1
|
Liu Y, Huang T, Yap NA, Lim K, Ju LA. Harnessing the power of bioprinting for the development of next-generation models of thrombosis. Bioact Mater 2024; 42:328-344. [PMID: 39295733 PMCID: PMC11408160 DOI: 10.1016/j.bioactmat.2024.08.040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Revised: 08/07/2024] [Accepted: 08/29/2024] [Indexed: 09/21/2024] Open
Abstract
Thrombosis, a leading cause of cardiovascular morbidity and mortality, involves the formation of blood clots within blood vessels. Current animal models and in vitro systems have limitations in recapitulating the complex human vasculature and hemodynamic conditions, limiting the research in understanding the mechanisms of thrombosis. Bioprinting has emerged as a promising approach to construct biomimetic vascular models that closely mimic the structural and mechanical properties of native blood vessels. This review discusses the key considerations for designing bioprinted vascular conduits for thrombosis studies, including the incorporation of key structural, biochemical and mechanical features, the selection of appropriate biomaterials and cell sources, and the challenges and future directions in the field. The advancements in bioprinting techniques, such as multi-material bioprinting and microfluidic integration, have enabled the development of physiologically relevant models of thrombosis. The future of bioprinted models of thrombosis lies in the integration of patient-specific data, real-time monitoring technologies, and advanced microfluidic platforms, paving the way for personalized medicine and targeted interventions. As the field of bioprinting continues to evolve, these advanced vascular models are expected to play an increasingly important role in unraveling the complexities of thrombosis and improving patient outcomes. The continued advancements in bioprinting technologies and the collaboration between researchers from various disciplines hold great promise for revolutionizing the field of thrombosis research.
Collapse
Affiliation(s)
- Yanyan Liu
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW, 2008, Australia
| | - Tao Huang
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW, 2008, Australia
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW 2006, Australia
| | - Nicole Alexis Yap
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW, 2008, Australia
| | - Khoon Lim
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW 2006, Australia
- School of Medical Sciences, The University of Sydney, Darlington, NSW 2008, Australia
- The University of Sydney Nano Institute (Sydney Nano), The University of Sydney, Camperdown, NSW, 2006, Australia
| | - Lining Arnold Ju
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW, 2008, Australia
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW 2006, Australia
- The University of Sydney Nano Institute (Sydney Nano), The University of Sydney, Camperdown, NSW, 2006, Australia
- Heart Research Institute, Camperdown, Newtown, NSW 2042, Australia
| |
Collapse
|
2
|
Kapteijn MY, Yanovska M, Laghmani EH, Postma RJ, van Duinen V, Ünlü B, Queiroz K, van Zonneveld AJ, Versteeg HH, Rondon AMR. Modeling cancer-associated hypercoagulability using glioblastoma spheroids in microfluidic chips. Res Pract Thromb Haemost 2024; 8:102475. [PMID: 39268353 PMCID: PMC11391032 DOI: 10.1016/j.rpth.2024.102475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 05/02/2024] [Accepted: 06/11/2024] [Indexed: 09/15/2024] Open
Abstract
Background Cancer increases the risk of venous thromboembolism, and glioblastoma is one of the cancer types with the highest risk of venous thromboembolism (10%-30%). Tumor-intrinsic features are believed to affect vascular permeability and hypercoagulability, but novel models are required to study the pathophysiological dynamics underlying cancer-associated thrombosis at the molecular level. Objectives We have developed a novel cancer-on-a-chip model to examine the effects of glioblastoma cells on the deregulation of blood coagulation. Methods This was accomplished by coculturing vessel-forming human umbilical vein endothelial cells with glioblastoma spheroids overexpressing tissue factor (TF), the initiator of coagulation (U251 lentivirus, LV-TF) or an LV-control (U251 LV-Ctrl) in an OrganoPlate Graft platform. Results Using a modified thrombin generation assay inside the cancer-on-a-chip, we found that U251 LV-Ctrl and U251 LV-TF spheroids promoted an increased procoagulant state in plasma, as was shown by a 3.1- and 7.0-fold increase in endogenous thrombin potential, respectively. Furthermore, the anticoagulant drug rivaroxaban and TF coagulation-blocking antibody 5G9 inhibited the activation of blood coagulation in U251 LV-TF spheroid-containing graft plates, as was shown by a reduced endogenous thrombin potential (4.0- and 4.4-fold, respectively). Conclusion With this study, we present a novel 3-dimensional cancer-on-a-chip model that has the potential to be used in the discovery of new anticoagulant drugs and identification of optimal anticoagulant strategies for glioblastoma and other cancer types.
Collapse
Affiliation(s)
- Maaike Y Kapteijn
- Division of Thrombosis and Hemostasis, Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Monika Yanovska
- Division of Thrombosis and Hemostasis, Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - El Houari Laghmani
- Division of Thrombosis and Hemostasis, Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Rudmer J Postma
- Division of Nephrology, Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Vincent van Duinen
- Division of Nephrology, Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, the Netherlands
- Mimetas BV, Oegstgeest, the Netherlands
| | - Betül Ünlü
- Division of Thrombosis and Hemostasis, Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | | | - Anton Jan van Zonneveld
- Division of Nephrology, Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Henri H Versteeg
- Division of Thrombosis and Hemostasis, Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Araci M R Rondon
- Division of Thrombosis and Hemostasis, Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
3
|
Wang M, Lu X, Han L, Diaz JA, Raju S, Kassab GS. Venous thromboembolism swine model with reflux-induced venous hypertension. JVS Vasc Sci 2024; 5:100200. [PMID: 38766270 PMCID: PMC11101933 DOI: 10.1016/j.jvssci.2024.100200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 02/16/2024] [Indexed: 05/22/2024] Open
Abstract
Objective This study describes a novel swine model of venous thromboembolism (VTE) with reflux-induced venous hypertension. Methods Six pigs underwent disruption of the tricuspid chordae tendineae to create reflux and venous hypertension in the femoral vein. The vein was traumatized 2 to 3 weeks later by repeated withdrawal of a slightly overinflated occlusion balloon across the lumen, followed by balloon occlusion of the outflow. A small amount of thrombin was injected into the traumatized vein segment immediately after outflow occlusion. Thrombosis of the traumatized vein evolved into an organized thrombus seven weeks later. The histological features of the harvested post-thrombotic femoral vein were studied with hematoxylin and eosin and Trichrome stains. Results In all six pigs, initial disruption of the chordae tendineae was successfully performed to create tricuspid reflux and venous hypertension. After two-stage sequential procedures, a thrombus formed in the target femoral vein segment. Histology of the harvested thrombotic vein showed features of an organizing thrombus with collagen formation and fibrosis. Conclusions The novel swine VTE model may serve as a platform for developing and testing human-sized therapeutic procedures and devices in translational venous research. Clinical Relevance This study describes a swine model of VTE created by incorporating all three elements of Virchow's triad. The model uniquely incorporates reflux-induced venous hypertension, which may be used in studying venous insufficiency and VTE in those with systemic venous hypertension. Likewise, this model may serve as a platform for development and evaluation of diagnostic imaging or therapeutic procedures and devices in subjects with systemic venous hypertension.
Collapse
Affiliation(s)
| | - Xiao Lu
- California Medical Innovations Institute, San Diego, CA
| | - Ling Han
- California Medical Innovations Institute, San Diego, CA
| | - José A. Diaz
- Surgical Research Division, Vanderbilt University Medical Center, Nashville, TN
| | - Seshadri Raju
- The Rane Center at St. Dominic's Hospital, Jackson, MS
| | | |
Collapse
|
4
|
Roberts SH, Zaghloul MS, Ismail U, Rowe RA, Engel C, Meade R, Elizondo-Benedetto S, Genin GM, Zayed MA. In Vivo Porcine Model of Acute Iliocaval Deep Vein Thrombosis. J Endovasc Ther 2024:15266028241231513. [PMID: 38357736 DOI: 10.1177/15266028241231513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2024]
Abstract
CLINICAL IMPACT The study establishes a rapid, technically straightforward, and reproducible porcine large animal model for acute iliocaval deep vein thrombosis (DVT). The procedure can be performed with basic endovascular skillsets. With its procedural efficiency and consistency, the platform is promising for comparative in vivo testing of venous thrombectomy devices in a living host, and for future verification and validation studies to determine efficacy of novel thrombectomy devices relative to predicates.
Collapse
Affiliation(s)
- Sophia H Roberts
- Section of Vascular Surgery, Department of Surgery, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
- Cardiovascular Research Innovation in Surgery and Engineering Center, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Mohamed S Zaghloul
- Section of Vascular Surgery, Department of Surgery, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
- Cardiovascular Research Innovation in Surgery and Engineering Center, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | | | | | - Connor Engel
- Section of Vascular Surgery, Department of Surgery, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
- Cardiovascular Research Innovation in Surgery and Engineering Center, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Rodrigo Meade
- Section of Vascular Surgery, Department of Surgery, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
- Cardiovascular Research Innovation in Surgery and Engineering Center, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Santiago Elizondo-Benedetto
- Section of Vascular Surgery, Department of Surgery, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
- Cardiovascular Research Innovation in Surgery and Engineering Center, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Guy M Genin
- Cardiovascular Research Innovation in Surgery and Engineering Center, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
- Caeli Vascular, Inc., St. Louis, MO, USA
- Department of Biomedical Engineering, McKelvey School of Engineering, Washington University in St. Louis, St. Louis, MO, USA
- NSF Science and Technology Center for Engineering Mechanobiology, McKelvey School of Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - Mohamed A Zayed
- Section of Vascular Surgery, Department of Surgery, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
- Cardiovascular Research Innovation in Surgery and Engineering Center, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
- Caeli Vascular, Inc., St. Louis, MO, USA
- Department of Biomedical Engineering, McKelvey School of Engineering, Washington University in St. Louis, St. Louis, MO, USA
- Division of Molecular Cell Biology, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
- Department of Radiology, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| |
Collapse
|
5
|
Samridhi, Setia A, Mehata AK, Priya V, Pradhan A, Prasanna P, Mohan S, Muthu MS. Nanoparticles for Thrombus Diagnosis and Therapy: Emerging Trends in Thrombus-theranostics. Nanotheranostics 2024; 8:127-149. [PMID: 38328614 PMCID: PMC10845253 DOI: 10.7150/ntno.92184] [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: 11/13/2023] [Accepted: 12/09/2023] [Indexed: 02/09/2024] Open
Abstract
Cardiovascular disease is one of the chief factors that cause ischemic stroke, myocardial infarction, and venous thromboembolism. The elements that speed up thrombosis include nutritional consumption, physical activity, and oxidative stress. Even though the precise etiology and pathophysiology remain difficult topics that primarily rely on traditional medicine. The diagnosis and management of thrombosis are being developed using discrete non-invasive and non-surgical approaches. One of the emerging promising approach is ultrasound and photoacoustic imaging. The advancement of nanomedicines offers concentrated therapy and diagnosis, imparting efficacy and fewer side effects which is more significant than conventional medicine. This study addresses the potential of nanomedicines as theranostic agents for the treatment of thrombosis. In this article, we describe the factors that lead to thrombosis and its consequences, as well as summarize the findings of studies on thrombus formation in preclinical and clinical models and also provide insights on nanoparticles for thrombus imaging and therapy.
