1
|
Davis J, Maranto M, Kennedy J, Wang X, Azhar M, Jain A, Evans CE. Transforming Growth Factors in Venous Thrombus Formation and Resolution. Arterioscler Thromb Vasc Biol 2025; 45:643-653. [PMID: 40109257 PMCID: PMC12018122 DOI: 10.1161/atvbaha.124.322395] [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] [Indexed: 03/22/2025]
Abstract
Deep vein thrombosis (DVT) and pulmonary embolism are vascular occlusive disorders categorized under the term venous thromboembolism. Venous thromboembolism affects ≈900 000 people per year in the United States alone. Understanding of the multifaceted process of DVT has improved in recent years, and current DVT treatments reduce thrombus propagation, but they also increase bleeding risk and fail to accelerate natural venous thrombus resolution. Multiple inflammatory cytokines regulate the development and subsequent resolution of DVT. One family of cytokines involved in DVT and venous thrombus resolution is the TGF-β (transforming growth factor-β) family. A comprehensive understanding of the control of venous thrombus formation and resolution by the TGF-β family could lead to the development of novel treatments for DVT that target ≥1 of the TGF-β isoforms. The aim of this review is to describe studies of the roles of the TGF-β isoforms in venous thrombus formation and resolution and to highlight opportunities for future research. TGF-β isoforms include TGF-β1, TGF-β2, and TGF-β3. TGF-β1 has a well-characterized role in the positive regulation of venous thrombus formation and the negative regulation of venous thrombus resolution. Further research is necessary, however, to understand the potential roles of TGF-β2 and TGF-β3 in venous thrombus formation and resolution. Given that TGF-β1 expression increases during venous thrombosis and that inhibition or knockdown of TGF-β1 reduces thrombus burden, TGF-β1 represents a potential diagnostic marker for DVT and a putative target for therapies that aim to prevent or treat DVT.
Collapse
Affiliation(s)
- Jonathan Davis
- Cardiovascular Translational Research Center, University of South Carolina School of Medicine, Columbia, South Carolina, USA
| | - Molly Maranto
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Columbia, South Carolina, USA
| | - Jonathan Kennedy
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, South Carolina, USA
| | - Xiaoqin Wang
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, South Carolina, USA
| | - Mohamad Azhar
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, South Carolina, USA
| | - Abhishek Jain
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas, USA
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston Methodist Hospital, Houston, Texas, USA
- Department of Medical Physiology, Texas A&M Health Science Center, Bryan, Texas, USA
| | - Colin E. Evans
- Cardiovascular Translational Research Center, University of South Carolina School of Medicine, Columbia, South Carolina, USA
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, South Carolina, USA
- Department of Biomedical Engineering, University of South Carolina, Columbia, South Carolina, USA
- Institute on Cardiovascular Disease Research, University of South Carolina, Columbia, USA
| |
Collapse
|
2
|
Adinata A, Hara T, Achyar AC, Suzuki Y, Hirata KI, Otake H, Emoto N. Usefulness of serial in vivo imaging to directly assess the role of inflammation in thrombus resolution and organization. Biochem Biophys Res Commun 2025; 747:151293. [PMID: 39793399 DOI: 10.1016/j.bbrc.2025.151293] [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: 12/31/2024] [Accepted: 01/05/2025] [Indexed: 01/13/2025]
Abstract
Deep vein thrombosis (DVT) remains a significant health problem. Although animal models have provided significant insights into the DVT pathophysiology, time-course assessment in a same animal is technically limited. Recently, we reported a novel murine saphenous DVT model for in vivo visualization of spatiotemporal dynamics of inflammatory cells. This study further shed a light on the resolution and organization process of DVT using serial in vivo imaging technique. Similar with ferric chloride-induced thrombus model, our saphenous DVT model allowed serial in vivo imaging with fluorescence microscopy. However, unlike ferric chloride-induced thrombus model, we observed a significant decrease of DVT burden. Red blood cells area gradually decreased followed by fibrin and collagen deposition over time, although ferric chloride model induced platelet-rich arterial thrombus. Histological assessment revealed that neutrophils influx peaked 3 h after DVT induction, followed by macrophages' migration at 120 h' post-induction, indicating similar organization process with traditional stasis-induced DVT model. Ly6G/Ly6C positive cells at 3 h predicted the reduction of DVT burden (r > 0.8; P < 0.01), suggesting that inflammatory response at acute phase plays pivotal role in DVT resolution. MMP-9 expression was observed and colocalized with neutrophils at early timepoints in both traditional stasis-induced DVT model and our femoral imaging models. Taken together, our in vivo imaging model might allow better understanding of the resolution and organization processes in DVT.
