1
|
Mathews R, Setthavongsack N, Le-Cook A, Kaempf A, Loftis JM, Woltjer RL, Lorentz CU, Revenko A, Hinds MT, Nguyen KP. Role of platelet count in a murine stasis model of deep vein thrombosis. Platelets 2024; 35:2290916. [PMID: 38099327 PMCID: PMC10805383 DOI: 10.1080/09537104.2023.2290916] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 11/29/2023] [Indexed: 12/18/2023]
Abstract
Platelets are core components of thrombi but their effect on thrombus burden during deep vein thrombosis (DVT) has not been fully characterized. We examined the role of thrombopoietin-altered platelet count on thrombus burden in a murine stasis model of DVT. To modulate platelet count compared to baseline, CD1 mice were pretreated with thrombopoietin antisense oligonucleotide (THPO-ASO, 56% decrease), thrombopoietin mimetic (TPO-mimetic, 36% increase), or saline (within 1%). Thrombi and vein walls were examined on postoperative days (POD) 3 and 7. Thrombus weights on POD 3 were not different between treatment groups (p = .84). The mean thrombus weights on POD 7 were significantly increased in the TPO-mimetic cohort compared to the THPO-ASO (p = .005) and the saline (p = .012) cohorts. Histological grading at POD 3 revealed a significantly increased smooth muscle cell presence in the thrombi and CD31 positive channeling in the vein wall of the TPO-mimetic cohort compared to the saline and THPO-ASO cohorts (p < .05). No differences were observed in histology on POD 7. Thrombopoietin-induced increased platelet count increased thrombus weight on POD 7 indicating platelet count may regulate thrombus burden during early resolution of venous thrombi in this murine stasis model of DVT.
Collapse
Affiliation(s)
- Rick Mathews
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, Oregon, USA
| | - Naly Setthavongsack
- Division of Neuropathology, Department of Pathology, Oregon Health and Science University, Portland, Oregon, USA
| | - Anh Le-Cook
- Research & Development Service, VA Portland Health Care System, Portland, Oregon, USA
| | - Andy Kaempf
- Biostatistics Shared Resource, Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon, USA
| | - Jennifer M Loftis
- Research & Development Service, VA Portland Health Care System, Portland, Oregon, USA
- Department of Psychiatry, Oregon Health and Science University, Portland, Oregon, USA
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, Oregon, USA
| | - Randall L Woltjer
- Division of Neuropathology, Department of Pathology, Oregon Health and Science University, Portland, Oregon, USA
| | | | | | - Monica T Hinds
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, Oregon, USA
| | - Khanh P Nguyen
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, Oregon, USA
- Research & Development Service, VA Portland Health Care System, Portland, Oregon, USA
- Division of Vascular Surgery, Department of Surgery, Oregon Health and Science University, Portland, Oregon, USA
| |
Collapse
|
2
|
Shaydakov ME, Diaz JA, Eklöf B, Lurie F. Venous valve hypoxia as a possible mechanism of deep vein thrombosis: a scoping review. INT ANGIOL 2024; 43:309-322. [PMID: 38864688 DOI: 10.23736/s0392-9590.24.05170-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/13/2024]
Abstract
INTRODUCTION The pathogenesis of deep vein thrombosis (DVT) has been explained by an interplay between a changed blood composition, vein wall alteration, and blood flow abnormalities. A comprehensive investigation of these components of DVT pathogenesis has substantially promoted our understanding of thrombogenesis in the venous system. Meanwhile, the process of DVT initiation remains obscure. This systematic review aims to collect, analyze, and synthesize the published evidence to propose hypoxia as a possible trigger of DVT. EVIDENCE ACQUISITION An exhaustive literature search was conducted across multiple electronic databased including PubMed, EMBASE, Scopus, and Web of Science to identify studies pertinent to the research hypothesis. The search was aimed at exploring the connection between hypoxia, reoxygenation, and the initiation of deep vein thrombosis (DVT). The following key words were used: "deep vein thrombosis," "venous thrombosis," "venous thromboembolism," "hypoxia," "reoxygenation," "venous valve," and "venous endothelium." Reviews, case reports, editorials, and letters were excluded. EVIDENCE SYNTHESIS Based on the systematic search outcome, 156 original papers relevant to the issue were selected for detailed review. These studies encompassed a range of experimental and observational clinical research, focusing on various aspects of DVT, including the anatomical, physiological, and cellular bases of the disease. A number of studies suggested limitations in the traditional understanding of Virchow's triad as an acceptable explanation for DVT initiation. Emerging evidence points to more complex interactions and additional factors that may be critical in the early stages of thrombogenesis. The role of venous valves has been recognized but remains underappreciated, with several studies indicating that these sites may act as primary loci for thrombus formation. A collection of studies describes the effects of hypoxia on venous endothelial cells at the cellular and molecular levels. Hypoxia influences several pathways that regulate endothelial cell permeability, inflammatory response, and procoagulation activity, underpinning the endothelial dysfunction noted in DVT. CONCLUSIONS Hypoxia of the venous valve may serve as an independent hypothesis to outline the DVT triggering process. Further research projects in this field may discover new molecular pathways responsible for the disease and suggest new therapeutic targets.