Collapse
Affiliation(s)
- Samridhi
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi-221005, India
| | - Aseem Setia
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi-221005, India
| | - Abhishesh Kumar Mehata
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi-221005, India
| | - Vishnu Priya
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi-221005, India
| | - Aditi Pradhan
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi-221005, India
| | - Pragya Prasanna
- National Institute of Pharmaceutical Education and Research, Hajipur, Bihar, India
| | - Syam Mohan
- Substance Abuse and Toxicology Research Centre, Jazan University, Jazan 45142, Saudi Arabia
- School of Health Sciences, University of Petroleum and Energy Studies, Dehradun 248007, India
| | - Madaswamy S Muthu
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi-221005, India
| |
Collapse
|
6
|
Yokomori R, Shirai T, Tsukiji N, Oishi S, Sasaki T, Takano K, Suzuki-Inoue K. C-type lectin-like receptor-2 (CLEC-2) is a key regulator of kappa-carrageenan-induced tail thrombosis model in mice. Platelets 2023; 34:2281941. [PMID: 38010137 DOI: 10.1080/09537104.2023.2281941] [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: 05/18/2023] [Accepted: 11/03/2023] [Indexed: 11/29/2023]
Abstract
Kappa-carrageenan (KCG), which is used to induce thrombosis in laboratory animals for antithrombotic drug screening, can trigger platelet aggregation. However, the cell-surface receptor and related signaling pathways remain unclear. In this study, we investigated the molecular basis of KCG-induced platelet activation using light-transmittance aggregometry, flow cytometry, western blotting, and surface plasmon resonance assays using platelets from platelet receptor-deficient mice and recombinant proteins. KCG-induced tail thrombosis was also evaluated in mice lacking the platelet receptor. We found that KCG induces platelet aggregation with α-granule secretion, activated integrin αIIbβ3, and phosphatidylserine exposure. As this aggregation was significantly inhibited by the Src family kinase inhibitor and spleen tyrosine kinase (Syk) inhibitor, a tyrosine kinase-dependent pathway is required. Platelets exposed to KCG exhibited intracellular tyrosine phosphorylation of Syk, linker activated T cells, and phospholipase C gamma 2. KCG-induced platelet aggregation was abolished in platelets from C-type lectin-like receptor-2 (CLEC-2)-deficient mice, but not in platelets pre-treated with glycoprotein VI-blocking antibody, JAQ1. Surface plasmon resonance assays showed a direct association between murine/human recombinant CLEC-2 and KCG. KCG-induced thrombosis and thrombocytopenia were significantly inhibited in CLEC-2-deficient mice. Our findings show that KCG induces platelet activation via CLEC-2.
Collapse
Affiliation(s)
- Ryohei Yokomori
- Department of Clinical and Laboratory Medicine, Faculty of Medicine, University of Yamanashi, Kofu, Japan
| | - Toshiaki Shirai
- Department of Clinical and Laboratory Medicine, Faculty of Medicine, University of Yamanashi, Kofu, Japan
| | - Nagaharu Tsukiji
- Department of Clinical and Laboratory Medicine, Faculty of Medicine, University of Yamanashi, Kofu, Japan
| | - Saori Oishi
- Department of Clinical and Laboratory Medicine, Faculty of Medicine, University of Yamanashi, Kofu, Japan
| | - Tomoyuki Sasaki
- Department of Clinical and Laboratory Medicine, Faculty of Medicine, University of Yamanashi, Kofu, Japan
| | - Katsuhiro Takano
- Department of Transfusion and Cell Therapy, University of Yamanashi Hospital, Chuo, Japan
| | - Katsue Suzuki-Inoue
- Department of Clinical and Laboratory Medicine, Faculty of Medicine, University of Yamanashi, Kofu, Japan
- Department of Transfusion and Cell Therapy, University of Yamanashi Hospital, Chuo, Japan
| |
Collapse
|
7
|
Wang M, Lu X, Han L, Wang AM, Raju S, Kassab GS. Novel venous balloon for compliance measurement and stent sizing in a post-thrombotic swine model. Front Bioeng Biotechnol 2023; 11:1298621. [PMID: 38076433 PMCID: PMC10702604 DOI: 10.3389/fbioe.2023.1298621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 11/01/2023] [Indexed: 02/12/2024] Open
Abstract
Objective: Real-time accurate venous lesion characterization is needed during endovenous interventions for stent deployment. The goal of this study is to validate a novel device for venoplasty sizing and compliance measurements. Methods: A compliance measuring sizing balloon (CMSB) uses real-time electrical conductance measurements based on Ohm's Law to measure the venous size and compliance in conjunction with pressure measurement. The sizing accuracy and repeatability of the CMSB system were performed with phantoms on the bench and in a swine model with an induced post thrombotic (PT) stenosis in the common femoral vein of swine. Results: The accuracy and repeatability of the CMSB system were validated with phantom bench studies of known dimensions in the range of venous diameters. In 9 swine (6 experimental and 3 control animals), the luminal cross-sectional areas (CSA) increased heterogeneously along the PT stenosis when the CMSB system was inflated by stepwise pressures. The PT stenosis showed lower compliance compared to the non-PT vein segments (5 mm2 vs. 10 mm2 and 13 mm2 at a pressure change of 40 cm H2O). Compliance had no statistical difference between venous hypertension (VHT) and Control. Compliance at PT stenosis, however, was significantly smaller than that at Control and VHT (p < 0.05, ANOVA). Conclusion: The CMSB system provides accurate, repeatable, real-time measurements of CSA and compliance for assessment of venous lesions to guide interventions. These findings provide the impetus for future first-in-human studies.
Collapse
Affiliation(s)
| | - Xiao Lu
- California Medical Innovations Institute, San Diego, CA, United States
| | - Ling Han
- California Medical Innovations Institute, San Diego, CA, United States
| | - Amy M. Wang
- 3DT Holdings LLC, San Diego, CA, United States
| | - Seshadri Raju
- The Rane Center at St. Dominic’s Hospital, Jackson, MS, United States
| | - Ghassan S. Kassab
- California Medical Innovations Institute, San Diego, CA, United States
| |
Collapse
|
8
|
Fumadó Navarro J, Lomora M. Mechanoresponsive Drug Delivery Systems for Vascular Diseases. Macromol Biosci 2023; 23:e2200466. [PMID: 36670512 DOI: 10.1002/mabi.202200466] [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: 11/01/2022] [Revised: 01/16/2023] [Indexed: 01/22/2023]
Abstract
Mechanoresponsive drug delivery systems (DDS) have emerged as promising candidates to improve the current effectiveness and lower the side effects typically associated with direct drug administration in the context of vascular diseases. Despite tremendous research efforts to date, designing drug delivery systems able to respond to mechanical stimuli to potentially treat these diseases is still in its infancy. By understanding relevant biological forces emerging in healthy and pathological vascular endothelium, it is believed that better-informed design strategies can be deduced for the fabrication of simple-to-complex macromolecular assemblies capable of sensing mechanical forces. These responsive systems are discussed through insights into essential parameter design (composition, size, shape, and aggregation state) , as well as their functionalization with (macro)molecules that are intrinsically mechanoresponsive (e.g., mechanosensitive ion channels and mechanophores). Mechanical forces, including the pathological shear stress and exogenous stimuli (e.g., ultrasound, magnetic fields), used for the activation of mechanoresponsive DDS are also introduced, followed by in vitro and in vivo experimental models used to investigate and validate such novel therapies. Overall, this review aims to propose a fresh perspective through identified challenges and proposed solutions that could be of benefit for the further development of this exciting field.
Collapse
Affiliation(s)
- Josep Fumadó Navarro
- School of Biological and Chemical Sciences, University of Galway, University Road, Galway, H91 TK33, Ireland
- CÚRAM, SFI Research Centre for Medical Devices, University of Galway, Upper Newcastle, Galway, H91 W2TY, Ireland
| | - Mihai Lomora
- School of Biological and Chemical Sciences, University of Galway, University Road, Galway, H91 TK33, Ireland
- CÚRAM, SFI Research Centre for Medical Devices, University of Galway, Upper Newcastle, Galway, H91 W2TY, Ireland
| |
Collapse
|
9
|
Meijers JCM, Bakhtiari K, Zwiers A, Peters SLM. OKL-1111, A modified cyclodextrin as a potential universal reversal agent for anticoagulants. Thromb Res 2023; 227:17-24. [PMID: 37207560 DOI: 10.1016/j.thromres.2023.05.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 05/03/2023] [Accepted: 05/05/2023] [Indexed: 05/21/2023]
Abstract
BACKGROUND Antithrombotic therapy is inevitably associated with a risk for bleeding and these bleeding complications can be life-threatening. Recently, specific reversal agents were developed for the direct factor Xa and thrombin inhibitors (DOACs). However, next to the fact that these agents are relatively expensive, the use of selective reversal agents complicates treatment of bleeding patients in practice. In a series of screening experiments, we discovered a class of cyclodextrins with procoagulant properties. In this study we characterize a lead compound, OKL-1111, and demonstrate its potential use as a universal reversal agent. OBJECTIVES To assess the anticoagulant reversal properties of OKL-1111, in vitro and in vivo. METHODS The effect of OKL-1111 on coagulation in the absence and presence of DOACs was investigated in a thrombin generation assay. Its reversal effect on a variety of anticoagulants in vivo was investigated in a rat tail cut bleeding model. A possible prothrombotic action of OKL-1111 was assessed in a Wessler model in rabbits. RESULTS OKL-1111 concentration-dependently reversed the in vitro anticoagulant effects of dabigatran, rivaroxaban, apixaban and edoxaban in the thrombin generation assay. Also in the absence of a DOAC, OKL-1111 concentration-dependently accelerated coagulation in this assay, but did not initiate coagulation. The reversal effect was also seen for all DOACs in the rat tail cut bleeding model. In addition, when tested with other anticoagulants, OKL-1111 also reversed the anticoagulant effect of the vitamin K antagonist warfarin, the low molecular weight heparin enoxaparin, the pentasaccharide fondaparinux and the platelet inhibitor clopidogrel in vivo. OKL-1111 did not have prothrombotic effects in the Wessler model. CONCLUSION OKL-1111 is a procoagulant cyclodextrin with a currently unknown working mechanism that has potential to become a universal reversal agent for anticoagulants and platelet inhibitors.
Collapse
Affiliation(s)
- Joost C M Meijers
- Department of Molecular Hematology, Sanquin Research, Plesmanlaan 125, 1066 CX Amsterdam, the Netherlands; Amsterdam UMC, University of Amsterdam, Department of Experimental Vascular Medicine, Amsterdam Cardiovascular Sciences, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands.
| | - Kamran Bakhtiari
- Department of Molecular Hematology, Sanquin Research, Plesmanlaan 125, 1066 CX Amsterdam, the Netherlands
| | - Alex Zwiers
- Alveron Pharma BV, Gasstraat 20, 5349 AA Oss, the Netherlands
| | | |
Collapse
|
10
|
Madhani SI, Larco JA, Liu Y, Abbasi M, Shahid AH, Yasin O, Sears V, Morris J, Rosenthal M, Baron S, Savastano L. Construction and Validation of a Benchtop Model for Testing of Mechanical Thrombectomy Devices for Pulmonary Embolism. Cardiovasc Intervent Radiol 2023; 46:385-391. [PMID: 36482095 DOI: 10.1007/s00270-022-03326-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 11/19/2022] [Indexed: 12/14/2022]
Abstract
PURPOSE This study aims to define the process of designing and manufacturing 3D printed and glass models of the pulmonary artery (PA) and utilizing them in a test bed for evaluation of devices for mechanical thrombectomy of pulmonary embolism (PE). MATERIALS AND METHODS Patient derived computed tomography angiography (CTA) images of the PA were digitally converted into a hollowed-out structure and translated into clear 3D printed and glass models. A test bed was created using a peristaltic pump and silicone tubing connected to the models. Human clot analogs were then prepared and injected within the models. Thrombectomy testing was done using clinically used predicates and baseline characteristics of the models were evaluated by independent interventionalists. RESULTS The mean sizes of the main pulmonary artery (MPA) for the 3D printed model and glass model were 30.4 mm and 29.2 mm, mimicking those of the patient's PA obtained on CTA. Heterogeneous human clot analogs were created with fibrin composition ranging from 60 to 30%. Mechanical thrombectomy was successfully attempted by independent interventionalists. Both the 3D printed, and glass model were appraised as very good for multiple attributes. CONCLUSION A complete test bed using 3D printed and glass models of the PA with human clot analogs was created for testing of mechanical thrombectomy devices for PE.