Collapse
Affiliation(s)
- Aditya Adinata
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan; Laboratory of Clinical Pharmaceutical Science, Kobe Pharmaceutical University, Kobe, Japan
| | - Tetsuya Hara
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan; Laboratory of Clinical Pharmaceutical Science, Kobe Pharmaceutical University, Kobe, Japan.
| | - Arinal Chairul Achyar
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan; Laboratory of Clinical Pharmaceutical Science, Kobe Pharmaceutical University, Kobe, Japan
| | - Yoko Suzuki
- Laboratory of Clinical Pharmaceutical Science, Kobe Pharmaceutical University, Kobe, Japan
| | - Ken-Ichi Hirata
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Hiromasa Otake
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Noriaki Emoto
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan; Laboratory of Clinical Pharmaceutical Science, Kobe Pharmaceutical University, Kobe, Japan
| |
Collapse
|
3
|
Morrison L, Smoody B, Woltjer R, Hinds MT, Loftis JM, Wyatt CW, Nguyen KP. Ferumoxytol-enhanced MRI assessment of venous Thrombus resolution and macrophage content in a murine deep vein thrombosis model. Thromb Res 2024; 240:109063. [PMID: 38878741 PMCID: PMC11239555 DOI: 10.1016/j.thromres.2024.109063] [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: 02/26/2024] [Revised: 05/29/2024] [Accepted: 06/11/2024] [Indexed: 07/07/2024]
Abstract
BACKGROUND Imaging evaluation of acute deep vein thrombosis (DVT) or post-thrombotic syndrome (PTS) in animal or clinical models is limited to anatomical assessment of the location and extent of thrombi. We hypothesize that Fe-MRI, used to evaluate macrophage content in other inflammatory diseases, can be useful to evaluate the thromboinflammatory features after DVT over time. METHODS Nineteen wild-type CD-1 mice underwent surgical IVC ligation to induce DVT. Mice received either saline or 5 mg/kg of 14E11, a Factor XI inhibitor, before the procedure. Fe-MRI was performed on days 6-7 after ligation to evaluate thrombus volume, perfusion, and macrophage content via T2-weighted images. Mice were euthanized at days 3-15 after surgery. The thrombi and adjacent vein walls were excised, weighed, formalin-fixed, and paraffin-embedded for immunohistological analysis. Specimens were stained with specific antibodies to evaluate macrophage content, collagen deposition, neovascularization, and recanalization. Significance was determined using the Mann-Whitney U or Student's t-test. RESULTS After IVC-ligation in control mice, thrombus weights decreased by 59 % from day 3 to 15. Thrombus volumes peaked on day 5 before decreasing by 85 % by day 13. FXI inhibition led to reduced macrophage content in both thrombi (p = .008) and vein walls (p = .01), decreased thrombus volume (p = .03), and decreased thrombus mass (p = .01) compared to control mice. CCR2+ staining corroborated these findings, showing significantly reduced macrophage presence in the thrombi (p = .002) and vein wall (p = .002). CONCLUSIONS Fe-MRI T2 relaxation times can be used to characterize and quantify post-thrombotic changes of perfusion, macrophage content, and thrombus volume over time in a surgical mouse model of venous thrombosis. This approach could lead to better quantification of in vivo inflammation correlating monocyte and macrophage content within resolving thrombi and veins and may serve as a useful tool for research and clinically in the evaluation of the post-thrombotic environment.
Collapse
Affiliation(s)
- L Morrison
- Department of Surgery, Division of Vascular Surgery, School of Medicine, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA.
