1
|
Liu CH, Rethi L, Weng PW, Trung Nguyen H, Chuang AEY. Cutting-edge advances in nano/biomedicine: A review on transforming thrombolytic therapy. Biochem Pharmacol 2024; 229:116523. [PMID: 39251141 DOI: 10.1016/j.bcp.2024.116523] [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: 05/06/2024] [Revised: 09/03/2024] [Accepted: 09/05/2024] [Indexed: 09/11/2024]
Abstract
Thrombotic blockages within blood vessels give rise to critical cardiovascular disorders, including ischemic stroke, venous thromboembolism, and myocardial infarction. The current approach to the therapy of thrombolysis involves administering Plasminogen Activators (PA), but it is hindered by fast drug elimination, narrow treatment window, and the potential for bleeding complications. Leveraging nanomedicine to encapsulate and deliver PA offers a solution by improving the efficacy of therapy, safeguarding the medicine from proteinase biodegradation, and reducing unwanted effects in in vivo trials. In this review, we delve into the underlying venous as well as arterial thrombus pathophysiology and provide an overview of clinically approved PA used to address acute thrombotic conditions. We explore the existing challenges and potential directions within recent pivotal research on a variety of targeted nanocarriers, such as lipid, polymeric, inorganic, and biological carriers, designed for precise delivery of PA to specific sites. We also discuss the promising role of microbubbles and ultrasound-assisted Sono thrombolysis, which have exhibited enhanced thrombolysis in clinical studies. Furthermore, our review delves into approaches for the strategic development of nano-based carriers tailored for targeting thrombolytic action and efficient encapsulation of PA, considering the intricate interaction in biology systems as well as nanomaterials. In conclusion, the field of nanomedicine offers a valuable method for the exact and effective therapy of severe thrombus conditions, presenting a pathway toward improved patient outcomes and reduced complications.
Collapse
Affiliation(s)
- Chia-Hung Liu
- Department of Urology, School of Medicine, College of Medicine, Taipei Medical University, 250 Wu-Hsing Street, Taipei 11031, Taiwan; TMU Research Center of Urology and Kidney, Taipei Medical University, 250 Wu-Hsing Street, Taipei 11031, Taiwan; Department of Urology, Shuang Ho Hospital, Taipei Medical University, 291 Zhongzheng Road, Zhonghe District, New Taipei City 23561, Taiwan
| | - Lekshmi Rethi
- International Ph.D. Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan; Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan
| | - Pei-Wei Weng
- Department of Orthopedics, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan; Department of Orthopedics, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Hieu Trung Nguyen
- Department of Orthopedics and Trauma, Faculty of Medicine, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, 700000, Viet Nam
| | - Andrew E-Y Chuang
- International Ph.D. Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan; Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan; Cell Physiology and Molecular Image Research Center, Taipei Medical University-Wan Fang Hospital, Taipei 11696, Taiwan.
| |
Collapse
|
2
|
Schulman S, Makatsariya A, Khizroeva J, Bitsadze V, Kapanadze D. The Basic Principles of Pathophysiology of Venous Thrombosis. Int J Mol Sci 2024; 25:11447. [PMID: 39519000 PMCID: PMC11547114 DOI: 10.3390/ijms252111447] [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: 09/07/2024] [Revised: 10/19/2024] [Accepted: 10/20/2024] [Indexed: 11/16/2024] Open
Abstract
The past few decades have brought tremendous insight into the molecular and pathophysiological mechanisms responsible for thrombus generation. For a clinician, it is usually sufficient to explain the incident of deep vein thrombosis (DVT) with provoking factors such as trauma with vascular injury, immobilization, hormonal factors, or inherited or acquired coagulation defects. About half of DVTs are, however, lacking such triggers and are called unprovoked. Venous stasis and hypoxia at the valve sinus level may start a chain of reactions. The concept of immunothrombosis has added a new dimension to the old etiological triad of venous stasis, vessel wall injury, and changes in blood components. This is particularly important in COVID-19, where hyperinflammation, cytokines, and neutrophil extracellular traps are associated with the formation of microthrombi in the lungs. To better understand the mechanisms behind DVT and reach beyond the above-mentioned simplifications, animal models and clinical epidemiological studies have brought insight into the complex interplay between leukocytes, platelets, endothelium, cytokines, complements, and coagulation factors and inhibitors. These pathways and the interplay will be reviewed here, as well as the roles of cancer, anticancer drugs, and congenital thrombophilic defects on the molecular level in hypercoagulability and venous thromboembolism.
Collapse
Affiliation(s)
- Sam Schulman
- Department of Medicine, Thrombosis and Atherosclerosis Research Institute, McMaster University, 1280 Main St W, Hamilton, ON L8S 4L8, Canada
- Department of Obstetrics, Gynecology and Perinatal Medicine, The I.M. Sechenov First Moscow State Medical University (Sechenov University), Trubetskaya Str 8-2, 119435 Moscow, Russia; (A.M.); (J.K.); (V.B.)
| | - Alexander Makatsariya
- Department of Obstetrics, Gynecology and Perinatal Medicine, The I.M. Sechenov First Moscow State Medical University (Sechenov University), Trubetskaya Str 8-2, 119435 Moscow, Russia; (A.M.); (J.K.); (V.B.)
| | - Jamilya Khizroeva
- Department of Obstetrics, Gynecology and Perinatal Medicine, The I.M. Sechenov First Moscow State Medical University (Sechenov University), Trubetskaya Str 8-2, 119435 Moscow, Russia; (A.M.); (J.K.); (V.B.)
| | - Victoria Bitsadze
- Department of Obstetrics, Gynecology and Perinatal Medicine, The I.M. Sechenov First Moscow State Medical University (Sechenov University), Trubetskaya Str 8-2, 119435 Moscow, Russia; (A.M.); (J.K.); (V.B.)
| | - Daredzhan Kapanadze
- Center of Pathology of Pregnancy and Hemostasis «Medlabi», Tbilisi 340112, Georgia;
| |
Collapse
|
3
|
Rayes J, Brill A. Hot under the clot: venous thrombogenesis is an inflammatory process. Blood 2024; 144:477-489. [PMID: 38728383 DOI: 10.1182/blood.2023022522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 04/29/2024] [Accepted: 05/01/2024] [Indexed: 05/12/2024] Open
Abstract
ABSTRACT Venous thrombosis (VT) is a serious medical condition in which a blood clot forms in deep veins, often causing limb swelling and pain. Current antithrombotic therapies carry significant bleeding risks resulting from targeting essential coagulation factors. Recent advances in this field have revealed that the cross talk between the innate immune system and coagulation cascade is a key driver of VT pathogenesis, offering new opportunities for potential therapeutic interventions without inducing bleeding complications. This review summarizes and discusses recent evidence from preclinical models on the role of inflammation in VT development. We highlight the major mechanisms by which endothelial cell activation, Weibel-Palade body release, hypoxia, reactive oxygen species, inflammasome, neutrophil extracellular traps, and other immune factors cooperate to initiate and propagate VT. We also review emerging clinical data describing anti-inflammatory approaches as adjuncts to anticoagulation in VT treatment. Finally, we identify key knowledge gaps and future directions that could maximize the benefit of anti-inflammatory therapies in VT. Identifying and targeting the inflammatory factors driving VT, either at the endothelial cell level or within the clot, may pave the way for new therapeutic possibilities for improving VT treatment and reducing thromboembolic complications without increasing bleeding risk.
Collapse
Affiliation(s)
- Julie Rayes
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Alexander Brill
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
4
|
Watson C, Saaid H, Vedula V, Cardenas JC, Henke PK, Nicoud F, Xu XY, Hunt BJ, Manning KB. Venous Thromboembolism: Review of Clinical Challenges, Biology, Assessment, Treatment, and Modeling. Ann Biomed Eng 2024; 52:467-486. [PMID: 37914979 DOI: 10.1007/s10439-023-03390-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 10/17/2023] [Indexed: 11/03/2023]
Abstract
Venous thromboembolism (VTE) is a massive clinical challenge, annually affecting millions of patients globally. VTE is a particularly consequential pathology, as incidence is correlated with extremely common risk factors, and a large cohort of patients experience recurrent VTE after initial intervention. Altered hemodynamics, hypercoagulability, and damaged vascular tissue cause deep-vein thrombosis and pulmonary embolism, the two permutations of VTE. Venous valves have been identified as likely locations for initial blood clot formation, but the exact pathway by which thrombosis occurs in this environment is not entirely clear. Several risk factors are known to increase the likelihood of VTE, particularly those that increase inflammation and coagulability, increase venous resistance, and damage the endothelial lining. While these risk factors are useful as predictive tools, VTE diagnosis prior to presentation of outward symptoms is difficult, chiefly due to challenges in successfully imaging deep-vein thrombi. Clinically, VTE can be managed by anticoagulants or mechanical intervention. Recently, direct oral anticoagulants and catheter-directed thrombolysis have emerged as leading tools in resolution of venous thrombosis. While a satisfactory VTE model has yet to be developed, recent strides have been made in advancing in silico models of venous hemodynamics, hemorheology, fluid-structure interaction, and clot growth. These models are often guided by imaging-informed boundary conditions or inspired by benchtop animal models. These gaps in knowledge are critical targets to address necessary improvements in prediction and diagnosis, clinical management, and VTE experimental and computational models.
Collapse
Affiliation(s)
- Connor Watson
- Department of Biomedical Engineering, The Pennsylvania State University, 122 Chemical and Biomedical Engineering Building, University Park, PA, 16802-4400, USA
| | - Hicham Saaid
- Department of Biomedical Engineering, The Pennsylvania State University, 122 Chemical and Biomedical Engineering Building, University Park, PA, 16802-4400, USA
| | - Vijay Vedula
- Department of Mechanical Engineering, Fu Foundation School of Engineering and Applied Science, Columbia University, New York, NY, USA
| | - Jessica C Cardenas
- Department of Surgery and the Center for Translational Injury Research, McGovern Medical School, University of Texas Health Science Center, Houston, TX, USA
| | - Peter K Henke
- Section of Vascular Surgery, Department of Surgery, University of Michigan Health System, Ann Arbor, MI, USA
| | - Franck Nicoud
- CNRS, IMAG, Université de Montpellier, Montpellier, France
- Institut Universitaire de France, Paris, France
| | - Xiao Yun Xu
- Department of Chemical Engineering, Imperial College London, London, UK
| | - Beverley J Hunt
- Department of Thrombosis and Haemostasis, King's College, London, UK
- Thrombosis and Haemophilia Centre, Guy's & St Thomas' NHS Trust, London, UK
| | - Keefe B Manning
- Department of Biomedical Engineering, The Pennsylvania State University, 122 Chemical and Biomedical Engineering Building, University Park, PA, 16802-4400, USA.
- Department of Surgery, Penn State Hershey Medical Center, Hershey, PA, USA.
| |
Collapse
|
5
|
Jakobsen SS, Frøkjaer JB, Fisker RV, Kristensen SR, Thorlacius-Ussing O, Larsen AC. Monocyte recruitment in venous pulmonary embolism at time of cancer diagnosis in upper gastrointestinal cancer patients. J Thromb Thrombolysis 2024; 57:11-20. [PMID: 37792208 PMCID: PMC10830795 DOI: 10.1007/s11239-023-02897-5] [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] [Accepted: 09/12/2023] [Indexed: 10/05/2023]
Abstract
Upper gastrointestinal cancer is frequently complicated by venous thromboembolisms (VTE), especially pulmonary embolisms (PE) increase the mortality rate. Monocytes are a part of the innate immune system and up-regulation may indicate an ongoing inflammatory response or infectious disease and has lately been associated with a moderate risk of suffering from VTE. This prospectively study aims to compare the incidence of pulmonary embolism with markers of coagulation and compare it to the absolute monocyte count. A consecutive cohort of 250 patients with biopsy proven upper gastrointestinal cancer (i.e. pancreas, biliary tract, esophagus and gastric cancer) where included at the time of cancer diagnosis and before treatment. All patients underwent bilateral compression ultrasonography for detection of deep vein thrombosis (DVT). Of these 143 had an additionally pulmonary angiografi (CTPA) with the staging computer tomography. 13 of 250 patients (5.2%) had a DVT and 11 of 143 (7.7%) had CTPA proven PE. PE was significantly more common among patients with elevated D-dimer (OR 11.62, 95%CI: 1.13-119, P = 0.039) and elevated absolute monocyte count (OR 7.59, 95%CI: 1.37-41.98, P = 0.020). Only patients with pancreatic cancer had a significantly higher risk of DVT (OR 11.03, 95%CI: 1.25-97.43, P = 0.031). The sensitivity of absolute monocyte count was 63.6 (95%CI: 30.8-89.1) and specificity 80.3 (95%CI: 72.5-86.7), with a negative predictive value of 96.4 (95%CI: 91-99) in PE. An increased absolute monocyte count was detected in patients suffering from PE but not DVT, suggesting a possible interaction with the innate immune system.