Collapse
Affiliation(s)
| | | | - Yang Liu
- Global Institute of Future Technologies, Shanghai Jiao Tong University, Shanghai, China
| | - Mehdi Abbasi
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | | | - Omar Yasin
- Department of Cardiology, Mayo Clinic, Rochester, MN, USA
| | - Victoria Sears
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | | | | | - Scott Baron
- Endovascular Engineering, Menlo Park, CA, USA
| | - Luis Savastano
- Department of Neurosurgery, University of California, San Francisco, CA, USA.
| |
Collapse
|
11
|
Ayyoub S, Orriols R, Oliver E, Ceide OT. Thrombosis Models: An Overview of Common In Vivo and In Vitro Models of Thrombosis. Int J Mol Sci 2023; 24:2569. [PMID: 36768891 PMCID: PMC9917341 DOI: 10.3390/ijms24032569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 01/19/2023] [Accepted: 01/21/2023] [Indexed: 02/03/2023] Open
Abstract
Occlusions in the blood vessels caused by blood clots, referred to as thrombosis, and the subsequent outcomes are leading causes of morbidity and mortality worldwide. In vitro and in vivo models of thrombosis have advanced our understanding of the complex pathways involved in its development and allowed the evaluation of different therapeutic approaches for its management. This review summarizes different commonly used approaches to induce thrombosis in vivo and in vitro, without detailing the protocols for each technique or the mechanism of thrombus development. For ease of flow, a schematic illustration of the models mentioned in the review is shown below. Considering the number of available approaches, we emphasize the importance of standardizing thrombosis models in research per study aim and application, as different pathophysiological mechanisms are involved in each model, and they exert varying responses to the same carried tests. For the time being, the selection of the appropriate model depends on several factors, including the available settings and research facilities, the aim of the research and its application, and the researchers' experience and ability to perform surgical interventions if needed.
Collapse
Affiliation(s)
- Sana Ayyoub
- Department of Pulmonary Medicine, Dr. Josep Trueta University Hospital de Girona, Santa Caterina Hospital de Salt and the Girona Biomedical Research Institute (IDIBGI), 17190 Girona, Spain
| | - Ramon Orriols
- Department of Pulmonary Medicine, Dr. Josep Trueta University Hospital de Girona, Santa Caterina Hospital de Salt and the Girona Biomedical Research Institute (IDIBGI), 17190 Girona, Spain
| | - Eduardo Oliver
- Centro de Investigaciones Biologicas Margarita Salas (CIB-CSIC), 28040 Madrid, Spain
- Centro de Investigación en Red de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| | - Olga Tura Ceide
- Department of Pulmonary Medicine, Dr. Josep Trueta University Hospital de Girona, Santa Caterina Hospital de Salt and the Girona Biomedical Research Institute (IDIBGI), 17190 Girona, Spain
- Biomedical Research Networking Centre on Respiratory Diseases (CIBERES), 28029 Madrid, Spain
| |
Collapse
|
12
|
Gromadziński L, Paukszto Ł, Lepiarczyk E, Skowrońska A, Lipka A, Makowczenko KG, Łopieńska-Biernat E, Jastrzębski JP, Holak P, Smoliński M, Majewska M. Pulmonary artery embolism: comprehensive transcriptomic analysis in understanding the pathogenic mechanisms of the disease. BMC Genomics 2023; 24:10. [PMID: 36624378 PMCID: PMC9830730 DOI: 10.1186/s12864-023-09110-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Accepted: 01/02/2023] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Pulmonary embolism (PE) is a severe disease that usually originates from deep vein thrombosis (DVT) of the lower extremities. This study set out to investigate the changes in the transcriptome of the pulmonary artery (PA) in the course of the PE in the porcine model. METHODS The study was performed on 11 male pigs: a thrombus was formed in each right femoral vein in six animals, and then was released to induce PE, the remaining five animals served as a control group. In the experimental animals total RNA was isolated from the PA where the blood clot lodged, and in the control group, from the corresponding PA segments. High-throughput RNA sequencing was used to analyse the global changes in the transcriptome of PA with induced PE (PA-E). RESULTS Applied multistep bioinformatics revealed 473 differentially expressed genes (DEGs): 198 upregulated and 275 downregulated. Functional Gene Ontology annotated 347 DEGs into 27 biological processes, 324 to the 11 cellular components and 346 to the 2 molecular functions categories. In the signaling pathway analysis, KEGG 'protein processing in endoplasmic reticulum' was identified for the mRNAs modulated during PE. The same KEGG pathway was also exposed by 8 differentially alternative splicing genes. Within single nucleotide variants, the 61 allele-specific expression variants were localised in the vicinity of the genes that belong to the cellular components of the 'endoplasmic reticulum'. The discovered allele-specific genes were also classified as signatures of the cardiovascular system. CONCLUSIONS The findings of this research provide the first thorough investigation of the changes in the gene expression profile of PA affected by an embolus. Evidence from this study suggests that the disturbed homeostasis in the biosynthesis of proteins in the endoplasmic reticulum plays a major role in the pathogenesis of PE.
Collapse
Affiliation(s)
- Leszek Gromadziński
- grid.412607.60000 0001 2149 6795Department of Cardiology and Internal Medicine, School of Medicine, Collegium Medicum, University of Warmia and Mazury in Olsztyn, Warszawska Str 30, 10-082 Olsztyn, Poland
| | - Łukasz Paukszto
- grid.412607.60000 0001 2149 6795Department of Botany and Nature Protection, University of Warmia and Mazury in Olsztyn, Plac Łódzki 1, 10-727 Olsztyn, Poland
| | - Ewa Lepiarczyk
- grid.412607.60000 0001 2149 6795Department of Human Physiology and Pathophysiology, School of Medicine, Collegium Medicum, University of Warmia and Mazury in Olsztyn, Warszawska Str 30, 10-082 Olsztyn, Poland
| | - Agnieszka Skowrońska
- grid.412607.60000 0001 2149 6795Department of Human Physiology and Pathophysiology, School of Medicine, Collegium Medicum, University of Warmia and Mazury in Olsztyn, Warszawska Str 30, 10-082 Olsztyn, Poland
| | - Aleksandra Lipka
- grid.412607.60000 0001 2149 6795Department of Gynecology, and Obstetrics, School of Medicine, Collegium Medicum, University of Warmia and Mazury in Olsztyn, Żołnierska Str 18, 10-561 Olsztyn, Poland
| | - Karol G. Makowczenko
- grid.412607.60000 0001 2149 6795Department of Animal Anatomy and Physiology, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, Oczapowskiego 1A, 10-719 Olsztyn, Poland
| | - Elżbieta Łopieńska-Biernat
- grid.412607.60000 0001 2149 6795Department of Biochemistry, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, Oczapowskiego Str 1A, 10-719 Olsztyn, Poland
| | - Jan P. Jastrzębski
- grid.412607.60000 0001 2149 6795Department of Plant Physiology, Genetics and Biotechnology, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, Oczapowskiego Str 1A, 10-719 Olsztyn-Kortowo, Poland
| | - Piotr Holak
- grid.412607.60000 0001 2149 6795Department of Surgery and Radiology With Clinic, Faculty of Veterinary Medicine, University of Warmia and Mazury in Olsztyn, Oczapowskiego Str 14, 10-719 Olsztyn, Poland
| | - Michał Smoliński
- grid.460107.4Clinic of Cardiology and Internal Diseases, University Clinical Hospital in Olsztyn, Warszawska Str 30, 10-082 Olsztyn, Poland
| | - Marta Majewska
- grid.412607.60000 0001 2149 6795Department of Human Physiology and Pathophysiology, School of Medicine, Collegium Medicum, University of Warmia and Mazury in Olsztyn, Warszawska Str 30, 10-082 Olsztyn, Poland
| |
Collapse
|
13
|
Ma S, Yin S, Zheng Y, Zang R. Establishment of a mouse model for ovarian cancer-associated venous thromboembolism. Exp Biol Med (Maywood) 2023; 248:26-35. [PMID: 36036485 PMCID: PMC9989150 DOI: 10.1177/15353702221118533] [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] Open
Abstract
Patients with ovarian cancer are at increased risk of venous thromboembolism (VTE), and the cumulative incidence is high, particularly at advanced stages of this disease. Nevertheless, it is challenging to investigate the molecular mechanisms of ovarian cancer-associated VTE (OC-VTE), mainly due to the lack of a well-developed animal model for this disease. We generated a mouse model for developing OC-VTE using ovarian cancer cell injection in combination with the inferior vena cava stenosis method. The rate of thrombosis in the OC-VTE group was 50%, compared with 0 in the control group. Moreover, we conducted a proteomic analysis using platelets from these models and revealed differentially expressed proteins between the OC-VTE and control groups, including upregulated and downregulated proteins. Gene Ontology analysis revealed that these differentially expressed proteins were mostly enriched in the biological process of negative regulation of fibrinolysis and the cellular component of the fibrinogen complex, both of which play key roles in thrombosis. In conclusion, this study lays the foundation for further investigation of the underlying mechanisms of how ovarian cancer promotes VTE formation.
Collapse
Affiliation(s)
- Sining Ma
- Ovarian Cancer Program, Department of Gynecologic Oncology, Zhongshan Hospital, Fudan University, Shanghai 200032, China.,Department of Obstetrics and Gynecology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Sheng Yin
- Ovarian Cancer Program, Department of Gynecologic Oncology, Zhongshan Hospital, Fudan University, Shanghai 200032, China.,Department of Obstetrics and Gynecology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yiyan Zheng
- Ovarian Cancer Program, Department of Gynecologic Oncology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Rongyu Zang
- Ovarian Cancer Program, Department of Gynecologic Oncology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| |
Collapse
|
14
|
Wang P, Pan Y, Yang C, Zhang L, Zhao Z, Ye K, Li L, Xia S, Lu X, Shi H, Li W, Yin M. TNFα activation and TGFβ blockage act synergistically for smooth muscle cell calcification in patients with venous thrombosis via TGFβ/ERK pathway. J Cell Mol Med 2022; 26:4479-4491. [PMID: 35808901 PMCID: PMC9357635 DOI: 10.1111/jcmm.17472] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 05/12/2022] [Accepted: 06/16/2022] [Indexed: 11/26/2022] Open
Abstract
Venous calcification has been observed in post‐thrombotic syndrome (PTS) patients; yet, the cell types and possible mechanisms regulating this process are still unclear. We evaluated the calcium deposition within the venous wall, the cell type involved in the calcified remodelling of the venous wall after thrombosis and explored possible mechanisms in vitro. Calcium deposition was found in human specimens of superficial thrombotic veins and was co‐localized with VSMCs markers αSMA and TAGLN (also known as SM22α). Besides, the expression of osteogenesis‐related genes was dramatically changed in superficial thrombotic veins. Moreover, the inhibition of the TGFβ signalling pathway after TNFα treatment effectively induced the expression of osteogenic phenotype markers, the calcium salt deposits and the obvious phosphorylation of ERK1/2 and JNK2 in the VSMCs calcification model. Supplementing TGFβ2 or blocking the activation of the ERK/MAPK signalling pathway prevented the transformation of VSMCs into osteoblast‐like cells in vitro. Taken together, VSMCs have an important role in venous calcification after thrombosis. Supplementing TGFβ2 or inhibiting the ERK/MAPK signalling pathway can reduce the appearance of VSMCs osteogenic phenotype. Our findings may present a novel therapeutic approach to prevent of vascular calcification after venous thrombosis.