| | - B Smoody
- Department of Radiology, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA; Advanced Imaging Research Center (AIRC), Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| | - R Woltjer
- Department of Pathology, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| | - M T Hinds
- Department of Biomedical Engineering, School of Medicine, Oregon Health & Science Advanced Imaging Research Center (AIRC), Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| | - J M Loftis
- Research and Development, Portland VA Health Care System, 3710 SW US Veterans Highway Road, Portland, OR 97239, USA; Departments of Psychiatry and Behavioral Neuroscience, School of Medicine, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| | - C W Wyatt
- Department of Pathology, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| | - K P Nguyen
- Department of Surgery, Division of Vascular Surgery, School of Medicine, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA; Department of Biomedical Engineering, School of Medicine, Oregon Health & Science Advanced Imaging Research Center (AIRC), Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA; Research and Development, Portland VA Health Care System, 3710 SW US Veterans Highway Road, Portland, OR 97239, USA
| |
Collapse
|
4
|
Li L, Xu X, Lv K, Zheng G, Wang H, Chen S, Huang L, Liu Y, Zhang Y, Tang Z, Zhang L, Wang J, Qiao J, Li H, Wang X, Yao G, Fang C. Asebogenin suppresses thrombus formation via inhibition of Syk phosphorylation. Br J Pharmacol 2023; 180:287-307. [PMID: 36166754 DOI: 10.1111/bph.15964] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 08/24/2022] [Accepted: 09/11/2022] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND AND PURPOSE Thrombosis is a major cause of morbidity and mortality worldwide. Platelet activation by exposed collagen through glycoprotein VI (GPVI) and formation of neutrophil extracellular traps (NETs) are critical pathogenic factors for arterial and venous thrombosis. Both events are regulated by spleen tyrosine kinase (Syk)-mediated signalling events. Asebogenin is a dihydrochalcone whose pharmacological effects remain largely unknown. This study aims to investigate the antithrombotic effects of asebogenin and the underlying molecular mechanisms. EXPERIMENTAL APPROACH Platelet aggregation was assessed using an aggregometer. Platelet P-selectin exposure, integrin activation and calcium mobilization were determined by flow cytometry. NETs formation was assessed by SYTOX Green staining and immunohistochemistry. Quantitative phosphoproteomics, microscale thermophoresis, in vitro kinase assay and molecular docking combined with dynamics simulation were performed to characterize the targets of asebogenin. The in vivo effects of asebogenin on arterial thrombosis were investigated using FeCl3 -induced and laser-induced injury models, whereas those of venous thrombosis were induced by stenosis of the inferior vena cava. KEY RESULTS Asebogenin inhibited a series of GPVI-induced platelet responses and suppressed NETs formation induced by proinflammatory stimuli. Mechanistically, asebogenin directly interfered with the phosphorylation of Syk at Tyr525/526, which is important for its activation. Further, asebogenin suppressed arterial thrombosis demonstrated by decreased platelet accumulation and fibrin generation and attenuated venous thrombosis determined by reduced neutrophil accumulation and NETs formation, without increasing bleeding risk. CONCLUSION AND IMPLICATIONS Asebogenin exhibits potent antithrombotic effects by targeting Syk and is a potential lead compound for the development of efficient and safe antithrombotic agents.
Collapse
Affiliation(s)
- Li Li
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xulin Xu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Keyu Lv
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Guijuan Zheng
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Hao Wang
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Shuai Chen
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Lang Huang
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yi Liu
- DeepKinase Biotechnologies Ltd., Beijing, China
| | | | - Zhaoming Tang
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Lili Zhang
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jinyu Wang
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.,The Key Laboratory of Oral and Maxillofacial Development and Regeneration of Hubei Province, Wuhan, Hubei, China
| | - Jianlin Qiao
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Hongliang Li
- Laboratory of Chinese Herbal Pharmacology, Department of Pharmacy, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei, China.,Biomedical Research Institute, School of Pharmaceutical Sciences and Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, Hubei, China
| | - Xuanbin Wang
- Laboratory of Chinese Herbal Pharmacology, Department of Pharmacy, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei, China.,Biomedical Research Institute, School of Pharmaceutical Sciences and Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, Hubei, China
| | - Guangmin Yao
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Chao Fang
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.,The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan, Hubei, China