Collapse
Affiliation(s)
- Sarah S Jakobsen
- Department of Gastrointestinal Surgery, Aalborg University Hospital, Hobrovej 18-22, 9000, Aalborg, Denmark
- Clinical Cancer Research Center, Aalborg University Hospital, 9000, Aalborg, Denmark
| | - Jens B Frøkjaer
- Department of Radiology, Aalborg University Hospital, 9000, Aalborg, Denmark
- Clinical Cancer Research Center, Aalborg University Hospital, 9000, Aalborg, Denmark
- Department of Clinical Medicine, Aalborg University, 9000, Aalborg, Denmark
| | - Rune V Fisker
- Department of Radiology, Aalborg University Hospital, 9000, Aalborg, Denmark
- Department of Nuclear Medicine, Aalborg University Hospital, 9000, Aalborg, Denmark
- Clinical Cancer Research Center, Aalborg University Hospital, 9000, Aalborg, Denmark
| | - Søren R Kristensen
- Department of Biochemistry, Aalborg University Hospital, 9000, Aalborg, Denmark
- Cardiovascular Research Center, Aalborg University, 9000, Aalborg, Denmark
| | - Ole Thorlacius-Ussing
- Department of Gastrointestinal Surgery, Aalborg University Hospital, Hobrovej 18-22, 9000, Aalborg, Denmark
- Clinical Cancer Research Center, Aalborg University Hospital, 9000, Aalborg, Denmark
- Department of Clinical Medicine, Aalborg University, 9000, Aalborg, Denmark
| | - Anders C Larsen
- Department of Gastrointestinal Surgery, Aalborg University Hospital, Hobrovej 18-22, 9000, Aalborg, Denmark.
- Clinical Cancer Research Center, Aalborg University Hospital, 9000, Aalborg, Denmark.
- Department of Clinical Medicine, Aalborg University, 9000, Aalborg, Denmark.
| |
Collapse
|
6
|
Hoofnagle MH, Hess A, Nalugo M, Ghosh S, Hughes SW, Fuchs A, Welsh JD, Kahn ML, Bochicchio GV, Randolph GJ, Leonard JM, Turnbull IR. Defects in vein valve PROX1/FOXC2 antithrombotic pathway in endothelial cells drive the hypercoagulable state induced by trauma and critical illness. J Trauma Acute Care Surg 2023; 95:197-204. [PMID: 37072887 PMCID: PMC10524206 DOI: 10.1097/ta.0000000000003945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/20/2023]
Abstract
OBJECTIVES Deep venous thrombosis (DVT) causes significant morbidity and mortality after trauma. Recently, we have shown that blood flow patterns at vein valves induce oscillatory stress genes, which maintain an anticoagulant endothelial phenotype that inhibits spontaneous clotting at vein valves and sinuses, is lost in the presence of DVT in human pathological samples, and is dependent on expression of the transcription factor FOXC2. We describe an assay, modifying our mouse multiple injury system, which shows evidence of clinically relevant microthrombosis and hypercoagulability applicable to the study of spontaneous DVT in trauma without requiring direct vascular injury or ligation. Finally, we investigated whether these model findings are relevant to a human model of critical illness by examining gene expression changes by quantitative polymerase chain reaction and immunofluorescence in veins collected from critically ill. METHODS C57/Bl6 mice were subjected to a modified mouse multiple injury model with liver crush injury, crush and pseudofracture of a single lower extremity, and 15% total blood volume hemorrhage. Serum was assayed for d-dimer at 2, 6, 24, and 48 hours after injury by enzyme-linked immunosorbent assay. For the thrombin clotting assay, veins of the leg were exposed, 100 μL of 1 mM rhodamine (6 g) was injected retro-orbitally, and 450 μg/mL thrombin was then applied to the surface of the vein with examination of real-time clot formation via in vivo immunofluorescence microscopy. Images were then examined for percentage area of clot coverage of visible mouse saphenous and common femoral vein. Vein valve specific knockout of FOXC2 was induced with tamoxifen treatment in PROX1 Ert2Cre FOXC2 fl/fl mice as previously described. Animals were then subjected to a modified mouse multiple injury model with liver crush injury, crush and pseudofracture of a single lower extremity, and 15% total blood volume hemorrhage. Twenty-four hours after injury, we examined the valve phenotype in naive versus multiple injury animals, with and without loss of the FOXC2 gene from the vein valve (FOXC2 del ) via the thrombin assay. Images were then examined for proximity of clot formation to the valve present at the junction of the mouse saphenous, tibial, and superficial femoral vein and presence of spontaneous microthrombi present in the veins before exposure to thrombin. Human vein samples were obtained from excess tissue preserved after harvest for elective cardiac surgery and from organ donors after organ procurement. Sections were submitted for paraffin embedding and then assayed by immunofluorescence for PROX1, FOXC2, thrombomodulin, endothelial protein C receptor, and von Willebrand's factor. All animal studies were reviewed and approved by the Institutional Animal Care and Use Committee, and all human studies reviewed and approved by the institutional review board. RESULTS After mouse multiple injuries, enzyme-linked immunosorbent assay for d-dimer showed evidence of products of fibrin breakdown consistent with formation of clot related to injury, fibrinolysis, and/or microthrombosis. The thrombin clotting assay demonstrated higher percentage area of vein covered with clot when exposed to thrombin in the multiple injury animals compared with uninjured (45% vs. 27% p = 0.0002) consistent with a phenotype of hypercoagulable state after trauma in our model system. Unmanipulated FoxC2 knockout mice manifest increased clotting at the vein valve as compared with unmanipulated wild type animals. After multiple injuries, wild type mice manifest increase clotting at the vein after thrombin exposure ( p = 0.0033), and equivalent to that of valvular knockout of FoxC2 (FoxC2del), recapitulating the phenotype seen in FoxC2 knockout animals. The combination of multiple injuries and FoxC2 knockout resulted in spontaneous microthrombi in 50% of the animals, a phenotype not observed with either multiple injuries or FoxC2 deficiency alone (χ 2 , p = 0.017). Finally, human vein samples demonstrated the protective vein valve phenotype of increased FOXC2 and PROX1 and showed decreased expression in the critically ill organ donor population by immunofluorescence imaging in organ donor samples. CONCLUSION We have established a novel model of posttrauma hypercoagulation that does not require direct restriction of venous flow or direct injury to the vessel endothelium to assay for hypercoagulability and can generate spontaneous microthrombosis when combined with valve-specific FOXC2 knockout. We find that multiple injuries induce a procoagulant phenotype that recapitulates the valvular hypercoagulability seen in FOXC2 knockout and, in critically ill human specimens, find evidence for loss of oscillatory shear stress-induced gene expression of FOXC2 and PROX1 in the valvular endothelium consistent with potential loss of DVT-protective valvular phenotype.
Collapse
Affiliation(s)
- Mark H Hoofnagle
- From the Section of Acute and Critical Care Surgery, Department of Surgery (M.H.H., A.H., S.G., S.-W.H., A.F., G.V.B., J.M.L., I.R.T.), Section of Vascular Surgery, Department of Surgery (M.N.), and Department of Pathology (G.J.R.), School of Medicine, Washington University in Saint Louis, St. Louis, Missouri; Osciflex LLC (J.D.W.); and Department of Medicine (M.L.K.), Cardiovascular Institute, University of Pennsylvania, Philadelphia, Pennsylvania
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
7
|
Bouck EG, Arvanitis M, Osburn WO, Sang Y, Reventun P, Ahmadzia HK, Smith NL, Lowenstein CJ, Wolberg AS. High risk oral contraceptive hormones do not directly enhance endothelial cell procoagulant activity in vitro. PLoS One 2023; 18:e0284333. [PMID: 37075041 PMCID: PMC10115293 DOI: 10.1371/journal.pone.0284333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 03/28/2023] [Indexed: 04/20/2023] Open
Abstract
BACKGROUND Oral contraceptive (OC) use increases venous thromboembolism risk 2-5-fold. Procoagulant changes can be detected in plasma from OC users even without thrombosis, but cellular mechanisms that provoke thrombosis have not been identified. Endothelial cell (EC) dysfunction is thought to initiate venous thromboembolism. It is unknown whether OC hormones provoke aberrant procoagulant activity in ECs. OBJECTIVE Characterize the effect of high-risk OC hormones (ethinyl estradiol [EE] and drospirenone) on EC procoagulant activity and the potential interplay with nuclear estrogen receptors ERα and ERβ and inflammatory processes. METHODS Human umbilical vein and dermal microvascular ECs (HUVEC and HDMVEC, respectively) were treated with EE and/or drospirenone. Genes encoding the estrogen receptors ERα and ERβ (ESR1 and ESR2, respectively) were overexpressed in HUVEC and HDMVEC via lentiviral vectors. EC gene expression was assessed by RT-qPCR. The ability of ECs to support thrombin generation and fibrin formation was measured by calibrated automated thrombography and spectrophotometry, respectively. RESULTS Neither EE nor drospirenone, alone or together, changed expression of genes encoding anti- or procoagulant proteins (TFPI, THBD, F3), integrins (ITGAV, ITGB3), or fibrinolytic mediators (SERPINE1, PLAT). EE and/or drospirenone did not increase EC-supported thrombin generation or fibrin formation, either. Our analyses indicated a subset of individuals express ESR1 and ESR2 transcripts in human aortic ECs. However, overexpression of ESR1 and/or ESR2 in HUVEC and HDMVEC did not facilitate the ability of OC-treated ECs to support procoagulant activity, even in the presence of a pro-inflammatory stimulus. CONCLUSIONS The OC hormones EE and drospirenone do not directly enhance thrombin generation potential of primary ECs in vitro.
Collapse
Affiliation(s)
- Emma G. Bouck
- Department of Pathology and Laboratory Medicine and UNC Blood Research Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | - Marios Arvanitis
- Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - William O. Osburn
- Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Yaqiu Sang
- Department of Pathology and Laboratory Medicine and UNC Blood Research Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | - Paula Reventun
- Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Homa K. Ahmadzia
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, George Washington University, Washington, DC, United States of America
| | - Nicholas L. Smith
- Department of Epidemiology, University of Washington, Seattle, WA, United States of America
- Kaiser Permanente Washington Health Research Institute, Kaiser Permanente Washington, Seattle, WA, United States of America
- Seattle Epidemiologic Research and Information Center, Department of Veterans Affairs Office of Research and Development, Seattle, WA, United States of America
| | - Charles J. Lowenstein
- Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Alisa S. Wolberg
- Department of Pathology and Laboratory Medicine and UNC Blood Research Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| |
Collapse
|
8
|
Platelet-Neutrophil Crosstalk in Thrombosis. Int J Mol Sci 2023; 24:ijms24021266. [PMID: 36674781 PMCID: PMC9861587 DOI: 10.3390/ijms24021266] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/02/2023] [Accepted: 01/04/2023] [Indexed: 01/11/2023] Open
Abstract
Platelets are essential for the formation of a haemostatic plug to prevent bleeding, while neutrophils are the guardians of our immune defences against invading pathogens. The interplay between platelets and innate immunity, and subsequent triggering of the activation of coagulation is part of the host system to prevent systemic spread of pathogen in the blood stream. Aberrant immunothrombosis and excessive inflammation can however, contribute to the thrombotic burden observed in many cardiovascular diseases. In this review, we highlight how platelets and neutrophils interact with each other and how their crosstalk is central to both arterial and venous thrombosis and in COVID-19. While targeting platelets and coagulation enables efficient antithrombotic treatments, they are often accompanied with a bleeding risk. We also discuss how novel approaches to reduce platelet-mediated recruitment of neutrophils could represent promising therapies to treat thrombosis without affecting haemostasis.
Collapse
|
9
|
Lisman T. Bleeding and thrombosis in cirrhosis. CARDIO-HEPATOLOGY 2023:165-202. [DOI: 10.1016/b978-0-12-817394-7.00010-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
10
|
Jing H, Wu X, Xiang M, Liu L, Novakovic VA, Shi J. Pathophysiological mechanisms of thrombosis in acute and long COVID-19. Front Immunol 2022; 13:992384. [PMID: 36466841 PMCID: PMC9709252 DOI: 10.3389/fimmu.2022.992384] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 10/27/2022] [Indexed: 08/02/2023] Open
Abstract
COVID-19 patients have a high incidence of thrombosis, and thromboembolic complications are associated with severe COVID-19 and high mortality. COVID-19 disease is associated with a hyper-inflammatory response (cytokine storm) mediated by the immune system. However, the role of the inflammatory response in thrombosis remains incompletely understood. In this review, we investigate the crosstalk between inflammation and thrombosis in the context of COVID-19, focusing on the contributions of inflammation to the pathogenesis of thrombosis, and propose combined use of anti-inflammatory and anticoagulant therapeutics. Under inflammatory conditions, the interactions between neutrophils and platelets, platelet activation, monocyte tissue factor expression, microparticle release, and phosphatidylserine (PS) externalization as well as complement activation are collectively involved in immune-thrombosis. Inflammation results in the activation and apoptosis of blood cells, leading to microparticle release and PS externalization on blood cells and microparticles, which significantly enhances the catalytic efficiency of the tenase and prothrombinase complexes, and promotes thrombin-mediated fibrin generation and local blood clot formation. Given the risk of thrombosis in the COVID-19, the importance of antithrombotic therapies has been generally recognized, but certain deficiencies and treatment gaps in remain. Antiplatelet drugs are not in combination with anticoagulant treatments, thus fail to dampen platelet procoagulant activity. Current treatments also do not propose an optimal time for anticoagulation. The efficacy of anticoagulant treatments depends on the time of therapy initiation. The best time for antithrombotic therapy is as early as possible after diagnosis, ideally in the early stage of the disease. We also elaborate on the possible mechanisms of long COVID thromboembolic complications, including persistent inflammation, endothelial injury and dysfunction, and coagulation abnormalities. The above-mentioned contents provide therapeutic strategies for COVID-19 patients and further improve patient outcomes.