Collapse
Affiliation(s)
- Penghui Wang
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Yiqing Pan
- Department of Histoembryology, Genetics and Developmental Biology, Shanghai Key Laboratory of Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chenghao Yang
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Linjie Zhang
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Zhen Zhao
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Kaichuang Ye
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Lei Li
- Department of Histoembryology, Genetics and Developmental Biology, Shanghai Key Laboratory of Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shoubing Xia
- Department of Histoembryology, Genetics and Developmental Biology, Shanghai Key Laboratory of Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xinwu Lu
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China.,Vascular Center of Shanghai JiaoTong University, Shanghai, China
| | - Huihua Shi
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Weimin Li
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Minyi Yin
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China.,Vascular Center of Shanghai JiaoTong University, Shanghai, China
| |
Collapse
|
15
|
Mutch NJ, Walters S, Gardiner EE, McCarty OJT, De Meyer SF, Schroeder V, Meijers JCM. Basic science research opportunities in thrombosis and hemostasis: Communication from the SSC of the ISTH. J Thromb Haemost 2022; 20:1496-1506. [PMID: 35352482 PMCID: PMC9325489 DOI: 10.1111/jth.15718] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 03/23/2022] [Accepted: 03/23/2022] [Indexed: 11/30/2022]
Abstract
Bleeding and thrombosis are major clinical problems with high morbidity and mortality. Treatment modalities for these diseases have improved in recent years, but there are many clinical questions remaining and a need to advance diagnosis, management, and therapeutic options. Basic research plays a fundamental role in understanding normal and disease processes, yet this sector has observed a steady decline in funding prospects thereby hindering support for studies of mechanisms of disease and therapeutic development opportunities. With the financial constraints faced by basic scientists, the ISTH organized a basic science task force (BSTF), comprising Scientific and Standardization Committee subcommittee chairs and co-chairs, to identify research opportunities for basic science in hemostasis and thrombosis. The goal of the BSTF was to develop a set of recommended priorities to build support in the thrombosis and hemostasis community and to inform ISTH basic science programs and policy making. The BSTF identified three principal opportunity areas that were of significant overarching relevance: mechanisms causing bleeding, innate immunity and thrombosis, and venous thrombosis. Within these, five fundamental research areas were highlighted: blood rheology, platelet biogenesis, cellular contributions to thrombosis and hemostasis, structure-function protein analyses, and visualization of hemostasis. This position paper discusses the importance and relevance of these opportunities and research areas, and the rationale for their inclusion. These findings have implications for the future of fundamental research in thrombosis and hemostasis to make transformative scientific discoveries and tackle key clinical questions. This will permit better understanding, prevention, diagnosis, and treatment of hemostatic and thrombotic conditions.
Collapse
Affiliation(s)
- Nicola J. Mutch
- Aberdeen Cardiovascular & Diabetes CentreInstitute of Medical SciencesSchool of MedicineMedical Sciences and NutritionUniversity of AberdeenAberdeenUK
| | | | - Elizabeth E. Gardiner
- John Curtin School of Medical ResearchThe Australian National UniversityCanberraAustralian Capital TerritoryAustralia
| | - Owen J. T. McCarty
- Departments of Biomedical Engineering and MedicineOregon Health & Science UniversityPortlandOregonUSA
| | - Simon F. De Meyer
- Laboratory for Thrombosis ResearchKU Leuven Campus Kulak KortrijkKortrijkBelgium
| | - Verena Schroeder
- Department for BioMedical Research (DBMR)University of BernBernSwitzerland
| | - Joost C. M. Meijers
- Department of Molecular HematologySanquin ResearchAmsterdamthe Netherlands
- Department of Experimental Vascular Medicine, Amsterdam Cardiovascular SciencesAmsterdam UMCUniversity of AmsterdamAmsterdamthe Netherlands
| |
Collapse
|
16
|
Dai Y, Zhou Q, Liu Y, Chen X, Li F, Yu B, Zhang Y, Kou J. Ruscogenin alleviates deep venous thrombosis and pulmonary embolism induced by inferior vena cava stenosis inhibiting MEK/ERK/Egr-1/TF signaling pathway in mice. Curr Pharm Des 2022; 28:2001-2009. [PMID: 35619253 DOI: 10.2174/1381612828666220526120515] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 04/07/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND Ruscogenin (RUS) has anti-inflammatory and antithrombotic effects, while its potential effects on deep venous thrombosis (DVT) and pulmonary embolism (PE) remain unclear. OBJECTIVE We aimed to elucidate the effects of RUS on DVT and PE induced by the inferior vena cava stenosis (IVCS) model and investigate the underlying mechanism. METHODS Male C57/BL6 mice were used to explore whether IVCS model could be complicated with deep venous thrombosis and pulmonary embolism. Then, Effects of RUS on DVT and PE related inflammatory factors and coagulation were examined using H&E staining, ELISA, and real-time PCR. Western blot analysis was used to examine the effects of RUS on MEK/ERK/Egr-1/TF signaling pathway in PE. RESULTS IVCS model induced DVT and complied with PE 48 h after surgery. Administration of RUS (0.01, 0.1, 1 mg/kg) inhibited DVT, decreased biomarker D-Dimer, cardiac troponin I, N-Terminal probrain natriuretic peptide in plasma to ameliorate PE induced by IVCS model. Meanwhile, RUS reduced tissue factor and fibrinogen content of lung tissue, inhibited P-selectin and C-reactive protein activity in plasma, and suppressed the expressions of interleukin-6 and interleukin-1β in mice. Furthermore, RUS suppressed the phosphorylation of ERK1/2 and MEK1/2, decreasing the expressions of Egr-1 and TF in the lung. CONCLUSION IVCS model contributed to the development of DVT and PE in mice and was associated with increased inflammation. RUS showed therapeutic effects by inhibiting inflammation as well as suppressing the activation of MEK/ERK/Egr-1/TF signaling pathway.
Collapse
Affiliation(s)
- Yujie Dai
- Department of Pharmacology of Chinese Material Medica, School of Traditional Pharmacy, China Pharmaceutical University, 639 Longmian Road, Nanjing 211198, PR China
| | - Qianliu Zhou
- Department of Pharmacology of Chinese Material Medica, School of Traditional Pharmacy, China Pharmaceutical University, 639 Longmian Road, Nanjing 211198, PR China
| | - Yuankai Liu
- Department of Pharmacology of Chinese Material Medica, School of Traditional Pharmacy, China Pharmaceutical University, 639 Longmian Road, Nanjing 211198, PR China
| | - Xiaojun Chen
- Department of Pharmacology of Chinese Material Medica, School of Traditional Pharmacy, China Pharmaceutical University, 639 Longmian Road, Nanjing 211198, PR China
| | - Fang Li
- Department of Pharmacology of Chinese Material Medica, School of Traditional Pharmacy, China Pharmaceutical University, 639 Longmian Road, Nanjing 211198, PR China
| | - Boyang Yu
- Department of Pharmacology of Chinese Material Medica, School of Traditional Pharmacy, China Pharmaceutical University, 639 Longmian Road, Nanjing 211198, PR China
| | - Yuanyuan Zhang
- Department of Pharmacology of Chinese Material Medica, School of Traditional Pharmacy, China Pharmaceutical University, 639 Longmian Road, Nanjing 211198, PR China
| | - Junping Kou
- Department of Pharmacology of Chinese Material Medica, School of Traditional Pharmacy, China Pharmaceutical University, 639 Longmian Road, Nanjing 211198, PR China
| |
Collapse
|
17
|
Akkurt G, Alimoğullari M, Kartal B, Altay ÇM, Alimoğullari E, Çayli S. The Effectiveness of Long-term Use of Low-Molecular-Weight Heparin on Venous Thromboembolism After Sleeve Gastrectomy in Rats. Bariatr Surg Pract Patient Care 2022. [DOI: 10.1089/bari.2020.0094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Gökhan Akkurt
- Ankara City Hospital, General Surgery Unit, Ankara, Turkey
| | - Mustafa Alimoğullari
- Department of General Surgery, Yenimahalle Research and Training Hospital, Ankara, Turkey
| | - Bahar Kartal
- Department of Histology, Faculty of Medicine, Yıldırım Beyazıt University, Ankara, Turkey
| | - Çetin Murat Altay
- Department of Radiology, Ersin Arslan Research and Training Hospital, Gaziantep, Turkey
| | - Ebru Alimoğullari
- Department of Histology, Faculty of Medicine, Yıldırım Beyazıt University, Ankara, Turkey
| | - Sevil Çayli
- Department of Histology, Faculty of Medicine, Yıldırım Beyazıt University, Ankara, Turkey
| |
Collapse
|
18
|
Ovine Iliac Vein Model for Endovascular Thrombectomy of Acute Deep Venous Thrombosis. J Vasc Interv Radiol 2022; 33:249-254.e1. [PMID: 35221045 DOI: 10.1016/j.jvir.2021.10.037] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 10/06/2021] [Accepted: 10/13/2021] [Indexed: 10/19/2022] Open
Abstract
An ovine iliac vein thrombosis model was devised to test a wall-contacting rotational thrombectomy device. Thrombosis was successfully induced in 9 sheep with an average clot length of 31 mm ± 12 and >60% vessel occlusion on angiography. The thrombus was subsequently removed, maintaining normal intraoperative pulmonary arterial pressure (5.9 mm Hg ± 3.6) and complete distal reperfusion after thrombectomy. Additionally, the sheep were without signs of vascular trauma or embolic complications on gross necropsy and histopathologic analysis. The findings from this study support the use of an ovine iliac deep vein thrombosis model for testing of a lower extremity thrombectomy device.
Collapse
|
19
|
Shahneh F, Christian Probst H, Wiesmann SC, A-Gonzalez N, Ruf W, Steinbrink K, Raker VK, Becker C. Inflammatory Monocyte Counts Determine Venous Blood Clot Formation and Resolution. Arterioscler Thromb Vasc Biol 2022; 42:145-155. [PMID: 34911360 DOI: 10.1161/atvbaha.121.317176] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Monocytes are thought to be involved in venous thrombosis but the role of individual monocyte subpopulations on thrombus formation, clot inflammation, and degradation is an important unresolved issue. We investigate the role of inflammatory Ly6Chi monocytes in deep vein thrombosis and their potential therapeutic impact. METHODS Frequencies and compositions of blood monocytes were analyzed by flow cytometry in CCR2-/- (C-C chemokine receptor type 2) and wild-type mice of different ages and after treatment with the NR4A1 (nuclear receptor group 4 family A member 1, Nur77) agonist CnsB (cytosporone B). TF (tissue factor) sufficient and deficient Ly6Chi monocytes were adoptively transferred into aged CCR2-/- mice. Thrombus formation and size were followed by ultrasound over a 3-week period after surgical reduction of blood flow (stenosis) in the inferior vena cava. RESULTS Reduced numbers of peripheral monocytes in aged (>30 w) CCR2-/- mice are accompanied by reduced thrombus formation after inferior vena cava ligation. Reducing the number of inflammatory Ly6Chi monocytes in wild-type mice by CsnB treatment before ligation, similarly suspends clotting, while later treatment (d1 or d4) reduces thrombus growth and accelerates resolution. We describe how changes in inflammatory monocyte numbers affect the gradual differentiation of monocytes in thrombi and show that only tissue factor-competent Ly6Chi monocytes restore thrombosis in aged CCR2-/- mice. CONCLUSIONS We conclude that the number of inflammatory Ly6Chi monocytes controls deep vein thrombosis formation, growth, and resolution and can be therapeutically manipulated with a NR4A1 agonist at all disease stages.
