1
|
Long J, Chen J, Huang G, Chen Z, Zhang H, Zhang Y, Duan Q, Wu B, He J. The differences of fibrinogen levels in various types of hemorrhagic transformations. Front Neurol 2024; 15:1364875. [PMID: 39119563 PMCID: PMC11306044 DOI: 10.3389/fneur.2024.1364875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 07/11/2024] [Indexed: 08/10/2024] Open
Abstract
Introduction Hemorrhagic transformation (HT) is a serious complication that can occur spontaneously after an acute ischemic stroke (AIS) or after a thrombolytic/mechanical thrombectomy. Our study aims to explore the potential correlations between fibrinogen levels and the occurrence of spontaneous HT (sHT) and HT after mechanical thrombectomy (tHT). Methods A total of 423 consecutive AIS patients diagnosed HT who did not undergone thrombolysis and 423 age- and sex-matched patients without HT (non-HT) were enrolled. Fibrinogen levels were measured within 24 h of admission after stroke. The cohorts were trisected according to fibrinogen levels. The HT were further categorized into hemorrhagic infarction (HI) or parenchymal hematoma (PH) based on their imaging characteristics. Results In sHT cohort, fibrinogen levels were higher in HT patients than non-HT patients (p < 0.001 versus p = 0.002). High fibrinogen levels were associated with the severity of HT. HT patients without atrial fibrillation (AF) had higher levels of fibrinogen compared to non-HT (median 3.805 vs. 3.160, p < 0.001). This relationship did not differ among AF patients. In tHT cohort, fibrinogen levels were lower in HT patients than non-HT patients (p = 0.002). Lower fibrinogen levels were associated with the severity of HT (p = 0.004). The highest trisection of fibrinogen both in two cohorts were associated with HT [sHT cohort: OR = 2.515 (1.339-4.725), p = 0.016; that cohort: OR = 0.238 (0.108-0.523), p = 0.003]. Conclusion Our study suggests that lower fibrinogen level in sHT without AF and higher fibrinogen level in tHT are associated with more severe HT.
Collapse
Affiliation(s)
- Jingfang Long
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Department of Neurology, Wenzhou Central Hospital, Wenzhou, Zhejiang, China
| | - Jiahao Chen
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Guiqian Huang
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- School of Mental Health, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhen Chen
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Heyu Zhang
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Ye Zhang
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Qi Duan
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Beilan Wu
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jincai He
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
2
|
Ebrahimi R, Nasri F, Kalantari T. Coagulation and Inflammation in COVID-19: Reciprocal Relationship between Inflammatory and Coagulation Markers. Ann Hematol 2024; 103:1819-1831. [PMID: 38349409 DOI: 10.1007/s00277-024-05630-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Accepted: 01/16/2024] [Indexed: 05/14/2024]
Abstract
The coronavirus disease 2019 (COVID-19) is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), formerly known as 2019-nCoV. Numerous cellular and biochemical issues arise after COVID-19 infection. The severe inflammation that is caused by a number of cytokines appears to be one of the key hallmarks of COVID-19. Additionally, people with severe COVID-19 have coagulopathy and fulminant thrombotic events. We briefly reviewed the COVID-19 disease at the beginning of this paper. The inflammation and coagulation markers and their alterations in COVID-19 illness are briefly discussed in the parts that follow. Next, we talked about NETosis, which is a crucial relationship between coagulation and inflammation. In the end, we mentioned the two-way relationship between inflammation and coagulation, as well as the factors involved in it. We suggest that inflammation and coagulation are integrated systems in COVID-19 that act on each other in such a way that not only inflammation can activate coagulation but also coagulation can activate inflammation.
Collapse
Affiliation(s)
- Rasoul Ebrahimi
- Division of Laboratory Hematology and Blood Banking, Department of Medical Laboratory Sciences, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Fatemeh Nasri
- Division of Laboratory Hematology and Blood Banking, Department of Medical Laboratory Sciences, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Tahereh Kalantari
- Division of Laboratory Hematology and Blood Banking, Department of Medical Laboratory Sciences, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
3
|
Arderiu G, Bejar MT, Civit-Urgell A, Peña E, Badimon L. Crosstalk of human coronary perivascular adipose-derived stem cells with vascular cells: role of tissue factor. Basic Res Cardiol 2024; 119:291-307. [PMID: 38430261 DOI: 10.1007/s00395-024-01037-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 01/31/2024] [Accepted: 02/01/2024] [Indexed: 03/03/2024]
Abstract
The coronary perivascular adipose tissue (cPVAT) has been associated to the burden of cardiovascular risk factors and to the underlying vessel atherosclerotic plaque severity. Although the "outside to inside" hypothesis of PVAT-derived-adipokine regulation of vessel function is currently accepted, whether the resident mesenchymal stem cells (ASCs) in PVAT have a regulatory role on the underlying vascular arterial smooth muscle cells (VSMCs) is not known. Here, we investigated the interactions between resident PVAT-ASCs and VSMCs. ASCs were obtained from PVAT overlying the left anterior descending (LAD) coronary artery of hearts removed at heart transplant operations. PVAT was obtained both from patients with non-ischemic and ischemic heart disease as the cause of heart transplant. ASCs were isolated from PVAT, phenotypically characterized by flow cytometry, functionally tested for proliferation, and differentiation. Crosstalk between ASCs and VSMCs was investigated by co-culture studies. ASCs were detected in the adventitia of the LAD-PVAT showing differentiation capacity and angiogenic potential. ASCs obtained from PVAT of non-ischemic and ischemic hearts showed different tissue factor (TF) expression levels, different VSMCs recruitment capacity through the axis ERK1/2-ETS1 signaling and different angiogenic potential. Induced upregulation of TF in ASCs isolated from ischemic PVAT rescued their angiogenic capacity in subcutaneously implanted plugs in mice, whereas silencing TF in ASCs decreased the proangiogenic capacity of non-ischemic ASCs. The results indicate for the first time a novel mechanism of regulation of VSMCs by PVAT-ASCs in angiogenesis, mediated by TF expression in ASCs. Regulation of TF in ASCs may become a therapeutic intervention to increase cardiac protection.
Collapse
Affiliation(s)
- Gemma Arderiu
- Cardiovascular-Program, Institut de Recerca Sant Pau, IIB-Sant Pau, Carrer Sant Quintí, 77-79, 08041, Barcelona, Spain.
- Ciber CV, Instituto Carlos III, Madrid, Spain.
| | - Maria Teresa Bejar
- Cardiovascular-Program, Institut de Recerca Sant Pau, IIB-Sant Pau, Carrer Sant Quintí, 77-79, 08041, Barcelona, Spain
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Puddicombe Way, Cambridge, CB2 0AW, UK
| | - Anna Civit-Urgell
- Cardiovascular-Program, Institut de Recerca Sant Pau, IIB-Sant Pau, Carrer Sant Quintí, 77-79, 08041, Barcelona, Spain
- Facultat de Farmàcia i Ciències de l'Alimentació, Universitat de Barcelona (UB), Barcelona, Spain
| | - Esther Peña
- Cardiovascular-Program, Institut de Recerca Sant Pau, IIB-Sant Pau, Carrer Sant Quintí, 77-79, 08041, Barcelona, Spain
- Ciber CV, Instituto Carlos III, Madrid, Spain
| | - Lina Badimon
- Cardiovascular-Program, Institut de Recerca Sant Pau, IIB-Sant Pau, Carrer Sant Quintí, 77-79, 08041, Barcelona, Spain
- Ciber CV, Instituto Carlos III, Madrid, Spain
| |
Collapse
|
4
|
Tando S, Kimura T, Mizuhara R, Yuki N, Yoshioka A, Takahashi H, Yasuda R, Itoh K. An autopsy case of intravascular large B-cell lymphoma showing a rapid transition to embolic strokes with occlusion of the major cerebral arteries. Neuropathology 2024; 44:135-146. [PMID: 37559506 DOI: 10.1111/neup.12940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 06/22/2023] [Accepted: 07/24/2023] [Indexed: 08/11/2023]
Abstract
Intravascular large B-cell lymphoma can induce central nervous system manifestations, including strokes, due to small-vessel occlusion caused by lymphoma cells. However, involvement in large-sized vessels is rare. Here, we present an unusual autopsy case of an 88-year-old man showing a rapid transition from multiple strokes due to small vessel occlusion, typical of intravascular lymphoma, to progressive embolic strokes caused by the occlusion of major cerebral arteries. Magnetic resonance angiography demonstrated the major cerebral arteries associated with those multiple progressive strokes, including the right posterior cerebral artery, left anterior cerebral artery, and right middle cerebral artery, but the detectability was poor. A random skin biopsy at the abdomen confirmed the diagnosis of intravascular large B-cell lymphoma. The patient died 106 days after hospitalization despite intensive treatment. An autopsy revealed broad liquefactive necrosis in the area governed by the major cerebral arteries and multiple small infarctions caused by intravascular lymphoma cells in the small-sized vessels. In addition, the major cerebral arteries showed multiple thromboembolism with partial organization and clusters of intravascular lymphoma cells. Notably, those cells were shown aggregated and attached along the vascular wall of the basilar artery, which might have caused focal hypercoagulation in the near vessels. This aggregation might have disseminated widely in the other major cerebral arteries. Moreover, the cluster of intravascular lymphoma cells in the basilar artery was positive for tumor necrosis factor α, and similar histopathology findings were observed in the splenic veins. However, the pathogenesis of this rare phenomenon involving these cells remains unknown. From a clinical perspective, we should consider the possibility that intravascular lymphoma cells may provoke similar progressive embolic strokes.
Collapse
Affiliation(s)
- So Tando
- Department of Pathology and Applied Neurobiology, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Kyoto, Japan
| | - Tadashi Kimura
- Departments of Neurology, National Hospital Organization Maizuru Medical Center, Maizuru, Japan
| | - Ryo Mizuhara
- Departments of Neurology, National Hospital Organization Maizuru Medical Center, Maizuru, Japan
| | - Natsuko Yuki
- Department of Neurology, Kyoto Kizugawa Hospital, Joyo, Japan
| | - Akira Yoshioka
- Department of Neurology, Kyoto Kizugawa Hospital, Joyo, Japan
| | - Hisashi Takahashi
- Department of Pathology and Applied Neurobiology, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Kyoto, Japan
- Department of Neurology, North Medical Center Kyoto Prefectural University of Medicine, Yosano-Gun, Japan
| | - Rei Yasuda
- Department of Neurology, North Medical Center Kyoto Prefectural University of Medicine, Yosano-Gun, Japan
| | - Kyoko Itoh
- Department of Pathology and Applied Neurobiology, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Kyoto, Japan
| |
Collapse
|
5
|
Meng S, Xu B, Yang W, Zhao M. Microparticle-associated tissue factor activity correlates with the inflammatory response in septic disseminated intravascular coagulation patients. PeerJ 2024; 12:e16636. [PMID: 38213768 PMCID: PMC10782946 DOI: 10.7717/peerj.16636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 11/18/2023] [Indexed: 01/13/2024] Open
Abstract
Background Sepsis is often accompanied by the formation of disseminated intravascular coagulation (DIC). Microparticles can exert their procoagulant and proinflammatory properties in a variety of ways. The purpose of this study was to investigate the relationship between microparticle-associated tissue factor activity (TF+-MP activity) and the inflammatory response. Methods Data from a total of 31 DIC patients with sepsis and 31 non-DIC patients with sepsis admitted to the ICU of the First Affiliated Hospital of Harbin Medical University from December 2017 to March 2019 were collected. Blood samples were collected and DIC scores were calculated on the day of enrollment. The hospital's clinical laboratory completed routine blood, procalcitonin, and C-reactive protein tests. TF+-MP activity was measured using a tissue factor-dependent FXa generation assay. Interleukin-1β (IL-1β) and tumor necrosis factor-α (TNF-α) levels were determined using ELISA kits. Results Compared with the non-DIC group, the DIC group had higher levels of leukocytes, neutrophils, procalcitonin, C-reactive protein, IL-1β, and TNF-α, and more severe inflammatory reactions. TF+-MP activity in the DIC group was higher than that in the non-DIC group. In sepsis patients, TF+-MP activity was strongly correlated with inflammatory response indices and DIC scores. Conclusion TF+-MP activity may play a major role in promoting inflammatory response in septic DIC.
Collapse
Affiliation(s)
- Shishuai Meng
- Intensive Care Unit, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Bin Xu
- Department of Cardiology, Cardiology, Harbin First Hospital, Harbin, Heilongjiang, China
| | - Wei Yang
- Intensive Care Unit, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Mingyan Zhao
- Intensive Care Unit, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| |
Collapse
|
6
|
De Nardi AC, Coy-Canguçu A, Saito A, Florio MF, Marti G, Degasperi GR, Orsi FA. Immunothrombosis and its underlying biological mechanisms. Hematol Transfus Cell Ther 2024; 46:49-57. [PMID: 37451977 PMCID: PMC10935458 DOI: 10.1016/j.htct.2023.05.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 03/31/2023] [Accepted: 05/15/2023] [Indexed: 07/18/2023] Open
Abstract
The evolutionary conserved link between coagulation and innate immunity is a biological process characterized by the thrombosis formation stimulus of immune cells and specific thrombosis-related molecules. In physiological settings, the relationship between the immune system and thrombosis facilitates the recognition of pathogens and damaged cells and inhibits pathogen proliferation. However, when deregulated, the interplay between hemostasis and innate immunity becomes a pathological process named immunothrombosis, which is at the basis of several infectious and inflammation-related thrombotic disorders, including coronavirus disease 2019 (COVID-19). In advanced stages, alterations in both coagulation and immune cell function due to extreme inflammation lead to an increase in blood coagulability, with high rates of thrombosis and mortality. Therefore, understanding underlying mechanisms in immunothrombosis has become decisive for the development of more efficient therapies to treat and prevent thrombosis in COVID-19 and in other thrombotic disorders. In this review, we outline the existing knowledge on the molecular and cellular processes involved in immunothrombosis, focusing on the role of neutrophil extracellular traps (NETs), platelets and the coagulation pathway. We also describe how the deregulation of hemostasis is associated with pathological conditions and can significantly aggravate a patient's condition, using COVID-19 as a clinical model.