Collapse
Affiliation(s)
- Haijiao Jing
- Department of Hematology, The First Hospital, Harbin Medical University, Harbin, China
| | - Xiaoming Wu
- Department of Hematology, The First Hospital, Harbin Medical University, Harbin, China
| | - Mengqi Xiang
- Department of Hematology, The First Hospital, Harbin Medical University, Harbin, China
| | - Langjiao Liu
- Department of Hematology, The First Hospital, Harbin Medical University, Harbin, China
| | - Valerie A. Novakovic
- Department of Research, VA Boston Healthcare System, Harvard Medical School, Boston, MA, United States
| | - Jialan Shi
- Department of Hematology, The First Hospital, Harbin Medical University, Harbin, China
- Department of Research, VA Boston Healthcare System, Harvard Medical School, Boston, MA, United States
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
11
|
Galli E, Maggio E, Pomero F. Venous Thromboembolism in Sepsis: From Bench to Bedside. Biomedicines 2022; 10:biomedicines10071651. [PMID: 35884956 PMCID: PMC9313423 DOI: 10.3390/biomedicines10071651] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 07/04/2022] [Accepted: 07/04/2022] [Indexed: 12/22/2022] Open
Abstract
Septic patients were commonly affected by coagulation disorders; thus, they are at high risk of thrombotic complications. In the last decades, novel knowledge has emerged about the interconnected and reciprocal influence of immune and coagulation systems. This phenomenon is called immunothrombosis, and it indicates an effective response whereby immune cells and the coagulation cascade cooperate to limit pathogen invasion and endothelial damage. When this network becomes dysregulated due to a systemic inflammatory activation, as occurs during sepsis, it can result in pathological thrombosis. Endothelium, platelets and neutrophils are the main characters involved in this process, together with the TF and coagulation cascade, playing a critical role in both the host defense and in thrombogenesis. A deeper understanding of this relationship may allow us to answer the growing need for clinical instruments to establish the thrombotic risk and treatments that consider more the connection between coagulation and inflammation. Heparin remains the principal therapeutical response to this phenomenon, although not sufficiently effective. To date, no other significant alternatives have been found yet. In this review, we discuss the role of sepsis-related inflammation in the development and resolution of venous thromboembolism and its clinical implications, from bench to bedside.
Collapse
Affiliation(s)
- Eleonora Galli
- Internal Medicine Residency Program, University of Turin, 10100 Turin, TO, Italy;
- Department of Internal Medicine, M. and P. Ferrero Hospital, 12060 Verduno, CN, Italy;
| | - Elena Maggio
- Department of Internal Medicine, M. and P. Ferrero Hospital, 12060 Verduno, CN, Italy;
| | - Fulvio Pomero
- Department of Internal Medicine, M. and P. Ferrero Hospital, 12060 Verduno, CN, Italy;
- Correspondence: ; Tel.: +39-01721408100
| |
Collapse
|
12
|
Jin FM, Wang M, Wu XM, Xiao H, Wang DX, Wang GM, Zhang CG, Zhao HR. Effects of wasp venom on venous thrombosis in rats. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2022; 25:822-826. [PMID: 36033945 PMCID: PMC9392573 DOI: 10.22038/ijbms.2022.63219.13962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 06/21/2022] [Indexed: 11/24/2022]
Abstract
OBJECTIVES This study aimed to investigate the potential effects of wasp venom (WV) from Vespa magnifica on antithrombosis in rats with inferior vena cava (IVC) thrombosis. MATERIALS AND METHODS The thrombosis rat model was established by improving the IVC stenosis, in which rats were subjected to IVC ligation for 75 min. Rats were administered argatroban (IP) or WV (s.c.) for 4 hr after IVC thrombosis. The weight, inhibition rate, and pathological morphology of the thrombosis induced by IVC ligation and the variation in four coagulation parameters, coagulation factors, and CD61+CD62P+ were simultaneously determined in IVC rats. RESULTS The thrombus formed as a result of IVC ligation was stable. Compared with the control group, the weight of the thrombus was significantly reduced in the argatroban group. Thrombus weight was reduced by treatment with 0.6, 0.2, and 0.05 mg/kg WV, with inhibition rates of 52.19%, 35.32%, and 28.98%, respectively. Inflammatory cells adhered to and infiltrated the vessel wall in the IVC group more than in the sham group. However, the pathological morphology and CD61+CD62P+ of the WV treatment groups tended to be normal. CONCLUSION We improved the model of IVC thrombosis to be suitable for evaluation of antithrombotic drugs. Our findings demonstrated that WV could inhibit IVC thrombosis associated with reducing coagulation factors V and CD61+CD62p expression in rats.
Collapse
Affiliation(s)
- Fan-mao Jin
- Lishui City People’s Hospital, Lishui, Zhejiang 323000, People’s Republic of China,These authors contributed equally to this work
| | - Mei Wang
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, Dali University, Dali, China,National-Local Joint Engineering Research Center of Entomoceutics, Dali University, Dali, China,These authors contributed equally to this work
| | - Xiu-mei Wu
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, Dali University, Dali, China,National-Local Joint Engineering Research Center of Entomoceutics, Dali University, Dali, China
| | - Huai Xiao
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, Dali University, Dali, China
| | - De-xiao Wang
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, Dali University, Dali, China,Genetic Testing Center, The First Affiliated Hospital of Dali University, Dali University, Dali, China
| | - Guang-ming Wang
- Genetic Testing Center, The First Affiliated Hospital of Dali University, Dali University, Dali, China
| | - Cheng-gui Zhang
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, Dali University, Dali, China,National-Local Joint Engineering Research Center of Entomoceutics, Dali University, Dali, China,Corresponding authors: Cheng-gui Zhang. Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, Dali University, Dali, China; National-Local Joint Engineering Research Center of Entomoceutics, Dali University, Dali, China. ; Hai-rong Zhao. Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, Dali University, Dali, China; Genetic Testing Center, The First Affiliated Hospital of Dali University, Dali University, Dali, China; National-Local Joint Engineering Research Center of Entomoceutics, Dali University, Dali, China.
| | - Hai-rong Zhao
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, Dali University, Dali, China,National-Local Joint Engineering Research Center of Entomoceutics, Dali University, Dali, China,Genetic Testing Center, The First Affiliated Hospital of Dali University, Dali University, Dali, China,Corresponding authors: Cheng-gui Zhang. Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, Dali University, Dali, China; National-Local Joint Engineering Research Center of Entomoceutics, Dali University, Dali, China. ; Hai-rong Zhao. Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, Dali University, Dali, China; Genetic Testing Center, The First Affiliated Hospital of Dali University, Dali University, Dali, China; National-Local Joint Engineering Research Center of Entomoceutics, Dali University, Dali, China.
| |
Collapse
|
13
|
Marar TT, Matzko CN, Wu J, Esmon CT, Sinno T, Brass LF, Stalker TJ, Tomaiuolo M. Thrombin spatial distribution determines protein C activation during hemostasis and thrombosis. Blood 2022; 139:1892-1902. [PMID: 34890454 PMCID: PMC8952187 DOI: 10.1182/blood.2021014338] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 12/07/2021] [Indexed: 11/20/2022] Open
Abstract
Rebalancing the hemostatic system by targeting endogenous anticoagulant pathways, like the protein C (PC) system, is being tested as a means of improving hemostasis in patients with hemophilia. Recent intravital studies of hemostasis demonstrated that, in some vascular contexts, thrombin activity is sequestered in the extravascular compartment. These findings raise important questions about the context-dependent contribution of activated PC (APC) to the hemostatic response, because PC activation occurs on the surface of endothelial cells. We used a combination of pharmacologic, genetic, imaging, and computational approaches to examine the relationships among thrombin spatial distribution, PC activation, and APC anticoagulant function. We found that inhibition of APC activity, in mice either harboring the factor V Leiden mutation or infused with an APC-blocking antibody, significantly enhanced fibrin formation and platelet activation in a microvascular injury model, consistent with the role of APC as an anticoagulant. In contrast, inhibition of APC activity had no effect on hemostasis after penetrating injury of the mouse jugular vein. Computational studies showed that differences in blood velocity, injury size, and vessel geometry determine the localization of thrombin generation and, consequently, the extent of PC activation. Computational predictions were tested in vivo and showed that when thrombin generation occurred intravascularly, without penetration of the vessel wall, inhibition of APC significantly increased fibrin formation in the jugular vein. Together, these studies show the importance of thrombin spatial distribution in determining PC activation during hemostasis and thrombosis.
Collapse
Affiliation(s)
- Tanya T Marar
- Department of Medicine, University of Pennsylvania, Philadelphia, PA
- Cardeza Center for Hemostasis, Thrombosis, and Vascular Biology, Cardeza Foundation for Hematologic Research, Thomas Jefferson University, Philadelphia, PA
| | - Chelsea N Matzko
- Department of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Jie Wu
- Department of Medicine, University of Pennsylvania, Philadelphia, PA
| | | | - Talid Sinno
- Department of Chemical and Biomolecular Engineering, University of Pennsylvania, Philadelphia, PA; and
| | - Lawrence F Brass
- Department of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Timothy J Stalker
- Department of Medicine, University of Pennsylvania, Philadelphia, PA
- Cardeza Center for Hemostasis, Thrombosis, and Vascular Biology, Cardeza Foundation for Hematologic Research, Thomas Jefferson University, Philadelphia, PA
| | - Maurizio Tomaiuolo
- Department of Medicine, University of Pennsylvania, Philadelphia, PA
- Vickie and Jack Farber Vision Research Center, Wills Eye Hospital, Philadelphia, PA
| |
Collapse
|
14
|
Navaneeth Krishna RP, Jain A. In silico analyses of blood flow and oxygen transport in human micro-veins and valves. Clin Hemorheol Microcirc 2022; 81:81-96. [PMID: 35034895 DOI: 10.3233/ch-211345] [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: 11/15/2022]
Abstract
BACKGROUND Almost 95% of the venous valves are micron scale found in veins smaller than 300μm diameter. The fluid dynamics of blood flow and transport through these micro venous valves and their contribution to thrombosis is not yet well understood or characterized due to difficulty in making direct measurements in murine models. OBJECTIVE The unique flow patterns that may arise in physiological and pathological non-actuating micro venous valves are predicted. METHODS Computational fluid and transport simulations are used to model blood flow and oxygen gradients in a microfluidic vein. RESULTS The model successfully recreates the typical non-Newtonian vortical flow within the valve cusps seen in preclinical experimental models and in clinic. The analysis further reveals variation in the vortex strengths due to temporal changes in blood flow. The cusp oxygen is typically low from the main lumen, and it is regulated by systemic venous flow. CONCLUSIONS The analysis leads to a clinically-relevant hypothesis that micro venous valves may not create a hypoxic environment needed for endothelial inflammation, which is one of the main causes of thrombosis. However, incompetent micro venous valves are still locations for complex fluid dynamics of blood leading to low shear regions that may contribute to thrombosis through other pathways.
Collapse
Affiliation(s)
| | - Abhishek Jain
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, USA.,Department of Medical Physiology, College of Medicine, Texas A&M Health Science Center, USA.,Department of Cardiovascular Sciences, Houston Methodist Academic Institute, Houston, USA
| |
Collapse
|
15
|
Wong DJ, Park DD, Park SS, Haller CA, Chen J, Dai E, Liu L, Mandhapati AR, Eradi P, Dhakal B, Wever WJ, Hanes M, Sun L, Cummings RD, Chaikof EL. A PSGL-1 glycomimetic reduces thrombus burden without affecting hemostasis. Blood 2021; 138:1182-1193. [PMID: 33945603 PMCID: PMC8570056 DOI: 10.1182/blood.2020009428] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 04/19/2021] [Indexed: 11/20/2022] Open
Abstract
Events mediated by the P-selectin/PSGL-1 pathway play a critical role in the initiation and propagation of venous thrombosis by facilitating the accumulation of leukocytes and platelets within the growing thrombus. Activated platelets and endothelium express P-selectin, which binds P-selectin glycoprotein ligand-1 (PSGL-1) that is expressed on the surface of all leukocytes. We developed a pegylated glycomimetic of the N terminus of PSGL-1, PEG40-GSnP-6 (P-G6), which proved to be a highly potent P-selectin inhibitor with a favorable pharmacokinetic profile for clinical translation. P-G6 inhibits human and mouse platelet-monocyte and platelet-neutrophil aggregation in vitro and blocks microcirculatory platelet-leukocyte interactions in vivo. Administration of P-G6 reduces thrombus formation in a nonocclusive model of deep vein thrombosis with a commensurate reduction in leukocyte accumulation, but without disruption of hemostasis. P-G6 potently inhibits the P-selectin/PSGL-1 pathway and represents a promising drug candidate for the prevention of venous thrombosis without increased bleeding risk.
