1
|
Chang JJ, Brew K, Hamilton JA, Kumar V, Diaz JA, Takayama S. Bioprinted Micro-Clots for Kinetic Analysis of Endothelial Cell-Mediated Fibrinolysis. Adv Healthc Mater 2025; 14:e2403043. [PMID: 39887618 PMCID: PMC11912099 DOI: 10.1002/adhm.202403043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 01/17/2025] [Indexed: 02/01/2025]
Abstract
Vascular hypo-fibrinolysis is a historically underappreciated and understudied aspect of venous thromboembolism (VTE). This paper describes the development of a micro-clot dissolution assay for quantifying the fibrinolytic capacity of endothelial cells - a key driver of VTE development. This assay is enabled using aqueous two-phase systems (ATPS) to bioprint microscale fibrin clots over human umbilical vein endothelial cells (HUVECs). Importantly, these micro-clots are orders of magnitude smaller than conventional fibrin constructs and allow HUVEC-produced plasminogen activators to mediate visually quantifiable fibrinolysis. Using live-cell time-lapse imaging, micro-clot dissolution by HUVECs is tracked, and fibrinolysis kinetics are quantified. The sensitivity of cell-driven fibrinolysis to various stimuli is rapidly tested. The physiological relevance of this convenient high-throughput assay is illustrated through treatments with lipopolysaccharide (LPS) and rosuvastatin that elicit anti- and pro-fibrinolytic responses, respectively. Furthermore, treatment with baricitinib, an anti-inflammatory therapeutic found to increase cardiovascular risks after market approval, provokes an anti-fibrinolytic response - which highlights the potential role of endothelial cells in increasing VTE risk for patients receiving this drug. This endothelial cell fibrinolysis assay provides a high-throughput and versatile drug testing platform - potentially allowing for early preclinical identification of therapeutics that may beneficially enhance or adversely impair endothelial fibrinolysis.
Collapse
Affiliation(s)
- Jonathan J. Chang
- Wallace H. Coulter Department of Biomedical EngineeringGeorgia Institute of Technology and Emory UniversityAtlantaGA30332USA
- The Parker H. Petit Institute of Bioengineering and BioscienceGeorgia Institute of TechnologyAtlantaGA30332USA
| | - Kelsey Brew
- Wallace H. Coulter Department of Biomedical EngineeringGeorgia Institute of Technology and Emory UniversityAtlantaGA30332USA
| | - Jamie A.G. Hamilton
- Wallace H. Coulter Department of Biomedical EngineeringGeorgia Institute of Technology and Emory UniversityAtlantaGA30332USA
| | - Varun Kumar
- Wallace H. Coulter Department of Biomedical EngineeringGeorgia Institute of Technology and Emory UniversityAtlantaGA30332USA
| | - José A. Diaz
- Division of Surgical ResearchVanderbilt University Medical CenterNashvilleTN37232USA
| | - Shuichi Takayama
- Wallace H. Coulter Department of Biomedical EngineeringGeorgia Institute of Technology and Emory UniversityAtlantaGA30332USA
- The Parker H. Petit Institute of Bioengineering and BioscienceGeorgia Institute of TechnologyAtlantaGA30332USA
| |
Collapse
|
2
|
Johnson TA, Mukhopadhyay S, Buzza MS, Brooks JA, Sarkar R, Antalis TM. Regulation of macrophage fibrinolysis during venous thrombus resolution. Thromb Res 2024; 243:109149. [PMID: 39317013 PMCID: PMC11486561 DOI: 10.1016/j.thromres.2024.109149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 08/26/2024] [Accepted: 09/09/2024] [Indexed: 09/26/2024]
Abstract
BACKGROUND Venous thromboembolism (VTE), which includes pulmonary embolism (PE) and deep vein thrombosis (DVT), is a serious cardiovascular disease with significant mortality and morbidity. Clinically, patients with faster resolution of a venous thrombi have improved prognosis. Urokinase-plasminogen activator (uPA), produced by macrophages, is a key mediator of fibrinolysis required for resolving venous thrombi and restoring vascular integrity. The major macrophage protein, plasminogen activator inhibitor type-2 (PAI-2), was originally identified as an inhibitor of uPA and is implicated in the modulation of pathways affecting fibrinolytic uPA activity, however its direct role in blocking uPA-mediated clot lysis is not known. OBJECTIVE To determine the contribution of macrophage PAI-2 in inhibiting uPA-mediated fibrinolysis during resolution of DVT. METHODS Using a murine model of venous thrombosis and resolution, we determined histological changes and molecular features of fibrin degradation in venous thrombi from WT mice and mice genetically deficient in PAI-2 and PAI-1, and determined the fibrinolytic activities of macrophages from these genotypes ex vivo. RESULTS Acceleration of venous thrombus resolution by PAI-2-/- mice increases fibrin degradation in venous thrombi showing a pattern similar to genetic deficiency of PAI-1, the major attenuator of fibrinolysis. PAI-2 deficiency was not associated with increased macrophage infiltration into thrombi or changes in macrophage PAI-1 expression. uPA-initiated fibrinolysis by macrophages in vitro could be accelerated by PAI-1 deficiency, but not PAI-2 deficiency. CONCLUSION PAI-2 has an alternate anti-fibrinolytic activity that is macrophage uPA independent, where PAI-1 is the dominant uPA inhibitor during DVT resolution.
Collapse
Affiliation(s)
- Tierra A Johnson
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Subhradip Mukhopadhyay
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Marguerite S Buzza
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Jacob A Brooks
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Rajabrata Sarkar
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Toni M Antalis
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Research & Development Service, VA Maryland Health Care System, Baltimore, MD, 21201, USA.
| |
Collapse
|
3
|
Henke PK, Nicklas JM, Obi A. Immune cell-mediated venous thrombus resolution. Res Pract Thromb Haemost 2023; 7:102268. [PMID: 38193054 PMCID: PMC10772895 DOI: 10.1016/j.rpth.2023.102268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 10/23/2023] [Accepted: 11/07/2023] [Indexed: 01/10/2024] Open
Abstract
Herein, we review the current processes that govern experimental deep vein thrombus (DVT) resolution. How the human DVT resolves at the molecular and cellular level is not well known due to limited specimen availability. Experimentally, the thrombus resolution resembles wound healing, with early neutrophil-mediated actions followed by monocyte/macrophage-mediated events, including neovascularization, fibrinolysis, and eventually collagen replacement. Potential therapeutic targets are described, and coupling with site-directed approaches to mitigate off-target effects is the long-term goal. Similarly, timing of adjunctive agents to accelerate DVT resolution is an area that is only starting to be considered. There is much critical research that is needed in this area.
Collapse
Affiliation(s)
- Peter K. Henke
- Department of Surgery, University of Michigan Health System, Frankel Cardiovascular Center, Ann Arbor, Michigan, USA
| | - John M. Nicklas
- Department of Medicine, Brown University Medical School, Providence, Rhode Island, USA
| | - Andrea Obi
- Department of Surgery, University of Michigan Health System, Frankel Cardiovascular Center, Ann Arbor, Michigan, USA
| |
Collapse
|
4
|
Knight JS, Kanthi Y. Mechanisms of immunothrombosis and vasculopathy in antiphospholipid syndrome. Semin Immunopathol 2022; 44:347-362. [PMID: 35122116 PMCID: PMC8816310 DOI: 10.1007/s00281-022-00916-w] [Citation(s) in RCA: 88] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 01/19/2022] [Indexed: 12/14/2022]
Abstract
Antiphospholipid syndrome (APS) is an autoimmune thrombophilia propelled by circulating antiphospholipid antibodies that herald vascular thrombosis and obstetrical complications. Antiphospholipid antibodies recognize phospholipids and phospholipid-binding proteins and are not only markers of disease but also key drivers of APS pathophysiology. Thrombotic events in APS can be attributed to various conspirators including activated endothelial cells, platelets, and myeloid-lineage cells, as well as derangements in coagulation and fibrinolytic systems. Furthermore, recent work has especially highlighted the role of neutrophil extracellular traps (NETs) and the complement system in APS thrombosis. Beyond acute thrombosis, patients with APS can also develop an occlusive vasculopathy, a long-term consequence of APS characterized by cell proliferation and infiltration that progressively expands the intima and leads to organ damage. This review will highlight known pathogenic factors in APS and will also briefly discuss similarities between APS and the thrombophilic coagulopathy of COVID-19.