| |
Collapse
|
5
|
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
|
6
|
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 2022; 33:78-85. [PMID: 34563699 DOI: 10.1016/j.jvir.2021.09.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [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
|
7
|
Investigation of ultrasonic chronic total occlusion system on a rabbit model. JOURNAL OF SURGERY AND MEDICINE 2021. [DOI: 10.28982/josam.936595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
8
|
Li W, Kessinger CW, Orii M, Lee H, Wang L, Weinberg I, Jaff MR, Reed GL, Libby P, Tawakol A, Henke PK, Jaffer FA. Time-Restricted Salutary Effects of Blood Flow Restoration on Venous Thrombosis and Vein Wall Injury in Mouse and Human Subjects. Circulation 2021; 143:1224-1238. [PMID: 33445952 PMCID: PMC7988304 DOI: 10.1161/circulationaha.120.049096] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Accepted: 12/11/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND Up to 50% of patients with proximal deep vein thrombosis (DVT) will develop the postthrombotic syndrome characterized by limb swelling and discomfort, hyperpigmentation, skin ulcers, and impaired quality of life. Although catheter-based interventions enabling the restoration of blood flow (RBF) have demonstrated little benefit on postthrombotic syndrome, the impact on the acuity of the thrombus and mechanisms underlying this finding remain obscure. In experimental and clinical studies, we examined whether RBF has a restricted time window for improving DVT resolution. METHODS First, experimental stasis DVT was generated in C57/BL6 mice (n=291) by inferior vena cava ligation. To promote RBF, mice underwent mechanical deligation with or without intravenous recombinant tissue plasminogen activator administered 2 days after deligation. RBF was assessed over time by ultrasonography and intravital microscopy. Resected thrombosed inferior vena cava specimens underwent thrombus and vein wall histological and gene expression assays. Next, in a clinical study, we conducted a post hoc analysis of the ATTRACT (Acute Venous Thrombosis: Thrombus Removal with Adjunctive Catheter-Directed Thrombolysis) pharmacomechanical catheter-directed thrombolysis (PCDT) trial (NCT00790335) to assess the effects of PCDT on Venous Insufficiency Epidemiological and Economic Study quality-of-life and Villalta scores for specific symptom-onset-to-randomization timeframes. RESULTS Mice that developed RBF by day 4, but not later, exhibited reduced day 8 thrombus burden parameters and reduced day 8 vein wall fibrosis and inflammation, compared with controls. In mice without RBF, recombinant tissue plasminogen activator administered at day 4, but not later, reduced day 8 thrombus burden and vein wall fibrosis. It is notable that, in mice already exhibiting RBF by day 4, recombinant tissue plasminogen activator administration did not further reduce thrombus burden or vein wall fibrosis. In the ATTRACT trial, patients receiving PCDT in an intermediate symptom-onset-to-randomization timeframe of 4 to 8 days demonstrated maximal benefits in Venous Insufficiency Epidemiological and Economic Study quality-of-life and Villalta scores (between-group difference=8.41 and 1.68, respectively, P<0.001 versus patients not receiving PCDT). PCDT did not improve postthrombotic syndrome scores for patients having a symptom-onset-to-randomization time of <4 days or >8 days. CONCLUSIONS Taken together, these data illustrate that, within a restricted therapeutic window, RBF improves DVT resolution, and PCDT may improve clinical outcomes. Further studies are warranted to examine the value of time-restricted RBF strategies to reduce postthrombotic syndrome in patients with DVT.
Collapse
Affiliation(s)
- Wenzhu Li
- Cardiovascular Research Center, Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Current Affiliations: W.L. - Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; C.W.K. - Masonic Medical Research Institute, Utica, New York, USA; M.O. - Department of Radiology, Iwate Medical University Hospital, Morioka, Japan; L.W. -Cardiovascular Division, Hubei Renmin Hospital, Wuhan University, Wuhan, China
| | - Chase W. Kessinger
- Cardiovascular Research Center, Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Current Affiliations: W.L. - Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; C.W.K. - Masonic Medical Research Institute, Utica, New York, USA; M.O. - Department of Radiology, Iwate Medical University Hospital, Morioka, Japan; L.W. -Cardiovascular Division, Hubei Renmin Hospital, Wuhan University, Wuhan, China
| | - Makoto Orii
- Cardiovascular Research Center, Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Current Affiliations: W.L. - Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; C.W.K. - Masonic Medical Research Institute, Utica, New York, USA; M.O. - Department of Radiology, Iwate Medical University Hospital, Morioka, Japan; L.W. -Cardiovascular Division, Hubei Renmin Hospital, Wuhan University, Wuhan, China
| | - Hang Lee
- Biostatistics Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Lang Wang