Collapse
Affiliation(s)
- Arthur Cunha De Nardi
- Pontifícia Universidade Católica de Campinas (PUCC), Faculdade de Medicina, Campinas, Brazil
| | - Andréa Coy-Canguçu
- Pontifícia Universidade Católica de Campinas (PUCC), Faculdade de Medicina, Campinas, Brazil.
| | - Atena Saito
- Pontifícia Universidade Católica de Campinas (PUCC), Faculdade de Medicina, Campinas, Brazil
| | - Maria Fernanda Florio
- Pontifícia Universidade Católica de Campinas (PUCC), Faculdade de Medicina, Campinas, Brazil
| | - Giovanna Marti
- Pontifícia Universidade Católica de Campinas (PUCC), Faculdade de Medicina, Campinas, Brazil
| | - Giovanna R Degasperi
- Pontifícia Universidade Católica de Campinas (PUCC), Faculdade de Medicina, Campinas, Brazil
| | - Fernanda A Orsi
- Universidade Estadual de Campinas (UNICAMP), Faculdade de Ciências Médicas, Departamento de Patologia, Campinas, Brazil
| |
Collapse
|
7
|
Seirin-Lee S, Matsubara D, Yanase Y, Kunieda T, Takahagi S, Hide M. Mathematical-based morphological classification of skin eruptions corresponding to the pathophysiological state of chronic spontaneous urticaria. COMMUNICATIONS MEDICINE 2023; 3:171. [PMID: 38049619 PMCID: PMC10696082 DOI: 10.1038/s43856-023-00404-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 11/10/2023] [Indexed: 12/06/2023] Open
Abstract
BACKGROUND Chronic spontaneous urticaria (CSU) is one of the most intractable human-specific skin diseases. However, as no experimental animal model exists, the mechanism underlying disease pathogenesis in vivo remains unclear, making the establishment of a curative treatment challenging. METHODS A novel approach combining mathematical modelling, in vitro experiments and clinical data analysis was used to infer the pathological state of CSU patients from geometric features of the skin eruptions. RESULTS Based on our hierarchical mathematical modelling, the eruptions of CSU were classified into five categories, each with distinct histamine, basophils, mast cells and coagulation factors network signatures. The analysis of 105 real CSU patients with this classification by six individual dermatologists achieved 87.6% agreement. Furthermore, our network analysis revealed that the coagulation status likely determines boundary/area pattern of wheals, while the state of spontaneous histamine release from mast cells may contribute to the divergence of size and outline of the eruptions. CONCLUSIONS Our multi-faceted approach was accurate in defining pathophysiological states of disease based on geometric features offering the potential to improve the accuracy of CSU diagnosis and better management of the disease in the clinic.
Collapse
Affiliation(s)
- Sungrim Seirin-Lee
- Kyoto University Institute for the Advanced Study of Human Biology (ASHBi), KUIAS, Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto, 606-8501, Japan.
- Department of Mathematical Medicine, Graduate School of Medicine, Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto, 606-8501, Japan.
- JST CREST, 4-1-8 Honcho, Kawaguchi, Saitama, 332-0012, Japan.
| | - Daiki Matsubara
- Department of Dermatology, Institute of Biomedical & Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Yuhki Yanase
- JST CREST, 4-1-8 Honcho, Kawaguchi, Saitama, 332-0012, Japan
- Department of Pharmacotherapy, Graduate School of Biomedical Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Takuma Kunieda
- Department of Pharmacotherapy, Graduate School of Biomedical Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Shunsuke Takahagi
- JST CREST, 4-1-8 Honcho, Kawaguchi, Saitama, 332-0012, Japan.
- Department of Dermatology, Institute of Biomedical & Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan.
| | - Michihiro Hide
- Department of Dermatology, Institute of Biomedical & Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
- Department of Dermatology, Hiroshima City Hiroshima Citizens Hospital, Motomachi, Naka-ku, Hiroshima, 730-8518, Japan
| |
Collapse
|
8
|
Li S, Wang H, Shao Q. The central role of neutrophil extracellular traps (NETs) and by-products in COVID-19 related pulmonary thrombosis. Immun Inflamm Dis 2023; 11:e949. [PMID: 37647446 PMCID: PMC10461423 DOI: 10.1002/iid3.949] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 06/26/2023] [Accepted: 07/08/2023] [Indexed: 09/01/2023] Open
Abstract
Extracellular trap networks (neutrophil extracellular traps [NETs]) of polymorphonuclear neutrophils are mesh-like substances that prevent the spread of pathogens. They primarily consist of DNA skeletons, histones, granule components, and cytoplasmic proteins. NETs formation requires a certain environment and there are different pathways for NETs production. However, it is still not clear how severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) promotes NETs. NETs exert antiinflammatory effects through immune response, while they can also lead to certain adverse outcomes, such as the development of immunothrombosis. Coronavirus disease 2019 (COVID-19) is an inflammatory reaction affecting various organs caused by SARS-CoV-2, especially the lungs. NETs production and disease severity are linked with unique neutrophil clusters by single-cell RNA sequencing. NETs might exert an anti-inflammatory role in the initial stage of lung tissue inflammation. Nevertheless, numerous studies and cases have shown that they can also result in pulmonary thrombosis. There is mounting evidence that NETs are tightly related with COVID-19 pulmonary thrombosis, and many studies on the mechanisms are involved. The role and mechanism of NETs in the development of pulmonary thrombosis will be the main topics of this manuscript. Additionally, we address the potential targeting of NETs in COVID-19 patients.
Collapse
Affiliation(s)
- Shi Li
- Department of ImmunologySchool of Medicine, Jiangsu UniversityZhenjiangJiangsuChina
| | - Hui Wang
- Department of ImmunologySchool of Medicine, Jiangsu UniversityZhenjiangJiangsuChina
| | - Qixiang Shao
- Department of ImmunologySchool of Medicine, Jiangsu UniversityZhenjiangJiangsuChina
- Department of Medical Microbiology and Immunology, Institute of Medical Genetics and Reproductive Immunity, School of Medical Science and Laboratory MedicineJiangsu College of NursingHuai'anJiangsuChina
| |
Collapse
|
9
|
Krüger-Genge A, Köhler S, Laube M, Haileka V, Lemm S, Majchrzak K, Kammerer S, Schulz C, Storsberg J, Pietzsch J, Küpper JH, Jung F. Anti-Cancer Prodrug Cyclophosphamide Exerts Thrombogenic Effects on Human Venous Endothelial Cells Independent of CYP450 Activation-Relevance to Thrombosis. Cells 2023; 12:1965. [PMID: 37566045 PMCID: PMC10416884 DOI: 10.3390/cells12151965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 05/09/2023] [Accepted: 07/13/2023] [Indexed: 08/12/2023] Open
Abstract
Cancer patients are at a very high risk of serious thrombotic events, often fatal. The causes discussed include the detachment of thrombogenic particles from tumor cells or the adverse effects of chemotherapeutic agents. Cytostatic agents can either act directly on their targets or, in the case of a prodrug approach, require metabolization for their action. Cyclophosphamide (CPA) is a widely used cytostatic drug that requires prodrug activation by cytochrome P450 enzymes (CYP) in the liver. We hypothesize that CPA could induce thrombosis in one of the following ways: (1) damage to endothelial cells (EC) after intra-endothelial metabolization; or (2) direct damage to EC without prior metabolization. In order to investigate this hypothesis, endothelial cells (HUVEC) were treated with CPA in clinically relevant concentrations for up to 8 days. HUVECs were chosen as a model representing the first place of action after intravenous CPA administration. No expression of CYP2B6, CYP3A4, CYP2C9 and CYP2C19 was found in HUVEC, but a weak expression of CYP2C18 was observed. CPA treatment of HUVEC induced DNA damage and a reduced formation of an EC monolayer and caused an increased release of prostacyclin (PGI2) and thromboxane (TXA) associated with a shift of the PGI2/TXA balance to a prothrombotic state. In an in vivo scenario, such processes would promote the risk of thrombus formation.
Collapse
Affiliation(s)
- Anne Krüger-Genge
- Department of Healthcare, Biomaterials and Cosmeceuticals, Fraunhofer Institute for Applied Polymer Research (IAP), 14476 Potsdam, Germany
| | - Susanne Köhler
- Institute of Biotechnology, Molecular Cell Biology, Brandenburg University of Technology Cottbus-Senftenberg, 01968 Senftenberg, Germany
| | - Markus Laube
- Department of Radiopharmaceutical and Chemical Biology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, 01328 Dresden, Germany
| | - Vanessa Haileka
- Institute of Biotechnology, Molecular Cell Biology, Brandenburg University of Technology Cottbus-Senftenberg, 01968 Senftenberg, Germany
| | - Sandy Lemm
- Department of Radiopharmaceutical and Chemical Biology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, 01328 Dresden, Germany
- Faculty of Chemistry and Food Chemistry, School of Science, Technische Universität Dresden, 01069 Dresden, Germany
| | - Karolina Majchrzak
- Institute of Biotechnology, Molecular Cell Biology, Brandenburg University of Technology Cottbus-Senftenberg, 01968 Senftenberg, Germany
| | - Sarah Kammerer
- Institute of Biotechnology, Molecular Cell Biology, Brandenburg University of Technology Cottbus-Senftenberg, 01968 Senftenberg, Germany
| | - Christian Schulz
- Institute of Biotechnology, Molecular Cell Biology, Brandenburg University of Technology Cottbus-Senftenberg, 01968 Senftenberg, Germany
- Brandenburg University of Technology Cottbus-Senftenberg, Fraunhofer Project Group PZ-Syn of the Fraunhofer Institute for Cell Therapy and Immunology, Branch Bioanalytics and Bioprocesses (IZI-BB), 14476 Potsdam, Germany
| | - Joachim Storsberg
- Department of Healthcare, Biomaterials and Cosmeceuticals, Fraunhofer Institute for Applied Polymer Research (IAP), 14476 Potsdam, Germany
- Faculty of Medicine, Private University in the Principality of Liechtenstein (UFL), 9495 Triesen, Liechtenstein
| | - Jens Pietzsch
- Department of Radiopharmaceutical and Chemical Biology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, 01328 Dresden, Germany
- Faculty of Chemistry and Food Chemistry, School of Science, Technische Universität Dresden, 01069 Dresden, Germany
| | - Jan-Heiner Küpper
- Institute of Biotechnology, Molecular Cell Biology, Brandenburg University of Technology Cottbus-Senftenberg, 01968 Senftenberg, Germany
| | - Friedrich Jung
- Institute of Biotechnology, Molecular Cell Biology, Brandenburg University of Technology Cottbus-Senftenberg, 01968 Senftenberg, Germany
| |
Collapse
|
10
|
Koltsova EM, Martyanov AA, Podoplelova NA. Procoagulant Properties of Extracellular Vesicles in Normal and Pathological Pregnancy. BIOCHEMISTRY (MOSCOW), SUPPLEMENT SERIES A: MEMBRANE AND CELL BIOLOGY 2023. [DOI: 10.1134/s1990747822060071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/30/2023]
|
11
|
Ye SL, Li WD, Li WX, Xiao L, Ran F, Chen MM, Li XQ, Sun LL. The regulatory role of exosomes in venous thromboembolism. Front Cell Dev Biol 2022; 10:956880. [PMID: 36092737 PMCID: PMC9449368 DOI: 10.3389/fcell.2022.956880] [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: 05/31/2022] [Accepted: 07/26/2022] [Indexed: 11/25/2022] Open
Abstract
Exosomes are nanoscale endocytic vesicles, 30-150 nm in diameter, secreted by most cells. They mainly originate from multivesicular bodies formed by intracellular invagination of lysosomal microparticles, and released into the extracellular matrix after fusion of multivesicular bodies with cell membrane. Studies have shown that exosomes contain a variety of active molecules, such as proteins, lipids and RNAs (such as mRNA, miRNA, lncRNA, circRNA, etc.), which regulate the behavior of recipient cells and serve as circulating biomarkers of diseases, including thrombosis. Therefore, exosome research is important for the diagnosis, treatment, therapeutic monitoring, and prognosis of thrombosis in that it can reveal the counts, surface marker expression, protein, and miRNA cargo involved. Recent studies have shown that exosomes can be used as therapeutic vectors for tissue regeneration and as alternative vectors for drug delivery. In this review, we summarize the physiological and biochemical characteristics, isolation, and identification of exosomes. Moreover, we focus on the role of exosomes in thrombosis, specifically venous thromboembolism, and their potential clinical applications, including as biomarkers and therapeutic vectors for thrombosis.
Collapse
Affiliation(s)
- Sheng-Lin Ye
- Department of Vascular Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Wen-Dong Li
- Department of Vascular Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Wei-Xiao Li
- Department of Vascular Surgery, Henan Provincial People’s Hospital, Zhengzhou, China
| | - Lun Xiao
- Department of Vascular Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Feng Ran
- Department of Vascular Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Meng-Meng Chen
- School of Electronic Engineering, Nanjing Xiaozhuang University, Nanjing, China
| | - Xiao-Qiang Li
- Department of Vascular Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Li-Li Sun
- Department of Vascular Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| |
Collapse
|
12
|
Expression of tissue factor and TF-mediated integrin regulation in HTR-8/SVneo trophoblast cells. J Reprod Immunol 2022; 150:103473. [PMID: 35030354 DOI: 10.1016/j.jri.2022.103473] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 10/16/2021] [Accepted: 01/03/2022] [Indexed: 11/22/2022]
Abstract
Placenta is a crucial source of Tissue Factor (TF) to initiate coagulation. As far as the TF is concern, aberrant expression of TF has been reported to have a significant role in thrombosis, inflammation, cancer metastasis and atherosclerosis. It is evident that TF and TF-FVIIa complex has major roles in the disease process beyond hemostasis and thrombosis. On the other hand, TF-FVII-dependent signaling primarily activates PAR2 and inducing pro-angiogenic and immune-modulating cytokines in tumor environment. However, the role of TF has not been delineated in placental functions. Integrin typically binds to the extracellular matrix which in turn mediate cell-cell adhesion and cell behavior for migration. Dysregulation of integrin expression affects cell interaction, proliferation, and migration. Therefore, this study aims to ascertain the expression of TF in HTR-8/SVneo trophoblast cell line and its role in signal transduction of integrin (ITGα1, ITGα2, ITGβ1) regulation concerning the invasion of trophoblasts. We have used RT-PCR and Western blot for the gene and protein expression analysis respectively. In addition, cell migration assays, MTT, and DAPI were performed to examine migration, cytotoxicity and apoptosis effect of FVIIa. The results suggest that the gene and protein level expressions of TF were predominant in HTR-8/SVneo cell line. Further, the cytotoxicity and apoptosis in HTR-8/SVneo cells were not observed when treated with FVIIa. The cells treated with FVIIa shown a dose-dependent up-regulation of integrin(s) (**p < 0.01, *p < 0.05) when compared to control. Migration of the HTR-8/SVneo cells was observed without any apoptosis in FVIIa-treated cells when compared to that of control. On the whole, these observations delineated the TF-FVIIa interaction in modulating the TF-dependent integrin signal transduction in HTR-8/SVneo trophoblast cell line.