Collapse
Affiliation(s)
- Fatemeh Shahneh
- Department of Dermatology (F.S.), University Medical Center Mainz, Johannes Gutenberg-University Mainz, Germany
- Center for Thrombosis and Hemostasis (F.S., W.R.), University Medical Center Mainz, Johannes Gutenberg-University Mainz, Germany
| | - Hans Christian Probst
- Institute for Immunology (H.C.P.), University Medical Center Mainz, Johannes Gutenberg-University Mainz, Germany
| | - Sabine C Wiesmann
- Institute of Immunology (S.C.W., N.A.-G) and Westfälische Wilhelms-University Münster, Germany
| | - Noelia A-Gonzalez
- Institute of Immunology (S.C.W., N.A.-G) and Westfälische Wilhelms-University Münster, Germany
| | - Wolfram Ruf
- Center for Thrombosis and Hemostasis (F.S., W.R.), University Medical Center Mainz, Johannes Gutenberg-University Mainz, Germany
| | - Kerstin Steinbrink
- Department of Dermatology, Westfälische Wilhelms-University Münster, Germany (K.S., V.K.R., C.B.)
| | - Verena K Raker
- Department of Dermatology, Westfälische Wilhelms-University Münster, Germany (K.S., V.K.R., C.B.)
| | - Christian Becker
- Department of Dermatology, Westfälische Wilhelms-University Münster, Germany (K.S., V.K.R., C.B.)
| |
Collapse
|
20
|
Mohammed BM, Cheng Q, Ivanov IS, Gailani D. Murine Models in the Evaluation of Heparan Sulfate-Based Anticoagulants. Methods Mol Biol 2022; 2303:789-805. [PMID: 34626423 PMCID: PMC8552346 DOI: 10.1007/978-1-0716-1398-6_59] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Evaluating prospective anticoagulant therapies in animal thrombosis and bleeding models are standard pre-clinical approaches. Mice are frequently used for initial evaluations because a variety of models have been developed in this well-characterized species, and mice are relatively inexpensive to maintain. Because mice seem to be resistant to forming "spontaneous" thrombosis, vessel injury is used to induce intravascular clot formation. For the purpose of testing heparin-based drugs, we adapted a well-established model in which thrombus formation in the carotid artery is induced by exposing the vessel to ferric chloride. For studying anticoagulant effects on venous thrombosis, we use a model in which the inferior vena cava is ligated and the size of the resulting clots are measured. The most common adverse effect of anticoagulation therapy is bleeding. We describe a simple tail bleeding time that has been used for many years to study the effects of anticoagulants on hemostasis. We also describe a more reproducible, but more technically challenging, saphenous vein bleeding model that is also used for this purpose.
Collapse
Affiliation(s)
- Bassem M Mohammed
- Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, Doisy Research Center, St. Louis, MO, USA
| | - Qiufang Cheng
- Department of Pathology, Microbiology and Immunology, Vanderbilt University, Nashville, TN, USA
| | - Ivan S Ivanov
- Department of Pathology, Microbiology and Immunology, Vanderbilt University, Nashville, TN, USA
| | - David Gailani
- Department of Pathology, Microbiology and Immunology, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
21
|
Khaja MS, Obi AT, Sharma AM, Cuker A, McCann SS, Thukral S, Matson JT, Hofmann LV, Charalel R, Kanthi Y, Meek ME, Meissner MH, White SB, Williams DM, Vedantham S. Optimal Medical Therapy Following Deep Venous Interventions: Proceedings from the Society of Interventional Radiology Foundation Research Consensus Panel. J Vasc Interv Radiol 2021; 33:78-85. [PMID: 34563699 DOI: 10.1016/j.jvir.2021.09.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 09/11/2021] [Accepted: 09/15/2021] [Indexed: 12/17/2022] Open
Abstract
The optimal medical management of patients following endovascular deep venous interventions remains ill-defined. As such, the Society of Interventional Radiology Foundation (SIRF) convened a multidisciplinary group of experts in a virtual Research Consensus Panel (RCP) to develop a prioritized research agenda regarding antithrombotic therapy following deep venous interventions. The panelists presented the gaps in knowledge followed by discussion and ranking of research priorities based on clinical relevance, overall impact, and technical feasibility. The following research topics were identified as high priority: 1) characterization of biological processes leading to in-stent stenosis/rethrombosis; 2) identification and validation of methods to assess venous flow dynamics and their effect on stent failure; 3) elucidation of the role of inflammation and anti-inflammatory therapies; and 4) clinical studies to compare antithrombotic strategies and improve venous outcome assessment. Collaborative, multicenter research is necessary to answer these questions and thereby enhance the care of patients with venous disease.
Collapse
Affiliation(s)
- Minhaj S Khaja
- Division of Vascular and Interventional Radiology, Department of Radiology and Medical Imaging, University of Virginia Health, Charlottesville, Virginia.
| | - Andrea T Obi
- Section of Vascular Surgery, Department of Surgery, University of Michigan, Ann Arbor, Michigan
| | - Aditya M Sharma
- Division of Cardiovascular Medicine, Department of Medicine, University of Virginia Health, Charlottesville, Virginia
| | - Adam Cuker
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Sara S McCann
- Division of Vascular and Interventional Radiology, Department of Radiology and Medical Imaging, University of Virginia Health, Charlottesville, Virginia
| | - Siddhant Thukral
- School of Medicine, University of Missouri-Kansas City, Kansas City, Missouri
| | - John T Matson
- Division of Vascular and Interventional Radiology, Department of Radiology and Medical Imaging, University of Virginia Health, Charlottesville, Virginia
| | - Lawrence V Hofmann
- Department of Radiology, Stanford University School of Medicine, Stanford, California
| | - Resmi Charalel
- Department of Radiology, Division of Interventional Radiology, New York Presbyterian Hospital/Weill Cornell Medicine, New York, New York
| | - Yogendra Kanthi
- Laboratory of Vascular Thrombosis and Inflammation, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Mary E Meek
- Division of Interventional Radiology, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Mark H Meissner
- Department of Surgery, University of Washington School of Medicine, Seattle, Washington
| | - Sarah B White
- Department of Radiology, Division of Vascular and Interventional Radiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - David M Williams
- Division of Vascular and Interventional Radiology, Department of Radiology, University of Michigan, Ann Arbor, Michigan
| | - Suresh Vedantham
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, Missouri
| |
Collapse
|
22
|
Alkarithi G, Duval C, Shi Y, Macrae FL, Ariëns RAS. Thrombus Structural Composition in Cardiovascular Disease. Arterioscler Thromb Vasc Biol 2021; 41:2370-2383. [PMID: 34261330 PMCID: PMC8384252 DOI: 10.1161/atvbaha.120.315754] [Citation(s) in RCA: 92] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Thrombosis is a major complication of cardiovascular disease, leading to myocardial infarction, acute ischemic stroke, or venous thromboembolism. Thrombosis occurs when a thrombus forms inside blood vessels disrupting blood flow. Developments in thrombectomy to remove thrombi from vessels have provided new opportunities to study thrombus composition which may help to understand mechanisms of disease and underpin improvements in treatments. We aimed to review thrombus compositions, roles of components in thrombus formation and stability, and methods to investigate thrombi. Also, we summarize studies on thrombus structure obtained from cardiovascular patients and animal models. Thrombi are composed of fibrin, red blood cells, platelets, leukocytes, and neutrophil extracellular traps. These components have been analyzed by several techniques, including scanning electron microscopy, laser scanning confocal microscopy, histochemistry, and immunohistochemistry; however, each technique has advantages and limitations. Thrombi are heterogenous in composition, but overall, thrombi obtained from myocardial infarction are composed of mainly fibrin and other components, including platelets, red blood cells, leukocytes, and cholesterol crystals. Thrombi from patients with acute ischemic stroke are characterized by red blood cell- and platelet-rich regions. Thrombi from patients with venous thromboembolism contain mainly red blood cells and fibrin with some platelets and leukocytes. Thrombus composition from patients with myocardial infarction is influenced by ischemic time. Animal thrombosis models are crucial to gain further mechanistic information about thrombosis and thrombus structure, with thrombi being similar in composition compared with those from patients. Further studies on thrombus composition and function are key to improve treatment and clinical outcome of thrombosis.
Collapse
Affiliation(s)
- Ghadir Alkarithi
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (G.A., C.D., Y.S., F.L.M., R.A.S.A.).,Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia (G.A.)
| | - Cédric Duval
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (G.A., C.D., Y.S., F.L.M., R.A.S.A.)
| | - Yu Shi
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (G.A., C.D., Y.S., F.L.M., R.A.S.A.)
| | - Fraser L Macrae
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (G.A., C.D., Y.S., F.L.M., R.A.S.A.)
| | - Robert A S Ariëns
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (G.A., C.D., Y.S., F.L.M., R.A.S.A.)
| |
Collapse
|
23
|
Gromadziński L, Paukszto Ł, Skowrońska A, Holak P, Smoliński M, Łopieńska-Biernat E, Lepiarczyk E, Lipka A, Jastrzębski JP, Majewska M. Transcriptomic Profiling of Femoral Veins in Deep Vein Thrombosis in a Porcine Model. Cells 2021; 10:1576. [PMID: 34206566 PMCID: PMC8304794 DOI: 10.3390/cells10071576] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 06/13/2021] [Accepted: 06/17/2021] [Indexed: 12/20/2022] Open
Abstract
Deep vein thrombosis (DVT) is a severe disease affecting the human venous system, accompanied by high morbidity and mortality rates caused by early and late complications. The study aimed at analyzing the changes in the transcriptome of the femoral vein caused by DVT in the porcine model based on the formation of the thrombus in vivo. The study was performed on 11 castrated male pigs: A thrombus was formed in each left femoral vein in six animals; the remaining five served as a control group. Total RNA was isolated from the left femoral veins of the experimental and control animals. High-throughput RNA sequencing was used to analyze the global changes in the transcriptome of veins with induced DVT. Applied multistep bioinformatics revealed 1474 differentially expressed genes (DEGs): 1019 upregulated and 455 downregulated. Functional Gene Ontology annotated 1220 of DEGs into 225 biological processes, 30 molecular functions and 40 cellular components categories. KEGG analysis disclosed TNF, NF-κB and apoptosis pathways' overexpression in DVT samples. A thorough analysis of the detected DEGs indicated that a dysregulated inflammatory response and disturbed balance between clotting and anti-clotting factors play a crucial role in the process of DVT.
Collapse
Affiliation(s)
- Leszek Gromadziński
- Department of Cardiology and Internal Medicine, School of Medicine, Collegium Medicum, University of Warmia and Mazury in Olsztyn, 10-082 Olsztyn, Poland
| | - Łukasz Paukszto
- Department of Plant Physiology, Genetics and Biotechnology, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, 10-719 Olsztyn, Poland; (Ł.P.); (J.P.J.)
| | - Agnieszka Skowrońska
- Department of Human Physiology and Pathophysiology, School of Medicine, Collegium Medicum, University of Warmia and Mazury in Olsztyn, 10-082 Olsztyn, Poland; (A.S.); (E.L.)
| | - Piotr Holak
- Department of Surgery and Radiology with Clinic, Faculty of Veterinary Medicine, University of Warmia and Mazury in Olsztyn, 10-719 Olsztyn, Poland;
| | - Michał Smoliński
- Clinic of Cardiology and Internal Diseases, University Clinical Hospital in Olsztyn, 10-082 Olsztyn, Poland;
| | - Elżbieta Łopieńska-Biernat
- Department of Biochemistry, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, 10-719 Olsztyn, Poland;
| | - Ewa Lepiarczyk
- Department of Human Physiology and Pathophysiology, School of Medicine, Collegium Medicum, University of Warmia and Mazury in Olsztyn, 10-082 Olsztyn, Poland; (A.S.); (E.L.)
| | - Aleksandra Lipka
- Department of Gynecology and Obstetrics, School of Medicine, Collegium Medicum, University of Warmia and Mazury in Olsztyn, 10-561 Olsztyn, Poland;
| | - Jan Paweł Jastrzębski
- Department of Plant Physiology, Genetics and Biotechnology, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, 10-719 Olsztyn, Poland; (Ł.P.); (J.P.J.)
| | - Marta Majewska
- Department of Human Physiology and Pathophysiology, School of Medicine, Collegium Medicum, University of Warmia and Mazury in Olsztyn, 10-082 Olsztyn, Poland; (A.S.); (E.L.)
| |
Collapse
|
24
|
Gromadziński L, Skowrońska A, Holak P, Smoliński M, Lepiarczyk E, Żurada A, Majewski MK, Skowroński MT, Majewska M. A New Experimental Porcine Model of Venous Thromboembolism. J Clin Med 2021; 10:1862. [PMID: 33923100 PMCID: PMC8123404 DOI: 10.3390/jcm10091862] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/19/2021] [Accepted: 04/24/2021] [Indexed: 12/22/2022] Open
Abstract
Venous thromboembolism (VTE), including deep vein thrombosis (DVT) and pulmonary embolism (PE), is a severe disease affecting the human venous system, accompanied by high morbidity and mortality rates. The aim of the study was to establish a new porcine VTE model based on the formation of the thrombus in vivo. The study was performed on 10 castrated male pigs: thrombus was formed in each closed femoral vein and then successfully released from the right femoral vein into the circulation of animals. In six pigs PE was confirmed via both computed tomography pulmonary angiography and an autopsy. Our research presents a novel experimental porcine model of VTE that involves inducing DVT and PE in the same animal in vivo, making it suitable for advanced clinical research and testing of future therapies.