Collapse
Affiliation(s)
- Jason S Knight
- Division of Rheumatology, University of Michigan, 1150 West Medical Center Drive, Ann Arbor, MI, 48109, USA.
| | - Yogendra Kanthi
- Division of Intramural Research National Heart, Lung, and Blood Institute, Bethesda, MD, USA
| |
Collapse
|
5
|
Abstract
Fibrinolysis is of paramount importance in maintaining or regaining the patency of veins and pulmonary arteries obstructed by thrombi. Growing experimental and clinical evidence indicates that impaired fibrinolysis mediated by multiple complex mechanisms is involved in venous thromboembolism (VTE). Global plasma fibrin clot lysis markers, especially clot lysis time, have been reported to predict recurrent deep-vein thrombosis and pulmonary embolism. The current overview summarizes available data linking fibrinolysis to VTE and its long-term sequelae.
Collapse
Affiliation(s)
- Anetta Undas
- Institute of Cardiology, Jagiellonian University Medical College, Krakow, Poland.,Krakow Centre for Medical Research and Technologies, John Paul II Hospital, Krakow, Poland
| |
Collapse
|
6
|
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: 49] [Impact Index Per Article: 9.8] [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
|
7
|
Diaz JA, Saha P, Cooley B, Palmer OR, Grover SP, Mackman N, Wakefield TW, Henke PK, Smith A, Lal BK. Choosing a Mouse Model of Venous Thrombosis. Arterioscler Thromb Vasc Biol 2020; 39:311-318. [PMID: 30786739 DOI: 10.1161/atvbaha.118.311818] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Murine models are widely used valuable tools to study deep vein thrombosis. Leading experts in venous thrombosis research came together through the American Venous Forum to develop a consensus on maximizing the utility and application of available mouse models of venous thrombosis. In this work, we provide an algorithm for model selection, with discussion of the advantages, disadvantages, and applications of the main mouse models of venous thrombosis. Additionally, we provide a detailed surgical description of the models with guidelines to validate surgical technique.
Collapse
Affiliation(s)
- Jose A Diaz
- From the Department of Surgery, Vascular Surgery, University of Michigan, Ann Arbor (J.A.D., O.R.P., T.W.W., P.K.H.)
| | - Prakash Saha
- Academic Department of Vascular Surgery, King's College London, UK (P.S., A.S.)
| | - Brian Cooley
- Department of Medicine, Division of Hematology/Oncology, University of North Carolina at Chapel Hill (B.C., S.P.G., N.M.)
| | - Olivia R Palmer
- From the Department of Surgery, Vascular Surgery, University of Michigan, Ann Arbor (J.A.D., O.R.P., T.W.W., P.K.H.)
| | - Steven P Grover
- Department of Medicine, Division of Hematology/Oncology, University of North Carolina at Chapel Hill (B.C., S.P.G., N.M.)
| | - Nigel Mackman
- Department of Medicine, Division of Hematology/Oncology, University of North Carolina at Chapel Hill (B.C., S.P.G., N.M.)
| | - Thomas W Wakefield
- From the Department of Surgery, Vascular Surgery, University of Michigan, Ann Arbor (J.A.D., O.R.P., T.W.W., P.K.H.)
| | - Peter K Henke
- From the Department of Surgery, Vascular Surgery, University of Michigan, Ann Arbor (J.A.D., O.R.P., T.W.W., P.K.H.)
| | - Alberto Smith
- Academic Department of Vascular Surgery, King's College London, UK (P.S., A.S.)
| | - Brajesh K Lal
- Department of Surgery, University of Maryland, College Park (B.K.L.)
| |
Collapse
|
8
|
Mukhopadhyay S, Johnson TA, Duru N, Buzza MS, Pawar NR, Sarkar R, Antalis TM. Fibrinolysis and Inflammation in Venous Thrombus Resolution. Front Immunol 2019; 10:1348. [PMID: 31258531 PMCID: PMC6587539 DOI: 10.3389/fimmu.2019.01348] [Citation(s) in RCA: 99] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 05/28/2019] [Indexed: 12/24/2022] Open
Abstract
Clinical observations and accumulating laboratory evidence support a complex interplay between coagulation, inflammation, innate immunity and fibrinolysis in venous thromboembolism (VTE). VTE, which includes deep vein thrombosis (DVT) and pulmonary embolism (PE), and the subsequent complications of post-thrombotic syndrome (PTS), are significant causes of morbidity and mortality in patients. Clinical risk factors for VTE include cancer, major trauma, surgery, sepsis, inflammatory bowel disease, paralysis, prolonged periods of immobility, and aging. Abnormalities in venous blood flow or stasis initiates the activation of endothelial cells, and in concert with platelets, neutrophils and monocytes, propagates VTE in an intact vein. In addition, inflammatory cells play crucial roles in thrombus recanalization and restoration of blood flow via fibrinolysis and vascular remodeling. Faster resolution of the thrombus is key for improved disease prognosis. While in the clinical setting, anticoagulation therapy is successful in preventing propagation of venous thrombi, current therapies are not designed to inhibit inflammation, which can lead to the development of PTS. Animal models of DVT have provided many insights into the molecular and cellular mechanisms involved in the formation, propagation, and resolution of venous thrombi as well as the roles of key components of the fibrinolytic system in these processes. Here, we review the recent advances in our understanding of fibrinolysis and inflammation in the resolution of VTE.
Collapse
Affiliation(s)
- Subhradip Mukhopadhyay
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, United States
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Tierra A. Johnson
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Nadire Duru
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Marguerite S. Buzza
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, United States
- University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, United States
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Nisha R. Pawar
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Rajabrata Sarkar
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, United States
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Toni M. Antalis
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, United States
- University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, United States
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
9
|
Diaz JA, Saha P, Cooley B, Palmer OR, Grover SP, Mackman N, Wakefield TW, Henke PK, Smith A, Lal BK. Choosing a mouse model of venous thrombosis: a consensus assessment of utility and application. J Thromb Haemost 2019; 17:699-707. [PMID: 30927321 DOI: 10.1111/jth.14413] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Murine models are widely used valuable tools to study deep vein thrombosis (VT). Leading experts in VT research came together through the American Venous Forum to develop a consensus on maximizing the utility and application of available mouse models of VT. In this work, we provide an algorithm for model selection, with discussion of the advantages, disadvantages, and applications of the main mouse models of VT. Additionally, we provide a detailed surgical description of the models with guidelines to validate surgical technique.
Collapse
|
10
|
A novel solution phase PAI-1/uPA-biotin complex assay for the measurement of active PAI-1 in plasma. Anal Biochem 2018; 563:35-39. [PMID: 30278155 DOI: 10.1016/j.ab.2018.09.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 09/24/2018] [Accepted: 09/26/2018] [Indexed: 11/23/2022]
Abstract
We devised a new assay procedure to use biotinylated uPA to trace the active PAI-1 levels in the plasma. We show here that the potency of inhibitory monoclonal antibody 33B8 measured with the new assay is consistent with its in vivo efficacy in PAI-1 inactivation. We also found that among the three monoclonal antibodies tested, the traditional solid phase assay caused mechanism dependent significant right shift of IC50 values. As our new assay avoids the use of non-physiological large quantities of uPA, we conclude that it is a better measure of pharmacodynamic effects of anti-PAI-1 antibodies in vivo.