Collapse
Affiliation(s)
- Maxim E Shaydakov
- Division of Vascular Surgery, University of Pittsburgh Medical Center, Pittsburg, PA, USA -
| | - Jose A Diaz
- Division of Surgical Research, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | - Fedor Lurie
- Jobst Vascular Institute, ProMedica Health System, Toledo, OH, USA
| |
Collapse
|
3
|
Sun B, Cheng X, Zhang M, Shi Q, Zhao X, Wang X, Zhang Y. Dynamic observation of circRNA and mRNA profiles in a rat model of deep vein thrombosis. Exp Ther Med 2023; 26:467. [PMID: 37664678 PMCID: PMC10469585 DOI: 10.3892/etm.2023.12166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 07/20/2023] [Indexed: 09/05/2023] Open
Abstract
The goal of the present study was to identify different transcriptome expression profiles involved in the pathogenesis of deep vein thrombosis (DVT), and to illustrate the diagnostic and therapeutic potential of circular RNAs (circRNAs) and mRNAs in DVT progression. A Sprague-Dawley rat model of DVT was successfully established through the stenosis method and samples were sequenced at four time points (1, 6 and 12 h, and 3 days after ligation) to observe the dynamic changes in circRNAs and mRNAs during DVT progression. RNA sequencing was used to analyze the circRNA and mRNA expression profiles, and associated functions and pathways, in the blood of DVT rats at the four time points. In addition, Short Time Series Expression Miner (STEM) analysis was performed to explore temporal gene expression. Differential expression of 1,680, 4,018, 3,724, and 3,036 circRNAs, and 400, 1,176, 373, and 573 mRNAs was observed in the 1, 6 and 12 h, and 3-day groups, respectively, compared with the sham group (fold change >2.0 or <-2.0, P<0.05). Functional enrichment analysis indicated that differentially expressed mRNAs were associated with the following terms: Immune response, cell activation, blood stasis facilitated organelle, extracellular membrane-bounded organelle, and blood microparticle, oxygen transporter activity. STEM analysis indicated that the expression of 366 circRNAs in circRNA profile 45 and 270 mRNAs in mRNA profile 45 was consistent with thrombus progression. Enrichment analysis was performed on mRNA profile 45. The main Gene Ontology annotations were chromosome segregation, mitotic sister chromatid segregation, cell cycle process, and ligand-dependent nuclear receptor transcription coactivator activity. Pathway enrichment analysis identified the platelet-associated pathway, immune-associated pathway, and inflammation-relation pathway. According to the enriched platelet-associated pathways, four mRNAs and ten candidate circRNAs were selected for reverse transcription-quantitative PCR verification. The expression of nine of the ten circRNAs and all four mRNAs was consistent with the sequencing results. In summary, differentially expressed circRNAs and mRNAs are dynamically involved in DVT development. Dysregulated transcriptome profiles and the corresponding functions and pathways may provide mechanistic insights into DVT diagnosis and treatment.
Collapse
Affiliation(s)
- Baolan Sun
- Department of Laboratory, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Xi Cheng
- Department of Gynecology and Obstetrics, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Mu Zhang
- Department of Ophthalmology, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Qin Shi
- Department of Gynecology and Obstetrics, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Xinxin Zhao
- Department of Gynecology and Obstetrics, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Xudong Wang
- Department of Laboratory, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Yuquan Zhang
- Department of Gynecology and Obstetrics, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| |
Collapse
|
4
|
Jin FM, Wang M, Wu XM, Xiao H, Wang DX, Wang GM, Zhang CG, Zhao HR. Effects of wasp venom on venous thrombosis in rats. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2022; 25:822-826. [PMID: 36033945 PMCID: PMC9392573 DOI: 10.22038/ijbms.2022.63219.13962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 06/21/2022] [Indexed: 11/24/2022]
Abstract
OBJECTIVES This study aimed to investigate the potential effects of wasp venom (WV) from Vespa magnifica on antithrombosis in rats with inferior vena cava (IVC) thrombosis. MATERIALS AND METHODS The thrombosis rat model was established by improving the IVC stenosis, in which rats were subjected to IVC ligation for 75 min. Rats were administered argatroban (IP) or WV (s.c.) for 4 hr after IVC thrombosis. The weight, inhibition rate, and pathological morphology of the thrombosis induced by IVC ligation and the variation in four coagulation parameters, coagulation factors, and CD61+CD62P+ were simultaneously determined in IVC rats. RESULTS The thrombus formed as a result of IVC ligation was stable. Compared with the control group, the weight of the thrombus was significantly reduced in the argatroban group. Thrombus weight was reduced by treatment with 0.6, 0.2, and 0.05 mg/kg WV, with inhibition rates of 52.19%, 35.32%, and 28.98%, respectively. Inflammatory cells adhered to and infiltrated the vessel wall in the IVC group more than in the sham group. However, the pathological morphology and CD61+CD62P+ of the WV treatment groups tended to be normal. CONCLUSION We improved the model of IVC thrombosis to be suitable for evaluation of antithrombotic drugs. Our findings demonstrated that WV could inhibit IVC thrombosis associated with reducing coagulation factors V and CD61+CD62p expression in rats.
Collapse
Affiliation(s)
- Fan-mao Jin
- Lishui City People’s Hospital, Lishui, Zhejiang 323000, People’s Republic of China,These authors contributed equally to this work
| | - Mei Wang
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, Dali University, Dali, China,National-Local Joint Engineering Research Center of Entomoceutics, Dali University, Dali, China,These authors contributed equally to this work
| | - Xiu-mei Wu
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, Dali University, Dali, China,National-Local Joint Engineering Research Center of Entomoceutics, Dali University, Dali, China
| | - Huai Xiao
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, Dali University, Dali, China
| | - De-xiao Wang
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, Dali University, Dali, China,Genetic Testing Center, The First Affiliated Hospital of Dali University, Dali University, Dali, China
| | - Guang-ming Wang
- Genetic Testing Center, The First Affiliated Hospital of Dali University, Dali University, Dali, China
| | - Cheng-gui Zhang
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, Dali University, Dali, China,National-Local Joint Engineering Research Center of Entomoceutics, Dali University, Dali, China,Corresponding authors: Cheng-gui Zhang. Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, Dali University, Dali, China; National-Local Joint Engineering Research Center of Entomoceutics, Dali University, Dali, China. ; Hai-rong Zhao. Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, Dali University, Dali, China; Genetic Testing Center, The First Affiliated Hospital of Dali University, Dali University, Dali, China; National-Local Joint Engineering Research Center of Entomoceutics, Dali University, Dali, China.
| | - Hai-rong Zhao
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, Dali University, Dali, China,National-Local Joint Engineering Research Center of Entomoceutics, Dali University, Dali, China,Genetic Testing Center, The First Affiliated Hospital of Dali University, Dali University, Dali, China,Corresponding authors: Cheng-gui Zhang. Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, Dali University, Dali, China; National-Local Joint Engineering Research Center of Entomoceutics, Dali University, Dali, China. ; Hai-rong Zhao. Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, Dali University, Dali, China; Genetic Testing Center, The First Affiliated Hospital of Dali University, Dali University, Dali, China; National-Local Joint Engineering Research Center of Entomoceutics, Dali University, Dali, China.