Collapse
Affiliation(s)
- Daniel J Wong
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Diane D Park
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Simon S Park
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Carolyn A Haller
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Jiaxuan Chen
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Erbin Dai
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Liying Liu
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Appi R Mandhapati
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Pradheep Eradi
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Bibek Dhakal
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Walter J Wever
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Melinda Hanes
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Lijun Sun
- Center for Drug Discovery and Translational Research, Department of Surgery, Beth Israel Deaconess Medical Center and
| | - Richard D Cummings
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
- Harvard Medical School Center for Glycoscience, Harvard Medical School, Boston, MA
| | - Elliot L Chaikof
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| |
Collapse
|
16
|
Lin J, Sorrells MG, Lam WA, Neeves KB. Physical forces regulating hemostasis and thrombosis: Vessels, cells, and molecules in illustrated review. Res Pract Thromb Haemost 2021; 5:e12548. [PMID: 34278188 PMCID: PMC8279127 DOI: 10.1002/rth2.12548] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 04/21/2021] [Accepted: 04/29/2021] [Indexed: 01/31/2023] Open
Abstract
This illustrated review focuses on the physical forces that regulate hemostasis and thrombosis. These phenomena span from the vessel to the cellular to the molecular scales. Blood is a complex fluid with a viscosity that varies with how fast it flows and the size of the vessel through which it flows. Blood flow imposes forces on the vessel wall and blood cells that dictates the kinetics, structure, and stability of thrombi. The mechanical properties of blood cells create a segmented flowing fluid whereby red blood cells concentrate in the vessel core and platelets marginate to the near-wall region. At the vessel wall, shear stresses are highest, which requires a repertoire of receptors with different bond kinetics to roll, tether, adhere, and activate on inflamed endothelium and extracellular matrices. As a thrombus grows and then contracts, forces regulate platelet aggregation as well as von Willebrand factor function and fibrin mechanics. Forces can also originate from platelets as they respond to the external forces and sense the stiffness of their local environment.
Collapse
Affiliation(s)
- Jessica Lin
- Wallace H. Coulter Department of Biomedical EngineeringGeorgia Institute of TechnologyEmory UniversityAtlantaGAUSA
| | - Matthew G. Sorrells
- Department of Chemical and Biological EngineeringColorado School of MinesGoldenCOUSA
| | - Wilbur A. Lam
- Wallace H. Coulter Department of Biomedical EngineeringGeorgia Institute of TechnologyEmory UniversityAtlantaGAUSA
- Division of Pediatric Hematology/OncologyDepartment of PediatricsAflac Cancer Center and Blood Disorder Service of Children’s Healthcare of AtlantaEmory University School of MedicineAtlantaGAUSA
| | - Keith B. Neeves
- Department of BioengineeringUniversity of Colorado DenverAnschutz Medical CampusAuroraCOUSA
- Department of Pediatrics, Section of Hematology, Oncology, and Bone Marrow Transplant, Hemophilia and Thrombosis CenterUniversity of Colorado DenverAnschutz Medical CampusAuroraCOUSA
| |
Collapse
|
17
|
Abstract
The association between inflammation, infection, and venous thrombosis has long been recognized; yet, only in the last decades have we begun to understand the mechanisms through which the immune and coagulation systems interact and reciprocally regulate one another. These interconnected networks mount an effective response to injury and pathogen invasion, but if unregulated can result in pathological thrombosis and organ damage. Neutrophils, monocytes, and platelets interact with each other and the endothelium in host defense and also play critical roles in the formation of venous thromboembolism. This knowledge has advanced our understanding of both human physiology and pathophysiology, as well as identified mechanisms of anticoagulant resistance and novel therapeutic targets for the prevention and treatment of thrombosis. In this review, we discuss the contributions of inflammation and infection to venous thromboembolism.
Collapse
Affiliation(s)
- Meaghan E. Colling
- Hematology Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
- Clinical Division of Haematology and Haemostaseology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Benjamin E. Tourdot
- Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Yogendra Kanthi
- Laboratory of Vascular Thrombosis and Inflammation, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
18
|
Kanji R, Kubica J, Navarese EP, Gorog DA. Endogenous fibrinolysis-Relevance to clinical thrombosis risk assessment. Eur J Clin Invest 2021; 51:e13471. [PMID: 33296082 DOI: 10.1111/eci.13471] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Revised: 12/05/2020] [Accepted: 12/06/2020] [Indexed: 12/11/2022]
Abstract
The development of an obstructive luminal thrombus is pathological and considered a failure of endogenous fibrinolysis. The consequences may be fatal, or result in lasting downstream organ damage. Therefore, assessment of endogenous fibrinolytic status in an individual may identify those at risk of occlusive thrombus formation and provide prognostic information. Arterial thrombi are more platelet rich and more resistant to fibrinolysis than venous thrombi. Several recent studies using global tests of fibrinolysis in patients with acute coronary syndromes (ACS) have shown that despite dual antiplatelet therapy, patients with impaired fibrinolytic status have an increased risk of adverse cardiovascular events, compared with those with effective fibrinolytic function. Such data add significantly to the predictive value of established cardiovascular risk factors and conventional biomarkers. Most data reported have been obtained with the Global Thrombosis Test and the turbidimetric plasma clot lysis assay. A few small studies in patients with ischaemic stroke suggest a similar predictive role of fibrinolytic status assessment in these patients. Studies reporting an association between impaired fibrinolysis and future venous thrombotic events are limited, and in the form of case-control studies. Viscoelastic assays may have a role in the prediction of venous thromboembolic risk. Assays of fibrinolytic function should be used to obtain a more accurate risk of future thrombotic events, particularly in the setting of ACS. The availability of point-of-care tests helps facilitate this and should encourage future studies to assess personalised antithrombotic treatment combinations to optimise fibrinolytic status and reduce thrombosis risk.
Collapse
Affiliation(s)
- Rahim Kanji
- Faculty of Medicine, National Heart and Lung Institute, Imperial College, London, UK.,Cardiology Department, East and North Hertfordshire NHS Trust, Stevenage, UK
| | - Jacek Kubica
- Department of Cardiology and Internal Medicine, Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz, Poland
| | - Eliano P Navarese
- Department of Cardiology and Internal Medicine, Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz, Poland.,Faculty of Medicine, University of Alberta, Edmonton, Canada
| | - Diana A Gorog
- Faculty of Medicine, National Heart and Lung Institute, Imperial College, London, UK.,Cardiology Department, East and North Hertfordshire NHS Trust, Stevenage, UK.,School of Life and Medical Sciences, Postgraduate Medical School, University of Hertfordshire, Stevenage, UK
| |
Collapse
|
19
|
Abstract
Post-thrombotic syndrome (PTS) is a chronic venous insufficiency manifestation following an episode of deep-vein thrombosis (DVT). It is an important and frequent long-term adverse event of proximal DVT affecting 20-50% of patients. This position paper integrates data guiding clinicians in deciding PTS diagnosis, treatment and follow-up.
Collapse
Affiliation(s)
- Adriana Visonà
- Angiology Unit, Azienda ULSS 2 Marca Trevigiana, Castelfranco Veneto, Italy
| | - Isabelle Quere
- Médecine Vasculaire, Université de Montpellier, Montpellier, France
| | - Lucia Mazzolai
- Division of Angiology, Heart and Vessel Department, Lausanne University Hospital, Lausanne, Switzerland
| | - Maria Amitrano
- Internal Medicine Unit, Moscati Hospital, Avellino, Italy
| | - Marzia Lugli
- Department of Vascular Surgery, International Center of Deep Venous Surgery, Hesperia Hospital, Modena, Italy
| | - Juraj Madaric
- Clinic of Angiology, Comenius University and National Institute of Cardiovascular Diseases, Bratislava, Slovakia
| | | | | |
Collapse
|
20
|
Current use of rivaroxaban in elderly patients with venous thromboembolism (VTE). J Thromb Thrombolysis 2021; 52:863-871. [PMID: 33674983 DOI: 10.1007/s11239-021-02415-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/16/2021] [Indexed: 12/12/2022]
Abstract
Venous thromboembolism (VTE), which is characterized by pulmonary embolism and deep vein thrombosis, has become a serious public concern. Notably, over half of the patients with VTE are over 70 years of age, but elderly patients are at high risk of anti-coagulation and bleeding, which increase with age. Moreover, risk factors and frailty also show a difference between elderly patients and ordinary patients diagnosed with VTE. Rivaroxaban is a direct inhibitor of activated factor Xa and has the advantage of predictable pharmacodynamics and pharmacokinetics, no coagulation monitoring, and few drug interactions. As a first-line therapy for VTE, this drug is more advantageous than traditional therapy and exhibits good efficacy and safety for ordinary patients. However, the effectiveness and safety of rivaroxaban in elderly patients have not been fully elucidated. This article reviewed the use of rivaroxaban in elderly patients, including drug interactions, monitoring, reversal agents of rivaroxaban, and the use of small dosages of rivaroxaban in elderly patients.
Collapse
|
21
|
Rajeeva Pandian NK, Walther BK, Suresh R, Cooke JP, Jain A. Microengineered Human Vein-Chip Recreates Venous Valve Architecture and Its Contribution to Thrombosis. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 16:e2003401. [PMID: 33205630 PMCID: PMC7791597 DOI: 10.1002/smll.202003401] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 10/01/2020] [Indexed: 05/03/2023]
Abstract
Deep vein thrombosis (DVT) and its consequences are lethal, but current models cannot completely dissect its determinants-endothelium, flow, and blood constituents-together called Virchow's triad. Most models for studying DVT forego assessment of venous valves that serve as the primary sites of DVT formation. Therefore, the knowledge of DVT formed at the venous cusps has remained obscure due to lack of experimental models. Here, organ-on-chip methodology is leveraged to create a Vein-Chip platform integrating fully vascularized venous valves and its hemodynamic, as seen in vivo. These Vein-Chips reveal that vascular endothelium of valve cusps adapts to the locally disturbed microenvironment by expressing a different phenotype from the regions of uniform flow. This spatial adaptation of endothelial function recreated on the in vitro Vein-Chip platform is shown to protect the vein from thrombosis from disturbed flow in valves, but interestingly, cytokine stimulation reverses the effect and switches the valve endothelium to becoming prothrombotic. The platform eventually modulates the three factors of Virchow's triad and provides a systematic approach to investigate the determinants of fibrin and platelet dynamics of DVT. Therefore, this Vein-Chip offers a new preclinical approach to study venous pathophysiology and show effects of antithrombotic drug treatment.
Collapse
Affiliation(s)
| | - Brandon K Walther
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, College Station, TX, 77843, USA
- Department of Cardiovascular Sciences, Houston Methodist Institute for Academic Medicine and Houston Methodist Research Institute, 6670 Bertner Ave., Houston, TX, 77030, USA
| | - Rishi Suresh
- Texas A&M Health Science Center, College of Medicine, Bryan, TX, 77807, USA
| | - John P Cooke
- Department of Cardiovascular Sciences, Houston Methodist Institute for Academic Medicine and Houston Methodist Research Institute, 6670 Bertner Ave., Houston, TX, 77030, USA
| | - Abhishek Jain
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, College Station, TX, 77843, USA
- Department of Medical Physiology, College of Medicine, Texas A&M Health Science Center, Bryan, TX, 77808, USA
| |
Collapse
|
22
|
Henke P, Sharma S, Wakefield T, Myers D, Obi A. Insights from experimental post-thrombotic syndrome and potential for novel therapies. Transl Res 2020; 225:95-104. [PMID: 32442728 PMCID: PMC7487018 DOI: 10.1016/j.trsl.2020.05.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 04/20/2020] [Accepted: 05/12/2020] [Indexed: 12/20/2022]
Abstract
Post-thrombotic syndrome (PTS) is an end stage manifestation of deep vein thrombosis. This is an inherently inflammatory process, with consequent fibrosis. Multiple cellular types are involved, and are likely driven by leukocytes. Herein, we review the current gaps in therapy, and insights from rodent models of venous thrombosis that suggest possible targets to treat and prevent PTS.
Collapse
Affiliation(s)
- Peter Henke
- From the University of Michigan Health System, Frankel Cardiovascular Center, Ann Arbor, MI.
| | - Sriganesh Sharma
- From the University of Michigan Health System, Frankel Cardiovascular Center, Ann Arbor, MI
| | - Thomas Wakefield
- From the University of Michigan Health System, Frankel Cardiovascular Center, Ann Arbor, MI
| | - Dan Myers
- From the University of Michigan Health System, Frankel Cardiovascular Center, Ann Arbor, MI
| | - Andrea Obi
- From the University of Michigan Health System, Frankel Cardiovascular Center, Ann Arbor, MI
| |
Collapse
|
23
|
Abstract
Focusing on the current state of the art, this article (a) describes recent advances in the understanding of the pathogenesis of venous thromboembolism (VTE), (b) discusses current approaches for the prevention, diagnosis and treatment of VTE, (c) outlines the role of aspirin for VTE prevention and treatment, and (d) highlights the unmet needs in VTE management and describes novel approaches to address them.