Collapse
|
11
|
Reinke AA, Li SH, Warnock M, Shaydakov ME, Guntaka NS, Su EJ, Diaz JA, Emal CD, Lawrence DA. Dual-reporter high-throughput screen for small-molecule in vivo inhibitors of plasminogen activator inhibitor type-1 yields a clinical lead candidate. J Biol Chem 2018; 294:1464-1477. [PMID: 30510136 DOI: 10.1074/jbc.ra118.004885] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 11/30/2018] [Indexed: 12/15/2022] Open
Abstract
Plasminogen activator inhibitor type-1 (PAI-1) is a serine protease inhibitor (serpin) implicated in numerous pathological processes, including coronary heart disease, arterial and venous thrombosis, and chronic fibrotic diseases. These associations have made PAI-1 an attractive pharmaceutical target. However, the complexity of the serpin inhibitory mechanism, the inherent metastability of serpins, and the high-affinity association of PAI-1 with vitronectin in vivo have made it difficult to identify pharmacologically effective small-molecule inhibitors. Moreover, the majority of current small-molecule PAI-1 inhibitors are poor pharmaceutical candidates. To this end and to find leads that can be efficiently applied to in vivo settings, we developed a dual-reporter high-throughput screen (HTS) that reduced the rate of nonspecific and promiscuous hits and identified leads that inhibit human PAI-1 in the high-protein environments present in vivo Using this system, we screened >152,000 pure compounds and 27,000 natural product extracts (NPEs), reducing the apparent hit rate by almost 10-fold compared with previous screening approaches. Furthermore, screening in a high-protein environment permitted the identification of compounds that retained activity in both ex vivo plasma and in vivo Following lead identification, subsequent medicinal chemistry and structure-activity relationship (SAR) studies identified a lead clinical candidate, MDI-2268, having excellent pharmacokinetics, potent activity against vitronectin-bound PAI-1 in vivo, and efficacy in a murine model of venous thrombosis. This rigorous HTS approach eliminates promiscuous candidate leads, significantly accelerates the process of identifying PAI-1 inhibitors that can be rapidly deployed in vivo, and has enabled identification of a potent lead compound.
Collapse
Affiliation(s)
- Ashley A Reinke
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109
| | - Shih-Hon Li
- Department of Pathology, University of Michigan, Ann Arbor, Michigan 48109
| | - Mark Warnock
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109
| | - Maxim E Shaydakov
- Section of Vascular Surgery, Department of Surgery, University of Michigan, Ann Arbor, Michigan 48109
| | | | - Enming J Su
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109
| | - Jose A Diaz
- Section of Vascular Surgery, Department of Surgery, University of Michigan, Ann Arbor, Michigan 48109
| | - Cory D Emal
- Department of Chemistry, Eastern Michigan University, Ypsilanti, Michigan 48197
| | - Daniel A Lawrence
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109.
| |
Collapse
|
12
|
Palmer OR, Shaydakov ME, Rainey JP, Lawrence DA, Greve JM, Diaz JA. Update on the electrolytic IVC model for pre-clinical studies of venous thrombosis. Res Pract Thromb Haemost 2018; 2:266-273. [PMID: 30046728 PMCID: PMC6055493 DOI: 10.1002/rth2.12074] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Accepted: 12/12/2017] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND The electrolytic inferior vena cava model (EIM) is a murine venous thrombosis (VT) model that produces a non-occlusive thrombus. The thrombus forms in the direction of blood flow, as observed in patients. The EIM is valuable for investigations of therapeutics due to the presence of continuous blood flow. However, the equipment used to induce thrombosis in the original model description was expensive and has since been discontinued. Further, the fibrinolytic system had not been previously studied in the EIM. OBJECTIVES We aimed to provide an equipment alternative. Additionally, we further characterized the model through mapping the current and time dependency of thrombus resolution dynamics, and investigated the fibrinolytic system from acute to chronic VT. RESULTS A voltage to current converter powered by a direct current power supply was constructed and validated, providing an added benefit of significantly reducing costs. The current and time dependency of thrombus volume dynamics was assessed by MRI, demonstrating the flexibility of the EIM to investigate both pro-thrombotic and anti-thrombotic conditions. Additionally, the fibrinolytic system was characterized in EIM. Centripetal distribution of plasminogen was observed over time, with peak staining at day 6 post thrombus induction. Both active circulating plasminogen activator inhibitor-1 (PAI-1) and vein wall gene expression of PAI-1 peaked at day 2, coinciding with a relative decrease in tissue plasminogen activator and urokinase plasminogen activator. CONCLUSIONS The EIM is a valuable model of VT that can now be performed at low cost and may be beneficial in investigations of the fibrinolytic system.
Collapse
Affiliation(s)
- Olivia R. Palmer
- Department of Biomedical EngineeringUniversity of MichiganAnn ArborMIUSA
- Department of Surgery, Vascular SurgeryUniversity of MichiganAnn ArborMIUSA
| | - Maxim E. Shaydakov
- Department of SurgeryUT Health San AntonioSan AntonioTXUSA
- Department of Surgery, Vascular SurgeryUniversity of MichiganAnn ArborMIUSA
| | - Joshua P. Rainey
- Department of Surgery, Vascular SurgeryUniversity of MichiganAnn ArborMIUSA
| | | | - Joan M. Greve
- Department of Biomedical EngineeringUniversity of MichiganAnn ArborMIUSA
| | - José A. Diaz
- Department of Surgery, Vascular SurgeryUniversity of MichiganAnn ArborMIUSA
| |
Collapse
|
13
|
Hisada Y, Mackman N. Mouse models of cancer-associated thrombosis. Thromb Res 2017; 164 Suppl 1:S48-S53. [PMID: 29306575 DOI: 10.1016/j.thromres.2017.12.018] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Revised: 12/22/2017] [Accepted: 12/27/2017] [Indexed: 10/18/2022]
Abstract
Cancer patients have an increased risk of venous thromboembolism (VTE) compared with the general population. Mouse models are used to better understand the mechanisms of cancer-associated thrombosis. Several mouse models of cancer-associated thrombosis have been developed that use different mouse strains, tumors, tumor sites and thrombosis models. In this review, we summarize these different models. These models have been used to determine the role of different pathways in cancer-associated thrombosis. For instance, they have revealed roles for tumor-derived tissue factor-positive microvesicles and neutrophil extracellular traps in thrombosis in tumor-bearing mice. A better understanding of the mechanisms of cancer-associated thrombosis may allow the development of new therapies to reduce thrombosis in cancer patients.
Collapse
Affiliation(s)
- Yohei Hisada
- Department of Medicine, Division of Hematology and Oncology, Thrombosis and Hemostasis Program, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; K. G. Jebsen Thrombosis Research and Expertise Center, University of Tromsø, Tromsø, Norway
| | - Nigel Mackman
- Department of Medicine, Division of Hematology and Oncology, Thrombosis and Hemostasis Program, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
14
|
Abstract
Deep vein thrombosis (DVT) and its devastating complication, pulmonary embolism, are a severe health problem with high mortality. Mechanisms of thrombus formation in veins remain obscure. Lack of mobility (e.g., after surgery or long-haul flights) is one of the main factors leading to DVT. The pathophysiological consequence of the lack of mobility is blood flow stagnation in venous valves. Here, a model is described that mimics such flow disturbance as a thrombosis-driving factor. In this model, partial flow restriction (stenosis) in the inferior vena cava (IVC) is created. Closure of about 90% of the IVC lumen for 48 h results in development of thrombi structurally similar to those in humans. The similarities are: i) most of the thrombus volume is red, i.e., consists of red blood cells and fibrin, ii) presence of a white part (lines of Zahn), iii) non-denuded endothelial monolayer, iv) elevated plasma D-Dimer levels, and v) possibility to prevent thrombosis by low molecular weight heparin. Limitations include variable size of thrombi and the fact that a certain percentage of wild-type mice (0 - 35%) may not produce a thrombus. In addition to visual observation and measurement, thrombi may be visualized by non-invasive technologies, such as ultrasonography, which allows for monitoring the dynamics of thrombus development. At shorter time points (1 - 6 h), intravital microscopy may be applied to directly observe events (e.g., recruitment of cells to the vessel wall) preceding thrombus formation. Use of this method by several teams around the world has made it possible to uncover basic mechanisms of DVT initiation and identify potential targets that might be beneficial for its prevention.
Collapse
Affiliation(s)
- Holly Payne
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham
| | - Alexander Brill
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham;
| |
Collapse
|
15
|
Albadawi H, Witting AA, Pershad Y, Wallace A, Fleck AR, Hoang P, Khademhosseini A, Oklu R. Animal models of venous thrombosis. Cardiovasc Diagn Ther 2017; 7:S197-S206. [PMID: 29399523 DOI: 10.21037/cdt.2017.08.10] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Venous thrombosis (VT) is a prevalent clinical condition with significant adverse sequela or mortality. Anticoagulation and pharmacologic or pharmacomechanical thrombolytic therapies are the mainstays of VT treatment. An understanding of thrombosis biology will allow for more effective VT-tailored diagnosis and therapy. In vivo models of thrombosis provide indispensable tools to study the pathogenesis of thrombus formation and to evaluate novel therapeutic or preventive adjuncts for VT management or prevention. In this article, we review the most prominent in vivo models of VT created in rodents and swine species and outline how each model can serve as a useful tool to promote our understanding of VT pathogenesis and to examine novel therapies.