Collapse
|
13
|
Barale C, Melchionda E, Morotti A, Russo I. Prothrombotic Phenotype in COVID-19: Focus on Platelets. Int J Mol Sci 2021; 22:ijms222413638. [PMID: 34948438 PMCID: PMC8705811 DOI: 10.3390/ijms222413638] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 12/16/2021] [Accepted: 12/17/2021] [Indexed: 12/15/2022] Open
Abstract
COVID-19 infection is associated with a broad spectrum of presentations, but alveolar capillary microthrombi have been described as a common finding in COVID-19 patients, appearing as a consequence of a severe endothelial injury with endothelial cell membrane disruption. These observations clearly point to the identification of a COVID-19-associated coagulopathy, which may contribute to thrombosis, multi-organ damage, and cause of severity and fatality. One significant finding that emerges in prothrombotic abnormalities observed in COVID-19 patients is that the coagulation alterations are mainly mediated by the activation of platelets and intrinsically related to viral-mediated endothelial inflammation. Beyond the well-known role in hemostasis, the ability of platelets to also release various potent cytokines and chemokines has elevated these small cells from simple cell fragments to crucial modulators in the blood, including their inflammatory functions, that have a large influence on the immune response during infectious disease. Indeed, platelets are involved in the pathogenesis of acute lung injury also by promoting NET formation and affecting vascular permeability. Specifically, the deposition by activated platelets of the chemokine platelet factor 4 at sites of inflammation promotes adhesion of neutrophils on endothelial cells and thrombogenesis, and it seems deeply involved in the phenomenon of vaccine-induced thrombocytopenia and thrombosis. Importantly, the hyperactivated platelet phenotype along with evidence of cytokine storm, high levels of P-selectin, D-dimer, and, on the other hand, decreased levels of fibrinogen, von Willebrand factor, and thrombocytopenia may be considered suitable biomarkers that distinguish the late stage of COVID-19 progression in critically ill patients.
Collapse
Affiliation(s)
| | | | | | - Isabella Russo
- Correspondence: ; Tel.: +39-011-6705447; Fax: +39-011-9038639
| |
Collapse
|
14
|
Intersection of regulatory pathways controlling hemostasis and hemochorial placentation. Proc Natl Acad Sci U S A 2021; 118:2111267118. [PMID: 34876522 DOI: 10.1073/pnas.2111267118] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/29/2021] [Indexed: 11/18/2022] Open
Abstract
Hemochorial placentation is characterized by the development of trophoblast cells specialized to interact with the uterine vascular bed. We utilized trophoblast stem (TS) cell and mutant rat models to investigate regulatory mechanisms controlling trophoblast cell development. TS cell differentiation was characterized by acquisition of transcript signatures indicative of an endothelial cell-like phenotype, which was highlighted by the expression of anticoagulation factors including tissue factor pathway inhibitor (TFPI). TFPI localized to invasive endovascular trophoblast cells of the rat placentation site. Disruption of TFPI in rat TS cells interfered with development of the endothelial cell-like endovascular trophoblast cell phenotype. Similarly, TFPI was expressed in human invasive/extravillous trophoblast (EVT) cells situated within first-trimester human placental tissues and following differentiation of human TS cells. TFPI was required for human TS cell differentiation to EVT cells. We next investigated the physiological relevance of TFPI at the placentation site. Genome-edited global TFPI loss-of-function rat models revealed critical roles for TFPI in embryonic development, resulting in homogeneous midgestation lethality prohibiting analysis of the role of TFPI as a regulator of the late-gestation wave of intrauterine trophoblast cell invasion. In vivo trophoblast-specific TFPI knockdown was compatible with pregnancy but had profound effects at the uterine-placental interface, including restriction of the depth of intrauterine trophoblast cell invasion while leading to the accumulation of natural killer cells and increased fibrin deposition. Collectively, the experimentation implicates TFPI as a conserved regulator of invasive/EVT cell development, uterine spiral artery remodeling, and hemostasis at the maternal-fetal interface.
Collapse
|
15
|
Kalita B, Saviola AJ, Samuel SP, Mukherjee AK. State-of-the-art review - A review on snake venom-derived antithrombotics: Potential therapeutics for COVID-19-associated thrombosis? Int J Biol Macromol 2021; 192:1040-1057. [PMID: 34656540 PMCID: PMC8514616 DOI: 10.1016/j.ijbiomac.2021.10.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 10/03/2021] [Accepted: 10/04/2021] [Indexed: 12/30/2022]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent responsible for the Coronavirus Disease-2019 (COVID-19) pandemic, has infected over 185 million individuals across 200 countries since December 2019 resulting in 4.0 million deaths. While COVID-19 is primarily associated with respiratory illnesses, an increasing number of clinical reports indicate that severely ill patients often develop thrombotic complications that are associated with increased mortality. As a consequence, treatment strategies that target COVID-associated thrombosis are of utmost clinical importance. An array of pharmacologically active compounds from natural products exhibit effects on blood coagulation pathways, and have generated interest for their potential therapeutic applications towards thrombotic diseases. In particular, a number of snake venom compounds exhibit high specificity on different blood coagulation factors and represent excellent tools that could be utilized to treat thrombosis. The aim of this review is to provide a brief summary of the current understanding of COVID-19 associated thrombosis, and highlight several snake venom compounds that could be utilized as antithrombotic agents to target this disease.
Collapse
Affiliation(s)
- Bhargab Kalita
- Department of Molecular Biology and Biotechnology, Tezpur University, Tezpur 784028, Assam, India; National Centre for Cell Science, Pune 411007, Maharashtra, India
| | - Anthony J Saviola
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Stephen P Samuel
- Queen Elizabeth Hospital King's Lynn NHS Foundation Trust, King's Lynn, Norfolk PE30 4ET, UK
| | - Ashis K Mukherjee
- Department of Molecular Biology and Biotechnology, Tezpur University, Tezpur 784028, Assam, India; Institute of Advanced Study in Science and Technology, Guwahati 781035, Assam, India.
| |
Collapse
|
16
|
Brusov OS, Senko OV, Kodryan MS, Kuznetsova AV, Matveev IA, Oleichik IV, Karpova NS, Faktor MI, Aleshenko AV, Sizov SV. [Application of machine learning for predicting the outcome of treatment of patients with schizophrenia according to the indicators of «Thrombodynamics» test]. Zh Nevrol Psikhiatr Im S S Korsakova 2021; 121:45-53. [PMID: 34481435 DOI: 10.17116/jnevro202112108145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
OBJECTIVE To identify relationships between thrombodynamic values and the severity of the condition in patients with schizophrenia spectrum disorders (SSD) before and after treatment. MATERIAL AND METHODS The study included 92 patients in an acute state of schizophrenia or schizotypal disorder, aged 16 to 57 years (median age [Q1; Q3] - 25 years). All patients received complex psychopharmacotherapy adequate to their psychopathological state. The PANSS was used to assess the severity of symptoms in patients. The coagulation parameters were determined by the thrombodynamics test, in which the growth of fibrin clots in platelet free plasma are observed from special activator. The patient population was divided into two groups with weak and strong response to treatment. Data analysis included machine learning (ML) techniques: logistic regression, random forests, decision trees, support vector machines with radial basis functions, statistically weighted syndromes, permutation method. RESULTS An analysis using permutation method revealed statistically significant different thrombodynamics values between groups of patients with weak and strong responses. There are significant differences between thrombodynamics values: T1D, T2D, T2Tlag and DTlag, and values characterizing the severity of positive symptoms before and after treatment (T1PposTot, T2PposTot), severity of psychopathological symptoms before treatment (T1Ppsy1, T1Ppsy6, T1Ppsy13). All ML techniques showed the relationship between thrombodynamics values and response to treatment. The best statistical significance was for statistically weighted syndromes method. CONCLUSION The combination of the results of different ML techniques at a high level of statistical significance identifies the thrombodynamic predictors of weak effect of treatment of SSD.
Collapse
Affiliation(s)
- O S Brusov
- Mental Health Research Center of the Ministry of Science and Higher Education, Moscow, Russia
| | - O V Senko
- Federal Research Center «Computer Science and Control» of Russian Academy of Science, Moscow, Russia
| | | | - A V Kuznetsova
- Emanuel Institute of Biochemical Physics of Russian Academy of Science, Moscow, Russia
| | - I A Matveev
- Federal Research Center «Computer Science and Control» of Russian Academy of Science, Moscow, Russia
| | - I V Oleichik
- Mental Health Research Center of the Ministry of Science and Higher Education, Moscow, Russia
| | - N S Karpova
- Mental Health Research Center of the Ministry of Science and Higher Education, Moscow, Russia
| | - M I Faktor
- Mental Health Research Center of the Ministry of Science and Higher Education, Moscow, Russia
| | - A V Aleshenko
- Emanuel Institute of Biochemical Physics of Russian Academy of Science, Moscow, Russia
| | - S V Sizov
- Mental Health Research Center of the Ministry of Science and Higher Education, Moscow, Russia
| |
Collapse
|
17
|
Subrahmanian S, Borczuk A, Salvatore S, Fung KM, Merrill JT, Laurence J, Ahamed J. Tissue factor upregulation is associated with SARS-CoV-2 in the lungs of COVID-19 patients. J Thromb Haemost 2021; 19:2268-2274. [PMID: 34236752 PMCID: PMC8565501 DOI: 10.1111/jth.15451] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 07/02/2021] [Accepted: 07/06/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND A substantial proportion of patients infected with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) develop severe/critical coronavirus disease 2019 (COVID-19) characterized by acute respiratory distress syndrome (ARDS) with thrombosis. OBJECTIVES We tested the hypothesis that SARS-CoV-2--induced upregulation of tissue factor (TF) expression may be responsible for thrombus formation in COVID-19. METHODS We compared autopsy lung tissues from 11 patients with COVID-19--associated ARDS with samples from 6 patients with ARDS from other causes (non-COVID-19 ARDS) and 11 normal control lungs. RESULTS Dual RNA in situ hybridization for SARS-CoV-2 and TF identified sporadic clustered SARS-CoV-2 with prominent co-localization of SARS-CoV-2 and TF RNA. TF expression was 2-fold higher in COVID-19 than in non-COVID-19 ARDS lungs (P = .017) and correlated with the intensity of SARS-CoV-2 staining (R2 = .36, P = .04). By immunofluorescence, TF protein expression was 2.1-fold higher in COVID-19 versus non-COVID-19 ARDS lungs (P = .0048) and 11-fold (P < .001) higher than control lungs. Fibrin thrombi and thrombi positive for platelet factor 4 (PF4) were found in close proximity to regions expressing TF in COVID-19 ARDS lung, and correlated with TF expression (fibrin, R2 = .52, P < .001; PF4, R2 = .59, P < .001). CONCLUSIONS These data suggest that upregulation of TF expression is associated with thrombus formation in COVID-19 lungs and could be a key therapeutic target. Correlation of TF expression with SARS-CoV-2 in lungs of COVID-19 patients also raises the possibility of direct TF induction by the virus.
Collapse
Affiliation(s)
- Sandeep Subrahmanian
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation (OMRF), Oklahoma City, Oklahoma, USA
| | - Alain Borczuk
- Department of Pathology, Weill Cornell Medical College, New York, New York, USA
| | - Steven Salvatore
- Department of Pathology, Weill Cornell Medical College, New York, New York, USA
| | - Kar-Ming Fung
- Department of Pathology, University of Oklahoma (HSC), Oklahoma City, Oklahoma, USA
| | - Joan T Merrill
- Arthritis & Clinical Immunology Research Program, Oklahoma Medical Research Foundation (OMR), Oklahoma City, Oklahoma, USA
| | - Jeffrey Laurence
- Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Jasimuddin Ahamed
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation (OMRF), Oklahoma City, Oklahoma, USA
- Department of Pathology, University of Oklahoma (HSC), Oklahoma City, Oklahoma, USA
- Department of Physiology, University of Oklahoma (HSC), Oklahoma City, Oklahoma, USA
| |
Collapse
|
18
|
Burgelman M, Vandendriessche C, Vandenbroucke RE. Extracellular Vesicles: A Double-Edged Sword in Sepsis. Pharmaceuticals (Basel) 2021; 14:ph14080829. [PMID: 34451925 PMCID: PMC8399948 DOI: 10.3390/ph14080829] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 08/17/2021] [Accepted: 08/17/2021] [Indexed: 01/08/2023] Open
Abstract
Sepsis is defined as a life-threatening organ dysfunction caused by a dysregulated host response to an infection. Several studies on mouse and patient sepsis samples have revealed that the level of extracellular vesicles (EVs) in the blood is altered compared to healthy controls, but the different functions of EVs during sepsis pathology are not yet completely understood. Sepsis EVs are described as modulators of inflammation, lymphocyte apoptosis, coagulation and organ dysfunction. Furthermore, EVs can influence clinical outcome and it is suggested that EVs can predict survival. Both detrimental and beneficial roles for EVs have been described in sepsis, depending on the EV cellular source and the disease phase during which the EVs are studied. In this review, we summarize the current knowledge of EV sources and functions during sepsis pathology based on in vitro and mouse models, as well as patient samples.
Collapse
Affiliation(s)
- Marlies Burgelman
- VIB Center for Inflammation Research, 9052 Ghent, Belgium; (M.B.); (C.V.)
- Department of Biomedical Molecular Biology, Ghent University, 9000 Ghent, Belgium
| | - Charysse Vandendriessche
- VIB Center for Inflammation Research, 9052 Ghent, Belgium; (M.B.); (C.V.)
- Department of Biomedical Molecular Biology, Ghent University, 9000 Ghent, Belgium
| | - Roosmarijn E. Vandenbroucke
- VIB Center for Inflammation Research, 9052 Ghent, Belgium; (M.B.); (C.V.)