Collapse
Affiliation(s)
- Noel C Chan
- Thrombosis and Atherosclerosis Research Institute and McMaster University, Hamilton, Ontario, Canada
| | - Jeffrey I Weitz
- Thrombosis and Atherosclerosis Research Institute and McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
24
|
Shaydakov ME, Diaz JA, Comerota AJ, Lurie F. Targeted gene expression analysis of human deep veins. J Vasc Surg Venous Lymphat Disord 2020; 9:770-780.e7. [PMID: 32860957 DOI: 10.1016/j.jvsv.2020.08.025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 08/15/2020] [Indexed: 11/25/2022]
Abstract
OBJECTIVE Endothelial-derived molecules involved in thrombosis and hemostasis have been investigated mainly in arteries and in experimental animals. The actual presence and integral function of these molecules in the human deep venous system have received less attention. Our aim was to evaluate the expression of certain prothrombotic and antithrombotic genes in the normal human deep veins of the lower extremities. METHODS Macroscopically intact and competent valve-containing segments of human deep veins were prospectively collected from patients who had undergone above-knee amputation. Vein samples were separated into four zones: zone 1, postvalve (downstream, proximal) vein wall; zone 2, the valve cusp; zone 3, prevalve (upstream, distal) vein wall; and zone 4, vein wall within the valve cusp (cusp removed). Real-time quantitative polymerase chain reaction for principal genes involved in coagulation, fibrinolysis, and inflammation was performed to quantify messenger RNA. Selected protein gene products were measured by the western blot assay. One additional valve-containing segment underwent mass spectrometry analysis to investigate global differences in the proteome between the study zones. RESULTS Seventeen valve-containing vein segments were analyzed. Significant upregulation of antithrombotic (protein C receptor [PROCR], thrombomodulin [THBD], tissue factor pathway inhibitor [TFPI]), prothrombotic (con Willebrand factor [VWF]), and proinflammatory (selectin P [SELP], intercellular adhesion molecule 1 [ICAM1]) genes was found in the valve cusp compared with the vein wall (P < .05). PROCR and THBD demonstrated the highest level of upregulation in the valve cusp. PROCR, serpin peptidase inhibitor, clade E, member 1 (SERPINE1), and SELP were upregulated in the valve cusp at the protein level (P < .05). Messenger RNA composition in the vein wall within the valve cusp was similar to the prevalve and postvalve vein wall for all genes, except for two times overexpressed ICAM1 (P < .05). Substantial differences within the proteome between the study zones were observed with mass spectrometry. CONCLUSIONS The biological properties of the valve cusp, vein wall within the valve cusp, and vein wall beyond the valve cusp are different. The endothelium of the valve cusps of a normal competent deep venous valve may be naturally less thrombogenic compared with the vein wall. The endothelium of the valve cusp may have a higher potential to interact with white blood cells compared with the vein wall. Mass spectrometry demonstrates substantial differences in the proteome between the vein wall and the valve cusps that were not anticipated before. (J Vasc Surg Venous Lymphat Disord 2021;9:770-80.) CLINICAL RELEVANCE: Deep vein thrombosis (DVT) is a major cause of mortality, morbidity, and impaired quality of life. Multiple risk factors have been identified, although their relative weight and pathophysiologic interactions remain obscure. Many patients with multiple risk factors for DVT never develop this condition. Conversely, in numerous cases DVT cannot be attributed to any known clinical risk factor. The molecular mechanisms that initiate DVT are unclear. An improved understanding of the normal biology of human deep veins will serve as an important foundation for new hypotheses of the pathogenesis of DVT. The latter may suggest new projects on novel therapeutic strategies.
Collapse
Affiliation(s)
- Maxim E Shaydakov
- Conrad Jobst Research Vascular Laboratories, University of Michigan Medical School, Ann Arbor, Mich; Jobst Vascular Institute, ProMedica Toledo Hospital, Toledo, Ohio; Department of Surgery, Central Michigan University College of Medicine, Saginaw, Mich.
| | - Jose A Diaz
- Conrad Jobst Research Vascular Laboratories, University of Michigan Medical School, Ann Arbor, Mich; Division of Surgical Research, Surgical Sciences, Vanderbilt University Medical Center, Nashville, Tenn
| | - Anthony J Comerota
- Jobst Vascular Institute, ProMedica Toledo Hospital, Toledo, Ohio; Inova Heart and Vascular Institute, Inova Alexandria Hospital, Alexandria, Va
| | - Fedor Lurie
- Jobst Vascular Institute, ProMedica Toledo Hospital, Toledo, Ohio
| |
Collapse
|
25
|
Lizarralde-Iragorri MA, Shet AS. Sickle Cell Disease: A Paradigm for Venous Thrombosis Pathophysiology. Int J Mol Sci 2020; 21:ijms21155279. [PMID: 32722421 PMCID: PMC7432404 DOI: 10.3390/ijms21155279] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 07/23/2020] [Accepted: 07/24/2020] [Indexed: 02/06/2023] Open
Abstract
Venous thromboembolism (VTE) is an important cause of vascular morbidity and mortality. Many risk factors have been identified for venous thrombosis that lead to alterations in blood flow, activate the vascular endothelium, and increase the propensity for blood coagulation. However, the precise molecular and cellular mechanisms that cause blood clots in the venous vasculature have not been fully elucidated. Patients with sickle cell disease (SCD) demonstrate all the risk factors for venous stasis, activated endothelium, and blood hypercoagulability, making them particularly vulnerable to VTE. In this review, we will discuss how mouse models have elucidated the complex vascular pathobiology of SCD. We review the dysregulated pathways of inflammation and coagulation in SCD and how the resultant hypercoagulable state can potentiate thrombosis through down-regulation of vascular anticoagulants. Studies of VTE pathogenesis using SCD mouse models may provide insight into the intersection between the cellular and molecular processes involving inflammation and coagulation and help to identify novel mechanistic pathways.
Collapse
|
26
|
Welsh JD, Hoofnagle MH, Bamezai S, Oxendine M, Lim L, Hall JD, Yang J, Schultz S, Engel JD, Kume T, Oliver G, Jimenez JM, Kahn ML. Hemodynamic regulation of perivalvular endothelial gene expression prevents deep venous thrombosis. J Clin Invest 2020; 129:5489-5500. [PMID: 31710307 DOI: 10.1172/jci124791] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 09/18/2019] [Indexed: 12/12/2022] Open
Abstract
Deep venous thrombosis (DVT) and secondary pulmonary embolism cause approximately 100,000 deaths per year in the United States. Physical immobility is the most significant risk factor for DVT, but a molecular and cellular basis for this link has not been defined. We found that the endothelial cells surrounding the venous valve, where DVTs originate, express high levels of FOXC2 and PROX1, transcription factors known to be activated by oscillatory shear stress. The perivalvular venous endothelial cells exhibited a powerful antithrombotic phenotype characterized by low levels of the prothrombotic proteins vWF, P-selectin, and ICAM1 and high levels of the antithrombotic proteins thrombomodulin (THBD), endothelial protein C receptor (EPCR), and tissue factor pathway inhibitor (TFPI). The perivalvular antithrombotic phenotype was lost following genetic deletion of FOXC2 or femoral artery ligation to reduce venous flow in mice, and at the site of origin of human DVT associated with fatal pulmonary embolism. Oscillatory blood flow was detected at perivalvular sites in human veins following muscular activity, but not in the immobile state or after activation of an intermittent compression device designed to prevent DVT. These findings support a mechanism of DVT pathogenesis in which loss of muscular activity results in loss of oscillatory shear-dependent transcriptional and antithrombotic phenotypes in perivalvular venous endothelial cells, and suggest that prevention of DVT and pulmonary embolism may be improved by mechanical devices specifically designed to restore perivalvular oscillatory flow.
Collapse
Affiliation(s)
- John D Welsh
- Department of Medicine and Cardiovascular Institute, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Mark H Hoofnagle
- Department of Surgery, Division of Traumatology, Surgical Critical Care, and Emergency Surgery, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Sharika Bamezai
- Department of Medicine and Cardiovascular Institute, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Michael Oxendine
- Center for Vascular and Developmental Biology, Feinberg Cardiovascular Research Institute, Northwestern University, Chicago, Illinois, USA
| | - Lillian Lim
- Department of Medicine and Cardiovascular Institute, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Joshua D Hall
- Department of Mechanical and Industrial Engineering, University of Massachusetts Amherst, Amherst, Massachusetts, USA
| | - Jisheng Yang
- Department of Medicine and Cardiovascular Institute, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Susan Schultz
- Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - James Douglas Engel
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan, USA
| | - Tsutomu Kume
- Center for Vascular and Developmental Biology, Feinberg Cardiovascular Research Institute, Northwestern University, Chicago, Illinois, USA
| | - Guillermo Oliver
- Center for Vascular and Developmental Biology, Feinberg Cardiovascular Research Institute, Northwestern University, Chicago, Illinois, USA
| | - Juan M Jimenez
- Department of Mechanical and Industrial Engineering, University of Massachusetts Amherst, Amherst, Massachusetts, USA
| | - Mark L Kahn
- Department of Medicine and Cardiovascular Institute, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
27
|
Gutmann C, Siow R, Gwozdz AM, Saha P, Smith A. Reactive Oxygen Species in Venous Thrombosis. Int J Mol Sci 2020; 21:E1918. [PMID: 32168908 PMCID: PMC7139897 DOI: 10.3390/ijms21061918] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 03/02/2020] [Accepted: 03/04/2020] [Indexed: 01/03/2023] Open
Abstract
Reactive oxygen species (ROS) have physiological roles as second messengers, but can also exert detrimental modifications on DNA, proteins and lipids if resulting from enhanced generation or reduced antioxidant defense (oxidative stress). Venous thrombus (DVT) formation and resolution are influenced by ROS through modulation of the coagulation, fibrinolysis, proteolysis and the complement system, as well as the regulation of effector cells such as platelets, endothelial cells, erythrocytes, neutrophils, mast cells, monocytes and fibroblasts. Many conditions that carry an elevated risk of venous thrombosis, such as the Antiphospholipid Syndrome, have alterations in their redox homeostasis. Dietary and pharmacological antioxidants can modulate several important processes involved in DVT formation, but their overall effect is unknown and there are no recommendations regarding their use. The development of novel antioxidant treatments that aim to abrogate the formation of DVT or promote its resolution will depend on the identification of targets that enable ROS modulation confined to their site of interest in order to prevent off-target effects on physiological redox mechanisms. Subgroups of patients with increased systemic oxidative stress might benefit from unspecific antioxidant treatment, but more clinical studies are needed to bring clarity to this issue.
Collapse
Affiliation(s)
- Clemens Gutmann
- King’s British Heart Foundation Centre, King’s College London, 125 Coldharbour Lane, London SE5 9NU, UK;
| | - Richard Siow
- Vascular Biology & Inflammation Section, School of Cardiovascular Medicine & Sciences, British Heart Foundation of Research Excellence, King’s College London, SE1 9NH, UK;
| | - Adam M. Gwozdz
- Academic Department of Surgery, School of Cardiovascular Medicine & Sciences, British Heart Foundation of Research Excellence, King’s College London, London SE1 7EH, UK; (A.M.G.); (P.S.)
| | - Prakash Saha
- Academic Department of Surgery, School of Cardiovascular Medicine & Sciences, British Heart Foundation of Research Excellence, King’s College London, London SE1 7EH, UK; (A.M.G.); (P.S.)
| | - Alberto Smith
- Academic Department of Surgery, School of Cardiovascular Medicine & Sciences, British Heart Foundation of Research Excellence, King’s College London, London SE1 7EH, UK; (A.M.G.); (P.S.)
| |
Collapse
|
28
|
Wolberg AS. Modeling Venous Thrombosis In Vitro: More Than Just (Valve) Pocket Change. Arterioscler Thromb Vasc Biol 2019; 38:980-981. [PMID: 29695530 DOI: 10.1161/atvbaha.118.310919] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Alisa S Wolberg
- From the Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill.
| |
Collapse
|
29
|
Mukhopadhyay S, Johnson TA, Duru N, Buzza MS, Pawar NR, Sarkar R, Antalis TM. Fibrinolysis and Inflammation in Venous Thrombus Resolution. Front Immunol 2019; 10:1348. [PMID: 31258531 PMCID: PMC6587539 DOI: 10.3389/fimmu.2019.01348] [Citation(s) in RCA: 98] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 05/28/2019] [Indexed: 12/24/2022] Open
Abstract
Clinical observations and accumulating laboratory evidence support a complex interplay between coagulation, inflammation, innate immunity and fibrinolysis in venous thromboembolism (VTE). VTE, which includes deep vein thrombosis (DVT) and pulmonary embolism (PE), and the subsequent complications of post-thrombotic syndrome (PTS), are significant causes of morbidity and mortality in patients. Clinical risk factors for VTE include cancer, major trauma, surgery, sepsis, inflammatory bowel disease, paralysis, prolonged periods of immobility, and aging. Abnormalities in venous blood flow or stasis initiates the activation of endothelial cells, and in concert with platelets, neutrophils and monocytes, propagates VTE in an intact vein. In addition, inflammatory cells play crucial roles in thrombus recanalization and restoration of blood flow via fibrinolysis and vascular remodeling. Faster resolution of the thrombus is key for improved disease prognosis. While in the clinical setting, anticoagulation therapy is successful in preventing propagation of venous thrombi, current therapies are not designed to inhibit inflammation, which can lead to the development of PTS. Animal models of DVT have provided many insights into the molecular and cellular mechanisms involved in the formation, propagation, and resolution of venous thrombi as well as the roles of key components of the fibrinolytic system in these processes. Here, we review the recent advances in our understanding of fibrinolysis and inflammation in the resolution of VTE.