Collapse
Affiliation(s)
- Hassan Albadawi
- Department of Radiology, Division of Vascular & Interventional Radiology, Mayo Clinic, Phoenix, AZ, USA
| | - Avery A Witting
- Department of Radiology, Division of Vascular & Interventional Radiology, Mayo Clinic, Phoenix, AZ, USA
| | - Yash Pershad
- Department of Radiology, Division of Vascular & Interventional Radiology, Mayo Clinic, Phoenix, AZ, USA
| | - Alex Wallace
- Department of Radiology, Mayo Clinic, Phoenix, AZ, USA
| | | | - Peter Hoang
- Department of Radiology, Mayo Clinic, Phoenix, AZ, USA
| | - Ali Khademhosseini
- Biomaterials Innovation Research Center, Brigham and Women's Hospital & Harvard Medical School, Cambridge, MA, USA.,Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Rahmi Oklu
- Department of Radiology, Division of Vascular & Interventional Radiology, Mayo Clinic, Phoenix, AZ, USA.,Biomaterials Innovation Research Center, Brigham and Women's Hospital & Harvard Medical School, Cambridge, MA, USA
| |
Collapse
|
16
|
Jin QQ, Sun JH, Du QX, Lu XJ, Zhu XY, Fan HL, Hölscher C, Wang YY. Integrating microRNA and messenger RNA expression profiles in a rat model of deep vein thrombosis. Int J Mol Med 2017; 40:1019-1028. [PMID: 28848993 PMCID: PMC5593457 DOI: 10.3892/ijmm.2017.3105] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2017] [Accepted: 08/21/2017] [Indexed: 01/10/2023] Open
Abstract
Deep vein thrombosis (DVT) is a disease involving multiple genes and systems. MicroRNAs (miRNAs) represent a class of non-coding small RNAs that post-transcriptionally suppress their target genes. The expression patterns of miRNA and messenger RNA (mRNA) in DVT remain poorly characterized. The aim of the present study was to evaluate miRNA and mRNA expression profiles in a stasis-induced DVT rat model. Male SD rats were randomly divided into three groups as follows: DVT, sham and control. The inferior vena cava (IVC) of rats was ligated to construct stasis-induced DVT models. Rats were sacrificed three days after ligation, and morphological changes in the vein tissues were observed by hematoxylin and eosin and Masson staining. The miRNA and mRNA expression profiles were evaluated by microarrays, followed by bioinformatics analysis. The microarray analysis identified 22 miRNAs and 487 mRNAs that were significantly differentially expressed between the experimental and control groups, and between the experimental and sham groups, but not between the control and sham groups (P≤0.05; ≥2.0-fold change). By subsequent bioinformatics analysis, a 19 miRNA-98 mRNAs network was constructed in the stasis-induced DVT rat model. Notably, the majority of these miRNAs and mRNAs are reported to be expressed by endothelial cells (ECs) and are associated with the function of ECs. The results provide evidence indicating that the regulatory association of miRNA and mRNA points to key roles played by ECs in thrombosis. These findings advance our understanding of the molecular regulatory mechanisms underlying the pathophysiology of DVT.
Collapse
Affiliation(s)
- Qian-Qian Jin
- Department of Forensic Pathology, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Jun-Hong Sun
- Department of Forensic Pathology, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Qiu-Xiang Du
- Department of Forensic Pathology, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Xiao-Jun Lu
- Department of Forensic Pathology, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Xi-Yan Zhu
- Department of Forensic Pathology, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Hao-Liang Fan
- Department of Forensic Pathology, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Christian Hölscher
- Biochemical and Life Sciences, Lancaster University, Lancaster, Lancashire LA1 4YQ, UK
| | - Ying-Yuan Wang
- Department of Forensic Pathology, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| |
Collapse
|
17
|
Wong CJ, Koch M, Behling-Kelly EL. Development of a plasminogen activator inhibitor (PAI-1) assay and comparison of plasma PAI-1 activity in hyperlipidemic/dyslipidemic dogs with either hyperadrenocorticism or diabetes mellitus, and healthy dogs. Res Vet Sci 2017; 111:1-8. [DOI: 10.1016/j.rvsc.2016.11.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Revised: 10/19/2016] [Accepted: 11/05/2016] [Indexed: 01/11/2023]
|
18
|
Tsai FC, Lin YC, Chang SH, Chang GJ, Hsu YJ, Lin YM, Lee YS, Wang CL, Yeh YH. Differential left-to-right atria gene expression ratio in human sinus rhythm and atrial fibrillation: Implications for arrhythmogenesis and thrombogenesis. Int J Cardiol 2016; 222:104-112. [PMID: 27494721 DOI: 10.1016/j.ijcard.2016.07.103] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Revised: 07/05/2016] [Accepted: 07/07/2016] [Indexed: 12/26/2022]
Abstract
BACKGROUND Atrial fibrillation (AF) causes atrial remodeling, and the left atrium (LA) is the favored substrate for maintaining AF. It remains unclear if AF remodels both atria differently and contributes to LA arrhythmogenesis and thrombogenesis. Therefore, we wished to characterize the transcript profiles in the LA and right atrium (RA) in sinus rhythm (SR) and AF respectively. METHODS Paired LA and RA appendages acquired from patients receiving cardiac surgery were used for ion-channel- and whole-exome-based transcriptome analysis. The ultrastructure was evaluated by immunohistochemistry. RESULTS Twenty-two and twenty ion-channels and transporters were differentially expressed between the LA and RA in AF and SR, respectively. Among these, 15 genes were differentially expressed in parallel between AF and SR. AF was associated with increased LA/RA expression ratio in 9 ion channel-related genes, including genes related to calcium handling. In microarray, AF was associated with a differential LA/RA gene expression ratio in 309 genes, and was involved in atherosclerosis-related signaling. AF was associated with the upregulation of thrombogenesis-related genes in the LA appendage, including P2Y12, CD 36 and ApoE. Immunohistochemistry showed higher expressions of collagen-1, oxidative stress and TGF-β1 in the RA compared to the LA. CONCLUSIONS AF was associated with differential LA-to-RA gene expression related to specific ion channels and pathways as well as upregulation of thrombogenesis-related genes in the LA appendage. Targeting the molecular mechanisms underlying the LA-to-RA difference and AF-related remodeling in the LA appendage may help provide new therapeutic options in treating AF and preventing thromboembolism in AF.
Collapse
Affiliation(s)
- Feng-Chun Tsai
- Division of Cardiac Surgery, Chang-Gung Memorial Hospital, Chang-Gung University College of Medicine, Taoyuan, Taiwan
| | - Yen-Chen Lin
- Cardiovascular Division, Chang-Gung Memorial Hospital, Chang-Gung University College of Medicine, Taoyuan, Taiwan
| | - Shang-Hung Chang
- Cardiovascular Division, Chang-Gung Memorial Hospital, Chang-Gung University College of Medicine, Taoyuan, Taiwan
| | - Gwo-Jyh Chang
- Graduate Institute of Clinical Medical Sciences, Chang-Gung University College of Medicine, Chang-Gung University, Taiwan
| | - Yu-Juei Hsu
- Division of Nephrology, Department of Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Yuan-Min Lin
- School of Dentistry, National Yang-Ming University, Taipei, Taiwan
| | - Yun-Shien Lee
- Department of Biotechnology, Ming-Chuan University, Taoyuan, Taiwan
| | - Chun-Li Wang
- Cardiovascular Division, Chang-Gung Memorial Hospital, Chang-Gung University College of Medicine, Taoyuan, Taiwan
| | - Yung-Hsin Yeh
- Cardiovascular Division, Chang-Gung Memorial Hospital, Chang-Gung University College of Medicine, Taoyuan, Taiwan.