- Cardiovascular Research Center, Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Current Affiliations: W.L. - Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; C.W.K. - Masonic Medical Research Institute, Utica, New York, USA; M.O. - Department of Radiology, Iwate Medical University Hospital, Morioka, Japan; L.W. -Cardiovascular Division, Hubei Renmin Hospital, Wuhan University, Wuhan, China
| | - Ido Weinberg
- Vascular Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Michael R. Jaff
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Guy L. Reed
- Department of Medicine, University of Arizona, College of Medicine, Phoenix, Arizona, USA
| | - Peter Libby
- Cardiovascular Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Ahmed Tawakol
- Cardiovascular Research Center, Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Peter K. Henke
- Section of Vascular Surgery, Department of Surgery, University of Michigan, Ann Arbor, Michigan, USA
| | - Farouc A. Jaffer
- Cardiovascular Research Center, Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
9
|
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
|
10
|
Lv X, Jiang C, Wu Z, Jiang W, Wang G. Complex cerebral aneurysms: intra-luminal reconstruction using Pipeline flow-diverting stent and the obliteration mechanism. Neuroradiol J 2020; 33:91-97. [PMID: 31822193 PMCID: PMC7140306 DOI: 10.1177/1971400919894879] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
OBJECTIVES Patients with complex cerebral aneurysms can now be treated intravascularly with the help of flow-diverting stents. The primary purpose of this article is to document the clinical and angiographic outcomes in 80 patients who were treated with the Pipeline flow-diverting stent (PFS; Medtronic, Dublin, Ireland) and the obliteration mechanism was discussed. PATIENTS AND METHODS Between October 2015 and October 2019, 80 patients with 90 complex (undefined neck, large/giant, blood blister-like, and recurrent side-wall) cerebral aneurysms treated with the PFS were retrospectively reviewed. Forty-five patients were women and 35 were men, with a mean age of 52 years. Large or giant aneurysms were defined as 10 mm or larger and small aneurysms were defined as less than 10 mm at the largest diameter measured on angiogram. RESULTS Forty-one aneurysms (45.6%) were large or giant, 41 (45.6%) were small, four (4.4%) were recurrent side-wall aneurysms and four (4.4%) were blood blister-like aneurysms. In total, 87 PFSs were placed in 80 patients with 90 aneurysms. In six patients, coexisting proximal stenosis of parent artery was also covered with PFS without balloon angioplasty. Adjunct coils were placed in 31 aneurysms (34%). One patient died of intracerebral hematoma after thrombolysis. There was one intrastent occlusion at six-month follow-up without any symptoms. The morbidity and the mortality rate is 0% and 1.3% (95% confidence interval (CI), 0%-3.7%). Control angiography was available in 74 (92.5%) patients with 83 aneurysms, and the aneurysm occlusion rate was 98.8% (95% CI, 96.5%-100%) in 6 to 12 months. CONCLUSION For wide-necked saccular, large/giant, blood blister-like aneurysms and recurrent side-wall aneurysms, PFS is a valid and safe treatment option.
Collapse
Affiliation(s)
- Xianli Lv
- Neurosurgery Department, Beijing
Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University,
China
| | - Chuhan Jiang
- Neurosurgery Department, Beijing
Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University,
China
| | - Zhongxue Wu
- Neurosurgery Department, Beijing
Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University,
China
| | - Weijian Jiang
- New Era Stroke Care and Research
Institute, The PLA Rocket Force General Hospital, Beijing
| | - Guihuai Wang
- Neurosurgery Department, Beijing
Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University,
China
| |
Collapse
|
11
|
Resolvin D4 attenuates the severity of pathological thrombosis in mice. Blood 2020; 134:1458-1468. [PMID: 31300403 DOI: 10.1182/blood.2018886317] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 07/03/2019] [Indexed: 12/31/2022] Open
Abstract
Deep vein thrombosis (DVT) is a common cardiovascular disease with a major effect on quality of life, and safe and effective therapeutic measures to efficiently reduce existent thrombus burden are scarce. Using a comprehensive targeted liquid chromatography-tandem mass spectrometry-based metabololipidomics approach, we established temporal clusters of endogenously biosynthesized specialized proresolving mediators (SPMs) and proinflammatory and prothrombotic lipid mediators during DVT progression in mice. Administration of resolvin D4 (RvD4), an SPM that was enriched at the natural onset of thrombus resolution, significantly reduced thrombus burden, with significantly less neutrophil infiltration and more proresolving monocytes in the thrombus, as well as an increased number of cells in an early apoptosis state. Moreover, RvD4 promoted the biosynthesis of other D-series resolvins involved in facilitating resolution of inflammation. Neutrophils from RvD4-treated mice were less susceptible to an ionomycin-induced release of neutrophil extracellular traps (NETs), a meshwork of decondensed chromatin lined with histones and neutrophil proteins critical for DVT development. These results suggest that delivery of SPMs, specifically RvD4, modulates the severity of thrombo-inflammatory disease in vivo and improves thrombus resolution.