Collapse
Affiliation(s)
- Subhradip Mukhopadhyay
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, United States
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Tierra A. Johnson
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Nadire Duru
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Marguerite S. Buzza
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, United States
- University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, United States
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Nisha R. Pawar
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Rajabrata Sarkar
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, United States
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Toni M. Antalis
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, United States
- University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, United States
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
30
|
Al-Hillan A, Bajwa RS, Cheema A, Ezeume A, Gor S, Mahida H, Kountz DS. Venous thromboembolism prevention through the use of novel Factor Xa inhibitors. Postgrad Med 2018; 131:89-95. [PMID: 30394165 DOI: 10.1080/00325481.2019.1542925] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
The risk of a venous thromboembolic event is not limited to the pre-hospital, hospital, or immediate post-hospital period. Because of challenges with data collection, the risk of venous thromboembolism (VTE) up to 3 months post-hospitalization for patients with acute and serious medical problems in the setting of chronic disease and/or risk factors for VTE is probably under reported. The growing acceptance and indications of direct oral anticoagulants (DOACs) now includes an indication for VTE prevention for one of the Factor Xa agents. In this paper, the authors explore the issue of VTE in the extended post-hospital period and strategies to provide protection from these morbid and potentially mortal events with oral anticoagulants.
Collapse
Affiliation(s)
- A Al-Hillan
- a Department of Medicine , Jersey Shore University Medical Center , Neptune , NJ , USA
| | - R S Bajwa
- a Department of Medicine , Jersey Shore University Medical Center , Neptune , NJ , USA
| | - A Cheema
- a Department of Medicine , Jersey Shore University Medical Center , Neptune , NJ , USA
| | - A Ezeume
- a Department of Medicine , Jersey Shore University Medical Center , Neptune , NJ , USA
| | - S Gor
- a Department of Medicine , Jersey Shore University Medical Center , Neptune , NJ , USA
| | - H Mahida
- a Department of Medicine , Jersey Shore University Medical Center , Neptune , NJ , USA
| | - D S Kountz
- a Department of Medicine , Jersey Shore University Medical Center , Neptune , NJ , USA
| |
Collapse
|
31
|
Geng X, Cha B, Mahamud MR, Srinivasan RS. Intraluminal valves: development, function and disease. Dis Model Mech 2018; 10:1273-1287. [PMID: 29125824 PMCID: PMC5719258 DOI: 10.1242/dmm.030825] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The circulatory system consists of the heart, blood vessels and lymphatic vessels, which function in parallel to provide nutrients and remove waste from the body. Vascular function depends on valves, which regulate unidirectional fluid flow against gravitational and pressure gradients. Severe valve disorders can cause mortality and some are associated with severe morbidity. Although cardiac valve defects can be treated by valve replacement surgery, no treatment is currently available for valve disorders of the veins and lymphatics. Thus, a better understanding of valves, their development and the progression of valve disease is warranted. In the past decade, molecules that are important for vascular function in humans have been identified, with mouse studies also providing new insights into valve formation and function. Intriguing similarities have recently emerged between the different types of valves concerning their molecular identity, architecture and development. Shear stress generated by fluid flow has also been shown to regulate endothelial cell identity in valves. Here, we review our current understanding of valve development with an emphasis on its mechanobiology and significance to human health, and highlight unanswered questions and translational opportunities.
Collapse
Affiliation(s)
- Xin Geng
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Boksik Cha
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Md Riaj Mahamud
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA.,Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - R Sathish Srinivasan
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA .,Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| |
Collapse
|
32
|
Karasu A, Šrámek A, Rosendaal FR, van der Geest RJ, van Hylckama Vlieg A. Aging of the venous valves as a new risk factor for venous thrombosis in the elderly: the BATAVIA study. J Thromb Haemost 2018; 16:96-103. [PMID: 29065242 DOI: 10.1111/jth.13880] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Indexed: 11/29/2022]
Abstract
Essentials Risk of venous thrombosis (VT) related to valve thickness and valvular reflux in unknown. Venous valves and reflux were measured by ultrasonography in cases and controls aged 70+. Risk of VT was associated with increased valve thickness and valvular reflux >1second. Thickening of valves is a generic process: there was no difference between right and left legs. SUMMARY Background Increasing age is the strongest risk factor for venous thrombosis (VT). Increasing age has been related to a thickening of the venous valves and a decreased valvular function. The association between valve thickness and the risk of VT is not known. Objectives To assess the association between increased valve thickness and valve closure time (VCT) and the risk of VT. Methods Analyses were performed in the BATAVIA study, including 70 cases aged 70 + with a first VT and 96 controls. We performed an ultrasound examination of the valves in the popliteal veins. The valves were imaged with a 9 MHz linear probe using B-mode ultrasonography. VCT was measured as an indicator for valve function using an automatic inflatable cuff. To estimate the risk of VT, valve thickness was dichotomized at the 90th percentile as measured in controls and VCT was dichotomized at 1 s. Results Mean valve thickness of controls was similar in the left (0.36 mm, 95% CI 0.34-0.37) and right (0.36 mm, 95% CI 0.35-0.38) leg. In 45 cases a valve was observed in the contralateral leg with a mean valve thickness of 0.39 mm (95% CI 0.36-0.42). Cases had an increased valve thickness compared with controls: mean difference 0.028 mm (95%CI 0.001-0.055). Valve thickness > 90th percentile increased the risk of VT 2.9-fold. Mean VCT in controls was 0.38 s, in contralateral leg of cases 0.58 s. VCT > 1 s increased the risk of VT 2.8-fold (95% CI 0.8-10.4). Conclusions Risk of VT was associated with increased valve thickness and valvular reflux of > 1 s.
Collapse
Affiliation(s)
- A Karasu
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, the Netherlands
| | - A Šrámek
- Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| | - F R Rosendaal
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, the Netherlands
- Department of Thrombosis and Haemostasis Research Center, Leiden University Medical Center, Leiden, the Netherlands
| | - R J van der Geest
- Division of Image Processing, Leiden University Medical Center, Leiden, the Netherlands
| | - A van Hylckama Vlieg
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
33
|
Stone J, Hangge P, Albadawi H, Wallace A, Shamoun F, Knuttien MG, Naidu S, Oklu R. Deep vein thrombosis: pathogenesis, diagnosis, and medical management. Cardiovasc Diagn Ther 2017; 7:S276-S284. [PMID: 29399531 DOI: 10.21037/cdt.2017.09.01] [Citation(s) in RCA: 175] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Deep vein thrombosis (DVT) is a major preventable cause of morbidity and mortality worldwide. Venous thromboembolism (VTE), which includes DVT and pulmonary embolism (PE), affects an estimated 1 per 1,000 people and contributes to 60,000-100,000 deaths annually. Normal blood physiology hinges on a delicate balance between pro- and anti-coagulant factors. Virchow's Triad distills the multitude of risk factors for DVT into three basic elements favoring thrombus formation: venous stasis, vascular injury, and hypercoagulability. Clinical, biochemical, and radiological tests are used to increase the sensitivity and specificity for diagnosing DVT. Anticoagulation therapy is essential for the treatment of DVT. With few exceptions, the standard therapy for DVT has been vitamin K-antagonists (VKAs) such as warfarin with heparin or fractionated heparin bridging. More recently, a number of large-scale clinical trials have validated the use of direct oral anticoagulants (DOACs) in place of warfarin in select cases. In this review, we summarize the pathogenesis, diagnosis, and medical management of DVT, with particular emphasis on anticoagulation therapy and the role of DOACs in the current treatment algorithm.
Collapse
Affiliation(s)
- Jonathan Stone
- Division of Interventional Radiology, Mayo Clinic, Phoenix, AZ, USA
| | - Patrick Hangge
- Division of Interventional Radiology, Mayo Clinic, Phoenix, AZ, USA
| | - Hassan Albadawi
- Division of Interventional Radiology, Mayo Clinic, Phoenix, AZ, USA
| | - Alex Wallace
- Division of Interventional Radiology, Mayo Clinic, Phoenix, AZ, USA
| | - Fadi Shamoun
- Division of Cardiovascular Diseases, Mayo Clinic, Phoenix, AZ, USA
| | - M Grace Knuttien
- Division of Interventional Radiology, Mayo Clinic, Phoenix, AZ, USA
| | - Sailendra Naidu
- Division of Interventional Radiology, Mayo Clinic, Phoenix, AZ, USA
| | - Rahmi Oklu
- Division of Interventional Radiology, Mayo Clinic, Phoenix, AZ, USA
| |
Collapse
|
34
|
Haynes LM, Orfeo T, Mann KG, Everse SJ, Brummel-Ziedins KE. Probing the Dynamics of Clot-Bound Thrombin at Venous Shear Rates. Biophys J 2017; 112:1634-1644. [PMID: 28445754 DOI: 10.1016/j.bpj.2017.03.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 02/28/2017] [Accepted: 03/01/2017] [Indexed: 02/07/2023] Open
Abstract
In closed system models of fibrin formation, exosite-mediated thrombin binding to fibrin contributes to clot stability and is resistant to inhibition by antithrombin/heparin while still susceptible to small, active-site inhibitors. Each molecule of fibrin can bind ∼1.6 thrombin molecules at low-affinity binding sites (Kd = 2.8 μM) and ∼0.3 molecules of thrombin at high-affinity binding sites (Kd = 0.15 μM). The goal of this study is to assess the stability of fibrin-bound thrombin under venous flow conditions and to determine both its accessibility and susceptibility to inhibition. A parallel-plate flow chamber (7 × 50 × 0.25 mm) for studying the stability of thrombin (0-1400 nM) adhered to a fibrin matrix (0.1-0.4 mg/mL fibrinogen, 10 nM thrombin) under a variety of venous flow conditions was developed using the thrombin-specific, fluorogenic substrate SN-59 (100 μM). The flow within this system is laminar (Re < 1) and reaction rates are driven by enzyme kinetics (Pe = 100, Da = 7000). A subpopulation of active thrombin remains stably adhered to a fibrin matrix over a range of venous shear rates (46-184 s-1) for upwards of 30 min, and this population is saturable at loads >500 nM and sensitive to the initial fibrinogen concentration. These observations were also supported by a mathematical model of thrombin adhesion to fibrin, which demonstrates that thrombin initially binds to the low-affinity thrombin binding sites before preferentially equilibrating to higher affinity sites. Antithrombin (2.6 μM) plus heparin (4 U/mL) inhibits 72% of the active clot-bound thrombin after ∼10 min at 92 s-1, while no inhibition is observed in the absence of heparin. Dabigatran (20 and 200 nM) inhibits (50 and 93%) clot-bound thrombin reversibly (87 and 66% recovery). This model illustrates that clot-bound thrombin stability is the result of a constant rearrangement of thrombin molecules within a dense matrix of binding sites.
Collapse
Affiliation(s)
- Laura M Haynes
- Department of Biochemistry, Robert Larner M.D. College of Medicine, University of Vermont, Colchester, Vermont
| | - Thomas Orfeo
- Department of Biochemistry, Robert Larner M.D. College of Medicine, University of Vermont, Colchester, Vermont
| | | | - Stephen J Everse
- Department of Biochemistry, Robert Larner M.D. College of Medicine, University of Vermont, Colchester, Vermont
| | - Kathleen E Brummel-Ziedins
- Department of Biochemistry, Robert Larner M.D. College of Medicine, University of Vermont, Colchester, Vermont.
| |
Collapse
|
35
|
|
36
|
Elizondo P, Fogelson AL. A Mathematical Model of Venous Thrombosis Initiation. Biophys J 2017; 111:2722-2734. [PMID: 28002748 DOI: 10.1016/j.bpj.2016.10.030] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Revised: 09/23/2016] [Accepted: 10/17/2016] [Indexed: 01/28/2023] Open
Abstract
We present a mathematical model for the initiation of venous thrombosis (VT) due to slow flow and the consequent activation of the endothelial cells (ECs) lining the vein, in the absence of overt mechanical disruption of the EC layer. It includes all reactions of the tissue factor (TF) pathway of coagulation through fibrin formation, incorporates the accumulation of blood cells on activated ECs, accounts for the flow-mediated delivery and removal of coagulation proteins and blood cells from the locus of the reactions, and accounts for the activity of major inhibitors including heparan-sulfate-accelerated antithrombin and activated protein C. The model reveals that the occurrence of robust thrombin generation (a thrombin burst) depends in a threshold manner on the density of TF on the activated ECs and on the concentration of thrombomodulin and the degree of heparan-sulfate accelerated antithrombin activity on those cells. Small changes in any of these in appropriate narrow ranges switches the response between "no burst" and "burst." The model predicts synergies among the inhibitors, both in terms of each inhibitor's multiple targets, and in terms of interactions between the different inhibitors. The model strongly suggests that the rate and extent of accumulation of activated monocytes, platelets, and MPs that can support the coagulation reactions has a powerful influence on whether a thrombin burst occurs and the thrombin response when it does. The slow rate of accumulation of cells supporting coagulation is one reason that the progress of VT is so much slower than that of arterial thrombosis initiated by subendothelial exposure.