| |
Collapse
|
19
|
Swine Model of Thrombotic Caval Occlusion Created by Autologous Thrombus Injection with Assistance of Intra-caval Net Knitting. Sci Rep 2015; 5:18546. [PMID: 26680253 PMCID: PMC4683581 DOI: 10.1038/srep18546] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Accepted: 11/20/2015] [Indexed: 01/14/2023] Open
Abstract
To evaluate the feasibility of a swine model of thrombotic inferior vena cava (IVC) occlusion (IVCO) created by autologous thrombus injection with assistance of intra-caval net knitting. Sixteen pigs were included and divided into two groups: Group A (n = 10), IVCO model created by knitting a caval net followed by autologous thrombus injection; Group B (n = 6), control model created by knitting a net and normal saline injection. Venography was performed to assess each model and the associated thrombotic occlusion. The vessels were examined histologically to analyse the pathological changes postoperatively. IVCO model was successfully created in 10 animals in Group A (100%). Immediate venography showed extensive clot burden in the IVC. Postoperative venography revealed partial caval occlusion at 7 days, and complete occlusion coupled with collateral vessels at 14 days. Histologically, Group A animals had significantly greater venous wall thickening, with CD163-positive and CD3-positive cell infiltration. Recanalization channels were observed at the margins of the thrombus. By contrast, no thrombotic occlusion of the IVC was observed in Group B. The thrombotic IVCO model can be reliably established in swine. The inflammatory reaction may contribute to the caval thrombus propagation following occlusion.
Collapse
|
20
|
Zhou TB. Signaling pathway factors expression in renal tissue of apoE-knockout mice. J Recept Signal Transduct Res 2015; 35:435-438. [PMID: 26096163 DOI: 10.3109/10799893.2014.1000465] [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] [Indexed: 11/13/2022]
Abstract
Apolipoprotein E (apoE) is regarded as one of the major plasma lipoproteins, and it plays an important role in the transport and metabolism of lipids. apoE can be found in multiple tissues, such as liver, kidney, jejunum, urinary bladder, ileum, colon, brain, adrenal glands, lung, ovary, spleen, pancreas, and testis, etc. As a secreted protein, it plays an important role in the systemic lipoprotein metabolism and vascular wall homeostasis and in the pathogenesis of renal diseases. apoE-knockout (apoE(-/-)) mice is a classic model of atherosclerosis and renal diseases. However, no review summed up the signaling pathway factors expression in renal tissue of apoE-knockout mice. The literatures were searched extensively and this review was performed to review the signaling pathway factors expression in renal tissue of apoE-knockout mice.
Collapse
Affiliation(s)
- Tian-Biao Zhou
- a Department of Nephrology , The Sixth Affiliated Hospital, Sun Yat-Sen University , Guangzhou , China
| |
Collapse
|
21
|
Peng X, Li J, Yu X, Tan R, Zhu L, Wang J, Wang R, Gu G, Liu Q, Ren L, Wang C, Hu Q. Therapeutic effectiveness of bone marrow-derived mesenchymal stem cell administration against acute pulmonary thromboembolism in a mouse model. Thromb Res 2015; 135:990-999. [PMID: 25712897 DOI: 10.1016/j.thromres.2015.02.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2014] [Revised: 01/31/2015] [Accepted: 02/08/2015] [Indexed: 02/04/2023]
Abstract
INSTRUCTION Acute pulmonary thromboembolism (APTE) is a common clinical condition associated with significant morbidity and mortality. Although promising, bone marrow-derived mesenchymal stem cell (BMSC) treatment for thrombus resolution remains controversial. The therapeutic effectiveness of BMSC against APTE has not been evaluated. This study aims to determine whether BMSCs administration is effective in mouse model. MATERIALS AND METHODS Therapeutic efficacy of female and male BMSCs were evaluated by applying serial sectioning analysis method for the whole lungs of APTE mice and calculating each thrombus size in volume. Plasmid construction and stable transfection were used to manipulate expression of glyceraldehyde-3-phosphate dehydrogenase (GAPDH) in both genders of BMSCs. Western blot were performed to detect GAPDH and urokinase plasminogen activator expression in BMSCs. RESULTS Our data showed, 1) compared with non-serial sectioning method, the serial sectioning method detected more thrombi, larger size ranges of thrombus area, and the volume of each individual thrombus. 2) BMSCs significantly decreased the thrombi size in APTE mice, with female BMSCs superior to male ones. 3) female BMSCs showed a higher GAPDH protein level and manipulations of GAPDH expression in female or male BMSCs profoundly affected their therapeutic efficacies as well as urokinase plasminogen activator expression. CONCLUSION This study indicates serial-sectioning analysis method is necessary for evaluating APTE and provides strong evidences for BMSCs possessing therapeutic effectiveness against APTE, with female BMSCs superior to male counterparts. GAPDH played a critical role in the superior function of female BMSCs, possibly by regulating the expression of urokinase plasminogen activator.
Collapse
Affiliation(s)
- Xiaochun Peng
- Department of Pathophysiology, Tongji School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Key Laboratory of Pulmonary Diseases of Ministry of Health, Tongji School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jiansha Li
- Key Laboratory of Pulmonary Diseases of Ministry of Health, Tongji School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xiangyuan Yu
- Department of Pathophysiology, Tongji School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Key Laboratory of Pulmonary Diseases of Ministry of Health, Tongji School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Rubin Tan
- Department of Pathophysiology, Tongji School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Key Laboratory of Pulmonary Diseases of Ministry of Health, Tongji School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Liping Zhu
- Department of Pathophysiology, Tongji School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Key Laboratory of Pulmonary Diseases of Ministry of Health, Tongji School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jun Wang
- Department of Physiology, Capital Medical University, Beijing 100730, China
| | - Rongshuai Wang
- Department of Forensic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Guoqiang Gu
- Department of Forensic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Qian Liu
- Department of Forensic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Liang Ren
- Department of Forensic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Chen Wang
- Department of Respiratory Medicine, Beijing Hospital, Ministry of Health, Beijing 100730, China.
| | - Qinghua Hu
- Department of Pathophysiology, Tongji School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Key Laboratory of Pulmonary Diseases of Ministry of Health, Tongji School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|
22
|
Diaz JA, Farris DM, Wrobleski SK, Myers DD, Wakefield TW. Inferior vena cava branch variations in C57BL/6 mice have an impact on thrombus size in an IVC ligation (stasis) model. J Thromb Haemost 2015; 13:660-4. [PMID: 25683196 DOI: 10.1111/jth.12866] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Accepted: 11/30/2014] [Indexed: 11/29/2022]
Abstract
BACKGROUND Animal models of venous thrombosis (VT) are critical tools for those investigating the VT mechanism. Recently, inferior vena cava (IVC) branches have been subject to debate, causing controversy in the field. OBJECTIVES To understand how the variability of IVC branches, in commonly used C57BL/6 mice, have an impact on thrombus formation in the IVC ligation model. METHODS C57BL/6 male mice (n = 46), 20-25 g, were subjected to the IVC ligation model with various interruptions of the IVC branches. Control animals (n = 50) had all branches interrupted. Two days after IVC ligation, thrombus weight (TW), as a parameter of thrombus size, was assessed. RESULTS We found four different anatomical patterns. Side branches were more prevalent on the mouse's right side (34%) compared with the left (20%). In mice where side branches were absent (21%), back branches appeared larger. Also, 25% of mice had both side branches. Controls that had all IVC branches interrupted had the most consistent and largest TW (32.6 mg to 34.7 mg) while groups that had no back branches interrupted had the smallest TW (3.6-9.7 mg), a 4 to 9-fold decrease. All groups with open back branches had significantly smaller TW (P < 0.05) than controls. CONCLUSIONS Variations in TW were observed based on different branch interruption patterns, compared with the fully ligated controls. Having two back branches was the most consistent anatomy and open back branches had the largest negative impact on thrombus size. This work confirms that the IVC branches significantly affect thrombus burden in C57BL/6 mice and further studies should be conducted in order to standardize this and other animal models of VT.