Collapse
|
12
|
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
|
13
|
Abstract
Deep vein thrombosis (DVT) is a disease with high prevalence and morbidity. It can lead to pulmonary embolism with severe respiratory insufficiency and risk of death. Mechanisms behind all stages of DVT, such as thrombosis commencement, propagation, and resolution, remain incompletely understood. Animal models represent an invaluable tool to explore these problems and identify new targets for DVT prevention and treatment. In this review, we discuss existing models of venous thrombosis, their advantages and disadvantages, and applicability to studying different aspects of DVT pathophysiology. We also speculate about requirements for an "ideal model" that would best recapitulate features of human DVT and discuss readouts of various models.
Collapse
Affiliation(s)
- Joana Campos
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham , Birmingham, UK
| | - Alexander Brill
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham , Birmingham, UK.,Department of Pathophysiology, Sechenov First Moscow State Medical University (Sechenov University) , Moscow, Russia.,Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham , The Midlands, UK
| |
Collapse
|
14
|
Wu G, Morelli J, Xiong Y, Liu X, Li X. Diffusion weighted cardiovascular magnetic resonance imaging for discriminating acute from non-acute deep venous Thrombus. J Cardiovasc Magn Reson 2019; 21:37. [PMID: 31286985 PMCID: PMC6615231 DOI: 10.1186/s12968-019-0552-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Accepted: 06/14/2019] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND The importance of discriminating acute from non-acute thrombus is highlighted. The study aims to investigate the feasibility of readout-segmented diffusion weighted (DW) cardiovascular magnetic resonance (CMR) for discrimination of acute from non-acute deep venous thrombus (DVT). METHODS For this prospective study from December 2015 to December 2017, 85 participants (mean age = 53 years, age range = 34~74) with DVT of lower extremities underwent readout-segmented DW CMR. DVT of ≤14 days were defined as acute (n = 55) and > 14 days as non-acute (n = 30). DVT visualization on b = 0, b = 800, and apparent diffusion coefficient (ADC) images were assessed using a 4-point scale (0~3, poor~excellent). DW CMR parameters were measured using region of interest (ROI). Relative signal intensity (rSI) and ADC were compared between acute and non-acute DVT using a Mann Whitney test. Sensitivity and specificity for ADC and rSI were calculated. RESULTS ADC maps had higher visualization scores than b = 0 and b = 800 images (2.7 ± 0.5, 2.5 ± 0.6, and 2.4 ± 0.6 respectively, P<0.05). The mean ADC was higher in acute DVT than non-acute DVT (0.56 ± 0.17 × 10- 3 vs. 0.22 ± 0.12 × 10- 3 mm2/s, P<0.001). Using 0.32 × 10- 3 mm2/s as the cutoff, sensitivity and specificity for ADC to discriminate acute from non-acute DVT were 93 and 90% respectively. Sensitivity and specificity were 73 and 60% for rSI on b = 0, and 75 and 63% for rSI on b = 800. CONCLUSIONS Readout segmented diffusion-weighted CMR derived ADC distinguishes acute from non-acute DVT. TRIAL REGISTRATION This study is retrospectively registered. TRIAL REGISTRATION NUMBER HUST-TJH-2015-146 .
Collapse
Affiliation(s)
- Gang Wu
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095, Jiefang Avenue, Wuhan, 430030 Hubei China
| | - John Morelli
- Department of Radiology, St. John’s Medical Center, Tulsa, OK USA
| | - Yan Xiong
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095, Jiefang Avenue, Wuhan, 430030 Hubei China
| | - Xuanlin Liu
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095, Jiefang Avenue, Wuhan, 430030 Hubei China
| | - Xiaoming Li
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095, Jiefang Avenue, Wuhan, 430030 Hubei China
| |
Collapse
|
15
|
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: 35] [Impact Index Per Article: 5.8] [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
|
16
|
Chinnadurai P, Bismuth J. Intraoperative Imaging and Image Fusion for Venous Interventions. Methodist Debakey Cardiovasc J 2018; 14:200-207. [PMID: 30410650 DOI: 10.14797/mdcj-14-3-200] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Advanced imaging for intraoperative evaluation of venous pathologies has played an increasingly significant role in this era of evolving minimally invasive surgical and interventional therapies. The evolution of dedicated venous stents and other novel interventional devices has mandated the need for advanced imaging tools to optimize safe and accurate device deployment. Most venous interventions are typically performed using a combination of standard 2-dimensional (2D) fluoroscopy, digital-subtraction angiography, and intravascular ultrasound imaging techniques. Latest generation computer tomography (CT) and magnetic resonance imaging (MRI) scanners have been shown to provide high-resolution 3D and 4D information about venous vasculature. In addition to morphological imaging, novel MRI techniques such as 3D time-resolved MR venography and 4D flow sequences can provide quantitative information and help visualize intricate flow patterns to better understand complex venous pathologies. Moreover, the high-fidelity information from multiple imaging techniques can be integrated using image fusion to overcome the limitations of current intraoperative imaging techniques. For example, the limitations of standard 2D fluoroscopy and luminal angiography can be compensated for by perivascular and soft-tissue information from MRI during complex venous interventions using image fusion techniques. Intraoperative dynamic evaluation of devices such as venous stents and real-time understanding of changes in flow patterns during venous interventions may be routinely available in future interventional suites with integrated multimodality CT or MR imaging capabilities. The purpose of this review is to discuss the outlook for intraoperative imaging and multimodality image fusion techniques and highlight their value during complex venous interventions.
