Collapse
Affiliation(s)
- Luke A Law
- Department of Anesthesiology, Mayo Clinic, Rochester, MN, United States
| | - Douglas K Graham
- Section of Hematology/Oncology, Department of Pediatrics, Emory University, Atlanta, GA, United States
| | - Jorge Di Paola
- Section of Hematology/Oncology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, United States.,University of Colorado Hemophilia and Thrombosis Center, Aurora, CO, United States
| | - Brian R Branchford
- Section of Hematology/Oncology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, United States.,University of Colorado Hemophilia and Thrombosis Center, Aurora, CO, United States
| |
Collapse
|
93
|
A clinically relevant and bias-controlled murine model to study acute traumatic coagulopathy. Sci Rep 2018; 8:5783. [PMID: 29636535 PMCID: PMC5893580 DOI: 10.1038/s41598-018-24225-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 03/20/2018] [Indexed: 12/30/2022] Open
Abstract
Acute traumatic coagulopathy (ATC) is an acute and endogenous mechanism triggered by the association of trauma and hemorrhage. Several animal models have been developed, but some major biases have not yet been identified. Our aim was to develop a robust and clinically relevant murine model to study this condition. Anesthetized adult Sprague Dawley rats were randomized into 4 groups: C, control; T, trauma; H, hemorrhage; TH, trauma and hemorrhage (n = 7 each). Trauma consisted of laparotomy associated with four-limb and splenic fractures. Clinical variables, ionograms, arterial and hemostasis blood tests were compared at 0 and 90 min. ATC and un-compensated shock were observed in group TH. In this group, the rise in prothrombin time and activated partial thromboplastin was 29 and 40%, respectively. Shock markers, compensation mechanisms and coagulation pathways were all consistent with human pathophysiology. The absence of confounding factors, such as trauma-related bleeding or dilution due to trans-capillary refill was verified. This ethic, cost effective and bias-controlled model reproduced the specific and endogenous mechanism of ATC and will allow to identify potential targets for therapeutics in case of trauma-related hemorrhage.
Collapse
|
94
|
Dahlbäck B, Guo LJ, Livaja‐Koshiar R, Tran S. Factor V-short and protein S as synergistic tissue factor pathway inhibitor (TFPIα) cofactors. Res Pract Thromb Haemost 2018; 2:114-124. [PMID: 30046712 PMCID: PMC6055574 DOI: 10.1002/rth2.12057] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 10/17/2017] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND FV-Short is a normal splice variant of Factor V (FV) having a short B domain, which exposes a high affinity-binding site for tissue factor pathway inhibitor α (TFPIα). FV-Short and TFPIα circulate in complex in plasma. OBJECTIVES The aim was to elucidate whether FV-Short affects TFPIα as inhibitor of coagulation FXa and to test whether the TFPIα-cofactor activity of protein S is influenced by FV-Short. METHODS Recombinant FV, wild-type FV-Short and a FV-Short thrombin-cleavage resistant variant were expressed and purified. The influence of FV and FV-Short variants and/or protein S on the FXa inhibitory activity of TFPIα was monitored both in a purified system and in a plasma-based thrombin generation assay. RESULTS FV-Short had intrinsically weak TFPIα-cofactor activity but with protein S present, FV-Short yielded efficient inactivation of FXa. Protein S alone did not promote full TFPIα-activity. Intact FV was inefficient at low protein S concentrations and had 10-fold lower activity compared to FV-Short at physiological protein S levels. Activation of FV-Short by thrombin resulted in the loss of the TFPIα-cofactor activity. The synergistic TFPIα-cofactor activity of FV-Short and protein S was also demonstrated in plasma using a thrombin generation assay. CONCLUSIONS FV-Short and protein S are highly efficient, synergistic cofactors to TFPIα in the regulation of FXa activity, whereas full length FV has lower activity. Our results suggest the formation of an efficient FXa-inhibitory complex between FV-Short, TFPIα and protein S on the surface of negatively charged phospholipids.