- Department of Biomedical Molecular Biology, Ghent University, 9000 Ghent, Belgium
- Correspondence: ; Tel.: +32-9-3313730
| |
Collapse
|
19
|
Zhang W, Sang L, Shi J, Zhong M, Jiang L, Song B, Kang L, Zhang Y, Zhang D, Yu Y, Zheng X. Association of D-dimer elevation with inflammation and organ dysfunction in ICU patients with COVID-19 in Wuhan, China: a retrospective observational study. Aging (Albany NY) 2021; 13:4794-4810. [PMID: 33591951 PMCID: PMC7950237 DOI: 10.18632/aging.202496] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 12/19/2020] [Indexed: 12/21/2022]
Abstract
Coronavirus disease 2019 (COVID-19)-associated coagulation dysfunction is gaining attention. In particular, dynamic changes in the D-dimer level may be related to disease progression. Here, we explored whether elevated D-dimer level was related to multiple organ failure and a higher risk of death. This study included 158 patients with COVID-19 who were admitted to the intensive care unit (ICU) at Jinyintan Hospital in Wuhan, China between January 20, 2020 and February 26, 2020. Clinical and laboratory data were collected. The relationship between D-dimer elevation and organ dysfunction was analyzed, as were dynamic changes in inflammation and lipid metabolism. Approximately 63.9% of patients with COVID-19 had an elevated D-dimer level on ICU admission. The 14 day ICU mortality rate was significantly higher in patients with a high D-dimer level than in those with a normal D-dimer level. Patients with a D-dimer level of 10-40μg/mL had similar organ function on ICU admission to those with a D-dimer level of 1.5-10μg/mL. However, patients with higher levels of D-dimer developed organ injuries within 7 days. Furthermore, significant differences in inflammation and lipid metabolism markers were observed between the two groups. In conclusion, the D-dimer level is closely related to COVID-19 severity and might influence the likelihood of rapid onset of organ injury after admission.
Collapse
Affiliation(s)
- Wang Zhang
- Department of Infectious Diseases, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, P.R. China
| | - Ling Sang
- Department of Critical Care Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Institute of Respiratory Health, Guangzhou, Guangdong, P.R. China
| | - Jiaran Shi
- Department of Cardiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, P.R. China
| | - Ming Zhong
- Department of Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, P.R. China
| | - Li Jiang
- Department of Critical Care Medicine, Xuanwu Hospital, Capital Medical University, Beijing, P.R. China
| | - Bin Song
- Department of Tuberculosis and Respiratory Disease, Jinyintan Hospital, Wuhan, Hubei, P.R. China
| | - Liang Kang
- Department of Critical Care Medicine, Jinyintan Hospital, Wuhan, Hubei, P.R. China
| | - Yun Zhang
- Department of Critical Care Medicine, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, P.R. China
| | - Dingyu Zhang
- Joint Laboratory of Infectious Diseases and Health, Wuhan Institute of Virology and Wuhan Jinyintan Hospital, Chinese Academy of Sciences, Hubei, P.R. China
- Research Center for Translational Medicine, Wuhan Jinyintan Hospital, Hubei, P.R. China
| | - Yunsong Yu
- Department of Infectious Diseases, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, P.R. China
| | - Xia Zheng
- Department of Critical Care Medicine, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, P.R. China
| |
Collapse
|
20
|
Orioli S, Henning Hansen CG, Arleth L. Ab initio determination of the shape of membrane proteins in a nanodisc. ACTA CRYSTALLOGRAPHICA SECTION D-STRUCTURAL BIOLOGY 2021; 77:176-193. [DOI: 10.1107/s2059798320015405] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 11/20/2020] [Indexed: 11/10/2022]
Abstract
New software, called Marbles, is introduced that employs SAXS intensities to predict the shape of membrane proteins embedded into membrane nanodiscs. To gain computational speed and efficient convergence, the strategy is based on a hybrid approach that allows one to account for the contribution of the nanodisc to the SAXS intensity through a semi-analytical model, while the embedded membrane protein is treated as a set of beads, similarly to as in well known ab initio methods. The reliability and flexibility of this approach is proved by benchmarking the code, implemented in C++ with a Python interface, on a toy model and two proteins with very different geometry and size.
Collapse
|
21
|
Brusov OS, Oleichik IV, Karpova NS, Faktor MI, Sizov SV. [Correlations between thrombodynamic parameters of coagulation and negative syndromes in patients with schizophrenia]. Zh Nevrol Psikhiatr Im S S Korsakova 2021; 120:86-91. [PMID: 33459546 DOI: 10.17116/jnevro202012012186] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
OBJECTIVE To identify a possible correlation between parameters of thrombodynamic coagulation and negative syndromes in patients with schizophrenia. MATERIAL AND METHODS The study included 148 female inpatients, aged 16 to 57 years, with the following mental disorders: schizophrenia with attack-like/attack-progressive/continuous type of course (F20.00-2), schizotypal disorder with affective fluctuations (F21.3-4). The thrombodynamics test (TD) was carried out on a T-2 thrombodynamics recorder (Hemacore LLC, Moscow, Russia). RESULTS A positive correlation was shown between the thrombodynamic parameters of clot growth rates (V, Vst, and Vi), clot size at the 30th minute (CS), and the total severity of negative syndromes (PANSS). There is a negative correlation between the time of spontaneous clots (Tsp) and the total severity of negative syndromes in patients. Positive correlations of V and Vst with scores on the fourth (Passive/apathetic social withdrawal), fifth (Difficulty in abstract thinking) and seventh (Stereotyped thinking) items of the PANSS negative subscale were revealed. There is a negative correlation between Tsp and the score on the 7th item, i.e. a shorter time for the appearance of spontaneous clots corresponds to a more pronounced Stereotyped thinking in patients. CONCLUSION For the first time, correlations between thrombodynamic indicators of hypercoagulation and negative syndromes in patients with schizophrenia are identified, which emphasizes the need to normalize hemostasis to prevent further aggravation of these disorders.
Collapse
Affiliation(s)
- O S Brusov
- Mental Health Research Center, Moscow, Russia
| | | | - N S Karpova
- Mental Health Research Center, Moscow, Russia
| | - M I Faktor
- Mental Health Research Center, Moscow, Russia
| | - S V Sizov
- Mental Health Research Center, Moscow, Russia
| |
Collapse
|
22
|
Shi J, Zhang W, Sang L, Qu Z, Zhong M, Jiang L, Song B, Kang L, Zhang Y, Wang X, Zhang D, Zheng X. Coagulation dysfunction in ICU patients with coronavirus disease 2019 in Wuhan, China: a retrospective observational study of 75 fatal cases. Aging (Albany NY) 2020; 13:1591-1607. [PMID: 33318314 PMCID: PMC7880373 DOI: 10.18632/aging.202223] [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: 08/24/2020] [Accepted: 10/27/2020] [Indexed: 12/19/2022]
Abstract
Coagulation dysfunction in critically ill patients with coronavirus disease 2019 (COVID-19) has not been well described, and the efficacy of anticoagulant therapy is unclear. In this study, we retrospectively reviewed 75 fatal COVID-19 cases who were admitted to the intensive care unit at Jinyintan Hospital (Wuhan, China). The median age of the cases was 67 (62-74) years, and 47 (62.7%) were male. Fifty patients (66.7%) were diagnosed with disseminated intra-vascular coagulation. Approximately 90% of patients had elevated D-dimer and fibrinogen degradation products, which decreased continuously after anticoagulant treatment and was accompanied by elevated albumin (all P<0.05). The median survival time of patients treated with anticoagulant was 9.0 (6.0-14.0) days compared with 7.0 (3.0-10.0) days in patients without anticoagulant therapy (P=0.008). After anticoagulation treatment, C-reactive protein levels decreased (P=0.004), as did high-sensitivity troponin (P=0.018), lactate dehydrogenase (P<0.001), and hydroxybutyrate dehydrogenase (P<0.001). In conclusion, coagulation disorders were widespread among fatal COVID-19 cases. Anticoagulant treatment partially improved hypercoagulability, prolonged median survival time, and may have postponed inflammatory processes and cardiac injury.
Collapse
Affiliation(s)
- Jiaran Shi
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, PR China
| | - Wang Zhang
- Department of Infectious Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, PR China
| | - Ling Sang
- Department of Critical Care Medicine, The First Affiliated Hospital of GuangZhou Medical University, GuangZhou Institute of Respiratory Health, Guangzhou, Guangdong, PR China
| | - Zhaohui Qu
- Department of Critical Care Medicine, Jinyintan Hospital, Wuhan, Hubei, PR China
| | - Ming Zhong
- Department of Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, PR China
| | - Li Jiang
- Department of Critical Care Medicine, Xuanwu Hospital, Capital Medical University, Beijing, PR China
| | - Bin Song
- Department of Tuberculosis and Respiratory Disease, Jinyintan Hospital, Wuhan, Hubei, PR China
| | - Liang Kang
- Department of Critical Care Medicine, Jinyintan Hospital, Wuhan, Hubei, PR China
| | - Yun Zhang
- Department of Critical Care Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, PR China
| | - Xingxiang Wang
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, PR China
| | - Dingyu Zhang
- Research Center for Translational Medicine, Wuhan Jinyintan Hospital, Hubei, PR China.,Joint Laboratory of Infectious Diseases and Health, Wuhan Institute of Virology and Wuhan Jinyintan Hospital, Chinese Academy of Sciences, Hubei, PR China
| | - Xia Zheng
- Department of Critical Care Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, PR China
| |
Collapse
|
23
|
Abstract
An ongoing global pandemic of viral pneumonia (coronavirus disease [COVID-19]), due to the virus SARS-CoV-2, has infected millions of people and remains a threat to many more. Most critically ill patients have respiratory failure and there is an international effort to understand mechanisms and predictors of disease severity. Coagulopathy, characterized by elevations in D-dimer and fibrin(ogen) degradation products (FDPs), is associated with critical illness and mortality in patients with COVID-19. Furthermore, increasing reports of microvascular and macrovascular thrombi suggest that hemostatic imbalances may contribute to the pathophysiology of SARS-CoV-2 infection. We review the laboratory and clinical findings of patients with COVID-19-associated coagulopathy, and prior studies of hemostasis in other viral infections and acute respiratory distress syndrome. We hypothesize that an imbalance between coagulation and inflammation may result in a hypercoagulable state. Although thrombosis initiated by the innate immune system is hypothesized to limit SARS-CoV-2 dissemination, aberrant activation of this system can cause endothelial injury resulting in loss of thromboprotective mechanisms, excess thrombin generation, and dysregulation of fibrinolysis and thrombosis. The role various components including neutrophils, neutrophil extracellular traps, activated platelets, microparticles, clotting factors, inflammatory cytokines, and complement play in this process remains an area of active investigation and ongoing clinical trials target these different pathways in COVID-19.
Collapse
Affiliation(s)
- Meaghan E Colling
- Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Yogendra Kanthi
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan, and Ann Arbor Veterans Administration Healthcare System, Ann Arbor, MI, USA
| |
Collapse
|
24
|
Monocyte procoagulant responses to anthrax peptidoglycan are reinforced by proinflammatory cytokine signaling. Blood Adv 2020; 3:2436-2447. [PMID: 31416821 DOI: 10.1182/bloodadvances.2019000513] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 06/17/2019] [Indexed: 01/03/2023] Open
Abstract
Disseminated intravascular coagulation is a frequent manifestation during bacterial infections and is associated with negative clinical outcomes. Imbalanced expression and activity of intravascular tissue factor (TF) is central to the development of infection-associated coagulopathies. Recently, we showed that anthrax peptidoglycan (PGN) induces disseminated intravascular coagulation in a nonhuman primate model of anthrax sepsis. We hypothesized that immune recognition of PGN by monocytes is critical for procoagulant responses to PGN and investigated whether and how PGN induces TF expression in primary human monocytes. We found that PGN induced monocyte TF expression in a large cohort of healthy volunteers similar to lipopolysaccharide stimulation. Both immune and procoagulant responses to PGN involve intracellular recognition after PGN internalization, as well as surface signaling through immune Fcγ receptors (FcγRs). In line with our hypothesis, blocking immune receptor function, both signaling and FcγR-mediated phagocytosis, significantly reduced but did not abolish PGN-induced monocyte TF expression, indicating that FcγR-independent internalization contributes to intracellular recognition of PGN. Conversely, when intracellular PGN recognition is abolished, TF expression was sensitive to inhibitors of FcγR signaling, indicating that surface engagement of monocyte immune receptors can promote TF expression. The primary procoagulant responses to PGN were further amplified by proinflammatory cytokines through paracrine and autocrine signaling. Despite intersubject variability in the study cohort, dual neutralization of tumor necrosis factor-α and interleukin-1β provided the most robust inhibition of the procoagulant amplification loop and may prove useful for reducing coagulopathies in gram-positive sepsis.
Collapse
|
25
|
Vermeulen JG, Burt F, van Heerden E, du Preez LL, Meiring M. Characterization of the inhibition mechanism of a tissuefactor inhibiting single-chain variable fragment: a combined computational approach. J Mol Model 2020; 26:87. [PMID: 32219568 DOI: 10.1007/s00894-020-4350-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 03/11/2020] [Indexed: 10/24/2022]
Abstract
The interaction of a single-chain variable fragment (scFv) directed against human tissue factor (TF) was predicted using an in silico approach with the aim to establish a most likely mechanism of inhibition. The structure of the TF inhibiting scFv (TFI-scFv) was predicted using homology modeling, and complementarity-determining regions (CDRs) were identified. The CDR was utilized to direct molecular docking between the homology model of TFI-scFv and the crystal structure of the extracellular domains of human tissue factor. The rigid-body docking model was refined by means of molecular dynamic (MD) simulations, and the most prevalent cluster was identified. MD simulations predicted improved interaction between TFI-scFv and TF and propose the formation of stable complex for duration of the 600-ns simulation. Analysis of the refined docking model suggests that the interactions between TFI-scFv would interfere with the allosterical activation of coagulation factor VII (FVII) by TF. This interaction would prevent the formation of the active TF:VIIa complex and in so doing inhibit the initiation phase of blood coagulation as observers during in vitro testing.