Collapse
Affiliation(s)
- Leszek Gromadziński
- Department of Cardiology and Internal Medicine, School of Medicine, Collegium Medicum, University of Warmia and Mazury in Olsztyn, Warszawska Str. 30, 10-082 Olsztyn, Poland;
| | - Agnieszka Skowrońska
- Department of Human Physiology and Pathophysiology, School of Medicine, Collegium Medicum, University of Warmia and Mazury in Olsztyn, Warszawska Str. 30, 10-082 Olsztyn, Poland; (A.S.); (E.L.); (M.K.M.)
| | - Piotr Holak
- Department of Surgery and Radiology with Clinic, Faculty of Veterinary Medicine, University of Warmia and Mazury in Olsztyn, Oczapowskiego Str. 14, 10-719 Olsztyn, Poland;
| | - Michał Smoliński
- University Clinical Hospital in Olsztyn, Clinic of Cardiology and Internal Diseases, Warszawska Str. 30, 10-082 Olsztyn, Poland;
| | - Ewa Lepiarczyk
- Department of Human Physiology and Pathophysiology, School of Medicine, Collegium Medicum, University of Warmia and Mazury in Olsztyn, Warszawska Str. 30, 10-082 Olsztyn, Poland; (A.S.); (E.L.); (M.K.M.)
| | - Anna Żurada
- Department of Radiology, School of Medicine, Collegium Medicum, University of Warmia and Mazury in Olsztyn, Warszawska Str. 30, 10-082 Olsztyn, Poland;
| | - Mariusz Krzysztof Majewski
- Department of Human Physiology and Pathophysiology, School of Medicine, Collegium Medicum, University of Warmia and Mazury in Olsztyn, Warszawska Str. 30, 10-082 Olsztyn, Poland; (A.S.); (E.L.); (M.K.M.)
| | - Mariusz Tomasz Skowroński
- Department of Basic and Preclinical Sciences, Institute for Veterinary Medicine, Nicolaus Copernicus University, Gagarina Str. 7, 87-100 Torun, Poland;
| | - Marta Majewska
- Department of Human Physiology and Pathophysiology, School of Medicine, Collegium Medicum, University of Warmia and Mazury in Olsztyn, Warszawska Str. 30, 10-082 Olsztyn, Poland; (A.S.); (E.L.); (M.K.M.)
| |
Collapse
|
25
|
The pathobiology of thrombosis, microvascular disease, and hemorrhage in the myeloproliferative neoplasms. Blood 2021; 137:2152-2160. [PMID: 33649757 DOI: 10.1182/blood.2020008109] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 01/11/2021] [Indexed: 12/19/2022] Open
Abstract
Thrombotic, vascular, and bleeding complications are the most common causes of morbidity and mortality in the Philadelphia chromosome-negative myeloproliferative neoplasms (MPNs). In these disorders, circulating red cells, leukocytes, and platelets, as well as some vascular endothelial cells, each have abnormalities that are cell-intrinsic to the MPN driver mutations they harbor (eg, JAK2 V617F). When these cells are activated in the MPNs, their interactions with each other create a highly proadhesive and prothrombotic milieu in the circulation that predisposes patients with MPN to venous, arterial, and microvascular thrombosis and occlusive disease. Bleeding problems in the MPNs are caused by the MPN blood cell-initiated development of acquired von Willebrand disease. The inflammatory state created by MPN stem cells in their microenvironment extends systemically to amplify the clinical thrombotic tendency and, at the same time, preferentially promote further MPN stem cell clonal expansion, thereby generating a vicious cycle that favors a prothrombotic state in these diseases.
Collapse
|
26
|
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: 20] [Impact Index Per Article: 4.0] [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
|
27
|
Schwein A, Magnus L, Chakfé N, Bismuth J. Critical Review of Large Animal Models for Central Deep Venous Thrombosis. Eur J Vasc Endovasc Surg 2020; 60:243-252. [PMID: 32359973 DOI: 10.1016/j.ejvs.2020.03.051] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 02/29/2020] [Accepted: 03/30/2020] [Indexed: 11/28/2022]
Abstract
OBJECTIVE To review the existing literature on large animal models of central venous thrombosis (CVT) and to evaluate its relevance in regard to the development and testing of dedicated therapeutics applicable to humans. METHODS A systematic literature search was conducted in PubMed and Embase. Articles describing an in vivo experimental protocol of CVT in large animals, involving the iliac vein and/or the vena cava and/or the brachiocephalic vein, were included. The primary aim of the study, animal characteristics, experimental protocol, and thrombus evaluation were recorded. RESULTS Thirty-eight papers describing more than 30 different protocols were included. Animals used were pigs (53%), dogs (21%), monkeys (24%), and cattle (3%). The median number of animals per study was 12. Animal sex, strain, and weight were missing in 18 studies (47%), seven studies (18%), and eight studies (21%), respectively. CVT was always induced by venous stasis: solely (55%), or in addition to hypercoagulability (37%) or endothelial damage (10%). The size of the vessel used for thrombus creation was measured in four studies (10%). Unexpected animal death occurred in nine studies (24%), ranging from 3% to 37% of the animals. Twenty-two studies (58%) in the acute phase and 31 studies in the chronic phase (82%) evaluated the presence or absence of the thrombus created, and its occlusive characteristic was reported, respectively, in five and 17 studies. Histological examination was performed in 24 studies (63%) with comparison to human thrombus in one study. CONCLUSION This review showed advantages and weaknesses of the existing large animal models of CVT. Future models should insist on more rigour and consistency in reporting animal characteristics, as well as evaluating and comparing the thrombus created to human thrombus.
Collapse
Affiliation(s)
- Adeline Schwein
- Department of Vascular Surgery and Kidney Transplantation, University Hospital of Strasbourg, and Groupe Européen de Recherche sur les Prothèses Appliquées à la Chirurgie Vasculaire, Strasbourg, France; Fédération de Médecine Translationnelle de Strasbourg, Department of Physiology, EA 3072, University Hospital of Strasbourg, Strasbourg, France.
| | - Louis Magnus
- Department of Vascular Surgery and Kidney Transplantation, University Hospital of Strasbourg, and Groupe Européen de Recherche sur les Prothèses Appliquées à la Chirurgie Vasculaire, Strasbourg, France
| | - Nabil Chakfé
- Department of Vascular Surgery and Kidney Transplantation, University Hospital of Strasbourg, and Groupe Européen de Recherche sur les Prothèses Appliquées à la Chirurgie Vasculaire, Strasbourg, France
| | - Jean Bismuth
- Houston Methodist DeBakey Heart & Vascular Centre, Houston, TX, USA
| |
Collapse
|
28
|
Nicklas JM, Gordon AE, Henke PK. Resolution of Deep Venous Thrombosis: Proposed Immune Paradigms. Int J Mol Sci 2020; 21:E2080. [PMID: 32197363 PMCID: PMC7139924 DOI: 10.3390/ijms21062080] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 03/14/2020] [Accepted: 03/15/2020] [Indexed: 12/12/2022] Open
Abstract
Venous thromboembolism (VTE) is a pathology encompassing deep vein thrombosis (DVT) and pulmonary embolism (PE) associated with high morbidity and mortality. Because patients often present after a thrombus has already formed, the mechanisms that drive DVT resolution are being investigated in search of treatment. Herein, we review the current literature, including the molecular mechanisms of fibrinolysis and collagenolysis, as well as the critical cellular roles of macrophages, neutrophils, and endothelial cells. We propose two general models for the operation of the immune system in the context of venous thrombosis. In early thrombus resolution, neutrophil influx stabilizes the tissue through NETosis. Meanwhile, macrophages and intact neutrophils recognize the extracellular DNA by the TLR9 receptor and induce fibrosis, a complimentary stabilization method. At later stages of resolution, pro-inflammatory macrophages police the thrombus for pathogens, a role supported by both T-cells and mast cells. Once they verify sterility, these macrophages transform into their pro-resolving phenotype. Endothelial cells both coat the stabilized thrombus, a necessary early step, and can undergo an endothelial-mesenchymal transition, which impedes DVT resolution. Several of these interactions hold promise for future therapy.
Collapse
Affiliation(s)
| | | | - Peter K. Henke
- School of Medicine, University of Michigan, 1500 East Medical Center Drive, Ann Arbor, MI 48109, USA; (J.M.N.); (A.E.G.)
| |
Collapse
|
29
|
Arce M, Pinto MP, Galleguillos M, Muñoz C, Lange S, Ramirez C, Erices R, Gonzalez P, Velasquez E, Tempio F, Lopez MN, Salazar-Onfray F, Cautivo K, Kalergis AM, Cruz S, Lladser Á, Lobos-González L, Valenzuela G, Olivares N, Sáez C, Koning T, Sánchez FA, Fuenzalida P, Godoy A, Contreras Orellana P, Leyton L, Lugano R, Dimberg A, Quest AFG, Owen GI. Coagulation Factor Xa Promotes Solid Tumor Growth, Experimental Metastasis and Endothelial Cell Activation. Cancers (Basel) 2019; 11:cancers11081103. [PMID: 31382462 PMCID: PMC6721564 DOI: 10.3390/cancers11081103] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 07/11/2019] [Accepted: 07/27/2019] [Indexed: 02/06/2023] Open
Abstract
Hypercoagulable state is linked to cancer progression; however, the precise role of the coagulation cascade is poorly described. Herein, we examined the contribution of a hypercoagulative state through the administration of intravenous Coagulation Factor Xa (FXa), on the growth of solid human tumors and the experimental metastasis of the B16F10 melanoma in mouse models. FXa increased solid tumor volume and lung, liver, kidney and lymph node metastasis of tail-vein injected B16F10 cells. Concentrating on the metastasis model, upon coadministration of the anticoagulant Dalteparin, lung metastasis was significantly reduced, and no metastasis was observed in other organs. FXa did not directly alter proliferation, migration or invasion of cancer cells in vitro. Alternatively, FXa upon endothelial cells promoted cytoskeleton contraction, disrupted membrane VE-Cadherin pattern, heightened endothelial-hyperpermeability, increased inflammatory adhesion molecules and enhanced B16F10 adhesion under flow conditions. Microarray analysis of endothelial cells treated with FXa demonstrated elevated expression of inflammatory transcripts. Accordingly, FXa treatment increased immune cell infiltration in mouse lungs, an effect reduced by dalteparin. Taken together, our results suggest that FXa increases B16F10 metastasis via endothelial cell activation and enhanced cancer cell-endothelium adhesion advocating that the coagulation system is not merely a bystander in the process of cancer metastasis.