Collapse
Affiliation(s)
- Björn Dahlbäck
- Department of Translational MedicineLund UniversitySkåne University HospitalMalmöSweden
| | - Li Jun Guo
- Department of Translational MedicineLund UniversitySkåne University HospitalMalmöSweden
| | - Ruzica Livaja‐Koshiar
- Department of Translational MedicineLund UniversitySkåne University HospitalMalmöSweden
| | - Sinh Tran
- Department of Translational MedicineLund UniversitySkåne University HospitalMalmöSweden
| |
Collapse
|
95
|
Wildhagen K, Lutgens E, Loubele S, Cate HT, Nicolaes G. The structure-function relationship of activated protein C. Thromb Haemost 2017; 106:1034-45. [DOI: 10.1160/th11-08-0522] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2011] [Accepted: 09/22/2011] [Indexed: 11/05/2022]
Abstract
SummaryProtein C is the central enzyme of the natural anticoagulant pathway and its activated form APC (activated protein C) is able to proteolyse non-active as well as active coagulation factors V and VIII. Proteolysis renders these cofactors inactive, resulting in an attenuation of thrombin formation and overall down-regulation of coagulation. Presences of the APC cofactor, protein S, thrombomodulin, endothelial protein C receptor and a phospholipid surface are important for the expression of anticoagulant APC activity. Notably, APC also has direct cytoprotective effects on cells: APC is able to protect the endothelial barrier function and expresses anti-inflammatory and anti-apoptotic activities. Exact molecular mechanisms have thus far not been completely described but it has been shown that both the protease activated receptor 1 and EPCR are essential for the cytoprotective activity of APC. Recently it was shown that also other receptors like sphingosine 1 phosphate receptor 1, Cd11b/CD18 and tyrosine kinase with immunoglobulin-like and EGFlike domains 2 are likewise important for APC signalling. Mutagenesis studies are being performed to map the various APC functions and interactions onto its 3D structure and to dissect anticoagulant and cytoprotective properties. The results of these studies have provided a wealth of structure-function information. With this review we describe the state-of-the-art of the intricate structure-function relationships of APC, a protein that harbours several important functions for the maintenance of both humoral and tissue homeostasis.Lessons from natural and engineered mutations
Collapse
|
96
|
Dienava-Verdoold I, Marchetti MR, te Boome LCJ, Russo L, Falanga A, Koene HR, Mertens K, Brinkman HJM. Platelet-mediated proteolytic down regulation of the anticoagulant activity of protein S in individuals with haematological malignancies. Thromb Haemost 2017; 107:468-76. [DOI: 10.1160/th11-07-0457] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2011] [Accepted: 12/08/2011] [Indexed: 11/05/2022]
Abstract
SummaryThe natural anticoagulant protein S contains a so-called thrombin-sensitive region (TSR), which is susceptible to proteolytic cleavage. We have previously shown that a platelet-associated protease is able to cleave protein S under physiological plasma conditions in vitro. The aim of the present study was to investigate the relation between platelet-associated protein S cleaving activity and in vivo protein S cleavage, and to evaluate the impact of in vivo protein S cleavage on its anticoagulant activity. Protein S cleavage in healthy subjects and in thrombocytopenic and thrombocythaemic patients was evaluated by immunological techniques. Concentration of cleaved and intact protein S was correlated to levels of activated protein C (APC)-dependent and APC-independent protein S anticoagulant activity. In plasma from healthy volunteers 25% of protein S is cleaved in the TSR. While in plasma there was a clear positive correlation between levels of intact protein S and both APC-dependent and APC-independent protein S anticoagulant activities, these correlations were absent for cleaved protein S. Protein S cleavage was significantly increased in patients with essential thrombocythaemia (ET) and significantly reduced in patients with chemotherapy-induced thrombocytopenia. In ET patients on cytoreductive therapy, both platelet count and protein S cleavage returned to normal values. Accordingly, platelet transfusion restored cleavage of protein S to normal values in patients with chemotherapy-induced thrombocytopenia. In conclusion, proteases from platelets seem to contribute to the presence of cleaved protein S in the circulation and may enhance the coagulation response in vivo by down regulating the anticoagulant activity of protein S.