Collapse
Affiliation(s)
- Jan-G Vermeulen
- Department of Microbial, Biochemical and Food Biotechnology, Faculty of Agricultural Sciences, University of the Free State, Bloemfontein, South Africa. .,Department of Haematology and Cell Biology, Faculty of Health Sciences, University of the Free State, Bloemfontein, South Africa.
| | - Felicity Burt
- Division of Virology, Faculty of Health Sciences, University of the Free State, Bloemfontein, South Africa.,National Health Laboratory Service, Universitas, Bloemfontein, South Africa
| | - Esta van Heerden
- Department of Microbial, Biochemical and Food Biotechnology, Faculty of Agricultural Sciences, University of the Free State, Bloemfontein, South Africa
| | - Louis Lategan du Preez
- Department of Microbial, Biochemical and Food Biotechnology, Faculty of Agricultural Sciences, University of the Free State, Bloemfontein, South Africa
| | - Muriel Meiring
- Department of Haematology and Cell Biology, Faculty of Health Sciences, University of the Free State, Bloemfontein, South Africa.,National Health Laboratory Service, Universitas, Bloemfontein, South Africa
| |
Collapse
|
26
|
Silva-Vaz P, Abrantes AM, Castelo-Branco M, Gouveia A, Botelho MF, Tralhão JG. Multifactorial Scores and Biomarkers of Prognosis of Acute Pancreatitis: Applications to Research and Practice. Int J Mol Sci 2020; 21:E338. [PMID: 31947993 PMCID: PMC6982212 DOI: 10.3390/ijms21010338] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 12/30/2019] [Accepted: 01/02/2020] [Indexed: 02/07/2023] Open
Abstract
Acute pancreatitis (AP) is a severe inflammation of the pancreas presented with sudden onset and severe abdominal pain with a high morbidity and mortality rate, if accompanied by severe local and systemic complications. Numerous studies have been published about the pathogenesis of AP; however, the precise mechanism behind this pathology remains unclear. Extensive research conducted over the last decades has demonstrated that the first 24 h after symptom onset are critical for the identification of patients who are at risk of developing complications or death. The identification of these subgroups of patients is crucial in order to start an aggressive approach to prevent mortality. In this sense and to avoid unnecessary overtreatment, thereby reducing the financial implications, the proper identification of mild disease is also important and necessary. A large number of multifactorial scoring systems and biochemical markers are described to predict the severity. Despite recent progress in understanding the pathophysiology of AP, more research is needed to enable a faster and more accurate prediction of severe AP. This review provides an overview of the available multifactorial scoring systems and biochemical markers for predicting severe AP with a special focus on their advantages and limitations.
Collapse
Affiliation(s)
- Pedro Silva-Vaz
- Health Sciences Research Centre, University of Beira Interior (CICS-UBI), 6200-506 Covilhã, Portugal;
- General Surgery Department, Hospital Local de Saúde de Castelo Branco, 6000-085 Castelo Branco, Portugal;
- Faculty of Health Sciences, University of Beira Interior, 6200-506 Covilhã, Portugal
| | - Ana Margarida Abrantes
- Coimbra Institute for Clinical and Biomedical Research (iCBR) area of Environment Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (A.M.A.); (M.F.B.); (J.G.T.)
- Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- Biophysics and Biomathematics Institute, IBILI-Faculty of Medicine of University of Coimbra, 3000-348 Coimbra, Portugal
| | - Miguel Castelo-Branco
- Health Sciences Research Centre, University of Beira Interior (CICS-UBI), 6200-506 Covilhã, Portugal;
- Faculty of Health Sciences, University of Beira Interior, 6200-506 Covilhã, Portugal
| | - António Gouveia
- General Surgery Department, Hospital Local de Saúde de Castelo Branco, 6000-085 Castelo Branco, Portugal;
- Faculty of Health Sciences, University of Beira Interior, 6200-506 Covilhã, Portugal
| | - Maria Filomena Botelho
- Coimbra Institute for Clinical and Biomedical Research (iCBR) area of Environment Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (A.M.A.); (M.F.B.); (J.G.T.)
- Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- Biophysics and Biomathematics Institute, IBILI-Faculty of Medicine of University of Coimbra, 3000-348 Coimbra, Portugal
| | - José Guilherme Tralhão
- Coimbra Institute for Clinical and Biomedical Research (iCBR) area of Environment Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (A.M.A.); (M.F.B.); (J.G.T.)
- Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- Biophysics and Biomathematics Institute, IBILI-Faculty of Medicine of University of Coimbra, 3000-348 Coimbra, Portugal
- Surgery Department, Centro Hospitalar e Universitário de Coimbra (CHUC), University Hospital, Faculty of Medicine, 3000-075 Coimbra, Portugal
| |
Collapse
|
27
|
Luchini A, Tidemand FG, Johansen NT, Campana M, Sotres J, Ploug M, Cárdenas M, Arleth L. Peptide Disc Mediated Control of Membrane Protein Orientation in Supported Lipid Bilayers for Surface-Sensitive Investigations. Anal Chem 2019; 92:1081-1088. [DOI: 10.1021/acs.analchem.9b04125] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Alessandra Luchini
- Niels Bohr Institute, University of Copenhagen, Universitetsparken 5, 2100 Copenhagen, Denmark
| | | | | | - Mario Campana
- ISIS-STFC, Rutherford Appleton Laboratory, Chilton, Oxon OX11 0QX, United Kingdom
| | - Javier Sotres
- Biofilms Research Center for Biointerfaces and Department of Biomedical Science, Faculty of Health and Society, Malmö University, Per Albin Hanssons Väg 35, 214 32 Malmö, Sweden
| | - Michael Ploug
- Biotech Research and Innovation Center, University of Copenhagen, Ole Maaløes Vej 5, 2200 Copenhagen, Denmark
- Finsen Laboratory, Rigshospitalet, Ole Maaløes Vej 5, 2200 Copenhagen, Denmark
| | - Marité Cárdenas
- Biofilms Research Center for Biointerfaces and Department of Biomedical Science, Faculty of Health and Society, Malmö University, Per Albin Hanssons Väg 35, 214 32 Malmö, Sweden
| | - Lise Arleth
- Niels Bohr Institute, University of Copenhagen, Universitetsparken 5, 2100 Copenhagen, Denmark
| |
Collapse
|
28
|
15-epi-lipoxin A4 inhibits TNF-α-induced tissue factor expression via the PI3K/AKT/ NF-κB axis in human umbilical vein endothelial cells. Biomed Pharmacother 2019; 117:109099. [DOI: 10.1016/j.biopha.2019.109099] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 06/04/2019] [Accepted: 06/04/2019] [Indexed: 01/03/2023] Open
|
29
|
Zarà M, Guidetti GF, Camera M, Canobbio I, Amadio P, Torti M, Tremoli E, Barbieri SS. Biology and Role of Extracellular Vesicles (EVs) in the Pathogenesis of Thrombosis. Int J Mol Sci 2019; 20:ijms20112840. [PMID: 31212641 PMCID: PMC6600675 DOI: 10.3390/ijms20112840] [Citation(s) in RCA: 109] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 06/07/2019] [Indexed: 02/06/2023] Open
Abstract
Extracellular vesicles (EVs) are well-established mediators of cell-to-cell communication. EVs can be released by every cell type and they can be classified into three major groups according to their biogenesis, dimension, density, and predominant protein markers: exosomes, microvesicles, and apoptotic bodies. During their formation, EVs associate with specific cargo from their parental cell that can include RNAs, free fatty acids, surface receptors, and proteins. The biological function of EVs is to maintain cellular and tissue homeostasis by transferring critical biological cargos to distal or neighboring recipient cells. On the other hand, their role in intercellular communication may also contribute to the pathogenesis of several diseases, including thrombosis. More recently, their physiological and biochemical properties have suggested their use as a therapeutic tool in tissue regeneration as well as a novel option for drug delivery. In this review, we will summarize the impact of EVs released from blood and vascular cells in arterial and venous thrombosis, describing the mechanisms by which EVs affect thrombosis and their potential clinical applications.
Collapse
Affiliation(s)
- Marta Zarà
- Unit of Heart-Brain Axis: Cellular and Molecular Mechanisms, Centro Cardiologico Monzino IRCCS, 20138 Milano, Italy.
| | | | - Marina Camera
- Department of Pharmacological and Biomolecular Sciences, University of Milan, 20133 Milano, Italy.
- Unit of Cell and Molecular Biology in Cardiovascular Diseases, Centro Cardiologico Monzino IRCCS, 20138 Milano, Italy.
| | - Ilaria Canobbio
- Department of Biology and Biotechnology, University of Pavia, 27100 Pavia, Italy.
| | - Patrizia Amadio
- Unit of Heart-Brain Axis: Cellular and Molecular Mechanisms, Centro Cardiologico Monzino IRCCS, 20138 Milano, Italy.
| | - Mauro Torti
- Department of Biology and Biotechnology, University of Pavia, 27100 Pavia, Italy.
| | - Elena Tremoli
- Scientific Direction, Centro Cardiologico Monzino IRCCS, 20138 Milano, Italy.
| | - Silvia Stella Barbieri
- Unit of Heart-Brain Axis: Cellular and Molecular Mechanisms, Centro Cardiologico Monzino IRCCS, 20138 Milano, Italy.
| |
Collapse
|
30
|
Korkmaz HI, Hahn NE, Jansen KM, Musters R, van Bezu J, van Wieringen WN, van Zuijlen P, Ulrich M, Niessen H, Krijnen P. Homocysteine-induced inverse expression of tissue factor and DPP4 in endothelial cells is related to NADPH oxidase activity. Physiol Int 2019; 106:29-38. [PMID: 30888218 DOI: 10.1556/2060.106.2019.05] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
PURPOSE We previously found that homocysteine (Hcy)-induced apoptosis in endothelial cells coincided with increased NADPH oxidase (NOX) activity. In addition, in ischemic endothelial cells present in the heart, we showed that loss of serine protease dipeptidyl peptidase IV (DPP4) expression was correlated with induction of tissue factor (TF) expression. Since Hcy can initiate thrombosis through the induction of TF expression, in this study, we evaluated whether the inverse relation of TF and DPP4 is also Hcy-dependent and whether NOX-mediated reactive oxygen species (ROS) is playing a role herein. METHODS Human umbilical vein endothelial cells (HUVECs) were incubated with 2.5 mM Hcy for 3 and 6 h. The effects of Hcy on DPP4 and TF expression and NOX2/p47phox-mediated nitrotyrosine (ROS) production were studied using digital-imaging microscopy. RESULTS In HUVECs, high levels of Hcy showed a significant increase of TF expression and a concomitant loss of DPP4 expression after 6 h. In addition, NOX subunits NOX2 and p47phox were also significantly increased after 6 h of Hcy incubation and coincided with nitrotyrosine (ROS) expression. Interestingly, inhibition of NOX-mediated nitrotyrosine (ROS) with the use of apocynin not only reduced these effects, but also counteracted the effects of Hcy on TF and DPP4 expression. CONCLUSION These results indicate that the inverse relation of TF and DPP4 in endothelial cells is also Hcy-dependent and related to NOX activity.
Collapse
Affiliation(s)
- H I Korkmaz
- 1 Department of Pathology, Amsterdam UMC, VUmc , Amsterdam, The Netherlands.,2 Amsterdam Cardiovascular Sciences (ACS), Amsterdam UMC, VUmc , Amsterdam, The Netherlands
| | - N E Hahn
- 1 Department of Pathology, Amsterdam UMC, VUmc , Amsterdam, The Netherlands.,2 Amsterdam Cardiovascular Sciences (ACS), Amsterdam UMC, VUmc , Amsterdam, The Netherlands
| | - K M Jansen
- 1 Department of Pathology, Amsterdam UMC, VUmc , Amsterdam, The Netherlands
| | - Rjp Musters
- 3 Department of Physiology, Amsterdam UMC, VUmc , Amsterdam, The Netherlands
| | - J van Bezu
- 3 Department of Physiology, Amsterdam UMC, VUmc , Amsterdam, The Netherlands
| | - W N van Wieringen
- 4 Department of Epidemiology and Biostatistics, Amsterdam UMC, VUmc , Amsterdam, The Netherlands.,5 Department of Mathematics, Amsterdam UMC, VUmc , Amsterdam, The Netherlands
| | - Ppm van Zuijlen
- 6 Department of Plastic, Reconstructive and Hand Surgery, MOVE Research Institute, Amsterdam UMC, VUmc , Amsterdam, The Netherlands.,7 Department of Plastic and Reconstructive Surgery, Burn Center, Red Cross Hospital , Beverwijk, The Netherlands.,8 Preclinical Research, Association of Dutch Burn Centres (ADBC) , Beverwijk, The Netherlands
| | - Mmw Ulrich
- 1 Department of Pathology, Amsterdam UMC, VUmc , Amsterdam, The Netherlands.,8 Preclinical Research, Association of Dutch Burn Centres (ADBC) , Beverwijk, The Netherlands.,9 Department of Molecular Cell Biology and Immunology, Amsterdam UMC, VUmc , Amsterdam, The Netherlands
| | - Hwm Niessen
- 1 Department of Pathology, Amsterdam UMC, VUmc , Amsterdam, The Netherlands.,2 Amsterdam Cardiovascular Sciences (ACS), Amsterdam UMC, VUmc , Amsterdam, The Netherlands.,10 Department of Cardiac Surgery, Amsterdam UMC, VUmc , Amsterdam, The Netherlands
| | - Paj Krijnen
- 1 Department of Pathology, Amsterdam UMC, VUmc , Amsterdam, The Netherlands.,2 Amsterdam Cardiovascular Sciences (ACS), Amsterdam UMC, VUmc , Amsterdam, The Netherlands
| |
Collapse
|
31
|
D'Angelo C, Franch O, Fernández-Paredes L, Oreja-Guevara C, Núñez-Beltrán M, Comins-Boo A, Reale M, Sánchez-Ramón S. Antiphospholipid Antibodies Overlapping in Isolated Neurological Syndrome and Multiple Sclerosis: Neurobiological Insights and Diagnostic Challenges. Front Cell Neurosci 2019; 13:107. [PMID: 30941020 PMCID: PMC6433987 DOI: 10.3389/fncel.2019.00107] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2018] [Accepted: 03/04/2019] [Indexed: 01/19/2023] Open
Abstract
Antiphospholipid syndrome (APS) is characterized by arterial and venous thrombosis, pregnancy morbidity and fetal loss caused by pathogenic autoantibodies directed against phospholipids (PL) and PL-cofactors. Isolated neurological APS may represent a significant diagnostic challenge, as epidemiological, clinical and neuroimaging features may overlap with those of multiple sclerosis (MS). In an open view, MS could be considered as an organ-specific anti-lipid (phospholipid and glycosphingolipid associated proteins) disease, in which autoreactive B cells and CD8+ T cells play a dominant role in its pathophysiology. In MS, diverse autoantibodies against the lipid-protein cofactors of the myelin sheath have been described, whose pathophysiologic role has not been fully elucidated. We carried out a review to select clinical studies addressing the prevalence of antiphospholipid (aPL) autoantibodies in the so-called MS-like syndrome. The reported prevalence ranged between 2% and 88%, particularly aCL and aβ2GPI, with predominant IgM isotype and suggesting worse MS prognosis. Secondarily, an updated summary of current knowledge on the pathophysiological mechanisms and events responsible for these conditions is presented. We draw attention to the clinical relevance of diagnosing isolated neurological APS. Prompt and accurate diagnosis and antiaggregant and anticoagulant treatment of APS could be vital to prevent or at least reduce APS-related morbidity and mortality.