Collapse
Affiliation(s)
| | - Jean Bismuth
- METHODIST DEBAKEY HEART & VASCULAR CENTER, HOUSTON METHODIST HOSPITAL, HOUSTON, TEXAS
| |
Collapse
|
17
|
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
|
18
|
Budnik I, Brill A. Immune Factors in Deep Vein Thrombosis Initiation. Trends Immunol 2018; 39:610-623. [PMID: 29776849 PMCID: PMC6065414 DOI: 10.1016/j.it.2018.04.010] [Citation(s) in RCA: 128] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 04/21/2018] [Accepted: 04/25/2018] [Indexed: 12/11/2022]
Abstract
Deep vein thrombosis (DVT) is a major origin of morbidity and mortality. While DVT has long been considered as blood coagulation disorder, several recent lines of evidence demonstrate that immune cells and inflammatory processes are involved in DVT initiation. Here, we discuss these mechanisms, in particular, the role of immune cells in endothelial activation, and the immune cascades leading to expression of adhesion receptors on endothelial cells. We analyze the specific recruitment and functional roles of different immune cells, such as mast cells and leukocytes, in DVT. Importantly, we also speculate how immune modulation could be used for DVT prevention with a lower risk of bleeding complications than conventional therapeutic approaches.
Collapse
Affiliation(s)
- Ivan Budnik
- Department of Pathophysiology, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Alexander Brill
- Department of Pathophysiology, Sechenov First Moscow State Medical University, Moscow, Russia; Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK.
| |
Collapse
|
19
|
Inhibition of prolyl hydroxylase domain proteins selectively enhances venous thrombus neovascularisation. Thromb Res 2018; 169:105-112. [PMID: 30031289 DOI: 10.1016/j.thromres.2018.07.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 06/30/2018] [Accepted: 07/09/2018] [Indexed: 11/21/2022]
Abstract
BACKGROUND Hypoxia within acute venous thrombi is thought to drive resolution through stabilisation of hypoxia inducible factor 1 alpha (HIF1α). Prolyl hydroxylase domain (PHD) isoforms are critical regulators of HIF1α stability. Non-selective inhibition of PHD isoforms with l-mimosine has been shown to increase HIF1α stabilisation and promote thrombus resolution. OBJECTIVE The aim of this study was to investigate the therapeutic potential of PHD inhibition in venous thrombus resolution. METHODS Thrombosis was induced in the inferior vena cava of mice using a combination of flow restriction and endothelial activation. Gene and protein expression of PHD isoforms in the resolving thrombus was measured by RT-PCR and immunohistochemistry. Thrombus resolution was quantified in mice treated with pan PHD inhibitors AKB-4924 and JNJ-42041935 or inducible all-cell Phd2 knockouts by micro-computed tomography, 3D high frequency ultrasound or endpoint histology. RESULTS Resolving venous thrombi demonstrated significant temporal gene expression profiles for PHD2 and PHD3 (P < 0.05), but not for PHD1. PHD isoform protein expression was localised to early and late inflammatory cell infiltrates. Treatment with selective pan PHD inhibitors, AKB-4924 and JNJ-42041935, enhanced thrombus neovascularisation (P < 0.05), but had no significant effect on overall thrombus resolution. Thrombus resolution or its markers, macrophage accumulation and neovascularisation, did not differ significantly in inducible all-cell homozygous Phd2 knockouts compared with littermate controls (P > 0.05). CONCLUSIONS This data suggests that PHD-mediated thrombus neovascularisation has a limited role in the resolution of venous thrombi. Directly targeting angiogenesis alone may not be a viable therapeutic strategy to enhance venous thrombus resolution.