Collapse
Affiliation(s)
| | - Aaron L Fogelson
- Department of Mathematics, University of Utah, Salt Lake City, Utah; Department of Bioengineering, University of Utah, Salt Lake City, Utah.
| |
Collapse
|
37
|
Jagadeeswaran P, Cooley BC, Gross PL, Mackman N. Animal Models of Thrombosis From Zebrafish to Nonhuman Primates: Use in the Elucidation of New Pathologic Pathways and the Development of Antithrombotic Drugs. Circ Res 2017; 118:1363-79. [PMID: 27126647 DOI: 10.1161/circresaha.115.306823] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Accepted: 11/30/2015] [Indexed: 12/23/2022]
Abstract
Thrombosis is a leading cause of morbidity and mortality worldwide. Animal models are used to understand the pathological pathways involved in thrombosis and to test the efficacy and safety of new antithrombotic drugs. In this review, we will first describe the central role a variety of animal models of thrombosis and hemostasis has played in the development of new antiplatelet and anticoagulant drugs. These include the widely used P2Y12 antagonists and the recently developed orally available anticoagulants that directly target factor Xa or thrombin. Next, we will describe the new players, such as polyphosphate, neutrophil extracellular traps, and microparticles, which have been shown to contribute to thrombosis in mouse models, particularly venous thrombosis models. Other mouse studies have demonstrated roles for the factor XIIa and factor XIa in thrombosis. This has spurred the development of strategies to reduce their levels or activities as a new approach for preventing thrombosis. Finally, we will discuss the emergence of zebrafish as a model to study thrombosis and its potential use in the discovery of novel factors involved in thrombosis and hemostasis. Animal models of thrombosis from zebrafish to nonhuman primates are vital in identifying pathological pathways of thrombosis that can be safely targeted with a minimal effect on hemostasis. Future studies should focus on understanding the different triggers of thrombosis and the best drugs to prevent each type of thrombotic event.
Collapse
Affiliation(s)
- Pudur Jagadeeswaran
- From the Department of Biological Sciences, University of North Texas, Denton (P.J.); Department of Pathology and Laboratory Medicine (B.C.C.), and Department of Medicine (N.M.), University of North Carolina, Chapel Hill; and Department of Medicine, McMaster University, Hamilton, Ontario, Canada (P.L.G.).
| | - Brian C Cooley
- From the Department of Biological Sciences, University of North Texas, Denton (P.J.); Department of Pathology and Laboratory Medicine (B.C.C.), and Department of Medicine (N.M.), University of North Carolina, Chapel Hill; and Department of Medicine, McMaster University, Hamilton, Ontario, Canada (P.L.G.)
| | - Peter L Gross
- From the Department of Biological Sciences, University of North Texas, Denton (P.J.); Department of Pathology and Laboratory Medicine (B.C.C.), and Department of Medicine (N.M.), University of North Carolina, Chapel Hill; and Department of Medicine, McMaster University, Hamilton, Ontario, Canada (P.L.G.)
| | - Nigel Mackman
- From the Department of Biological Sciences, University of North Texas, Denton (P.J.); Department of Pathology and Laboratory Medicine (B.C.C.), and Department of Medicine (N.M.), University of North Carolina, Chapel Hill; and Department of Medicine, McMaster University, Hamilton, Ontario, Canada (P.L.G.)
| |
Collapse
|
38
|
Byrnes JR, Wolberg AS. New findings on venous thrombogenesis. Hamostaseologie 2017; 37:25-35. [PMID: 27878206 PMCID: PMC5680039 DOI: 10.5482/hamo-16-09-0034] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 11/04/2016] [Indexed: 12/22/2022] Open
Abstract
Venous thrombosis (VT) is the third most common cause of cardiovascular death worldwide. Complications from VT and pulmonary embolism are the leading cause of lost disability-adjusted life years. Risks include genetic (e.g., non-O blood group, activated protein C resistance, hyperprothrombinemia) and acquired (e.g., age, surgery, cancer, pregnancy, immobilisation, female hormone use) factors. Pathophysiologic mechanisms that promote VT are incompletely understood, but involve abnormalities in blood coagulability, vessel function, and flow (so-called Virchow's Triad). Epidemiologic studies of humans, animal models, and biochemical and biophysical investigations have revealed contributions from extrinsic, intrinsic, and common pathways of coagulation, endothelial cells, leukocytes, red blood cells, platelets, cell-derived microvesicles, stasis-induced changes in vascular cells, and blood rheology. Knowledge of these mechanisms may yield new therapeutic targets. Characterisation of mechanisms that mediate VT formation and stability, particularly in aging, are needed to advance understanding of VT.
Collapse
Affiliation(s)
| | - Alisa S Wolberg
- Alisa S. Wolberg, Ph. D., Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, 819 Brinkhous-Bullitt Building, CB #7525, Chapel Hill, NC 27599-7525, United States, Phone: (919) 962-8943, Fax: (919) 966-6718, E-Mail:
| |
Collapse
|
39
|
Xu D, Xu R, He L, Xu T, Zhang Z, Han D, Du J. Comparison of Pathogenic Mechanisms Underlying Single and Recurrent Venous Thromboembolism Based on Gene Expression Profiling. Ann Vasc Surg 2016; 36:252-259. [PMID: 27423718 DOI: 10.1016/j.avsg.2016.05.088] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Revised: 05/16/2016] [Accepted: 05/23/2016] [Indexed: 11/19/2022]
Abstract
BACKGROUND Unprovoked venous thromboembolism (VTE), generally divided into single and recurrent categories, is a common leading cause of morbidity and mortality in a real-world population. This study was aimed to explore the similarities and differences in the mechanisms of single and recurrent VTE. METHODS Gene expression data (GSE19151) generated from 63 healthy controls, 32 single, and 38 recurrent VTE patients were analyzed. Differentially expressed genes (DEGs) were screened by Affy package and Kyoto Encyclopedia of Genes and Genomes and Gene Ontology enrichment analysis were performed using database for annotation, visualization, and integrated discovery. Based on the Search Tool for the Retrieval of Interacting Genes/Proteins database, protein-protein interaction network was visualized by Cytoscape, and modules were identified by CFinder. Finally, transcription factor regulatory networks were constructed. RESULTS Totally, 559 and 294 DEGs were obtained from recurrent and single VTE, respectively. There were 202 upregulated and 58 downregulated genes overlapped between them. Terms of regulation of actin cytoskeleton enriched by downregulated genes and oxidative phosphorylation enriched by upregulated genes were found in 2 types of VTE. Leukocyte transendothelial migration and Jak-STAT signaling pathway were found related with recurrent VTE. In addition, genes including signal transducer and activator of transcription 3 (STAT3) involving in the Jak-STAT signaling pathway were highly connected nodes. CONCLUSIONS Actin cytoskeleton and oxidative phosphorylation may be involved in the common mechanisms of recurrent VTE and single VTE. Leukocyte migration and Jak-STAT signaling pathway and their related genes may be important for the development and recurrence of VTE.
Collapse
Affiliation(s)
- Dejun Xu
- Department of Vascular and Lymphatic Surgery, Vascular and Lymphatic Surgery, China Japan Union Hospital of Jilin University, Changchun, Jilin, PR China
| | - Rutao Xu
- Department of Vascular Surgery, Henan Provincial People's Hospital, Zhengzhou, Henan, PR China
| | - Liu He
- Department of Anesthesiology, China Japan Union Hospital of Jilin University, Changchun, Jilin, PR China
| | - Tao Xu
- Department of Gastroenterology, The Fourth Clinical Hospital of Jilin University, Changchun, Jilin, PR China
| | - Zhenyu Zhang
- The Second Department of General Surgery, Armed Police Corps Hospital in Jilin Province, Changchun, Jilin, PR China
| | - Dongmei Han
- Department of Vascular and Lymphatic Surgery, Vascular and Lymphatic Surgery, China Japan Union Hospital of Jilin University, Changchun, Jilin, PR China
| | - Jianshi Du
- Department of Vascular and Lymphatic Surgery, Vascular and Lymphatic Surgery, China Japan Union Hospital of Jilin University, Changchun, Jilin, PR China.
| |
Collapse
|
40
|
Cote DJ, Smith TR. Venous thromboembolism in brain tumor patients. J Clin Neurosci 2016; 25:13-8. [DOI: 10.1016/j.jocn.2015.05.053] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Revised: 05/15/2015] [Accepted: 05/22/2015] [Indexed: 10/22/2022]
|
41
|
Cooley BC, Schmeling G. Murine model of large-vein electrolytic injury induction of thrombosis with slow resolution. Thromb Res 2016; 140:149-152. [PMID: 26860966 DOI: 10.1016/j.thromres.2016.01.023] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2015] [Revised: 01/17/2016] [Accepted: 01/27/2016] [Indexed: 10/22/2022]
Abstract
Deep vein thrombosis (DVT) and its sequela, pulmonary embolism, occur at a rate of 1 per 1000 person/year. Experimental models for evaluation of DVT have many short-comings, such as mechanical occlusion or stenosis to cause thrombosis, rather than the clinical scenario of thrombosis causing occlusion/stenosis. The goal of this study was to develop a model of flow-based large-vein thrombosis with resistance to resolution, to model clinical DVT behavior. Adult male C57Bl/6 mice underwent thrombus induction via an electrolytic injury to the femoral vein (3V positive current for 90s), with subsequent intra-vital fluorescence quantitation of platelet and fibrin accumulation through the first 60 min, and final histomorphometric volume evaluation at 1, 7, 14, and 28 days. Platelet accumulation at the injury site was comparable to a milder electrolytic injury, whereas fibrin was greatly augmented by 60 min in the more severe injury model. Thrombi showed persistent presence at 1 and 7 days, with remodeling to a stenotic fibrosis that encroached into the lumen at 14 and 28 days. The thrombotic/fibrotic volume within the femoral vein fell by 23% from 1 to 7 days, but had a residual presence at 28 days that was 31% the 1-day volume. This new model may provide an alternative approach to evaluating DVT persistence and therapeutic inhibition, to develop a better understanding of the clinical progression of DVT to thrombophlebitis.
Collapse
Affiliation(s)
- Brian C Cooley
- University of North Carolina at Chapel Hill, United States.
| | | |
Collapse
|
42
|
Venous Thromboembolism in Brain Tumor Patients. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 906:215-228. [DOI: 10.1007/5584_2016_117] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
|
43
|
Dydek EV, Chaikof EL. Simulated thrombin responses in venous valves. J Vasc Surg Venous Lymphat Disord 2015; 4:329-35. [PMID: 27318053 DOI: 10.1016/j.jvsv.2015.09.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Accepted: 09/15/2015] [Indexed: 10/22/2022]
Abstract
OBJECTIVE Venous thromboembolism frequently results in thrombi formation near or within the pocket of a venous valve due to recirculating hemodynamics, which has been largely attributed to hypoxia-induced tissue factor (TF) expression. Numerical models are now capable of assessing the spatiotemporal behavior of the TF-initiated coagulation cascade under nonuniform hemodynamics. The aim of this study was to use such a numerical simulation to analyze the degree and location of thrombin formation with respect to TF position in the presence of disturbed flow induced by an open venous valve. METHODS Thrombin formation was simulated using a computational model that captures the hemodynamics, kinetics, and chemical transport of 22 biochemical species. Disturbed flow is described by the presence of a valve in the equilibrium phase of the valve cycle with leaflets in a fully open position. Three different positions of TF downstream of the valve opening were investigated. RESULTS The critical amount of TF required to initiate a thrombotic response is reduced by up to 80% when it is positioned underneath the recirculating regions near the valve opening. In addition, because of the increased surface area of the open valve cusp in conjunction with recirculating hemodynamics, it was observed that thrombin is generated inside the valve pocket even when the exposed region of TF is downstream of the valve. CONCLUSIONS The presence of prothrombotic surface reactions in conjunction with recirculating hemodynamics provides an additional mechanism for thrombus formation in venous valves that does not require direct damage or dysfunction to the valve itself.
Collapse
Affiliation(s)
- E Victoria Dydek
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Mass
| | - Elliot L Chaikof
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Mass; Wyss Institute of Biologically Inspired Engineering of Harvard University, Boston, Mass; Division of Health Sciences Technology, Harvard-MIT, Cambridge, Mass.
| |
Collapse
|
44
|
Engbers MJ, Karasu A, Blom JW, Cushman M, Rosendaal FR, van Hylckama Vlieg A. Clinical features of venous insufficiency and the risk of venous thrombosis in older people. Br J Haematol 2015. [PMID: 26221838 DOI: 10.1111/bjh.13579] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Venous thrombosis is common in older age, with an incidence of 0·5-1% per year in those aged >70 years. Stasis of blood flow is an important contributor to the development of thrombosis and may be due to venous insufficiency in the legs. The risk of thrombosis associated with clinical features of venous insufficiency, i.e., varicose veins, leg ulcers and leg oedema, obtained with a standardized interview was assessed in the Age and Thrombosis Acquired and Genetic risk factors in the Elderly (AT-AGE) study. The AT-AGE study is a case-control study in individuals aged 70 years and older (401 cases with a first-time venous thrombosis and 431 control subjects). We calculated odds ratios (ORs) and corresponding 95% confidence intervals (CI) adjusted for age, sex and study centre. Varicose veins and leg ulcer were associated with a 1·6-fold (95% CI 1·2-2·3) and 3·3-fold increased risk of thrombosis (95% CI 1·6-6·7), respectively, while the risk was increased 3·0-fold (95% CI 2·1-4·5) in the presence of leg oedema. The risk of thrombosis was highest when all three risk factors occurred simultaneously (OR: 10·5; 95% CI 1·3-86·1). In conclusion, clinical features of venous insufficiency, i.e., varicose veins, leg ulcers and leg oedema, are risk factors for venous thrombosis in older people.