Collapse
Affiliation(s)
- Maximiliano Arce
- Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
- Advanced Center for Chronic Diseases (ACCDiS), Santiago, Chile
| | - Mauricio P Pinto
- Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
| | - Macarena Galleguillos
- Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
| | - Catalina Muñoz
- Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
| | - Soledad Lange
- Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
| | - Carolina Ramirez
- Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
| | - Rafaela Erices
- Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
- Vicerrectoría de Investigación, Universidad Mayor, Santiago 7510041, Chile
| | - Pamela Gonzalez
- Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
| | - Ethel Velasquez
- Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
- Comisión Chilena de Energía Nuclear (CCHEN), Santiago, Chile
| | - Fabián Tempio
- Institute of Biomedical Sciences, Faculty of Medicine, University de Chile, Santiago 8380453, Chile
| | - Mercedes N Lopez
- Institute of Biomedical Sciences, Faculty of Medicine, University de Chile, Santiago 8380453, Chile
- Millennium Institute on Immunology and Immunotherapy, Santiago 8331150, Chile
| | - Flavio Salazar-Onfray
- Institute of Biomedical Sciences, Faculty of Medicine, University de Chile, Santiago 8380453, Chile
- Millennium Institute on Immunology and Immunotherapy, Santiago 8331150, Chile
| | - Kelly Cautivo
- Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
| | - Alexis M Kalergis
- Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
- Millennium Institute on Immunology and Immunotherapy, Santiago 8331150, Chile
- Biomedical Research Consortium of Chile, Santiago 8331010, Chile
| | - Sebastián Cruz
- Laboratory of Immunoncology, Fundación Ciencia & Vida, Santiago, Chile
| | - Álvaro Lladser
- Millennium Institute on Immunology and Immunotherapy, Santiago 8331150, Chile
- Laboratory of Immunoncology, Fundación Ciencia & Vida, Santiago, Chile
| | - Lorena Lobos-González
- Advanced Center for Chronic Diseases (ACCDiS), Santiago, Chile
- Laboratory of Immunoncology, Fundación Ciencia & Vida, Santiago, Chile
- Regenerative Medicine Center, Faculty of Medicine, Clinica Alemana-Universidad Del Desarrollo, Santiago 7650568, Chile
| | - Guillermo Valenzuela
- Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
| | - Nixa Olivares
- Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
| | - Claudia Sáez
- Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
| | - Tania Koning
- Immunology Institute, Faculty of Medicine, Universidad Austral de Chile, Valdivia 5110566, Chile
| | - Fabiola A Sánchez
- Immunology Institute, Faculty of Medicine, Universidad Austral de Chile, Valdivia 5110566, Chile
| | - Patricia Fuenzalida
- Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
| | - Alejandro Godoy
- Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
- Department of Urology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14203, USA
| | - Pamela Contreras Orellana
- Advanced Center for Chronic Diseases (ACCDiS), Santiago, Chile
- Laboratory of Cellular Communication, ICBM, Faculty of Medicine, Universidad de Chile, Santiago 8380453, Chile
| | - Lisette Leyton
- Advanced Center for Chronic Diseases (ACCDiS), Santiago, Chile
- Laboratory of Cellular Communication, ICBM, Faculty of Medicine, Universidad de Chile, Santiago 8380453, Chile
| | - Roberta Lugano
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, 751 85 Uppsala, Sweden
| | - Anna Dimberg
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, 751 85 Uppsala, Sweden
| | - Andrew F G Quest
- Advanced Center for Chronic Diseases (ACCDiS), Santiago, Chile
- Laboratory of Cellular Communication, ICBM, Faculty of Medicine, Universidad de Chile, Santiago 8380453, Chile
| | - Gareth I Owen
- Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile.
- Advanced Center for Chronic Diseases (ACCDiS), Santiago, Chile.
- Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile.
- Millennium Institute on Immunology and Immunotherapy, Santiago 8331150, Chile.
| |
Collapse
|
30
|
Cao ZH, Green-Johnson JM, Buckley ND, Lin QY. Bioactivity of soy-based fermented foods: A review. Biotechnol Adv 2019; 37:223-238. [PMID: 30521852 DOI: 10.1016/j.biotechadv.2018.12.001] [Citation(s) in RCA: 125] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Revised: 09/29/2018] [Accepted: 12/02/2018] [Indexed: 12/13/2022]
Abstract
For centuries, fermented soy foods have been dietary staples in Asia and, now, in response to consumer demand, they are available throughout the world. Fermentation bestows unique flavors, boosts nutritional values and increases or adds new functional properties. In this review, we describe the functional properties and underlying action mechanisms of soy-based fermented foods such as Natto, fermented soy milk, Tempeh and soy sauce. When possible, the contribution of specific bioactive components is highlighted. While numerous studies with in vitro and animal models have hinted at the functionality of fermented soy foods, ascribing health benefits requires well-designed, often complex human studies with analysis of diet, lifestyle, family and medical history combined with long-term follow-ups for each subject. In addition, the contribution of the microbiome to the bioactivities of fermented soy foods, possibly mediated through direct action or bioactive metabolites, needs to be studied. Potential synergy or other interactions among the microorganisms carrying out the fermentation and the host's microbial community may also contribute to food functionality, but the details still require elucidation. Finally, safety evaluation of fermented soy foods has been limited, but is essential in order to provide guidelines for consumption and confirm lack of toxicity.
Collapse
Affiliation(s)
- Zhen-Hui Cao
- Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China
| | - Julia M Green-Johnson
- Faculty of Science, University of Ontario Institute of Technology (UOIT), Oshawa L1H 7K4, Canada
| | | | - Qiu-Ye Lin
- College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, China.
| |
Collapse
|
31
|
Amic F, Drmic D, Bilic Z, Krezic I, Zizek H, Peklic M, Klicek R, Pajtak A, Amic E, Vidovic T, Rakic M, Milkovic Perisa M, Horvat Pavlov K, Kokot A, Tvrdeic A, Boban Blagaic A, Zovak M, Seiwerth S, Sikiric P. Bypassing major venous occlusion and duodenal lesions in rats, and therapy with the stable gastric pentadecapeptide BPC 157, L-NAME and L-arginine. World J Gastroenterol 2018; 24:5366-5378. [PMID: 30598581 PMCID: PMC6305534 DOI: 10.3748/wjg.v24.i47.5366] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 11/26/2018] [Accepted: 12/01/2018] [Indexed: 02/06/2023] Open
Abstract
AIM To investigate whether duodenal lesions induced by major venous occlusions can be attenuated by BPC 157 regardless nitric oxide (NO) system involvement.
METHODS Male Wistar rats underwent superior anterior pancreaticoduodenal vein (SAPDV)-ligation and were treated with a bath at the ligated SAPDV site (BPC 157 10 μg, 10 ng/kg per 1 mL bath/rat; L-NAME 5 mg/kg per 1 mL bath/rat; L-arginine 100 mg/kg per 1 mL bath/rat, alone and/or together; or BPC 157 10 μg/kg instilled into the rat stomach, at 1 min ligation-time). We recorded the vessel presentation (filled/appearance or emptied/disappearance) between the 5 arcade vessels arising from the SAPDV on the ventral duodenum side, the inferior anterior pancreaticoduodenal vein (IAPDV) and superior mesenteric vein (SMV) as bypassing vascular pathway to document the duodenal lesions presentation; increased NO- and oxidative stress [malondialdehyde (MDA)]-levels in duodenum.
RESULTS Unlike the severe course in the SAPDV-ligated controls, after BPC 157 application, the rats exhibited strong attenuation of the mucosal lesions and serosal congestion, improved vessel presentation, increased interconnections, increased branching by more than 60% from the initial value, the IAPDV and SMV were not congested. Interestingly, after 5 min and 30 min of L-NAME and L-arginine treatment alone, decreased mucosal and serosal duodenal lesions were observed; their effect was worsened at 24 h, and no effect on the collateral vessels and branching was seen. Together, L-NAME+L-arginine antagonized each other’s response, and thus, there was an NO-related effect. With BPC 157, all SAPDV-ligated rats receiving L-NAME and/or L-arginine appeared similar to the rats treated with BPC 157 alone. Also, BPC 157 in SAPDV-ligated rats normalized levels of NO and MDA, two oxidative stress markers, in duodenal tissues.
CONCLUSION BPC 157, rapidly bypassing occlusion, rescued the original duodenal flow through IAPDV to SMV flow, an effect related to the NO system and reduction of free radical formation.