Collapse
Affiliation(s)
- J A Diaz
- Department of Surgery, Section of Vascular Surgery, Conrad Jobst Vascular Research Laboratories, Ann Arbor, MI, USA
| | | | | | | | | |
Collapse
|
23
|
Increased 18F-FDG uptake is predictive of rupture in a novel rat abdominal aortic aneurysm rupture model. Ann Surg 2015; 261:395-404. [PMID: 24651130 DOI: 10.1097/sla.0000000000000602] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
OBJECTIVE To determine whether F-fluorodeoxyglucose (F-FDG) micro-positron emission tomography (micro-PET) can predict abdominal aortic aneurysm (AAA) rupture. BACKGROUND An infrarenal AAA model is needed to study inflammatory mechanisms that drive rupture. F-FDG PET can detect vascular inflammation in animal models and patients. METHODS After exposing Sprague-Dawley rats to intra-aortic porcine pancreatic elastase (PPE) (12 U/mL), AAA rupture was induced by daily, subcutaneous, β-aminopropionitrile (BAPN, 300 mg/kg, N = 24) administration. Negative control AAA animals (N = 15) underwent daily saline subcutaneous injection after PPE exposure. BAPN-exposed animals that did not rupture served as positive controls [nonruptured AAA (NRAAA) 14d, N = 9]. Rupture was witnessed using radiotelemetry. Maximum standard uptakes for F-FDG micro-PET studies were determined. Aortic wall PAI-1, uPA, and tPA concentrations were determined by western blot analyses. Interleukin (IL)-1β, IL-6, IL-10, and MIP-2 were determined by Bio-Plex bead array. Neutrophil and macrophage populations per high-power field were quantified. Matrix metalloproteinase (MMP) activities were determined by zymography. RESULTS When comparing ruptured AAA (RAAA) to NRAAA 14d animals, increased focal F-FDG uptakes were detected at subsequent sites of rupture (P = 0.03). PAI-1 expression was significantly less in RAAA tissue (P = 0.01), with comparable uPA and decreased tPA levels (P = 0.02). IL-1β (P = 0.04), IL-6 (P = 0.001), IL-10 (P = 0.04), and MIP-2 (P = 0.02) expression, neutrophil (P = 0.02) and macrophage presence (P = 0.002), and MMP9 (P < 0.0001) activity were increased in RAAA tissue. CONCLUSIONS With this AAA rupture model, increased prerupture F-FDG uptake on micro-PET imaging was associated with increased inflammation in the ruptured AAA wall. F-FDG PET imaging may be used to monitor inflammatory changes before AAA rupture.
Collapse
|
24
|
Abstract
In this chapter, an overview of some of the prominent risk factors that contribute to the pathophysiology of venous thrombosis will be discussed. In 1856, Dr Rudolf Virchow developed the concept outlining the genesis of intravascular thrombosis. Dr Virchow hypothesized that circulatory stasis due to interrupted blood flow, changes in the blood leading to blood coagulation, and irritation or damage to the vascular endothelium would initiate acute venous thrombus generation. Presently, it is known that these above-mentioned risk factors are influenced by increasing age, gender, and obesity. The current chapter will focus on recent preclinical and clinical investigations that will give the reader insight into the prothrombotic mechanisms that lead to acute venous thrombosis.
Collapse
Affiliation(s)
- DD Myers
- Department of Surgery, Section of Vascular Surgery, University of Michigan, Ann Arbor, USA
- Unit for Laboratory Animal Medicine, University of Michigan, Ann Arbor, USA
| |
Collapse
|
25
|
Abstract
Galectin-3-binding protein (gal3bp) and its receptor/ligand, galectin-3 (gal3), are secreted proteins that initiate signaling cascades in several diseases, and recent human proteomic data suggest they may play a role in venous thrombosis (VT). We hypothesized that gal3bp and gal3 may promote VT. Using a mouse stasis model of VT, we found that gal3bp and gal3 were localized on vein wall, red blood cells, platelets, and microparticles, whereas leukocytes expressed gal3 only. Gal3 was dramatically increased during early VT and gal3bp:gal3 colocalized in the leukocyte/endothelial cell interface, where leukocytes were partially attached to the vein wall. Thrombus size correlated with elevated gal3 and interleukin-6 (IL-6) vein wall levels. Recombinant gal3 promoted VT and increased vein wall IL-6 mRNA. Although recombinant gal3 restored the VT size in gal3(-/-) mice, it had no effect on IL6(-/-) mice, suggesting that gal3:gal3bp promotes VT through IL-6. Moreover, significantly fewer activated neutrophils were present in the gal3(-/-) vein walls. In a group of human patients, elevated circulating gal3bp correlated with acute VT. In conclusion, gal3bp:gal3 play a critical role in VT, likely via IL-6 and PMN-mediated thrombotic mechanisms, and may be a potential biomarker in human VT.
Collapse
|
26
|
Obi AT, Diaz JA, Ballard-Lipka NL, Roelofs KJ, Farris DM, Lawrence DA, Henke PK, Wakefield TW. Low-molecular-weight heparin modulates vein wall fibrotic response in a plasminogen activator inhibitor 1-dependent manner. J Vasc Surg Venous Lymphat Disord 2014; 2:441-450.e1. [PMID: 25419511 PMCID: PMC4235166 DOI: 10.1016/j.jvsv.2014.02.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
BACKGROUND Treatment with low-molecular-weight heparin (LMWH) favorably alters the vein wall response to deep venous thrombosis (DVT), although the mechanisms remain unclear. Previous studies have suggested that LMWH alters the levels of circulating plasminogen activator inhibitor 1 (PAI-1), a known mediator of fibrosis, and may improve endogenous fibrinolysis. We hypothesized that LMWH favorably alters the vein wall response by binding of PAI-1 and acceleration of fibrinolysis. METHODS Wild-type and PAI-1 -/- mice underwent treatment with LMWH after induction of occlusive DVT. Vein wall and plasma were harvested and analyzed by enzyme-linked immunosorbent assay, zymography, real-time polymerase chain reaction, and immunohistochemistry. RESULTS Wild-type mice treated with LMWH exhibited diminished vein wall fibrosis (0.6 ± 0.6 vs 1.4 ± 0.2; P < .01; n = 5) and elevation of circulating PAI-1 (1776 ± 342 vs 567 ± 104 ρg/mL; P < .01; n = 5) compared with untreated controls after occlusive DVT. PAI-1-/- mice treated with LMWH were not similarly protected from fibrosis, despite improved thrombus resolution. Treatment with LMWH was associated with decreased intrathrombus interleukin-lβ (68.6 ± 31.0 vs 223.4 ± 28.9 ρg/mg total protein; P < .01; n = 5) but did not alter inflammatory cell recruitment to the vein wall. PAI-1 -/- mice exhibited significantly elevated intrathrombus (257.2 ± 51.5 vs 4.3 ± 3.8 ρg/mg total protein; n = 5) and vein wall interleukin-13 (187.2 ± 57.6 vs 9.9 ± 1.1 ρg/mg total protein; P < .05; n = 5) as well as vein wall F4/80 positively staining monocytes (53 ± 11 vs 16 ± 2 cells/5 high-power fields; P < .05; n = 4). CONCLUSIONS LMWH did not accelerate venous thrombosis resolution but did protect against vein wall fibrosis in a PAI-1-dependent manner in an occlusive DVT model. Lack of PAI-1 correlated with accelerated venous thrombosis resolution but no protection from fibrosis. PAI-1 inhibition as a treatment strategy for DVT is likely to accelerate clearance of the thrombus but may come at the expense of increased vein wall fibrosis. CLINICAL RELEVANCE The pathophysiologic mechanism of post-thrombotic syndrome is not well understood clinically or experimentally. In this study, we evaluated the effect of the prominent fibrinolytic mechanism, plasminogen activator inhibitor 1 (PAI-1), and low-molecular-weight heparin (LMWH) on vein wall injury after thrombosis. We show here that LMWH is protective from vein wall fibrosis, but this is abrogated in PAI-1-deleted mice. This is also correlated with monocyte vein wall influx. These data support the clinical observation that LMWH may be protective from post-thrombotic vein wall injury in a PAI-1-dependent manner.