| |
Collapse
|
5
|
Beck S, Hochreiter B, Schmid JA. Extracellular Vesicles Linking Inflammation, Cancer and Thrombotic Risks. Front Cell Dev Biol 2022; 10:859863. [PMID: 35372327 PMCID: PMC8970602 DOI: 10.3389/fcell.2022.859863] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 02/21/2022] [Indexed: 12/13/2022] Open
Abstract
Extracellular vesicles (EVs) being defined as lipid-bilayer encircled particles are released by almost all known mammalian cell types and represent a heterogenous set of cell fragments that are found in the blood circulation and all other known body fluids. The current nomenclature distinguishes mainly three forms: microvesicles, which are formed by budding from the plasma membrane; exosomes, which are released, when endosomes with intraluminal vesicles fuse with the plasma membrane; and apoptotic bodies representing fragments of apoptotic cells. Their importance for a great variety of biological processes became increasingly evident in the last decade when it was discovered that they contribute to intercellular communication by transferring nucleotides and proteins to recipient cells. In this review, we delineate several aspects of their isolation, purification, and analysis; and discuss some pitfalls that have to be considered therein. Further on, we describe various cellular sources of EVs and explain with different examples, how they link cancer and inflammatory conditions with thrombotic processes. In particular, we elaborate on the roles of EVs in cancer-associated thrombosis and COVID-19, representing two important paradigms, where local pathological processes have systemic effects in the whole organism at least in part via EVs. Finally, we also discuss possible developments of the field in the future and how EVs might be used as biomarkers for diagnosis, and as vehicles for therapeutics.
Collapse
Affiliation(s)
- Sarah Beck
- Institute of Vascular Biology and Thrombosis Research, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
- Institute of Experimental Biomedicine, University Hospital Würzburg and Rudolf Virchow Center for Integrative and Translational Bioimaging, University of Würzburg, Würzburg, Germany
- *Correspondence: Sarah Beck, ; Johannes A. Schmid,
| | - Bernhard Hochreiter
- Institute of Vascular Biology and Thrombosis Research, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Johannes A. Schmid
- Institute of Vascular Biology and Thrombosis Research, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
- *Correspondence: Sarah Beck, ; Johannes A. Schmid,
| |
Collapse
|
6
|
Henke PK, Obi AT. Advances in understanding the interplay between adaptive and innate immunity in experimental venous thrombus resolution. J Thromb Haemost 2021; 19:1387-1389. [PMID: 33595180 DOI: 10.1111/jth.15249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 01/08/2021] [Accepted: 01/11/2021] [Indexed: 11/27/2022]
Affiliation(s)
- Peter K Henke
- University of Michigan Health System, Ann Arbor, MI, USA
| | - Andrea T Obi
- University of Michigan Health System, Ann Arbor, MI, USA
| |
Collapse
|
7
|
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: 18] [Impact Index Per Article: 6.0] [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
|
8
|
Shi C, Yang L, Braun A, Anders HJ. Extracellular DNA-A Danger Signal Triggering Immunothrombosis. Front Immunol 2020; 11:568513. [PMID: 33117353 PMCID: PMC7575749 DOI: 10.3389/fimmu.2020.568513] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 09/09/2020] [Indexed: 12/14/2022] Open
Abstract
Clotting and inflammation are effective danger response patterns positively selected by evolution to limit fatal bleeding and pathogen invasion upon traumatic injuries. As a trade-off, thrombotic, and thromboembolic events complicate severe forms of infectious and non-infectious states of acute and chronic inflammation, i.e., immunothrombosis. Factors linked to thrombosis and inflammation include mediators released by platelet granules, complement, and lipid mediators and certain integrins. Extracellular deoxyribonucleic acid (DNA) was a previously unrecognized cellular component in the blood, which elicits profound proinflammatory and prothrombotic effects. Pathogens trigger the release of extracellular DNA together with other pathogen-associated molecular patterns. Dying cells in the inflamed or infected tissue release extracellular DNA together with other danger associated molecular pattern (DAMPs). Neutrophils release DNA by forming neutrophil extracellular traps (NETs) during infection, trauma or other forms of vascular injury. Fluorescence tissue imaging localized extracellular DNA to sites of injury and to intravascular thrombi. Functional studies using deoxyribonuclease (DNase)-deficient mouse strains or recombinant DNase show that extracellular DNA contributes to the process of immunothrombosis. Here, we review rodent models of immunothrombosis and the evolving evidence for extracellular DNA as a driver of immunothrombosis and discuss challenges and prospects for extracellular DNA as a potential therapeutic target.
Collapse
Affiliation(s)
- Chongxu Shi
- Renal Division, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Ludwig-Maximilians University Munich, Munich, Germany
| | - Luying Yang
- Renal Division, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Ludwig-Maximilians University Munich, Munich, Germany
| | - Attila Braun
- German Center for Lung Research, Walther-Straub-Institute for Pharmacology and Toxicology, Ludwig-Maximilians University Munich, Munich, Germany
| | - Hans-Joachim Anders
- Renal Division, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Ludwig-Maximilians University Munich, Munich, Germany
| |
Collapse
|
9
|
Shaydakov ME, Diaz JA, Comerota AJ, Lurie F. Targeted gene expression analysis of human deep veins. J Vasc Surg Venous Lymphat Disord 2020; 9:770-780.e7. [PMID: 32860957 DOI: 10.1016/j.jvsv.2020.08.025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 08/15/2020] [Indexed: 11/25/2022]
Abstract
OBJECTIVE Endothelial-derived molecules involved in thrombosis and hemostasis have been investigated mainly in arteries and in experimental animals. The actual presence and integral function of these molecules in the human deep venous system have received less attention. Our aim was to evaluate the expression of certain prothrombotic and antithrombotic genes in the normal human deep veins of the lower extremities. METHODS Macroscopically intact and competent valve-containing segments of human deep veins were prospectively collected from patients who had undergone above-knee amputation. Vein samples were separated into four zones: zone 1, postvalve (downstream, proximal) vein wall; zone 2, the valve cusp; zone 3, prevalve (upstream, distal) vein wall; and zone 4, vein wall within the valve cusp (cusp removed). Real-time quantitative polymerase chain reaction for principal genes involved in coagulation, fibrinolysis, and inflammation was performed to quantify messenger RNA. Selected protein gene products were measured by the western blot assay. One additional valve-containing segment underwent mass spectrometry analysis to investigate global differences in the proteome between the study zones. RESULTS Seventeen valve-containing vein segments were analyzed. Significant upregulation of antithrombotic (protein C receptor [PROCR], thrombomodulin [THBD], tissue factor pathway inhibitor [TFPI]), prothrombotic (con Willebrand factor [VWF]), and proinflammatory (selectin P [SELP], intercellular adhesion molecule 1 [ICAM1]) genes was found in the valve cusp compared with the vein wall (P < .05). PROCR and THBD demonstrated the highest level of upregulation in the valve cusp. PROCR, serpin peptidase inhibitor, clade E, member 1 (SERPINE1), and SELP were upregulated in the valve cusp at the protein level (P < .05). Messenger RNA composition in the vein wall within the valve cusp was similar to the prevalve and postvalve vein wall for all genes, except for two times overexpressed ICAM1 (P < .05). Substantial differences within the proteome between the study zones were observed with mass spectrometry. CONCLUSIONS The biological properties of the valve cusp, vein wall within the valve cusp, and vein wall beyond the valve cusp are different. The endothelium of the valve cusps of a normal competent deep venous valve may be naturally less thrombogenic compared with the vein wall. The endothelium of the valve cusp may have a higher potential to interact with white blood cells compared with the vein wall. Mass spectrometry demonstrates substantial differences in the proteome between the vein wall and the valve cusps that were not anticipated before. (J Vasc Surg Venous Lymphat Disord 2021;9:770-80.) CLINICAL RELEVANCE: Deep vein thrombosis (DVT) is a major cause of mortality, morbidity, and impaired quality of life. Multiple risk factors have been identified, although their relative weight and pathophysiologic interactions remain obscure. Many patients with multiple risk factors for DVT never develop this condition. Conversely, in numerous cases DVT cannot be attributed to any known clinical risk factor. The molecular mechanisms that initiate DVT are unclear. An improved understanding of the normal biology of human deep veins will serve as an important foundation for new hypotheses of the pathogenesis of DVT. The latter may suggest new projects on novel therapeutic strategies.