Collapse
Affiliation(s)
- Fedor Amic
- Department of Pharmacology, Medical Faculty, University of Zagreb, Zagreb 10000, Croatia
- Department of Pathology, Medical Faculty, University of Zagreb, Zagreb 10000, Croatia
| | - Domagoj Drmic
- Department of Pharmacology, Medical Faculty, University of Zagreb, Zagreb 10000, Croatia
- Department of Pathology, Medical Faculty, University of Zagreb, Zagreb 10000, Croatia
| | - Zdenko Bilic
- Department of Pharmacology, Medical Faculty, University of Zagreb, Zagreb 10000, Croatia
- Department of Pathology, Medical Faculty, University of Zagreb, Zagreb 10000, Croatia
| | - Ivan Krezic
- Department of Pharmacology, Medical Faculty, University of Zagreb, Zagreb 10000, Croatia
- Department of Pathology, Medical Faculty, University of Zagreb, Zagreb 10000, Croatia
| | - Helena Zizek
- Department of Pharmacology, Medical Faculty, University of Zagreb, Zagreb 10000, Croatia
- Department of Pathology, Medical Faculty, University of Zagreb, Zagreb 10000, Croatia
| | - Marina Peklic
- Department of Pharmacology, Medical Faculty, University of Zagreb, Zagreb 10000, Croatia
- Department of Pathology, Medical Faculty, University of Zagreb, Zagreb 10000, Croatia
| | - Robert Klicek
- Department of Pharmacology, Medical Faculty, University of Zagreb, Zagreb 10000, Croatia
- Department of Pathology, Medical Faculty, University of Zagreb, Zagreb 10000, Croatia
| | - Alen Pajtak
- Department of Pharmacology, Medical Faculty, University of Zagreb, Zagreb 10000, Croatia
- Department of Pathology, Medical Faculty, University of Zagreb, Zagreb 10000, Croatia
| | - Enio Amic
- Department of Pharmacology, Medical Faculty, University of Zagreb, Zagreb 10000, Croatia
- Department of Pathology, Medical Faculty, University of Zagreb, Zagreb 10000, Croatia
| | - Tinka Vidovic
- Department of Pharmacology, Medical Faculty, University of Zagreb, Zagreb 10000, Croatia
- Department of Pathology, Medical Faculty, University of Zagreb, Zagreb 10000, Croatia
| | - Mislav Rakic
- Department of Pharmacology, Medical Faculty, University of Zagreb, Zagreb 10000, Croatia
- Department of Pathology, Medical Faculty, University of Zagreb, Zagreb 10000, Croatia
| | - Marija Milkovic Perisa
- Department of Pharmacology, Medical Faculty, University of Zagreb, Zagreb 10000, Croatia
- Department of Pathology, Medical Faculty, University of Zagreb, Zagreb 10000, Croatia
| | - Katarina Horvat Pavlov
- Department of Pharmacology, Medical Faculty, University of Zagreb, Zagreb 10000, Croatia
- Department of Pathology, Medical Faculty, University of Zagreb, Zagreb 10000, Croatia
| | - Antonio Kokot
- Department of Pharmacology, Medical Faculty, University of Zagreb, Zagreb 10000, Croatia
- Department of Pathology, Medical Faculty, University of Zagreb, Zagreb 10000, Croatia
| | - Ante Tvrdeic
- Department of Pharmacology, Medical Faculty, University of Zagreb, Zagreb 10000, Croatia
- Department of Pathology, Medical Faculty, University of Zagreb, Zagreb 10000, Croatia
| | - Alenka Boban Blagaic
- Department of Pharmacology, Medical Faculty, University of Zagreb, Zagreb 10000, Croatia
- Department of Pathology, Medical Faculty, University of Zagreb, Zagreb 10000, Croatia
| | - Mario Zovak
- Department of Pharmacology, Medical Faculty, University of Zagreb, Zagreb 10000, Croatia
- Department of Pathology, Medical Faculty, University of Zagreb, Zagreb 10000, Croatia
| | - Sven Seiwerth
- Department of Pharmacology, Medical Faculty, University of Zagreb, Zagreb 10000, Croatia
- Department of Pathology, Medical Faculty, University of Zagreb, Zagreb 10000, Croatia
| | - Predrag Sikiric
- Department of Pharmacology, Medical Faculty, University of Zagreb, Zagreb 10000, Croatia
- Department of Pathology, Medical Faculty, University of Zagreb, Zagreb 10000, Croatia
| |
Collapse
|
32
|
Bonis A, Anderson L, Talhouarne G, Schueller E, Unke J, Krus C, Stokka J, Koepke A, Lehrer B, Schuh A, Andersen JJ, Cooper S. Cardiovascular resistance to thrombosis in 13-lined ground squirrels. J Comp Physiol B 2018; 189:167-177. [PMID: 30317383 DOI: 10.1007/s00360-018-1186-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 08/08/2018] [Accepted: 10/06/2018] [Indexed: 12/24/2022]
Abstract
13-lined ground squirrels (Ictidomys tridecemlineatus) enter hibernation as a survival strategy during extreme environmental conditions. Typical ground squirrel hibernation is characterized by prolonged periods of torpor with significantly reduced heart rate, blood pressure, and blood flow, interrupted every few weeks by brief interbout arousals (IBA) during which blood flow fluctuates dramatically. These physiological conditions should increase the risk of stasis-induced blood clots and myocardial ischemia. However, ground squirrels have adapted to survive repeated bouts of torpor and IBA without forming lethal blood clots or sustaining lethal ischemic myocardial damage. The purpose of this study was to determine if ground squirrels are resistant to thrombosis and myocardial ischemia during hibernation. Blood markers of coagulation, fibrinolysis, thrombosis, and ischemia, as well as histological markers of myocardial ischemia were measured throughout the annual hibernation cycle. Hibernating ground squirrels were also treated with isoprenaline to induce myocardial ischemia. Thrombin-antithrombin complex levels were significantly reduced (p < 0.05) during hibernation, while D-dimer level remained unchanged throughout the annual cycle, both consistent with an antithrombotic state. During torpor, the ground squirrels were in a hyperfibrinolytic state with an elevated ratio of tissue plasminogen activator complexed with plasminogen activator inhibitor to total plasminogen activator inhibitor (p < 0.05). Histological markers of myocardial ischemia were reversibly elevated during hibernation with no increase in markers of myocardial cell death in the blood. These data suggest that ground squirrels do not form major blood clots during hibernation through suppression of coagulation and a hyperfibrinolytic state. These animals also demonstrate myocardial resistance to ischemia.
Collapse
Affiliation(s)
- Alison Bonis
- Biology Department, University of Wisconsin-La Crosse, 1725 State St. La Crosse, La Crosse, WI, 54601, USA
| | - Leah Anderson
- Biology Department, University of Wisconsin-La Crosse, 1725 State St. La Crosse, La Crosse, WI, 54601, USA
| | - Gaëlle Talhouarne
- Biology Department, University of Wisconsin-La Crosse, 1725 State St. La Crosse, La Crosse, WI, 54601, USA
| | - Emily Schueller
- Biology Department, University of Wisconsin-La Crosse, 1725 State St. La Crosse, La Crosse, WI, 54601, USA
| | - Jenna Unke
- Biology Department, University of Wisconsin-La Crosse, 1725 State St. La Crosse, La Crosse, WI, 54601, USA
| | - Catherine Krus
- Biology Department, University of Wisconsin-La Crosse, 1725 State St. La Crosse, La Crosse, WI, 54601, USA
| | - Jordan Stokka
- Biology Department, University of Wisconsin-La Crosse, 1725 State St. La Crosse, La Crosse, WI, 54601, USA
| | - Anna Koepke
- Biology Department, University of Wisconsin-La Crosse, 1725 State St. La Crosse, La Crosse, WI, 54601, USA
| | - Brittany Lehrer
- Biology Department, University of Wisconsin-La Crosse, 1725 State St. La Crosse, La Crosse, WI, 54601, USA
| | - Anthony Schuh
- Biology Department, University of Wisconsin-La Crosse, 1725 State St. La Crosse, La Crosse, WI, 54601, USA
| | | | - Scott Cooper
- Biology Department, University of Wisconsin-La Crosse, 1725 State St. La Crosse, La Crosse, WI, 54601, USA.
| |
Collapse
|
33
|
Qiu X, Zhou J, Wang W, Zhao Z, Tang L, Sun S. Effect of a new inhibitor of factor Xa zifaxaban, on thrombosis in the inferior vena cava in rabbits. J Thromb Thrombolysis 2018; 47:80-86. [PMID: 30298304 DOI: 10.1007/s11239-018-1743-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
In recent years, oral factor Xa inhibitors have become a research focus as anticoagulant drugs. Zifaxaban is the first oral FXa inhibitor to enter clinical trials in China. The aim of this study was to determine the inhibitory effect of zifaxaban on thrombosisthrough a model ofinferior vena cava (IVC) thrombosis in rabbits. IVC thrombosis model was established by electrical injury and stenosis, and zifaxaban was administered (p.o.) for 5 consecutive days, then coagulation indicators and bleeding were observed. The results showed that zifaxaban had obvious inhibitory effects on FXa, and had a significant inhibitory effect on IVC thrombosis induced by electrical damage and stenosis. The effect of zifaxaban was similar to that of rivaroxaban, but the bleeding side-effects of zifaxaban were less severe than those of rivaroxaban. Zifaxaban could prolong the prothrombin time and activated partial thromboplastin time of plasma similar to that of other oral FXa inhibitors. Zifaxaban had a significant inhibitory effect on FXa, but it had no obvious effect on other coagulation factors, major anticoagulant factors or fibrinolytic indices. Our results suggest that zifaxaban had specific inhibitory effects on FXa and inhibited IVC thrombosis in rabbits with its hemorrhagic effect was less than that of rivaroxaban. Zifaxaban is ecpected to be developed as a new drug for the prevention of deep venous thrombosis, providing more medication options for patients with such disease, more research is required to support it in the future.
Collapse
Affiliation(s)
- Xiaomiao Qiu
- Tianjin Medical University School, Tianjin, 300070, People's Republic of China
| | - Junjun Zhou
- Department of Pharmacology, Dalian Medical University, Dalian, 116044, Liaoning, People's Republic of China
| | - Weiting Wang
- Tianjin Institute of Pharmaceutical Research New Drug Evaluation Co.Ltd, 308, Huiren Street, Binhai New Area, Tianjin, 300301, People's Republic of China
| | - Zhuanyou Zhao
- Tianjin Institute of Pharmaceutical Research New Drug Evaluation Co.Ltd, 308, Huiren Street, Binhai New Area, Tianjin, 300301, People's Republic of China
| | - Lida Tang
- Tianjin Institute of Pharmaceutical Research New Drug Evaluation Co.Ltd, 308, Huiren Street, Binhai New Area, Tianjin, 300301, People's Republic of China
| | - Shuangyong Sun
- Tianjin Institute of Pharmaceutical Research New Drug Evaluation Co.Ltd, 308, Huiren Street, Binhai New Area, Tianjin, 300301, People's Republic of China.
| |
Collapse
|
34
|
Qiu X, Wang W, Zhao Z, Sun S, Tang L. Pre-clinical pharmacodynamic study of a novel oral factor Xa inhibitor zifaxaban. Eur J Pharmacol 2018; 836:50-56. [PMID: 30125563 DOI: 10.1016/j.ejphar.2018.08.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2018] [Revised: 08/13/2018] [Accepted: 08/15/2018] [Indexed: 10/28/2022]
Abstract
Zifaxaban is an orally active, direct Factor Xa (FXa) inhibitor that is in development for the prevention and treatment of arterial and venous thrombosis. This study was conducted to investigate the biochemical and pharmacological activity of zifaxaban. In vitro activity was evaluated by enzyme, platelet aggregation, and clotting assays. In vivo effects were examined in venous thrombosis, arteriovenous-shunt thrombosis, carotid thrombosis, and bleeding models in rats. Zifaxaban competitively inhibits human FXa (IC50 = 11.1 nM) with > 10,000-fold greater selectivity than other serine proteases. It did not impair platelet aggregation induced by collagen, adenosine diphosphate (ADP) or arachidonic acid. It significantly prolonged clotting time, prothrombin time (PT), and activated partial thromboplastin time (APTT) in the plasma of humans, rabbits, and rats, with a relatively weak effect on thrombin time (TT). In venous thrombosis models in rats, zifaxaban strongly suppressed thrombus formation with ED50 values of 3.09 mg/kg, and its best efficacy time occurred at 2 h after administration. In arteriovenous-shunt thrombosis and carotid thrombosis models in rats, it inhibited thrombus formation in a dose-dependent manner. And in the rat tail bleeding assay, it showed a trend of less bleeding than rivaroxaban at doses that achieved the same antithrombotic effect. In conclusion, zifaxaban is a selective and direct FXa inhibitor and a promising oral anticoagulant for the prophylaxis and treatment of thromboembolic diseases.
Collapse
Affiliation(s)
- Xiaomiao Qiu
- Tianjin Medical University, Tianjin 300070, China
| | - Weiting Wang
- Tianjin Institute of Pharmaceutical Research New Drug Evaluation Co. Ltd, Tianjin 300193, China
| | - Zhuanyou Zhao
- Tianjin Institute of Pharmaceutical Research New Drug Evaluation Co. Ltd, Tianjin 300193, China
| | - Shuangyong Sun
- Tianjin Institute of Pharmaceutical Research New Drug Evaluation Co. Ltd, Tianjin 300193, China.
| | - Lida Tang
- Tianjin Institute of Pharmaceutical Research New Drug Evaluation Co. Ltd, Tianjin 300193, China.
| |
Collapse
|
35
|
Abstract
: The role of sex and gender in determining clinical presentation, diagnostic approach and outcomes of venous thromboembolism is not fully and systematically addressed, except for hormone-related events in women. A lack of knowledge is also apparent regarding drug prescription patterns, physician bias, enrolment in clinical studies and analysis of sex-related confounders in preclinical and clinical studies. As was shown for cardiovascular disease, ignoring sex and gender in medicine can have important impact on outcomes, including mortality. In this review, we seek to address some aspects of venous thromboembolism such as epidemiology and clinical presentation, recurrence, risk factors, animal studies, safety and efficacy of antithrombotic drugs, highlighting what is known and what is not regarding the role of sex and gender, and hoping to focus some interest and to promote the inclusion of these variables in all future studies on venous thromboembolism.
Collapse
|
36
|
Thrombosis-on-a-chip: Prospective impact of microphysiological models of vascular thrombosis. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2018; 5:29-34. [DOI: 10.1016/j.cobme.2017.12.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
37
|
Affiliation(s)
- Rahmi Oklu
- Division of Vascular & Interventional Radiology, Mayo Clinic, Phoenix, AZ, USA.
| |
Collapse
|