Collapse
Affiliation(s)
- Andrea T. Obi
- Section of Vascular Surgery, Department of Surgery, Conrad Jobst Vascular Research Laboratories, Ann Arbor, Mich
| | - Jose A. Diaz
- Section of Vascular Surgery, Department of Surgery, Conrad Jobst Vascular Research Laboratories, Ann Arbor, Mich
| | - Nicole L. Ballard-Lipka
- Section of Vascular Surgery, Department of Surgery, Conrad Jobst Vascular Research Laboratories, Ann Arbor, Mich
| | - Karen J. Roelofs
- Section of Vascular Surgery, Department of Surgery, Conrad Jobst Vascular Research Laboratories, Ann Arbor, Mich
| | - Diana M. Farris
- Section of Vascular Surgery, Department of Surgery, Conrad Jobst Vascular Research Laboratories, Ann Arbor, Mich
| | - Daniel A. Lawrence
- Department of Medicine, University of Michigan Medical School, Ann Arbor, Mich
| | - Peter K. Henke
- Section of Vascular Surgery, Department of Surgery, Conrad Jobst Vascular Research Laboratories, Ann Arbor, Mich
| | - Thomas W. Wakefield
- Section of Vascular Surgery, Department of Surgery, Conrad Jobst Vascular Research Laboratories, Ann Arbor, Mich
| |
Collapse
|
27
|
Gardiner EE, Medcalf RL. Is plasminogen activator inhibitor type 2 really a plasminogen activator inhibitor after all? J Thromb Haemost 2014; 12:1703-5. [PMID: 25142275 DOI: 10.1111/jth.12704] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Indexed: 11/30/2022]
Affiliation(s)
- E E Gardiner
- Australian Centre for Blood Diseases, Monash University, Melbourne, Victoria, Australia
| | | |
Collapse
|
28
|
Siefert SA, Chabasse C, Mukhopadhyay S, Hoofnagle MH, Strickland DK, Sarkar R, Antalis TM. Enhanced venous thrombus resolution in plasminogen activator inhibitor type-2 deficient mice. J Thromb Haemost 2014; 12:1706-16. [PMID: 25041188 PMCID: PMC4194171 DOI: 10.1111/jth.12657] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Accepted: 06/30/2014] [Indexed: 11/28/2022]
Abstract
BACKGROUND The resolution of deep vein thrombosis requires an inflammatory response and mobilization of proteases, such as urokinase-type plasminogen activator (uPA) and matrix metalloproteinases (MMPs), to degrade the thrombus and remodel the injured vein wall. Plasminogen activator inhibitor type 2 (PAI-2) is a serine protease inhibitor (serpin) with unique immunosuppressive and cell survival properties that was originally identified as an inhibitor of uPA. OBJECTIVE To investigate the role of PAI-2 in venous thrombus formation and resolution. METHODS Venous thrombus resolution was compared in wild-type C57BL/6, PAI-2(-/-) , and PAI-1(-/-) mice using the stasis model of deep vein thrombosis. Formed thrombi were harvested, thrombus weights were recorded, and tissue was analyzed for uPA and MMP activities, PAI-1 expression, and the nature of inflammatory cell infiltration. RESULTS We found that the absence of PAI-2 enhanced venous thrombus resolution, while thrombus formation was unaffected. Enhanced venous thrombus resolution in PAI-2(-/-) mice was associated with increased uPA activity and reduced levels of PAI-1, with no significant effect on MMP-2 and -9 activities. PAI-1 deficiency resulted in an increase in thrombus resolution similar to PAI-2 deficiency, but additionally reduced venous thrombus formation and altered MMP activity. PAI-2-deficient thrombi had increased levels of the neutrophil chemoattractant CXCL2, which was associated with early enhanced neutrophil recruitment. CONCLUSIONS These data identify PAI-2 as a novel regulator of venous thrombus resolution, which modulates several pathways involving both inflammatory and uPA activity mechanisms, distinct from PAI-1. Further examination of these pathways may lead to potential therapeutic prospects in accelerating thrombus resolution.
Collapse
Affiliation(s)
- S A Siefert
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, USA; Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, USA
| | | | | | | | | | | | | |
Collapse
|
29
|
Obi AT, Diaz JA, Ballard-Lipka NL, Roelofs KJ, Farris DM, Lawrence DA, Wakefield TW, Henke PK. Plasminogen activator-1 overexpression decreases experimental postthrombotic vein wall fibrosis by a non-vitronectin-dependent mechanism. J Thromb Haemost 2014; 12:1353-63. [PMID: 24943740 PMCID: PMC4127110 DOI: 10.1111/jth.12644] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Accepted: 06/12/2014] [Indexed: 12/30/2022]
Abstract
BACKGROUND Factors associated with postthrombotic syndrome are known clinically, but the underlying cellular processes at the vein wall are not well delineated. Prior work suggests that vein wall damage does not correlate with thrombus resolution but rather with plasminogen activator-1 (PAI-1) and matrix metalloproteinase (MMP) activity. OBJECTIVE We hypothesized that PAI-1 would confer post venous thrombosis (VT) vein wall protection via a vitronectin (Vn)-dependent mechanism. METHODS A stasis model of VT was used with harvest over 2 weeks, in wild-type, Vn(-/-) , and PAI-1-overexpressing mice (PAI-1 Tg). RESULTS PAI-1 Tg mice had larger VT at 6 and 14 days, compared to controls, but Vn(-/-) mice had no alteration of VT resolution. Gene deletion of Vn resulted in an increase in, rather than the expected decrease in, circulating PAI-1 activity. While both Vn(-/-) and PAI-1 Tg had attenuated intimal fibrosis, PAI-1 Tg had significantly less vein wall collagen and a compensatory increase in collagen III gene expression. Both Vn(-/-) and PAI-1 Tg vein wall had less monocyte chemotactic factor-1 and fewer macrophages (F4/80), with significantly less MMP-2 activity and decreased TIMP-1 antigen. Ex vivo assessment of transforming growth factor β-mediated fibrotic response showed that PAI-1 Tg vein walls had increased profibrotic gene expression (collagens I and III, MMP-2, and α-smooth muscle actin) compared with controls, opposite of the in vivo response. CONCLUSIONS The absence of Vn increases circulating PAI-1, which positively modulates vein wall fibrosis in a dose-dependent manner. Translationally, PAI-1 elevation may decrease vein wall damage after deep vein thrombosis, perhaps by decreasing macrophage-mediated activities.
Collapse
Affiliation(s)
- A T Obi
- Conrad Jobst Vascular Research Laboratory, Section of Vascular Surgery, Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Park KM, Lee SH. Anti-hyperlipidemic activity of Rhynchosia nulubilis seeds pickled with brown rice vinegar in mice fed a high-fat diet. Nutr Res Pract 2013; 7:453-9. [PMID: 24353830 PMCID: PMC3865267 DOI: 10.4162/nrp.2013.7.6.453] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2013] [Revised: 07/17/2013] [Accepted: 07/18/2013] [Indexed: 11/13/2022] Open
Abstract
The abnormal content of blood lipids often results in metabolic diseases, such as hyperlipidemia and obesity. Many agents, including natural sources from traditional food, have been developed to regulate the blood lipid contents. In this study, we examined the anti-hyperlipidemic activity of Rhynchosia nulubilis seeds pickled with brown rice vinegar (RNSpBRV), a Korean traditional pickled soybean food. Since RNSpBRV is made of R. nulubilis seeds (RNS) soaked in brown rice vinegar (BRV), we compared the anti-adipogenic activity between RNS, BRV and solid fraction of RNSpBRV (SF-RNSpBRV), liquid fraction of RNSpBRV (LF-RNSpBRV). For this, the inhibitory effect of lipid accumulation in 3T3-L1 adipocyte was checked by adding methanol extracts of mixed RNS and BRV, LF-RNSpBRV, and SF-RNSpBRV. The addition of each methanol extract up to 1 mg/ml showed no cytotoxicity on 3T3-L1 adipocyte, and approximately 20% of the lipid droplet formation was suppressed with the methanol extract of BRL or SF-RNSpBRV. The highest suppression (42.1%) was achieved with LF-RNSpBRV. In addition, mice fed a high fat diet (HFD) supplemented with 5% RNSpBRV powder led to increased high density lipoprotein (HDL) cholesterol and lower blood glucose, triglyceride, and total cholesterol compared to mice fed with a HFD diet only. Interestingly, the size of the epididymis cells gradually decreased in HFD + 1% RNSpBRV- and HFD + 5% RNSpBRV-fed mice if compared those of HFD-fed mice. Taken together, these results provide evidence that RNSpBRV has a regulatory role in lipid metabolism that is related to hyperlipidemia.