Collapse
Affiliation(s)
- Chiara D'Angelo
- Department of Clinical Immunology and IdISSC, Hospital Clínico San Carlos, Madrid, Spain.,Department of Immunology, Ophthalmology and ENT, Complutense University School of Medicine, Madrid, Spain.,Department of Medical, Oral and Biotechnological Sciences, University "G. d'Annunzio" Chieti-Pescara, Chieti, Italy
| | - Oriol Franch
- Department of Neurology, Hospital Ruber Internacional, Madrid, Spain
| | - Lidia Fernández-Paredes
- Department of Clinical Immunology and IdISSC, Hospital Clínico San Carlos, Madrid, Spain.,Department of Immunology, Ophthalmology and ENT, Complutense University School of Medicine, Madrid, Spain
| | | | - María Núñez-Beltrán
- Department of Clinical Immunology and IdISSC, Hospital Clínico San Carlos, Madrid, Spain
| | - Alejandra Comins-Boo
- Department of Clinical Immunology and IdISSC, Hospital Clínico San Carlos, Madrid, Spain.,Department of Immunology, Ophthalmology and ENT, Complutense University School of Medicine, Madrid, Spain
| | - Marcella Reale
- Department of Medical, Oral and Biotechnological Sciences, University "G. d'Annunzio" Chieti-Pescara, Chieti, Italy
| | - Silvia Sánchez-Ramón
- Department of Clinical Immunology and IdISSC, Hospital Clínico San Carlos, Madrid, Spain.,Department of Immunology, Ophthalmology and ENT, Complutense University School of Medicine, Madrid, Spain
| |
Collapse
|
32
|
Abstract
Venous thromboembolism is known to be associated with an increase in morbidity and mortality in patients with malignancy. Predictive laboratory biomarkers of venous thromboembolism (VTE) have long been sought after to improve outcomes and help guide clinical decision making. Previously studied biomarkers include C reactive protein (CRP), tissue factor expressing microparticles (TF MP), D-dimer, soluble P-selectin (sP-selectin), plasminogen activator inhibitor 1 (PAI-1), factor VIII, platelet count, and leukocyte counts. This chapter will focus on these possible biomarkers for cancer-associated thrombosis (CAT) with particular emphasis on the pathophysiology behind thrombosis formation as well as data from clinical studies in patients with malignancy. The incorporation of the above biomarkers into risk assessment tools to predict CAT will also be reviewed, as will risk factors for recurrent VTE in patients with malignancy. Further studies are ongoing to develop readily available biomarkers that can be incorporated into future risk assessment models with the goal of reducing morbidity and mortality due to cancer-associated thrombosis.
Collapse
Affiliation(s)
- Anjlee Mahajan
- Division of Hematology and Oncology, UC Davis School of Medicine, UC Davis Cancer Center, 4501 X Street, Sacramento, CA, 95817, USA.
| | - Ted Wun
- Division of Hematology and Oncology, UC Davis School of Medicine, UC Davis Cancer Center, 4501 X Street, Sacramento, CA, 95817, USA
- UC Davis School of Medicine, Clinical and Translational Sciences Center (CTSC), Sacramento, USA
| |
Collapse
|
33
|
Role of TF-Triggered Activation of the Coagulation Cascade in the Pathogenesis of Chronic Spontaneous Urticaria. CURRENT TREATMENT OPTIONS IN ALLERGY 2018. [DOI: 10.1007/s40521-018-0183-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
|
34
|
Göbel K, Eichler S, Wiendl H, Chavakis T, Kleinschnitz C, Meuth SG. The Coagulation Factors Fibrinogen, Thrombin, and Factor XII in Inflammatory Disorders-A Systematic Review. Front Immunol 2018; 9:1731. [PMID: 30105021 PMCID: PMC6077258 DOI: 10.3389/fimmu.2018.01731] [Citation(s) in RCA: 108] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 07/12/2018] [Indexed: 11/13/2022] Open
Abstract
Background The interaction of coagulation factors has been shown to go beyond their traditional roles in hemostasis and to affect the development of inflammatory diseases. Key molecular players, such as fibrinogen, thrombin, or factor XII have been mechanistically and epidemiologically linked to inflammatory disorders like multiple sclerosis (MS), rheumatoid arthritis (RA), and colitis. Objectives To systematically review the evidence for a role of coagulation factors, especially factor XII, fibrinogen, and thrombin in inflammatory disorders like MS, RA, and bowel disorders. Methods A systematic literature search was done in the PubMed database to identify studies about coagulation factors in inflammatory diseases. Original articles and reviews investigating the role of the kallikrein–kinin and the coagulation system in mouse and humans were included. Results We identified 43 animal studies dealing with inflammatory disorders and factors of the kallikrein–kinin or the coagulation system. Different immunological influences are described and novel molecular mechanisms linking coagulation and inflammation are reported. Conclusion A number of studies have highlighted coagulation factors to tip the balance between hemostasis and thrombosis and between protection from infection and extensive inflammation. To optimize the treatment of chronic inflammatory disorders by these factors, further studies are necessary.
Collapse
Affiliation(s)
- Kerstin Göbel
- Department of Neurology with Institute of Translational Neurology, University of Münster, Münster, Germany
| | - Susann Eichler
- Department of Neurology with Institute of Translational Neurology, University of Münster, Münster, Germany
| | - Heinz Wiendl
- Department of Neurology with Institute of Translational Neurology, University of Münster, Münster, Germany
| | - Triantafyllos Chavakis
- Department of Clinical Pathobiochemistry, Laboratory Medicine, Institute for Clinical Chemistry, University Clinic Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | | | - Sven G Meuth
- Department of Neurology with Institute of Translational Neurology, University of Münster, Münster, Germany
| |
Collapse
|
35
|
Design and Utility of a Point-of-Care Microfluidic Platform to Assess Hematocrit and Blood Coagulation. Cell Mol Bioeng 2018; 11:519-529. [PMID: 31105798 DOI: 10.1007/s12195-018-0541-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Purpose— To develop a small volume whole blood analyzer capable of measuring the hematocrit and coagulation kinetics of whole blood. Methods and Results— A co-planar microfluidic chamber designed to facilitate self-driven capillary action across an internal electrical chip was developed and used to measure the electric parameters of whole human blood that had been anticoagulated or allowed to clot. To promote blood clotting, select chip surfaces were coated with a prothrombin time (PT) reagent containing lipidated tissue factor (TF), which activates the extrinsic pathway of coagulation to promote thrombin generation and fibrin formation. Whole human blood was added to the microfluidic device, and voltage changes within the platform were measured and interpreted using basic resistor-capacitor (RC) circuit and fluid dynamics theory. Upon wetting of the sensing zone, a circuit between two co-planar electrodes within the sensing zone was closed to generate a rapid voltage drop from baseline. The voltage then rose due to sedimentation of red blood cells (RBC) in the sensing zone. For anticoagulated blood samples, the time for the voltage to return to baseline was dependent on hematocrit. In the presence of coagulation, the initiation of fibrin formation in the presence of the PT reagent prevented the return of voltage to baseline due to the reduced packing of RBCs in the sensing zone. Conclusions— The technology presented in this study has potential for monitoring the hematocrit and coagulation parameters of patient samples using a small volume of whole blood, suggesting it may hold clinical utility as a point-of-care test.
Collapse
|
36
|
Skjeflo EW, Christiansen D, Fure H, Ludviksen JK, Woodruff TM, Espevik T, Nielsen EW, Brekke OL, Mollnes TE. Staphylococcus aureus-induced complement activation promotes tissue factor-mediated coagulation. J Thromb Haemost 2018; 16:905-918. [PMID: 29437288 DOI: 10.1111/jth.13979] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Indexed: 12/13/2022]
Abstract
Essentials Complement, Toll-like receptors and coagulation cross-talk in the process of thromboinflammation. This is explored in a unique human whole-blood model of S. aureus bacteremia. Coagulation is here shown as a downstream event of C5a-induced tissue factor (TF) production. Combined inhibition of C5 and CD14 efficiently attenuated TF and coagulation. SUMMARY Background There is extensive cross-talk between the complement system, the Toll-like receptors (TLRs), and hemostasis. Consumptive coagulopathy is a hallmark of sepsis, and is often mediated through increased tissue factor (TF) expression. Objectives To study the relative roles of complement, TLRs and TF in Staphylococcus aureus-induced coagulation. Methods Lepirudin-anticoagulated human whole blood was incubated with the three S. aureus strains Cowan, Wood, and Newman. C3 was inhibited with compstatin, C5 with eculizumab, C5a receptor 1 (C5aR1) and activated factor XII with peptide inhibitors, CD14, TLR2 and TF with neutralizing antibodies, and TLR4 with eritoran. Complement activation was measured by ELISA. Coagulation was measured according to prothrombin fragment 1 + 2 (PTF1 + 2 ) determined with ELISA, and TF mRNA, monocyte surface expression and functional activity were measured with quantitative PCR, flow cytometry, and ELISA, respectively. Results All three strains generated substantial and statistically significant amounts of C5a, terminal complement complex, PTF1 + 2 , and TF mRNA, and showed substantial TF surface expression on monocytes and TF functional activity. Inhibition of C5 cleavage most efficiently and significantly inhibited all six markers in strains Cowan and Wood, and five markers in Newman. The effect of complement inhibition was shown to be completely dependent on C5aR1. The C5 blocking effect was equally potentiated when combined with blocking of CD14 or TLR2, but not TLR4. TF blocking significantly reduced PTF1 + 2 levels to baseline levels. Conclusions S. aureus-induced coagulation in human whole blood was mainly attributable to C5a-induced mRNA upregulation, monocyte TF expression, and plasma TF activity, thus underscoring complement as a key player in S. aureus-induced coagulation.
Collapse
Affiliation(s)
- E W Skjeflo
- Research Laboratory, Nordland Hospital, Bodø, Norway
- Faculty of Health Sciences, K. G. Jebsen TREC, UiT - The Arctic University of Norway, Tromsø, Norway
| | | | - H Fure
- Research Laboratory, Nordland Hospital, Bodø, Norway
| | - J K Ludviksen
- Research Laboratory, Nordland Hospital, Bodø, Norway
| | - T M Woodruff
- School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia
| | - T Espevik
- Center of Molecular Inflammation Research, and Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - E W Nielsen
- Faculty of Health Sciences, K. G. Jebsen TREC, UiT - The Arctic University of Norway, Tromsø, Norway
- Department of Anesthesiology, Nordland Hospital, Bodø, Norway
- Faculty of Nursing and Health Sciences, Nord University, Bodø, Norway
| | - O L Brekke
- Research Laboratory, Nordland Hospital, Bodø, Norway
- Faculty of Health Sciences, K. G. Jebsen TREC, UiT - The Arctic University of Norway, Tromsø, Norway
| | - T E Mollnes
- Research Laboratory, Nordland Hospital, Bodø, Norway
- Faculty of Health Sciences, K. G. Jebsen TREC, UiT - The Arctic University of Norway, Tromsø, Norway
- Center of Molecular Inflammation Research, and Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Immunology, Oslo University Hospital and K. G. Jebsen IRC, University of Oslo, Oslo, Norway
| |
Collapse
|
37
|
Duan J, Liang S, Yu Y, Li Y, Wang L, Wu Z, Chen Y, Miller MR, Sun Z. Inflammation-coagulation response and thrombotic effects induced by silica nanoparticles in zebrafish embryos. Nanotoxicology 2018; 12:470-484. [PMID: 29658397 PMCID: PMC6157531 DOI: 10.1080/17435390.2018.1461267] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Nowadays, nanotechnology environmental health and safety (nanoEHS) is gaining attention. We previously found that silica nanoparticles (SiNPs) could induce vascular endothelial damage. However, the subsequent toxicologic response to SiNPs-induced endothelial damage was still largely unknown. In this study, we explored the inflammation–coagulation response and thrombotic effects of SiNPs in endothelial cells and zebrafish embryos. For in vitro study, swollen mitochondria and autophagosome were observed in ultrastructural analysis. The cytoskeleton organization was disrupted by SiNPs in vascular endothelial cells. The release of proinflammatory and procoagulant cytokines including IL-6, IL-8, MCP-1, PECAM-1, TF and vWF, were markedly elevated in a dose-dependent manner. For in vivo study, based on the NOAEL for dosimetry selection, and using two transgenic zebrafish, Tg(mpo:GFP) and Tg(fli-1:EGFP), SiNPs-induced neutrophil-mediated inflammation and impaired vascular endothelial cells. With the dosage higher than NOAEL, SiNPs significantly decreased blood flow and velocity, exhibiting a blood hypercoagulable state in zebrafish embryos. The thrombotic effect was assessed by o-dianisidine staining, showed that an increasing of erythrocyte aggregation occurred in SiNPs-treated zebrafish. Microarray analysis was used to screen the possible genes for inflammation–coagulation response to SiNPs in zebrafish, and the JAK1/TF signaling pathway was further verified by qRT-PCR and Western blot assays. For in-deepth study, il6st was knocked down with specific morpholinos. The whole-mount in situ hybridization and qRT-PCR analysis showed that the expression jak1 and f3b were attenuated in il6st knockdown groups. In summary, our data demonstrated that SiNPs could induce inflammation–coagulation response and thrombotic effects via JAK1/TF signaling pathway.