Collapse
Affiliation(s)
- Maxim E Shaydakov
- Conrad Jobst Research Vascular Laboratories, University of Michigan Medical School, Ann Arbor, Mich; Jobst Vascular Institute, ProMedica Toledo Hospital, Toledo, Ohio; Department of Surgery, Central Michigan University College of Medicine, Saginaw, Mich.
| | - Jose A Diaz
- Conrad Jobst Research Vascular Laboratories, University of Michigan Medical School, Ann Arbor, Mich; Division of Surgical Research, Surgical Sciences, Vanderbilt University Medical Center, Nashville, Tenn
| | - Anthony J Comerota
- Jobst Vascular Institute, ProMedica Toledo Hospital, Toledo, Ohio; Inova Heart and Vascular Institute, Inova Alexandria Hospital, Alexandria, Va
| | - Fedor Lurie
- Jobst Vascular Institute, ProMedica Toledo Hospital, Toledo, Ohio
| |
Collapse
|
10
|
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: 10.5] [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
|
11
|
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
|
12
|
Collateral vein dynamics in mouse models of venous thrombosis: Pathways consistent with humans. Thromb Res 2019; 182:116-123. [DOI: 10.1016/j.thromres.2019.08.018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 07/29/2019] [Accepted: 08/17/2019] [Indexed: 01/31/2023]
|
13
|
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: 80] [Impact Index Per Article: 16.0] [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
|
14
|
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: 7.0] [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
|
15
|
Metz AK, Diaz JA, Obi AT, Wakefield TW, Myers DD, Henke PK. Venous Thrombosis and Post-Thrombotic Syndrome: From Novel Biomarkers to Biology. Methodist Debakey Cardiovasc J 2019; 14:173-181. [PMID: 30410646 DOI: 10.14797/mdcj-14-3-173] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Deep vein thrombosis (DVT) is a common disease that carries serious ramifications for patients, including pulmonary embolism and post-thrombotic syndrome (PTS). Although standard treatment for DVT is anticoagulation, this carries an added risk of bleeding and increased medication monitoring. Identifying those at risk for DVT and PTS can be difficult, and current research with murine models is helping to illuminate the biologic changes associated with these two disorders. Potential novel biomarkers for improving the diagnosis of DVT and PTS include ICAM-1, P-selectin, and cell-free DNA. Inhibition of factor XI, P- and E-selectin, and neutrophil extracellular traps holds promise for novel clinical treatment of DVT. Experimental research on PTS suggests potential cellular and mediator therapy targets of TLR9, MMP-2 and-9, PAI-1, and IL-6. Although many important concepts and mechanisms have been elucidated through research on DVT and PTS, more work must be done to translate experimental findings to the clinical arena. This review examines the currently used murine models of DVT, biomarkers involved in the pathophysiology and diagnosis of DVT and PTS, and potential pharmacologic targets for PTS treatment.
Collapse
|
16
|
Gallagher KA, Obi AT, Elfline MA, Hogikyan E, Luke CE, Henke S, Coleman D, Henke PK. Alterations in macrophage phenotypes in experimental venous thrombosis. J Vasc Surg Venous Lymphat Disord 2018; 4:463-71. [PMID: 27639001 DOI: 10.1016/j.jvsv.2016.03.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 03/12/2016] [Indexed: 10/21/2022]
Abstract
OBJECTIVE Macrophages are involved in venous thrombus (VT) resolution and vein wall remodeling. This study was undertaken to identify variations in macrophage phenotypes in thrombi and vein wall in multiple models of VT to clarify the natural history of macrophage polarization in clearance of VT. We also sought to demonstrate the feasibility of macrophage phenotyping in human VT. METHODS Established murine models of VT were used to mimic the clinical spectrum of human VT (stasis and nonstasis models). Vein wall and thrombi were isolated at acute (2 days) or chronic (6-21 days) time points and analyzed by Bio-Plex assay (Bio-Rad, Carlsbad, Calif) for cytokines (interleukin [IL]-1β, IL-6, IL-10, IL-12), by immunohistochemistry for "M1-like" (IL-12) or "M2-like" (arginase 1 [Arg-1]) markers, and by histology for intimal thickness and collagen content (Sirius red staining). Bone marrow was harvested from animals 2 days after undergoing sham, stasis, or nonstasis surgery. Macrophages were skewed toward M1 using lipopolysaccharide, and RNA analysis was done for inflammatory cytokine genes (IL-1β, IL-12). Human blood samples were similarly analyzed with reverse transcription polymerase chain reaction for macrophage polarization markers (CD206, inducible nitric oxide synthase, CCR2) and thrombi with immunohistochemistry (inducible nitric oxide synthase, Arg-1). RESULTS Stasis (chronic) and nonstasis (acute and chronic) thrombi were characterized by a predominance in anti-inflammatory (M2) macrophages (n = 4-5/group; P < .05). Larger thrombi were found in the stasis model at both time points (n = 3; P < .01), correlating with decreased intrathrombus inflammatory (M1) cytokines (IL-1β, P = .03; IL-12, P = .17; n = 4) and diminished inflammatory response of bone marrow-derived macrophages to lipopolysaccharide (IL-1β, P = .03; IL-12, P = .04; n = 4) compared with nonstasis model. Anti-inflammatory (M2 [Arg-1]) macrophage cell counts were elevated in the post-thrombotic vein wall of stasis mice compared with nonstasis mice (acute: n = 4, P < .05; chronic: n = 5, P < .01), consistent with increased intimal thickness (P < .01; n = 4-6) and collagen deposition chronically (P = .005; n = 12). M2-like thrombi (Arg-1, P < .05; n = 4-7) and circulating markers (CD206, P < .05; n = 9-17) decreased over time in human VT. CONCLUSIONS Experimental VT is characterized by an anti-inflammatory predominant macrophage phenotype, possibly impairing thrombus resolution, and is model dependent. Altering the M1/M2 macrophage balance may accelerate thrombus resolution and allow the development of translatable novel therapies to treat VT and to prevent post-thrombotic syndrome.