Collapse
|
20
|
Xu Y, Chandarajoti K, Zhang X, Pagadala V, Dou W, Hoppensteadt DM, Sparkenbaugh EM, Cooley B, Daily S, Key NS, Severynse-Stevens D, Fareed J, Linhardt RJ, Pawlinski R, Liu J. Synthetic oligosaccharides can replace animal-sourced low-molecular weight heparins. Sci Transl Med 2017; 9:eaan5954. [PMID: 28878012 PMCID: PMC6231235 DOI: 10.1126/scitranslmed.aan5954] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 08/07/2017] [Indexed: 12/12/2022]
Abstract
Low-molecular weight heparin (LMWH) is used clinically to treat clotting disorders. As an animal-sourced product, LMWH is a highly heterogeneous mixture, and its anticoagulant activity is not fully reversible by protamine. Furthermore, the reliability of the LMWH supply chain is a concern for regulatory agencies. We demonstrate the synthesis of heparin dodecasaccharides (12-mers) at the gram scale. In vitro experiments demonstrate that the anticoagulant activity of the 12-mers could be reversed using protamine. One of these, labeled as 12-mer-1, reduced the size of blood clots in the mouse model of deep vein thrombosis and attenuated circulating procoagulant markers in the mouse model of sickle cell disease. An ex vivo experiment demonstrates that the anticoagulant activity of 12-mer-1 could be reversed by protamine. 12-mer-1 was also examined in a nonhuman primate model to determine its pharmacodynamic parameters. A 7-day toxicity study in a rat model showed no toxic effects. The data suggest that a synthetic homogeneous oligosaccharide can replace animal-sourced LMWHs.
Collapse
Affiliation(s)
- Yongmei Xu
- Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Kasemsiri Chandarajoti
- Division of Hematology/Oncology, Department of Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Prince of Songkla University, Hat Yai, Songkhla 90112, Thailand
| | - Xing Zhang
- Department of Chemistry and Chemical Biology, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| | - Vijayakanth Pagadala
- Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Wenfang Dou
- Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA
| | | | - Erica M Sparkenbaugh
- Division of Hematology/Oncology, Department of Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Brian Cooley
- Department of Pathology and Laboratory Medicine, School of Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Sharon Daily
- Center for Global Health, RTI International, Research Triangle Park, NC 27709, USA
| | - Nigel S Key
- Division of Hematology/Oncology, Department of Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
| | | | - Jawed Fareed
- Department of Pathology, Loyola University Medical Center, Maywood, IL 60660, USA
| | - Robert J Linhardt
- Department of Chemistry and Chemical Biology, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY 12180, USA.
| | - Rafal Pawlinski
- Division of Hematology/Oncology, Department of Medicine, University of North Carolina, Chapel Hill, NC 27599, USA.
| | - Jian Liu
- Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA.
| |
Collapse
|
21
|
Byrnes JR, Wolberg AS. New findings on venous thrombogenesis. Hamostaseologie 2017; 37:25-35. [PMID: 27878206 PMCID: PMC5680039 DOI: 10.5482/hamo-16-09-0034] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 11/04/2016] [Indexed: 12/22/2022] Open
Abstract
Venous thrombosis (VT) is the third most common cause of cardiovascular death worldwide. Complications from VT and pulmonary embolism are the leading cause of lost disability-adjusted life years. Risks include genetic (e.g., non-O blood group, activated protein C resistance, hyperprothrombinemia) and acquired (e.g., age, surgery, cancer, pregnancy, immobilisation, female hormone use) factors. Pathophysiologic mechanisms that promote VT are incompletely understood, but involve abnormalities in blood coagulability, vessel function, and flow (so-called Virchow's Triad). Epidemiologic studies of humans, animal models, and biochemical and biophysical investigations have revealed contributions from extrinsic, intrinsic, and common pathways of coagulation, endothelial cells, leukocytes, red blood cells, platelets, cell-derived microvesicles, stasis-induced changes in vascular cells, and blood rheology. Knowledge of these mechanisms may yield new therapeutic targets. Characterisation of mechanisms that mediate VT formation and stability, particularly in aging, are needed to advance understanding of VT.
Collapse
Affiliation(s)
| | - Alisa S Wolberg
- Alisa S. Wolberg, Ph. D., Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, 819 Brinkhous-Bullitt Building, CB #7525, Chapel Hill, NC 27599-7525, United States, Phone: (919) 962-8943, Fax: (919) 966-6718, E-Mail:
| |
Collapse
|