Collapse
Affiliation(s)
- Junchao Duan
- a Department of Toxicology and Sanitary Chemistry, School of Public Health , Capital Medical University , Beijing , P.R. China.,b Beijing Key Laboratory of Environmental Toxicology , Capital Medical University , Beijing , P.R. China
| | - Shuang Liang
- a Department of Toxicology and Sanitary Chemistry, School of Public Health , Capital Medical University , Beijing , P.R. China.,b Beijing Key Laboratory of Environmental Toxicology , Capital Medical University , Beijing , P.R. China
| | - Yang Yu
- a Department of Toxicology and Sanitary Chemistry, School of Public Health , Capital Medical University , Beijing , P.R. China.,b Beijing Key Laboratory of Environmental Toxicology , Capital Medical University , Beijing , P.R. China
| | - Yang Li
- a Department of Toxicology and Sanitary Chemistry, School of Public Health , Capital Medical University , Beijing , P.R. China.,b Beijing Key Laboratory of Environmental Toxicology , Capital Medical University , Beijing , P.R. China
| | - Lijing Wang
- a Department of Toxicology and Sanitary Chemistry, School of Public Health , Capital Medical University , Beijing , P.R. China.,b Beijing Key Laboratory of Environmental Toxicology , Capital Medical University , Beijing , P.R. China
| | - Zehao Wu
- a Department of Toxicology and Sanitary Chemistry, School of Public Health , Capital Medical University , Beijing , P.R. China.,b Beijing Key Laboratory of Environmental Toxicology , Capital Medical University , Beijing , P.R. China
| | - Yueyue Chen
- a Department of Toxicology and Sanitary Chemistry, School of Public Health , Capital Medical University , Beijing , P.R. China.,b Beijing Key Laboratory of Environmental Toxicology , Capital Medical University , Beijing , P.R. China
| | - Mark R Miller
- c BHF Centre for Cardiovascular Science, Queens Medical Research Institute, The University of Edinburgh , Edinburgh , UK
| | - Zhiwei Sun
- a Department of Toxicology and Sanitary Chemistry, School of Public Health , Capital Medical University , Beijing , P.R. China.,b Beijing Key Laboratory of Environmental Toxicology , Capital Medical University , Beijing , P.R. China
| |
Collapse
|
38
|
Yanase Y, Takahagi S, Hide M. Chronic spontaneous urticaria and the extrinsic coagulation system. Allergol Int 2018; 67:191-194. [PMID: 28993062 DOI: 10.1016/j.alit.2017.09.003] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Accepted: 09/08/2017] [Indexed: 01/05/2023] Open
Abstract
Chronic spontaneous urticaria (CSU) is a common skin disorder characterized by daily or almost daily recurring skin edema and flare with itch. Recently, the activation of the blood coagulation cascade has been suggested to be involved in CSU, but the trigger of the coagulation cascade remains unclear. In this article, we review recent understanding of the relationship between the pathogenesis of CSU and extrinsic coagulation reactions. In CSU, vascular endothelial cells and eosinophils may play a role as TF-expressing cells for activating the extrinsic coagulation pathway. Moreover, the expression of TF on endothelial cells is synergistically enhanced by the activation of Toll-like receptors and histamine H1 receptors. The activated coagulation factors may induce plasma extravasation followed by degranulation of skin mast cells and edema formation recognized as wheal in CSU. Molecules involved in this cascade could be a target for new and more effective treatments of urticaria.
Collapse
|
39
|
Coleman LG, Maile R, Jones SW, Cairns BA, Crews FT. HMGB1/IL-1β complexes in plasma microvesicles modulate immune responses to burn injury. PLoS One 2018; 13:e0195335. [PMID: 29601597 PMCID: PMC5877880 DOI: 10.1371/journal.pone.0195335] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 03/20/2018] [Indexed: 01/08/2023] Open
Abstract
Modulating immune responses to sepsis and trauma remain one of the most difficult challenges in modern medicine. Large burn injuries (LBI) are a severe form of trauma associated with sepsis, immune impairment, and mortality. Immune dysfunction after LBI is complex, involving both enhanced and impaired immune activation. The release of Damage-Associated Molecular Patterns (DAMPs), such as HMGB1, and cytokines (e.g. IL-1β) creates an environment of immune dysfunction often leading to end organ failure and death. Both HMGB1 and IL-1β have been found to play critical roles in sepsis and post-burn immune dysfunction. HMGB1 and IL-1β have been shown previously to form potent complexes in vitro. We recently identified the presence of HMGB1/IL-1β heterocomplexes in human tissue. We now find HMGB1/IL-1β complexes in human and mouse plasma, and identify a synergistic role of HMGB1/IL-1β complexes in post-burn immune dysfunction. In both humans and mice, we found that HMGB1 was enriched in plasma microvesicles (MVs) after LBI. HMGB1 was found form complexes with IL-1β. Using flow cytometry of mouse plasma MVs, we identified an increase in an HMGB1+/IL-1β+ MVs. Using co-IP, HMGB1 was found to bind the pro-form of IL-1β in mouse and human plasma. Pro-IL-1β, which is traditionally considered inactive, became active when complexed with HMGB1. Human THP-1 monocytes treated with HMGB1-pro-IL-1β complexes showed increased transcription of LBI associated cytokines IL-6 and IFNβ along with suppression of iNOS, mimicking findings associated with LBI. These findings identify that HMGB1/IL-1β complexes released after burn injuries can modulate immune responses, and microvesicles are identified as a novel reservoir for these immune mediators. These complexes might serve as novel immune targets for the treatment of systemic immune responses due to LBI or other causes of sepsis.
Collapse
Affiliation(s)
- Leon G Coleman
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America.,Bowles Center for Alcohol Studies, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | - Robert Maile
- North Carolina Jaycee Burn Center, Department of Surgery, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America.,Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | - Samuel W Jones
- North Carolina Jaycee Burn Center, Department of Surgery, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | - Bruce A Cairns
- North Carolina Jaycee Burn Center, Department of Surgery, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America.,Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | - Fulton T Crews
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America.,Bowles Center for Alcohol Studies, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| |
Collapse
|
40
|
Hernández C, Orbe J, Roncal C, Alvarez-Hernandez M, de Lizarrondo SM, Alves MT, Mata JG, Páramo JA. Tissue factor expressed by microparticles is associated with mortality but not with thrombosis in cancer patients. Thromb Haemost 2017; 110:598-608. [DOI: 10.1160/th13-02-0122] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2013] [Accepted: 06/07/2013] [Indexed: 01/08/2023]
Abstract
SummaryA prothrombotic state is one of the hallmarks of malignancy and a major contributor to morbidity and mortality in cancer patients. Tissue factor (TF) is often overexpressed in malignancy and is a prime candidate in predicting the hypercoagulable state. Moreover, increased number of TF-exposing microparticles (MPs) in cancer patients may contribute to venous thromboembolism (VTE). We have conducted a prospective cohort study to determine whether elevated TF antigen, TF activity and TF associated to MPs (MPs-TF) are predictive of VTE and mortality in cancer patients. The studied population consisted of 252 cancer patients and 36 healthy controls. TF antigen and activity and MPs-TF were determined by ELISA and chromogenic assays. During a median follow-up of 10 months, 40 thrombotic events were recorded in 34 patients (13.5%), and 73 patients (28.9%) died. TF antigen and activity were significantly higher in patients than in controls (p<0.01) mainly in patients with advanced stages, whereas no differences were observed for TF activity of isolated MPs. We did not find a statistically significant association of TF variables with the risk of VTE. Multivariate analysis adjusting for age, sex, type of cancer and other confounding variables showed that TF activity (p<0.01) and MPs-TF activity (p<0.05) were independently associated with mortality. In conclusion, while TF variables were not associated with future VTE in cancer patients, we found a strong association of TF and MPs-TF activity with mortality, thus suggesting they might be good prognostic markers in cancer patients.
Collapse
|
41
|
Hosseini E, Ghasemzadeh M. Intravascular leukocyte migration through platelet thrombi: directing leukocytes to sites of vascular injury. Thromb Haemost 2017; 113:1224-35. [DOI: 10.1160/th14-08-0662] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2014] [Accepted: 01/13/2015] [Indexed: 12/15/2022]
Abstract
SummaryLeukocytes recruitment to thrombi supports an intimate cellular interaction leading to the enhancement of pro-coagulant functions and pro-inflammatory responses at site of vascular injury. Recent observations of neutrophil extracellular traps (NETs) formation and its mutual reactions with platelet thrombi adds more clinical interest to the growing body of knowledge in the field of platelet-leukocyte crosstalk. However, having considered thrombus as a barrier between leukocytes and injured endothelium, the full inflammatory roles of these cells during thrombosis is still ill defined. The most recent observation of neutrophils migration into the thrombi is a phenomenon that highlights the inflammatory functions of leukocytes at the site of injury. It has been hypothesised that leukocytes migration might be associated with the conveyance of highly reactive pro-inflammatory and/or procoagulant mediators to sites of vascular injury. In addition, the evidence of neutrophils migration into arterial thrombi following traumatic and ischaemia-reperfusion injury highlights the already described role of these cells in atherosclerosis. Regardless of the mechanisms behind leukocyte migration, whether these migrated cells benefit normal homeostasis by their involvement in wound healing and vascular rebuilding or they increase unwilling inflammatory responses, could be of interest for future researches that provide new insight into biological importance of leukocyte recruitment to thrombi.
Collapse
|
42
|
The Role of Gap Junction-Mediated Endothelial Cell-Cell Interaction in the Crosstalk between Inflammation and Blood Coagulation. Int J Mol Sci 2017; 18:ijms18112254. [PMID: 29077057 PMCID: PMC5713224 DOI: 10.3390/ijms18112254] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 10/21/2017] [Accepted: 10/24/2017] [Indexed: 12/29/2022] Open
Abstract
Endothelial cells (ECs) play a pivotal role in the crosstalk between blood coagulation and inflammation. Endothelial cellular dysfunction underlies the development of vascular inflammatory diseases. Recent studies have revealed that aberrant gap junctions (GJs) and connexin (Cx) hemichannels participate in the progression of cardiovascular diseases such as cardiac infarction, hypertension and atherosclerosis. ECs can communicate with adjacent ECs, vascular smooth muscle cells, leukocytes and platelets via GJs and Cx channels. ECs dynamically regulate the expression of numerous Cxs, as well as GJ functionality, in the context of inflammation. Alterations to either result in various side effects across a wide range of vascular functions. Here, we review the roles of endothelial GJs and Cx channels in vascular inflammation, blood coagulation and leukocyte adhesion. In addition, we discuss the relevant molecular mechanisms that endothelial GJs and Cx channels regulate, both the endothelial functions and mechanical properties of ECs. A better understanding of these processes promises the possibility of pharmacological treatments for vascular pathogenesis.
Collapse
|
43
|
Ettelaie C, Collier MEW, Featherby S, Greenman J, Maraveyas A. Peptidyl-prolyl isomerase 1 (Pin1) preserves the phosphorylation state of tissue factor and prolongs its release within microvesicles. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2017; 1865:12-24. [PMID: 28962834 DOI: 10.1016/j.bbamcr.2017.09.016] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 08/07/2017] [Accepted: 09/24/2017] [Indexed: 01/23/2023]
Abstract
The exposure and release of TF is regulated by post-translational modifications of its cytoplasmic domain. Here, the potential of Pin1 to interact with the cytoplasmic domain of TF, and the outcome on TF function was examined. MDA-MB-231 and transfected-primary endothelial cells were incubated with either Pin1 deactivator Juglone, or its control Plumbagin, as well as transfected with Pin1-specific or control siRNA. TF release into microvesicles following activation, and also phosphorylation and ubiquitination states of cellular-TF were then assessed. Furthermore, the ability of Pin1 to bind wild-type and mutant forms of overexpressed TF-tGFP was investigated by co-immunoprecipitation. Additionally, the ability of recombinant or cellular Pin1 to bind to peptides of the C-terminus of TF, synthesised in different phosphorylation states was examined by binding assays and spectroscopically. Finally, the influence of recombinant Pin1 on the ubiquitination and dephosphorylation of the TF-peptides was examined. Pre-incubation of Pin1 with Juglone but not Plumbagin, reduced TF release as microvesicles and was also achievable following transfection with Pin1-siRNA. This was concurrent with early ubiquitination and dephosphorylation of cellular TF at Ser253. Pin1 co-immunoprecipitated with overexpressed wild-type TF-tGFP but not Ser258→Ala or Pro259→Ala substituted mutants. Pin1 did interact with Ser258-phosphorylated and double-phosphorylated TF-peptides, with the former having higher affinity. Finally, recombinant Pin1 was capable of interfering with the ubiquitination and dephosphorylation of TF-derived peptides. In conclusion, Pin1 is a fast-acting enzyme which may be utilised by cells to protect the phosphorylation state of TF in activated cells prolonging TF activity and release, and therefore ensuring adequate haemostasis.
Collapse
Affiliation(s)
- Camille Ettelaie
- Biomedical Section, Department of Biological Sciences, University of Hull, Cottingham Road, Hull HU6 7RX, UK.
| | - Mary E W Collier
- Department of Cardiovascular Sciences, University of Leicester, Clinical Sciences Wing, Glenfield General Hospital, Leicester LE3 9QP, UK
| | - Sophie Featherby
- Biomedical Section, Department of Biological Sciences, University of Hull, Cottingham Road, Hull HU6 7RX, UK
| | - John Greenman
- Biomedical Section, Department of Biological Sciences, University of Hull, Cottingham Road, Hull HU6 7RX, UK
| | - Anthony Maraveyas
- Division of Cancer, Hull York Medical School University of Hull, Cottingham Road, Hull HU6 7RX, UK
| |
Collapse
|
44
|
Kral‐Pointner JB, Schrottmaier WC, Horvath V, Datler H, Hell L, Ay C, Niederreiter B, Jilma B, Schmid JA, Assinger A, Mackman N, Knapp S, Schabbauer G. Myeloid but not epithelial tissue factor exerts protective anti-inflammatory effects in acid aspiration-induced acute lung injury. J Thromb Haemost 2017; 15:1625-1639. [PMID: 28509332 PMCID: PMC5575489 DOI: 10.1111/jth.13737] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Indexed: 01/11/2023]
Abstract
Essentials Tissue factor (TF) represents a central link between hemostasis and inflammation. We studied the roles of myeloid and airway epithelial TF in acid-caused acute lung injury (ALI). TF on myeloid cells displays a non-coagulatory role regulating the inflammatory response in ALI. Airway epithelial TF contributes to hemostatic functions, but is dispensable in ALI pathogenesis. SUMMARY Introduction Acute lung injury (ALI) is a life-threatening condition characterized by damaged alveolar-capillary structures and activation of inflammatory and hemostatic processes. Tissue factor (TF) represents a crucial link between inflammation and coagulation, as inflammatory mediators induce myeloid TF expression, and TF initiates extrinsic coagulation. Objective As pulmonary inflammation stimulates TF expression and TF modulates immune responses, we aimed to elucidate its impact on ALI. In particular, we wanted to distinguish the contributions of TF expressed on airway epithelial cells and TF expressed on myeloid cells. Methods Mice with different cell type-specific TF deficiency and wild-type littermates were intratracheally treated with hydrochloric acid, and leukocyte recruitment, cytokine levels, thrombin-antithrombin (TAT) complexes and pulmonary protein-rich infiltrates were analyzed. Results Our data demonstrate that a lack of epithelial TF did not influence acute responses, as bronchoalveolar neutrophil accumulation 8 h after ALI induction was unaltered. However, it led to mild, prolonged inflammation, as pulmonary leukocyte and erythrocyte numbers were still increased after 24 h, whereas those in wild-type mice had returned to basal levels. In contrast, myeloid TF was primarily involved in regulating the acute phase of ALI without affecting local coagulation, as indicated by increased bronchoalveolar neutrophil infiltration, pulmonary interleukin-6 levels, and edema formation, but equal TAT complex formation, 8 h after ALI induction. This augmented inflammatory response associated with myeloid TF deficiency was confirmed in vitro, as lipopolysaccharide-stimulated TF-deficient alveolar macrophages released increased levels of chemokine (C-X-C motif) ligand 1 and tumor necrosis factor-α as compared with wild-type macrophages. Conclusion We conclude that myeloid TF dampens inflammation in acid-induced ALI.