Collapse
Affiliation(s)
- Katherine A Gallagher
- Section of Vascular Surgery, Department of Vascular Surgery, University of Michigan Medical School, Ann Arbor, Mich
| | - Andrea T Obi
- Section of Vascular Surgery, Department of Vascular Surgery, University of Michigan Medical School, Ann Arbor, Mich
| | - Megan A Elfline
- Section of Vascular Surgery, Department of Vascular Surgery, University of Michigan Medical School, Ann Arbor, Mich
| | - Emily Hogikyan
- Section of Vascular Surgery, Department of Vascular Surgery, University of Michigan Medical School, Ann Arbor, Mich
| | - Catherine E Luke
- Section of Vascular Surgery, Department of Vascular Surgery, University of Michigan Medical School, Ann Arbor, Mich
| | - Samuel Henke
- Section of Vascular Surgery, Department of Vascular Surgery, University of Michigan Medical School, Ann Arbor, Mich
| | - Dawn Coleman
- Section of Vascular Surgery, Department of Vascular Surgery, University of Michigan Medical School, Ann Arbor, Mich
| | - Peter K Henke
- Section of Vascular Surgery, Department of Vascular Surgery, University of Michigan Medical School, Ann Arbor, Mich.
| |
Collapse
|
17
|
Andraska EA, Luke CE, Elfline MA, Henke SP, Madapoosi SS, Metz AK, Hoinville ME, Wakefield TW, Henke PK, Diaz JA. Pre-Clinical Model to Study Recurrent Venous Thrombosis in the Inferior Vena Cava. Thromb Haemost 2018; 118:1048-1057. [PMID: 29695021 DOI: 10.1055/s-0038-1645855] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
BACKGROUND Patients undergoing deep vein thrombosis (VT) have over 30% recurrence, directly increasing their risk of post-thrombotic syndrome. Current murine models of inferior vena cava (IVC) VT model host one thrombosis event. OBJECTIVE We aimed to develop a murine model to study IVC recurrent VT in mice. MATERIALS AND METHODS An initial VT was induced using the electrolytic IVC model (EIM) with constant blood flow. This approach takes advantage of the restored vein lumen 21 days after a single VT event in the EIM demonstrated by ultrasound. We then induced a second VT 21 days later, using either EIM or an IVC ligation model for comparison. The control groups were a sham surgery and, 21 days later, either EIM or IVC ligation. IVC wall and thrombus were harvested 2 days after the second insult and analysed for IVC and thrombus size, gene expression of fibrotic markers, histology for collagen and Western blot for citrullinated histone 3 (Cit-H3) and fibrin. RESULTS Ultrasound confirmed the first VT and its progressive resolution with an anatomical channel allowing room for the second thrombus by day 21. As compared with a primary VT, recurrent VT has heavier walls with significant up-regulation of transforming growth factor-β (TGF-β), elastin, interleukin (IL)-6, matrix metallopeptidase 9 (MMP9), MMP2 and a thrombus with high citrullinated histone-3 and fibrin content. CONCLUSION Experimental recurrent thrombi are structurally and compositionally different from the primary VT, with a greater pro-fibrotic remodelling vein wall profile. This work provides a VT recurrence IVC model that will help to improve the current understanding of the biological mechanisms and directed treatment of recurrent VT.
Collapse
Affiliation(s)
- Elizabeth A Andraska
- Department of Surgery, Section of Vascular Surgery, Conrad Jobst Vascular Research Laboratories, University of Michigan, Ann Arbor, Michigan, United States
| | - Catherine E Luke
- Department of Surgery, Section of Vascular Surgery, Conrad Jobst Vascular Research Laboratories, University of Michigan, Ann Arbor, Michigan, United States
| | - Megan A Elfline
- Department of Surgery, Section of Vascular Surgery, Conrad Jobst Vascular Research Laboratories, University of Michigan, Ann Arbor, Michigan, United States
| | - Samuel P Henke
- Department of Surgery, Section of Vascular Surgery, Conrad Jobst Vascular Research Laboratories, University of Michigan, Ann Arbor, Michigan, United States
| | - Siddharth S Madapoosi
- Department of Surgery, Section of Vascular Surgery, Conrad Jobst Vascular Research Laboratories, University of Michigan, Ann Arbor, Michigan, United States
| | - Allan K Metz
- Department of Surgery, Section of Vascular Surgery, Conrad Jobst Vascular Research Laboratories, University of Michigan, Ann Arbor, Michigan, United States
| | - Megan E Hoinville
- Department of Surgery, Section of Vascular Surgery, Conrad Jobst Vascular Research Laboratories, University of Michigan, Ann Arbor, Michigan, United States
| | - Thomas W Wakefield
- Department of Surgery, Section of Vascular Surgery, Conrad Jobst Vascular Research Laboratories, University of Michigan, Ann Arbor, Michigan, United States
| | - Peter K Henke
- Department of Surgery, Section of Vascular Surgery, Conrad Jobst Vascular Research Laboratories, University of Michigan, Ann Arbor, Michigan, United States
| | - Jose A Diaz
- Department of Surgery, Section of Vascular Surgery, Conrad Jobst Vascular Research Laboratories, University of Michigan, Ann Arbor, Michigan, United States
| |
Collapse
|
18
|
Rys RN, Blostein MD, Lemarié CA. Deep Vein Thrombosis Induced by Stasis in Mice Monitored by High Frequency Ultrasonography. J Vis Exp 2018. [PMID: 29708541 DOI: 10.3791/57392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Venous thrombosis is a common condition affecting 1 - 2% of the population, with an annual incidence of 1 in 500. Venous thrombosis can lead to death through pulmonary embolism or results in the post-thrombotic syndrome, characterized by chronic leg pain, swelling, and ulceration, or in chronic pulmonary hypertension resulting in significant chronic respiratory compromise. This is the most common cardiovascular disease after myocardial infarction and ischemic stroke and is a clinical challenge for all medical disciplines, as it can complicate the course of other disorders such as cancer, systemic disease, surgery, and major trauma. Experimental models are necessary to study these mechanisms. The stasis model induces consistent thrombus size and a quantifiable amount of thrombus. However, it is necessary to systematically ligate side branches of the inferior vena cava to avoid variability in thrombus sizes and any erroneous data interpretation. We have developed a non-invasive technique to measure thrombus size using ultrasonography. Using this technique, we can assess thrombus development and resolution over time in the same animal. This approach limits the number of mice required for quantification of venous thrombosis consistent with the principle of replacement, reduction, and refinement of animals in research. We have demonstrated that thrombus weight and histological analysis of thrombus size correlate with measurement obtained with ultrasonography. Therefore, the current study describes how to induce deep vein thrombosis in mice using the inferior vena cava stasis model and how to monitor it using high frequency ultrasound.