Collapse
|
97
|
Laboratory assessment of Activated Protein C Resistance/Factor V-Leiden and performance characteristics of a new quantitative assay. Transfus Apher Sci 2017; 56:906-913. [PMID: 29162399 DOI: 10.1016/j.transci.2017.11.021] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Activated Protein C Resistance is mainly associated to a factor V mutation (RQ506), which induces a deficient inactivation of activated factor V by activated protein C, and is associated to an increased risk of venous and arterial thrombosis in affected individuals, caused by the prolonged activated factor V survival. Its prevalence is mainly in Caucasians (about 5%), and this mutation is absent in Africans and Asians. Presence of Factor V-Leiden is usually evidenced with clotting methods, using a two-step APTT assay performed without or with APC: prolongation of blood coagulation time is decreased if this factor is present. The R506Q Factor V-Leiden mutation is now usually characterized using molecular biology, and this technique tends to become the first intention assay for characterization of patients. Both techniques are qualitative, and allow classifying tested individuals as heterozygotes or homozygotes for the mutation, when present. A new quantitative assay for Factor V-Leiden, using a one-step clotting method, has been developed, and designed with highly purified human coagulation proteins. Clotting is triggered with human Factor Xa, in presence of calcium and phospholipids (mixture which favours APC action over clotting process). Diluted tested plasma, is supplemented with a clotting mixture containing human fibrinogen, prothrombin, and protein S at a constant concentration. APC is added, and clotting is initiated with calcium. Calibration is performed with a pool of plasmas from patients carrying the R506Q Factor V mutation, and its mixtures with normal plasma. Homozygous patients have clotting times of about <40sec; heterozygous patients have clotting times of about 40-60sec and normal individuals yield clotting times >70sec. Factor V-Leiden concentration is usually >75% in homozygous patients, 30-60% in heterozygous patients and below 5% in normal. The assay is insensitive to clotting factor deficiencies (II, VII, VIII: C, IX, X), dicoumarol or heparin therapies, and has no interference with lupus anticoagulant (LA). This new assay for Factor V-Leiden can be easily used in any coagulation laboratory, is performed as a single test, and is quantitative. This assay has a high robustness, is accurate and presents a good intra- (<3%) and inter-assay (<5%) variability. It contributes solving most of the laboratory issues faced when testing factor V-Leiden. Quantitation of Factor V-L could contribute to a better assessment of thrombotic risk in affected patients, as this complication is first associated to and caused by the presence of a defined amount of FVa.
Collapse
|
98
|
Fischer PM. Design of Small-Molecule Active-Site Inhibitors of the S1A Family Proteases as Procoagulant and Anticoagulant Drugs. J Med Chem 2017; 61:3799-3822. [DOI: 10.1021/acs.jmedchem.7b00772] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Peter M. Fischer
- School of Pharmacy and Centre for Biomolecular Sciences, University of Nottingham, Nottingham NG7 2RD, U.K
| |
Collapse
|
99
|
Roumenina LT, Rayes J, Frimat M, Fremeaux-Bacchi V. Endothelial cells: source, barrier, and target of defensive mediators. Immunol Rev 2017; 274:307-329. [PMID: 27782324 DOI: 10.1111/imr.12479] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Endothelium is strategically located at the interface between blood and interstitial tissues, placing thus endothelial cell as a key player in vascular homeostasis. Endothelial cells are in a dynamic equilibrium with their environment and constitute concomitantly a source, a barrier, and a target of defensive mediators. This review will discuss the recent advances in our understanding of the complex crosstalk between the endothelium, the complement system and the hemostasis in health and in disease. The first part will provide a general introduction on endothelial cells heterogeneity and on the physiologic role of the complement and hemostatic systems. The second part will analyze the interplay between complement, hemostasis and endothelial cells in physiological conditions and their alterations in diseases. Particular focus will be made on the prototypes of thrombotic microangiopathic disorders, resulting from complement or hemostasis dysregulation-mediated endothelial damage: atypical hemolytic uremic syndrome and thrombotic thrombocytopenic purpura. Novel aspects of the pathophysiology of the thrombotic microangiopathies will be discussed.
Collapse
Affiliation(s)
- Lubka T Roumenina
- INSERM UMRS 1138, Cordeliers Research Center, Université Pierre et Marie Curie (UPMC-Paris-6) and Université Paris Descartes Sorbonne Paris-Cité, Paris, France.
| | - Julie Rayes
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Marie Frimat
- INSERM UMR 995, Lille, France.,Nephrology Department, CHU Lille, Lille, France
| | - Veronique Fremeaux-Bacchi
- INSERM UMRS 1138, Cordeliers Research Center, Université Pierre et Marie Curie (UPMC-Paris-6) and Université Paris Descartes Sorbonne Paris-Cité, Paris, France.,Assistance Publique - Hôpitaux de Paris, Service d'Immunologie Biologique, Hôpital Européen Georges Pompidou, Paris, France
| |
Collapse
|
100
|
Gorbacheva LR, Kiseleva EV, Savinkova IG, Strukova SM. A new concept of action of hemostatic proteases on inflammation, neurotoxicity, and tissue regeneration. BIOCHEMISTRY (MOSCOW) 2017; 82:778-790. [DOI: 10.1134/s0006297917070033] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|