Collapse
Affiliation(s)
- J. B. Kral‐Pointner
- Institute for PhysiologyCenter for Physiology and PharmacologyMedical University of ViennaViennaAustria
| | - W. C. Schrottmaier
- Institute for PhysiologyCenter for Physiology and PharmacologyMedical University of ViennaViennaAustria
| | - V. Horvath
- Institute for PhysiologyCenter for Physiology and PharmacologyMedical University of ViennaViennaAustria
| | - H. Datler
- Institute for PhysiologyCenter for Physiology and PharmacologyMedical University of ViennaViennaAustria
| | - L. Hell
- Clinical Division of Hematology and HemostaseologyDepartment of Medicine IMedical University of ViennaViennaAustria
| | - C. Ay
- Clinical Division of Hematology and HemostaseologyDepartment of Medicine IMedical University of ViennaViennaAustria
| | - B. Niederreiter
- Division of RheumatologyInternal Medicine IIIMedical University of ViennaViennaAustria
| | - B. Jilma
- Departments of Clinical PharmacologyDepartment of Medicine IMedical University of ViennaViennaAustria
| | - J. A. Schmid
- Department for Vascular Biology and Thrombosis ResearchCenter for Physiology and PharmacologyMedical University of ViennaViennaAustria
| | - A. Assinger
- Institute for PhysiologyCenter for Physiology and PharmacologyMedical University of ViennaViennaAustria
| | - N. Mackman
- Division of Hematology/Oncology, Thrombosis and Hemostasis ProgramUNC McAllister Heart InstituteUniversity of North CarolinaChapel HillNCUSA
| | - S. Knapp
- CEMMResearch Center for Molecular Medicine of the Austrian Academy of SciencesViennaAustria
- Laboratory of Infection BiologyDepartment of Medicine IMedical University of ViennaViennaAustria
| | - G. Schabbauer
- Institute for PhysiologyCenter for Physiology and PharmacologyMedical University of ViennaViennaAustria
| |
Collapse
|
45
|
Hell L, Wisgrill L, Ay C, Spittler A, Schwameis M, Jilma B, Pabinger I, Altevogt P, Thaler J. Procoagulant extracellular vesicles in amniotic fluid. Transl Res 2017; 184:12-20.e1. [PMID: 28236427 PMCID: PMC6544525 DOI: 10.1016/j.trsl.2017.01.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2016] [Revised: 01/06/2017] [Accepted: 01/30/2017] [Indexed: 10/20/2022]
Abstract
Embolization of amniotic fluid (AF) into the blood circulation leads to disseminated intravascular coagulation (DIC). Procoagulant phosphatidylserine (PS)- and tissue factor (TF)-exposing extracellular vesicles (EVs) might play an important role in AF embolism-induced DIC. It was the aim of the present study to perform analyses of the procoagulant properties of AF with a panel of functional coagulation assays and flow cytometry. We applied a prothrombinase assay (that quantifies PS exposure on EVs), an EV-associated TF activity assay, a fibrin generation assay, a thrombin generation assay, a whole blood clotting model, and flow cytometry in AF and control plasma. We found that PS exposure on EVs was 21-fold increased in AF compared with plasma. Also, EV-associated TF activity was highly increased in AF compared with plasma. AF-derived EVs activated the blood coagulation cascade via PS and TF in the fibrin and thrombin generation assays. In a whole blood clotting model, AF-derived EVs significantly shortened the clotting time from 734 ± 139 seconds in the presence to 232 ± 139 seconds in the absence of an anti-TF antibody. The contact activation pathway via factor XII (FXII) was not affected. Applying flow cytometry, a subpopulation of PS+ and TF+ EVs was identified in AF but not in control plasma. In conclusion, we investigated the effect of AF on blood coagulation and found that PS+ and TF+ EVs determine their procoagulant potential. Taken together, our data further delineate the pathomechanisms underlying AF-induced coagulopathy.
Collapse
Affiliation(s)
- Lena Hell
- Clinical Division of Haematology and Haemostaseology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Lukas Wisgrill
- Clinical Division of Neonatology, Paediatric Intensive Care & Neuropaediatrics, Department of Paediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| | - Cihan Ay
- Clinical Division of Haematology and Haemostaseology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Andreas Spittler
- Department of Surgery, Medical University of Vienna, Vienna, Austria
| | - Michael Schwameis
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Bernd Jilma
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Ingrid Pabinger
- Clinical Division of Haematology and Haemostaseology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Peter Altevogt
- Tumor Immunology Programme, German Cancer Research Center, Heidelberg, Germany
| | - Johannes Thaler
- Clinical Division of Haematology and Haemostaseology, Department of Medicine I, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
46
|
Zhang Y, Li L, Zhao Y, Han H, Hu Y, Liang D, Yu B, Kou J. The Myosin II Inhibitor, Blebbistatin, Ameliorates FeCl3-induced Arterial Thrombosis via the GSK3β-NF-κB Pathway. Int J Biol Sci 2017; 13:630-639. [PMID: 28539835 PMCID: PMC5441179 DOI: 10.7150/ijbs.18485] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Accepted: 03/07/2017] [Indexed: 12/13/2022] Open
Abstract
Arterial thrombosis and its related diseases are major healthcare problems worldwide. Blebbistatin is an inhibitor of myosin II, which plays an important role in thrombosis. The aim of our study is to explore the effect and potential mechanism of blebbistatin on arterial thrombosis. A ferric chloride (FeCl3) solution at a concentration of 5% was used to induce carotid artery thrombosis in mice. Immunohistochemistry and immunofluorescence staining were used to detect the expression or activation of non-muscle myosin heavy chain IIA (NMMHC IIA), tissue factor (TF), GSK3β and NF-κB. Blebbistatin (1 mg/kg, i.p.) significantly reduced carotid artery thrombosis induced by FeCl3 solution in mice, inhibited NMMHC IIA expression and impeded TF expression via the GSK3β-NF-κB signalling pathway in mouse arterial vascular tissues. The present study demonstrates that blebbistatin may impede TF expression partly via the Akt/GSK3β-NF-κB signalling pathways in the endothelium in a FeCl3 model, shedding new insights into the pathogenesis of arterial thrombosis and providing new clues for the development of antithrombotic drugs.
Collapse
Affiliation(s)
- Yuanyuan Zhang
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Complex Prescription of TCM, China Pharmaceutical University, 639 Longmian Road, Nanjing 211198, China
| | - Long Li
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Complex Prescription of TCM, China Pharmaceutical University, 639 Longmian Road, Nanjing 211198, China
| | - Yazheng Zhao
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Complex Prescription of TCM, China Pharmaceutical University, 639 Longmian Road, Nanjing 211198, China
| | - Han Han
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Complex Prescription of TCM, China Pharmaceutical University, 639 Longmian Road, Nanjing 211198, China
| | - Yang Hu
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Complex Prescription of TCM, China Pharmaceutical University, 639 Longmian Road, Nanjing 211198, China
| | - Di Liang
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Complex Prescription of TCM, China Pharmaceutical University, 639 Longmian Road, Nanjing 211198, China
| | - Boyang Yu
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Complex Prescription of TCM, China Pharmaceutical University, 639 Longmian Road, Nanjing 211198, China
| | - Junping Kou
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Complex Prescription of TCM, China Pharmaceutical University, 639 Longmian Road, Nanjing 211198, China
| |
Collapse
|
47
|
Scarlatescu E, Tomescu D, Arama SS. Anticoagulant Therapy in Sepsis. The Importance of Timing. ACTA ACUST UNITED AC 2017; 3:63-69. [PMID: 29967873 PMCID: PMC5769917 DOI: 10.1515/jccm-2017-0011] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Accepted: 03/24/2017] [Indexed: 11/25/2022]
Abstract
Sepsis associated coagulopathy is due to the inflammation-induced activation of coagulation pathways concomitant with dysfunction of anticoagulant and fibrinolytic systems, leading to different degrees of haemostasis dysregulation. This response is initially beneficial, contributing to antimicrobial defence, but when control is lost coagulation activation leads to widespread microvascular thrombosis and subsequent organ failure. Large clinical trials of sepsis-related anticoagulant therapies failed to show survival benefits, but posthoc analysis of databases and several smaller studies showed beneficial effects of anticoagulants in subgroups of patients with early sepsis-induced disseminated intravascular coagulation. A reasonable explanation could be the difference in timing of anticoagulant therapy and patient heterogeneity associated with large trials. Proper selection of patients and adequate timing are required for treatment to be successful. The time when coagulation activation changes from advantageous to detrimental represents the right moment for the administration of coagulation-targeted therapy. In this way, the defence function of the haemostatic system is preserved, and the harmful effects of overwhelming coagulation activation are avoided.
Collapse
Affiliation(s)
- Ecaterina Scarlatescu
- Department of Anesthesiology and Intensive Care III, Fundeni Clinical Institute, Bucharest, Romania
| | - Dana Tomescu
- Department of Anesthesiology and Intensive Care III, Fundeni Clinical Institute, Bucharest, Romania.,University of Medicine and Pharmacy, "Carol Davila", Bucharest, Romania
| | | |
Collapse
|
48
|
Recent Progress in Research on the Pathogenesis of Pulmonary Thromboembolism: An Old Story with New Perspectives. BIOMED RESEARCH INTERNATIONAL 2017; 2017:6516791. [PMID: 28484717 PMCID: PMC5397627 DOI: 10.1155/2017/6516791] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Revised: 02/26/2017] [Accepted: 03/27/2017] [Indexed: 12/25/2022]
Abstract
Pulmonary thromboembolism (PTE) is part of a larger clinicopathological entity, venous thromboembolism. It is also a complex, multifactorial disorder divided into four major disease processes including venous thrombosis, thrombus in transit, acute pulmonary embolism, and pulmonary circulation reconstruction. Even when treated, some patients develop chronic thromboembolic pulmonary hypertension. PTE is also a common fatal type of pulmonary vascular disease worldwide, but earlier studies primarily focused on the pathological changes in the blood component of the disease. With contemporary advances in molecular and cellular biology, people are becoming increasingly aware of coagulation pathways, the function of vascular smooth muscle cells, microparticles, and the inflammatory pathways that play key roles in PTE. Combined hypoxia and immune research has revealed that PTE should be regarded as a class of complex diseases caused by multiple factors involving the vascular microenvironment and vascular cell dysfunction.
Collapse
|
49
|
Detection of tissue factor-positive extracellular vesicles by laser scanning confocal microscopy. Thromb Res 2016; 150:65-72. [PMID: 28043041 DOI: 10.1016/j.thromres.2016.12.021] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Revised: 12/21/2016] [Accepted: 12/22/2016] [Indexed: 01/15/2023]
Abstract
INTRODUCTION Increased levels of tissue factor-positive extracellular vesicles (TF+EVs) have been detected in the plasma of patients with various diseases, including cancer and endotoxemia. Levels of TF+EVs in plasma samples can be measured by antigen and activity assays. The aim of the present study was to visualize TF+EVs by laser scanning confocal microscopy (LSCM). METHODS EVs were isolated from the supernatant of two cultured human pancreatic cancer cell lines (Panc-1 and BxPc-3), from untreated or lipopolysaccharide (LPS) treated whole blood, and from plasma of pancreatic cancer patients. EV-TF activity was determined using an in-house assay. The EVs were labeled with 5(6)-carboxyfluorescein diacetate N-succinimidyl ester, which is converted to the impermeant green fluorescent molecule carboxyfluorescein inside the EVs. EVs were either captured using annexin V and detected using a fluorescent-labeled anti-TF antibody, or captured using an anti-TF antibody and detected using fluorescent-labeled annexin V. EVs were visualized by LSCM. RESULTS TF+EVs were easily detected from high TF-expressing BxPc-3 cells using annexin V capture, whereas the addition of tyramide amplification was required to detect TF+EVs from low TF-expressing Panc-1 cells. Visualization of TF+EVs in plasma from LPS treated whole human blood and in plasma from pancreatic cancer patients required either capture with annexin V and detection with a fluorescent-labeled anti-TF antibody with tyramide signal amplification, or capture with an anti-TF antibody and detection with a fluorescent-labeled annexin V. CONCLUSION LSCM enables visualization of TF+EVs in the supernatant from cultured cells and in clinical samples.
Collapse
|
50
|
Identification of Potential Biomarkers for Rhegmatogenous Retinal Detachment Associated with Choroidal Detachment by Vitreous iTRAQ-Based Proteomic Profiling. Int J Mol Sci 2016; 17:ijms17122052. [PMID: 27941623 PMCID: PMC5187852 DOI: 10.3390/ijms17122052] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Revised: 11/24/2016] [Accepted: 11/30/2016] [Indexed: 12/16/2022] Open
Abstract
Rhegmatogenous retinal detachment associated with choroidal detachment (RRDCD) is a complicated and serious type of rhegmatogenous retinal detachment (RRD). In this study, we identified differentially expressed proteins in the vitreous humors of RRDCD and RRD using isobaric tags for relative and absolute quantitation (iTRAQ) combined with nano-liquid chromatography-electrospray ion trap-mass spectrometry-mass spectrometry (nano-LC-ESI-MS/MS) and bioinformatic analysis. Our result shows that 103 differentially expressed proteins, including 54 up-regulated and 49 down-regulated proteins were identified in RRDCD. Gene ontology (GO) analysis suggested that most of the differentially expressed proteins were extracellular.The Kyoto encyclopedia of genes and genomes (KEGG) pathway analysis suggested that proteins related to complement and coagulation cascades were significantly enriched. iTRAQ-based proteomic profiling reveals that complement and coagulation cascades and inflammation may play important roles in the pathogenesis of RRDCD. This study may provide novel insights into the pathogenesis of RRDCD and offer potential opportunities for the diagnosis and treatment of RRDCD.
Collapse
|