Collapse
Affiliation(s)
- Ryan N Rys
- Lady Davis Institute for Medical Research, Jewish General Hospital, McGill University
| | - Mark D Blostein
- Lady Davis Institute for Medical Research, Jewish General Hospital, McGill University; Department of Medicine, Jewish General Hospital, McGill University
| | - Catherine A Lemarié
- Lady Davis Institute for Medical Research, Jewish General Hospital, McGill University; Department of Medicine, Jewish General Hospital, McGill University;
| |
Collapse
|
19
|
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: 2.0] [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
|
20
|
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: 5.0] [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
|
21
|
Tashima Y, Banno F, Akiyama M, Miyata T. Influence of ADAMTS13 deficiency on venous thrombosis in mice. Thromb Haemost 2017; 114:206-7. [DOI: 10.1160/th14-08-0656] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Accepted: 02/14/2015] [Indexed: 01/27/2023]
|
22
|
Jagadeeswaran P, Cooley BC, Gross PL, Mackman N. Animal Models of Thrombosis From Zebrafish to Nonhuman Primates: Use in the Elucidation of New Pathologic Pathways and the Development of Antithrombotic Drugs. Circ Res 2017; 118:1363-79. [PMID: 27126647 DOI: 10.1161/circresaha.115.306823] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Accepted: 11/30/2015] [Indexed: 12/23/2022]
Abstract
Thrombosis is a leading cause of morbidity and mortality worldwide. Animal models are used to understand the pathological pathways involved in thrombosis and to test the efficacy and safety of new antithrombotic drugs. In this review, we will first describe the central role a variety of animal models of thrombosis and hemostasis has played in the development of new antiplatelet and anticoagulant drugs. These include the widely used P2Y12 antagonists and the recently developed orally available anticoagulants that directly target factor Xa or thrombin. Next, we will describe the new players, such as polyphosphate, neutrophil extracellular traps, and microparticles, which have been shown to contribute to thrombosis in mouse models, particularly venous thrombosis models. Other mouse studies have demonstrated roles for the factor XIIa and factor XIa in thrombosis. This has spurred the development of strategies to reduce their levels or activities as a new approach for preventing thrombosis. Finally, we will discuss the emergence of zebrafish as a model to study thrombosis and its potential use in the discovery of novel factors involved in thrombosis and hemostasis. Animal models of thrombosis from zebrafish to nonhuman primates are vital in identifying pathological pathways of thrombosis that can be safely targeted with a minimal effect on hemostasis. Future studies should focus on understanding the different triggers of thrombosis and the best drugs to prevent each type of thrombotic event.
Collapse
Affiliation(s)
- Pudur Jagadeeswaran
- From the Department of Biological Sciences, University of North Texas, Denton (P.J.); Department of Pathology and Laboratory Medicine (B.C.C.), and Department of Medicine (N.M.), University of North Carolina, Chapel Hill; and Department of Medicine, McMaster University, Hamilton, Ontario, Canada (P.L.G.).
| | - Brian C Cooley
- From the Department of Biological Sciences, University of North Texas, Denton (P.J.); Department of Pathology and Laboratory Medicine (B.C.C.), and Department of Medicine (N.M.), University of North Carolina, Chapel Hill; and Department of Medicine, McMaster University, Hamilton, Ontario, Canada (P.L.G.)
| | - Peter L Gross
- From the Department of Biological Sciences, University of North Texas, Denton (P.J.); Department of Pathology and Laboratory Medicine (B.C.C.), and Department of Medicine (N.M.), University of North Carolina, Chapel Hill; and Department of Medicine, McMaster University, Hamilton, Ontario, Canada (P.L.G.)
| | - Nigel Mackman
- From the Department of Biological Sciences, University of North Texas, Denton (P.J.); Department of Pathology and Laboratory Medicine (B.C.C.), and Department of Medicine (N.M.), University of North Carolina, Chapel Hill; and Department of Medicine, McMaster University, Hamilton, Ontario, Canada (P.L.G.)