Collapse
Affiliation(s)
- Marissa J Engbers
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Alev Karasu
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Jeanet W Blom
- Department of Public Health and Primary Care, Leiden University Medical Center, Leiden, the Netherlands
| | - Mary Cushman
- Department of Medicine, University of Vermont Medical Centre, Burlington, VT, USA
| | - Frits R Rosendaal
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, the Netherlands.,Department of Thrombosis and Haemostasis, Leiden University Medical Center, Leiden, the Netherlands
| | | |
Collapse
|
45
|
Wolberg AS, Rosendaal FR, Weitz JI, Jaffer IH, Agnelli G, Baglin T, Mackman N. Venous thrombosis. Nat Rev Dis Primers 2015; 1:15006. [PMID: 27189130 DOI: 10.1038/nrdp.2015.6] [Citation(s) in RCA: 231] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Venous thromboembolism (VTE) encompasses deep-vein thrombosis (DVT) and pulmonary embolism. VTE is the leading cause of lost disability-adjusted life years and the third leading cause of cardiovascular death in the world. DVT leads to post-thrombotic syndrome, whereas pulmonary embolism can cause chronic pulmonary hypertension, both of which reduce quality of life. Genetic and acquired risk factors for thrombosis include non-O blood groups, factor V Leiden mutation, oral contraceptive use, hormone replacement therapy, advanced age, surgery, hospitalization and long-haul travel. A combination of blood stasis, plasma hypercoagulability and endothelial dysfunction is thought to trigger thrombosis, which starts most often in the valve pockets of large veins. Animal studies have revealed pathogenic roles for leukocytes, platelets, tissue factor-positive microvesicles, neutrophil extracellular traps and factors XI and XII. Diagnosis of VTE requires testing and exclusion of other pathologies, and typically involves laboratory measures (such as D-dimer) and diagnostic imaging. VTE is treated with anticoagulants and occasionally with thrombolytics to prevent thrombus extension and to reduce thrombus size. Anticoagulants are also used to reduce recurrence. New therapies with improved safety profiles are needed to prevent and treat venous thrombosis. For an illustrated summary of this Primer, visit: http://go.nature.com/8ZyCuY.
Collapse
Affiliation(s)
- Alisa S Wolberg
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, 819 Brinkhous-Bullitt Building, Chapel Hill, North Carolina 27599-7525, USA.,McAllister Heart Institute, University of North Carolina at Chapel Hill, USA
| | - Frits R Rosendaal
- Department of Clinical Epidemiology and Department of Thrombosis and Hemostasis, Leiden University Medical Center, The Netherlands.,K.G. Jensen Thrombosis Research and Expertise Center, University of Tromsø, Norway
| | - Jeffrey I Weitz
- Department of Medicine and Department of Biochemistry and Biomedical Sciences, McMaster University, and Thrombosis and Atherosclerosis Research Institute, Hamilton, Ontario, Canada
| | - Iqbal H Jaffer
- Department of Medicine and Department of Biochemistry and Biomedical Sciences, McMaster University, and Thrombosis and Atherosclerosis Research Institute, Hamilton, Ontario, Canada
| | - Giancarlo Agnelli
- Division of Internal and Cardiovascular Medicine, Stroke Unit, University of Perugia, Italy
| | - Trevor Baglin
- Department of Haematology, Addenbrooke's Hospital, Cambridge University Hospitals NHS Trust, Cambridge, UK
| | - Nigel Mackman
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, 819 Brinkhous-Bullitt Building, Chapel Hill, North Carolina 27599-7525, USA.,McAllister Heart Institute, University of North Carolina at Chapel Hill, USA.,K.G. Jensen Thrombosis Research and Expertise Center, University of Tromsø, Norway.,Department of Medicine, University of North Carolina at Chapel Hill, USA
| |
Collapse
|
46
|
Key NS, Connes P, Derebail VK. Negative health implications of sickle cell trait in high income countries: from the football field to the laboratory. Br J Haematol 2015; 170:5-14. [PMID: 25754217 DOI: 10.1111/bjh.13363] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Worldwide, sickle cell trait is a highly prevalent gene carrier state. While generally a benign condition with a normal life expectancy, it is becoming increasingly clear that the sickle trait is associated with certain adverse outcomes. This article will focus on three of these outcomes, namely exertional rhabdomyolysis and sudden death, chronic renal dysfunction, and venous thromboembolism. In each case, the epidemiological evidence for the association is reviewed, together with the existing data on potential underlying mechanisms. Because newborn screening programmes for sickle cell anaemia also identify those with sickle cell trait, it is imperative that further studies determine what, if any, preventive measures can be taken to reduce the burden of these uncommon but potentially morbid complications in affected individuals.
Collapse
Affiliation(s)
- Nigel S Key
- Division of Hematology/Oncology, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Philippe Connes
- Laboratoire CRIS EA647, Section 'Vascular Biology and Red Blood Cell', Université Claude Bernard Lyon 1, Lyon, France.,Institut Universitaire de France, Paris, France
| | - Vimal K Derebail
- Division of Nephrology, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
47
|
Kumar NG, Clark A, Roztocil E, Caliste X, Gillespie DL, Cullen JP. Fibrinolytic activity of endothelial cells from different venous beds. J Surg Res 2014; 194:297-303. [PMID: 25481530 DOI: 10.1016/j.jss.2014.09.028] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Revised: 09/05/2014] [Accepted: 09/23/2014] [Indexed: 10/24/2022]
Abstract
BACKGROUND Little is known about the molecular biology of endothelial cells from different venous vascular beds. As a result, our treatment of deep vein thrombosis and pulmonary artery embolism remain identical. As an initial step in understanding venous thromboembolic disease in the trauma and surgical patients, this study sought to investigate the balance between coagulation and fibrinolysis in the pulmonary and deep venous vascular beds and how trauma might influence this balance. MATERIALS AND METHODS Confluent human iliac vein endothelial cells (HIVECs) and human pulmonary artery endothelial cells (HPAECs), were cultured in the absence or presence of tumor necrosis factor (TNFα; 10 ng/mL) for 24 h. The expression of mediators of coagulation and fibrinolysis were determined by Western blot analysis, and plasminogen activator activity was determined by a fibrin clot degradation assay. RESULTS After TNFα stimulation, there was decreased expression of endothelial protein C receptor and thrombomodulin in both HIVECs and HPAECs. TNFα stimulation increased urokinase plasminogen activator expression in both HIVECs and HPAECs. There was an increase in the expression of tissue plasminogen activator and plasminogen activator inhibitor-1 in response to TNFα in HPAECs, but not in HIVECs. There was significantly greater clot degradation in the presence of both the conditioned media and cell extracts from HIVECs, when compared with HPAECs. CONCLUSIONS HPAECs and HIVECs react differently in terms of fibrinolytic potential when challenged with a cytokine associated with inflammation. These findings suggest that endothelial cells from distinct venous vascular beds may differentially regulate the fibrinolytic pathway.
Collapse
Affiliation(s)
- Neil G Kumar
- Division of Vascular Surgery, Department of Surgery, University of Rochester Medical Center, Rochester, New York
| | - Amanda Clark
- Division of Vascular Surgery, Department of Surgery, University of Rochester Medical Center, Rochester, New York
| | - Elisa Roztocil
- Division of Vascular Surgery, Department of Surgery, University of Rochester Medical Center, Rochester, New York
| | - Xzabia Caliste
- Division of Vascular Surgery, Department of Surgery, University of Rochester Medical Center, Rochester, New York
| | - David L Gillespie
- Division of Vascular Surgery, Department of Surgery, University of Rochester Medical Center, Rochester, New York
| | - John P Cullen
- Division of Vascular Surgery, Department of Surgery, University of Rochester Medical Center, Rochester, New York.
| |
Collapse
|
48
|
Saliba W, Zahalka W, Goldstein L, Ron G, Elias M. Padua prediction score and thrombin generation in hospitalized medical patients. Thromb Res 2014; 134:803-6. [PMID: 25087886 DOI: 10.1016/j.thromres.2014.07.022] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Revised: 07/12/2014] [Accepted: 07/14/2014] [Indexed: 11/24/2022]
Abstract
INTRODUCTION The Padua prediction score is a risk assessment model used to identify medical patients at high risk for venous thromboembolim (VTE).We aimed to assess the relationship between the severity of Padua score and thrombin generation as a measure of overall thrombotic activity. MATERIALS AND METHODS A total of 253 patients hospitalized in the medical wards, at the Haemek Medical Center, Israel, were enrolled in the study. Patients treated with anticoagulation, and those admitted for VTE were excluded. Padua score was classified into two categories; low-risk for VTE (<4 points), and high-risk for VTE (≥4 points). Thrombin generation was assessed by the Calibrated Automated Thrombogram (CAT) method. RESULTS Overall 187 (73.9%) patients had Padua score<4, and 66 (26.1%) patients had Padua score ≥4. Comparison of the thrombogram parameters between the two Padua score categories showed no significant difference; lag time (P=0.066), ETP (P=0.266), peak height (P=0.418), and time to peak (P=0.415). Among the individual Padua score risk factors, only active cancer was significantly associated with peak height, myocardial infarction or stroke with lag time, and none of the risk factors was significantly associated with ETP. Because of their low frequency, the association with previous VTE, known thrombophilia, hormonal treatment, and recent trauma or/and surgery was not assessed. CONCLUSIONS Single thrombin generation measurement obtained at the same time in acutely hospitalized patients didn't bear any correlation with the Padua prediction score. This finding should be interpreted with caution considering the underrepresentation of risk factors that may influence thrombin generation.
Collapse
Affiliation(s)
- Walid Saliba
- Department of Internal Medicine C, Ha'emek Medical Center, Afula, Israel; Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel.
| | - Wael Zahalka
- Department of Internal Medicine A, Ha'emek Medical Center, Afula, Israel
| | - Lee Goldstein
- Department of Internal Medicine C, Ha'emek Medical Center, Afula, Israel; Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel; Clinical Pharmacology Unit, Ha'emek Medical Center, Afula, Israel
| | - Gilat Ron
- Department of Internal Medicine C, Ha'emek Medical Center, Afula, Israel
| | - Mazen Elias
- Department of Internal Medicine C, Ha'emek Medical Center, Afula, Israel; Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
49
|
Costanzo L, Di Pino L, Ragusa M, Buccheri S, Sole A, Virgilio V, Tamburino C, Bellanca S. Prevalence of asymptomatic lower limb venous thrombosis in infertile women with thrombophilic disorders. Phlebology 2014; 30:449-54. [PMID: 24906906 DOI: 10.1177/0268355514539317] [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: 11/16/2022]
Abstract
OBJECTIVE We sought to assess the prevalence of asymptomatic venous thrombosis in infertile women with thrombophilic disorders (TDs). METHODS AND RESULTS A total of 73 infertile women with TDs underwent duplex ultrasound scan to evaluate superficial and deep venous circulation of lower limbs. A control group of 35 infertile women without TDs was included. A single TD was found in 13 (17.8%) subjects, and 40 (54.8%) women presented a combined defect (more than three alterations). No residual mural thrombosis (RT) was noted in any deep veins. We found RT in 48 (65.8%) patients of TD group, while no RT was found in the control group (p < 0.0001). None of the clinical and prothrombotic factors were predictors of RT (all p > 0.20), and frequency of TD did not correlate with multi-vessel RT (p = 0.252). CONCLUSIONS No signs of deep vein thrombosis but high prevalence of superficial RT is present in infertile women with TDs. Further studies are needed to assess the prognostic value of our findings.
Collapse
Affiliation(s)
- Luca Costanzo
- Cardiotoracovascular Department, Division of Angiology, Ferrarotto-Policlinic Hospital, University of Catania, Catania, Italy
| | - Luigi Di Pino
- Cardiotoracovascular Department, Division of Angiology, Ferrarotto-Policlinic Hospital, University of Catania, Catania, Italy
| | - Mario Ragusa
- Cardiotoracovascular Department, Division of Angiology, Ferrarotto-Policlinic Hospital, University of Catania, Catania, Italy
| | - Sergio Buccheri
- Cardiotoracovascular Department, Division of Angiology, Ferrarotto-Policlinic Hospital, University of Catania, Catania, Italy
| | - Andrea Sole
- Cardiotoracovascular Department, Division of Angiology, Ferrarotto-Policlinic Hospital, University of Catania, Catania, Italy
| | | | - Corrado Tamburino
- Cardiotoracovascular Department, Division of Angiology, Ferrarotto-Policlinic Hospital, University of Catania, Catania, Italy
| | - Salvatore Bellanca
- Conjugal Sterility Center, Policlinic Hospital, University of Catania, Catania, Italy
| |
Collapse
|
50
|
Linnemann B, Weingarz L, Schindewolf M, Schwonberg J, Weber A, Herrmann E, Lindhoff-Last E. Prevalence of established risk factors for venous thromboembolism according to age. J Vasc Surg Venous Lymphat Disord 2014; 2:131-9. [DOI: 10.1016/j.jvsv.2013.09.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2013] [Revised: 09/03/2013] [Accepted: 09/13/2013] [Indexed: 12/21/2022]
|