Collapse
Affiliation(s)
- Ki-Moon Park
- Department of Food Science and Biotechnology, Sungkyunkwan University, Suwon 440-746, Korea
| | - Seung Ho Lee
- Major of Nano-Bioengineering, Incheon National University, 119 Academy-ro, Yeonsu-gu, Incheon 406-772, Korea
| |
Collapse
|
31
|
Hyperlipidemia, tissue factor, coagulation, and simvastatin. Trends Cardiovasc Med 2013; 24:95-8. [PMID: 24016468 DOI: 10.1016/j.tcm.2013.07.003] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2013] [Revised: 07/02/2013] [Accepted: 07/02/2013] [Indexed: 11/20/2022]
Abstract
Hyperlipidemia affects millions of people worldwide and is a major risk factor for cardiovascular disease. People with hyperlipidemia have elevated levels of serum cholesterol and an increased risk of thrombosis. Studies have suggested that oxidized lipoproteins, such as oxidized low-density lipoprotein (oxLDL), contribute to the development of a pro-thrombotic state. In this review, we discuss our recent studies demonstrating a role for hematopoietic cell-derived tissue factor (TF) expression in the activation of coagulation and increased thrombosis associated with hyperlipidemia. In addition, we investigated the effect of simvastatin on TF expression and coagulation. We found that simvastatin reduced leukocyte TF expression, TF⁺ microparticles, and coagulation. These results and earlier studies suggest that the anti-coagulant activity of statins is due, in part, to their ability to reduce monocyte TF expression in patients with cardiovascular disease.
Collapse
|
32
|
Zhou TB. Signaling pathways of apoE and its role of gene expression in glomerulus diseases. J Recept Signal Transduct Res 2013; 33:73-78. [PMID: 23384034 DOI: 10.3109/10799893.2013.765466] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The roles of apolipoprotein E (apoE) in regulating plasma lipids and lipoproteins levels have been investigated for over several decades. However, in different tissues/cells, the role of apoE was different, such as that it was a risk factor for cancer, but some reports stated that apoE was a protective factor for renal diseases. At the moment, most of the studies find that apoE not only acts as a ligand for metabolism of lipids, but also plays as a factor to regulate lots of signaling pathways. There was rare review to sum up the signaling pathways for apoE, and there was also rare review to widely review the gene expression of apoE in glomerulus diseases. This review was performed to provide a relatively complete signaling pathways flowchart for apoE to the investigators who were interested in the roles of apoE in the pathogenesis of glomerulus diseases. In the past decades, some studies were also performed to explore the association of apoE gene expression with the risk of glomerulus diseases. However, the role of apoE in the pathogenesis of glomerulus diseases was controversial. Here, the signal transduction pathways of apoE and its role of gene expression in the pathogenesis of glomerulus diseases were reviewed.
Collapse
Affiliation(s)
- Tian-Biao Zhou
- Department of Pediatric Nephrology, The First Affiliated Hospital of Guangxi Medical University, Guangxi, China.
| |
Collapse
|
33
|
Diaz JA, Alvarado CM, Wrobleski SK, Slack DW, Hawley AE, Farris DM, Henke PK, Wakefield TW, Myers DD. The electrolytic inferior vena cava model (EIM) to study thrombogenesis and thrombus resolution with continuous blood flow in the mouse. Thromb Haemost 2013; 109:1158-69. [PMID: 23571406 DOI: 10.1160/th12-09-0711] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2012] [Accepted: 02/19/2013] [Indexed: 11/05/2022]
Abstract
Previously, we presented the electrolytic inferior vena cava (IVC) model (EIM) during acute venous thrombosis (VT). Here, we present our evaluation of the EIM for chronic VT time points in order to determine whether this model allows for the study of thrombus resolution. C57BL/6 mice (n=191) were utilised. In this model a copper-wire, inserted into a 25-gauge needle, is placed in the distal IVC and another subcutaneously. An electrical current (250 μAmp/15 minutes) activates the endothelial cells, inducing thrombogenesis. Ultrasound, thrombus weight (TW), vein wall leukocyte counts, vein wall thickness/fibrosis scoring, thrombus area and soluble P-selectin (sP-sel) were performed at baseline, days 1, 2, 4, 6, 9, 11 and 14, post EIM. A correlation between TW and sP-sel was also determined. A thrombus formed in each mouse undergoing EIM. Blood flow was documented by ultrasound at all time points. IVC thrombus size increased up to day 2 and then decreased over time, as shown by ultrasound, TW, and sP-sel levels. TW and sP-sel showed a strong positive correlation (r=0.48, p<0.0002). Vein wall neutrophils were the most common cell type present in acute VT (up to day 2) with monocytes becoming the most prevalent in chronic VT (from day 6 to day 14). Thrombus resolution was demonstrated by ultrasound, TW and thrombus area. In conclusion, the EIM produces a non-occlusive and consistent IVC thrombus, in the presence of constant blood flow, allowing for the study of VT at both acute and chronic time points. Thrombus resolution was demonstrated by all modalities utilised in this study.
Collapse
Affiliation(s)
- Jose A Diaz
- Department of Surgery, Section of Vascular Surgery, Conrad Jobst Vascular Research Laboratories, University of Michigan, North Campus Research Complex (NCRC), 2800 Plymouth Road, B26, R251N, Ann Arbor, MI 48105-0654, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Patterson KA, Zhang X, Wrobleski SK, Hawley AE, Lawrence DA, Wakefield TW, Myers DD, Diaz JA. Rosuvastatin reduced deep vein thrombosis in ApoE gene deleted mice with hyperlipidemia through non-lipid lowering effects. Thromb Res 2013; 131:268-76. [PMID: 23276528 PMCID: PMC3594437 DOI: 10.1016/j.thromres.2012.12.006] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2012] [Revised: 12/02/2012] [Accepted: 12/07/2012] [Indexed: 11/24/2022]
Abstract
INTRODUCTION Statins, particularly rosuvastatin, have recently become relevant in the setting of venous thrombosis. The objective of this study was to study the non-lipid lowering effects of rosuvastatin in venous thrombosis in mice with hyperlipidemia. MATERIALS AND METHODS An inferior vena cava ligation model of venous thrombosis in mice was utilized. Saline or 5mg/kg of rosuvastatin was administered by gavage 48hs previous to thrombosis. Blood, the inferior vena cava, thrombus, and liver were harvested 3, 6hours, and 2days post-thrombosis. Thrombus weight, inflammatory markers, and plasminogen activator inhibitor-1 expression and plasma levels were measured. Also, neutrophil migration to the IVC was assessed. RESULTS Rosuvastatin significantly decreased thrombus weight, plasminogen activator inhibitor-1 expression and plasma levels, expression of molecules related to the interleukin-6 pathway, and neutrophil migration into the vein wall. CONCLUSIONS This work supports the beneficial effects of rosuvastatin on venous thrombosis in mice with hyperlipidemia, due to its non-lipid lowering effects.
Collapse
Affiliation(s)
- K A Patterson
- Unit for Laboratory Animal Medicine, University of Michigan, Ann Arbor, MI 48103, USA
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Diaz JA, Obi AT, Myers DD, Wrobleski SK, Henke PK, Mackman N, Wakefield TW. Critical review of mouse models of venous thrombosis. Arterioscler Thromb Vasc Biol 2012; 32:556-62. [PMID: 22345593 DOI: 10.1161/atvbaha.111.244608] [Citation(s) in RCA: 198] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Deep vein thrombosis and pulmonary embolism are a significant health care concern, representing a major source of mortality and morbidity. In order to understand the pathophysiology of thrombogenesis and thrombus resolution, animal models are necessary. Mouse models of venous thrombosis contribute to our understanding of the initiation, propagation, and resolution of venous thrombus, as well as allow for the evaluation of new pharmaceutical approaches to prophylaxis and treatment of deep vein thrombosis. In this work we review the ferric chloride model, the inferior vena cava ligation model, the inferior vena cava stenosis models, and the electrolytic inferior vena cava model and compare their advantages and disadvantages.
Collapse
Affiliation(s)
- Jose A Diaz
- Department of Surgery, Conrad Jobst Vascular Research Laboratories, University of Michigan, A570 MSRB II, Dock #6, 1150 W. Medical Center Drive, Ann Arbor, MI 48109-0654, USA.
| | | | | | | | | | | | | |
Collapse
|