| |
Collapse
|
23
|
Culmer DL, Dunbar ML, Hawley AE, Sood S, Sigler RE, Henke PK, Wakefield TW, Magnani JL, Myers DD. E-selectin inhibition with GMI-1271 decreases venous thrombosis without profoundly affecting tail vein bleeding in a mouse model. Thromb Haemost 2017; 117:1171-1181. [PMID: 28300869 DOI: 10.1160/th16-04-0323] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2016] [Accepted: 02/21/2017] [Indexed: 12/31/2022]
Abstract
Selectins, such as E-selectin (CD62E), function in venous thrombosis by binding and activating immune cells to initiate the coagulation cascade. GMI-1271 is a small molecule antagonist that inhibits E-selectin activity. Here we determine whether inhibition of E-selectin is sufficient to decrease acute venous thrombosis and associated inflammatory events in both prophylactic and treatment protocols without significantly affecting haemostasis. Male C57BL/6 mice underwent surgery for experimental thrombosis induction and were harvested at peak thrombus formation in our animal model, two days post induction. Groups included non-thrombosed true controls, shams, controls, and prophylactic or treatment groups of GMI-1271 (10 mg/kg intraperitoneal BID (twice a day) and low-molecular-weight heparin (LMWH, Lovenox 6 mg/kg subcutaneously (SC), once a day (SID). Compared with control animals, prophylaxis or treatment with LMWH and GMI-1271 in a dose-dependent manner significantly decreased thrombosis. GMI-1271 significantly lowered tail bleeding times when compared to LMWH. GMI-1271 and LMWH prophylactically administered significantly decreased vein wall neutrophil cell extravasation. However, all treatment and prophylactic therapies significantly decreased vein wall monocyte extravasation versus controls. GMI-1271 prophylactic therapy significantly decreased intra-thrombus cell counts versus control animals and other treatment groups. Immunohistochemistry confirmed that both treatments with GMI-1271 and LMWH significantly decreased activated leukocyte migration. GMI-1271 therapy significantly decreased thrombus weight and resulted in significantly lower bleeding times than LMWH. GMI-1271 treated mice showed decreased local and systemic inflammatory effects while modulating neutrophil activation, suggesting that GMI-1271 is a viable therapeutic candidate for venous thrombosis prophylaxis and treatment.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Daniel D Myers
- Daniel D. Myers, Jr., DVM, MPH, DACLAM, University of Michigan, North Campus Research Complex, Building 26, Room 263N, 2800 Plymouth Road, Ann Arbor, MI 48109-2800, USA, Tel.: +1 734 763 0940, E-mail:
| |
Collapse
|
24
|
Simvastatin ameliorates deep vein thrombosis in rabbits by regulating the fibrinolytic system. Blood Coagul Fibrinolysis 2016; 27:531-41. [DOI: 10.1097/mbc.0000000000000567] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
25
|
Evans CE. Inducing femoral vein thrombosis under unrestricted flow: Comments on an alternative electrolytic mouse model. Thromb Res 2016; 140:153-154. [PMID: 26879583 DOI: 10.1016/j.thromres.2016.02.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2016] [Accepted: 02/02/2016] [Indexed: 10/22/2022]
Affiliation(s)
- Colin E Evans
- Department of Physiology, Development and Neuroscience, University of Cambridge, CB2 3EG, UK; British Heart Foundation Centre of Research Excellence, University of Cambridge, UK.
| |
Collapse
|
26
|
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: 1.0] [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
|
27
|
Obi AT, Stringer KA, Diaz JA, Finkel MA, Farris DM, Yeomans L, Wakefield T, Myers DD. 1D-¹H-nuclear magnetic resonance metabolomics reveals age-related changes in metabolites associated with experimental venous thrombosis. J Vasc Surg Venous Lymphat Disord 2015; 4:221-30. [PMID: 26993871 DOI: 10.1016/j.jvsv.2015.09.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Accepted: 09/23/2015] [Indexed: 01/12/2023]
Abstract
OBJECTIVE Age is a significant risk factor for the development of venous thrombosis (VT), but the mechanism(s) that underlie this risk remain(s) undefined and poorly understood. Aging is known to adversely influence inflammation and affect metabolism. Untargeted metabolomics permits an agnostic assessment of the physiological landscape and lends insight into the mechanistic underpinnings of clinical phenotypes. The objective of this exploratory study was to test the feasibility of a metabolomics approach for identifying potential metabolic mechanisms of age-related VT. METHODS We subjected whole blood samples collected from young and old nonthrombosed controls and VT mice 2 days after thrombus induction using the electrolytic inferior vena cava, to a methanol:chloroform extraction and assayed the resulting aqueous fractions using 1D-(1)H- nuclear magnetic resonance. Normalized mouse metabolite data were compared across groups using analysis of variance (ANOVA) with Holm-Sidak post-testing. In addition, associations between metabolite concentrations and parameters of thrombosis such as thrombus and vein wall weights, and markers of inflammation, vein wall P- and E-selectin levels, were assessed using linear regression. The relatedness of the found significant metabolites was visually assessed using a bioinformatics tool, Metscape, which generates compound-reaction-enzyme-gene networks to aid in the interpretation of metabolomics data. RESULTS Old mice with VT had a greater mean vein wall weight compared with young mice with VT (P < .05). Clot weight differences between old and young mice followed the same trend as vein wall weight (0.011 ± 0.04 g vs 0.008 ± 0.003 g; P = not significant). Glutamine (ANOVA, P < .01), proline (ANOVA, P < .01), and phenylalanine (ANOVA, P < .05) levels were increased in old VT mice compared with age-matched controls and young VT mice. Betaine and/or trimethylamine N-oxide levels were increased in aged mice compared with young animals. Vein wall weight was strongly associated with glutamine (P < .05), and phenylalanine (P < .01) concentrations and there was a trend toward an association with proline (P = .09) concentration. Vein wall P-selectin, but not E-selectin levels, were increased in old VT mice and were associated with the three found metabolites of age-related VT. Collectively, with the addition of glutamate, these metabolites form a single compound-reaction-enzyme-gene network that was generated by Metscape. CONCLUSIONS We used 1D-(1)H-nuclear magnetic resonance-metabolite profiling to identify, for the first time, in an experimental model, three potential metabolites, glutamine, phenylalanine, and proline, associated with age-related VT. These metabolites are metabolically related and their levels are associated with vein wall weight and P-selectin concentrations. In aggregate, these findings provide a "roadmap" of pathways that could be interrogated in future studies, which could include provocation of the glutamine, phenylalanine, and proline pathways in the vein wall. This study introduces metabolomics as a new approach to furthering knowledge about the mechanisms of age-related VT.
Collapse
Affiliation(s)
- Andrea T Obi
- Jobst Vascular Research Laboratories, Section of Vascular Surgery, Department of Surgery, University of Michigan Health System, Ann Arbor, Mich
| | - Kathleen A Stringer
- The NMR Metabolomics Laboratory and the Department of Clinical Pharmacy, College of Pharmacy, University of Michigan, Ann Arbor, Mich; Center for Computational Medicine and Bioinformatics, School of Medicine, University of Michigan, Ann Arbor, Mich
| | - Jose A Diaz
- Jobst Vascular Research Laboratories, Section of Vascular Surgery, Department of Surgery, University of Michigan Health System, Ann Arbor, Mich
| | - Michael A Finkel
- The NMR Metabolomics Laboratory and the Department of Clinical Pharmacy, College of Pharmacy, University of Michigan, Ann Arbor, Mich
| | - Diana M Farris
- Jobst Vascular Research Laboratories, Section of Vascular Surgery, Department of Surgery, University of Michigan Health System, Ann Arbor, Mich
| | - Larisa Yeomans
- The NMR Metabolomics Laboratory and the Department of Clinical Pharmacy, College of Pharmacy, University of Michigan, Ann Arbor, Mich
| | - Thomas Wakefield
- Jobst Vascular Research Laboratories, Section of Vascular Surgery, Department of Surgery, University of Michigan Health System, Ann Arbor, Mich
| | - Daniel D Myers
- Jobst Vascular Research Laboratories, Section of Vascular Surgery, Department of Surgery, University of Michigan Health System, Ann Arbor, Mich; Unit for Laboratory Animal Medicine, University of Michigan, Ann Arbor, Mich.
| |
Collapse
|
28
|
El-Sayed OM, Dewyer NA, Luke CE, Elfline M, Laser A, Hogaboam C, Kunkel SL, Henke PK. Intact Toll-like receptor 9 signaling in neutrophils modulates normal thrombogenesis in mice. J Vasc Surg 2015; 64:1450-1458.e1. [PMID: 26482993 DOI: 10.1016/j.jvs.2015.08.070] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Accepted: 08/17/2015] [Indexed: 12/21/2022]
Abstract
BACKGROUND Deletion of Toll-like receptor 9 (Tlr9) signaling, which is important for sterile inflammatory processes, results in impaired resolution of venous thrombosis (VT) in mice. The purpose of this study was to determine if deletion of Tlr9 affected sterile necrosis, apoptosis, and neutrophil extracellular trap (NET) production in VT. METHODS Stasis and nonstasis murine models of VT were used in wild-type (WT) and Tlr9-/- mice, with assessment of thrombus size and determination of NETs, necrosis, and apoptosis markers. Anti-polymorphonuclear neutrophil (PMN) and antiplatelet antibody strategies were used to determine the cellular roles and their roles in WT and Tlr9-/- mice. RESULTS At 2 days, stasis thrombi in Tlr9-/- mice were 62% larger (n = 6-10), with 1.4-fold increased uric acid levels, 1.7-fold more apoptotic cells, 2-fold increased citrullinated histones, 2-fold increased peptidylarginine deiminase 4 (PAD4), and 1.5-fold increased elastase and a 2.4-fold reduction in tissue factor pathway inhibitor compared with WT mice (all n = 4-7; P < .05). In contrast, the sizes of nonstasis thrombi were not significantly different in Tlr9-/- mice (n = 4-6), and they did not have elevated necrosis or NET markers. Stasis thrombus size was not reduced at the 2-day time point in WT or Tlr9-/- mice that received treatment with deoxyribonuclease I or in PAD4-/- mice, which are incapable of forming NETs. In Tlr9-/- mice undergoing PMN depletion (n = 8-10), stasis thrombus size was reduced 18% and was associated with 29-fold decreased citrullinated histones, 1.3-fold decreased elastase, and 1.5-fold increased tissue factor pathway inhibitor (all n = 6; P < .05). Last, platelet depletion (>90% reduction) did not significantly reduce stasis thrombus size in Tlr9-/- mice. CONCLUSIONS These data suggest that the thrombogenic model affects Tlr9 thrombogenic mechanisms and that functional Tlr9 signaling in PMNs, but not in platelets or NETs, is an important mechanism in early stasis experimental venous thrombogenesis.
Collapse
Affiliation(s)
- Osama M El-Sayed
- Department of Vascular Surgery, University of Michigan Medical School, Ann Arbor, Mich
| | - Nicholas A Dewyer
- Department of Vascular Surgery, University of Michigan Medical School, Ann Arbor, Mich
| | - Catherine E Luke
- Department of Vascular Surgery, University of Michigan Medical School, Ann Arbor, Mich
| | - Megan Elfline
- Department of Vascular Surgery, University of Michigan Medical School, Ann Arbor, Mich
| | - Adriana Laser
- Department of Vascular Surgery, University of Michigan Medical School, Ann Arbor, Mich
| | - Cory Hogaboam
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Mich
| | - Steven L Kunkel
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Mich
| | - Peter K Henke
- Department of Vascular Surgery, University of Michigan Medical School, Ann Arbor, Mich.
| |
Collapse
|
29
|
Dewyer NA, El-Sayed OM, Luke CE, Elfline M, Kittan N, Allen R, Laser A, Oostra C, Comerota A, Hogaboam C, Kunkel SL, Henke PK. Divergent effects of Tlr9 deletion in experimental late venous thrombosis resolution and vein wall injury. Thromb Haemost 2015; 114:1028-37. [PMID: 26179893 DOI: 10.1160/th14-12-1031] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2014] [Accepted: 05/07/2015] [Indexed: 12/19/2022]
Abstract
Deep-vein thrombosis (DVT) resolves via a sterile inflammatory response. Defining the inflammatory response of DVT may allow for new therapies that do not involve anticoagulation. Previously, we have shown that Toll-like receptor 9 (Tlr9) gene deleted mice had impaired venous thrombosis (VT) resolution. Here, we further characterise the role of Tlr9 signalling and sterile inflammation in chronic VT and vein wall responses. First, we found a human precedent exists with Tlr9+ cells present in chronic post thrombotic intraluminal tissue. Second, in a stasis VT mouse model, endogenous danger signal mediators of uric acid, HMGB-1, and neutrophil extracellular traps marker of citrullinated histone-3 (and extracellular DNA) were greater in Tlr9-/- thrombi as compared with wild-type (WT), corresponding with larger VT at 8 and 21 days. Fewer M1 type (CCR2+) monocyte/macrophages (MØ) were present in Tlr9-/- thrombi than WT controls at 8 days, suggesting an impaired inflammatory cell influx. Using bone marrow-derived monocyte (BMMØ) cell culture, we found decreased fibrinolytic gene expression with exposure to several endogenous danger signals. Next, adoptive transfer of cultured Tlr9+/+ BMMØ to Tlr9-/- mice normalised VT resolution at 8 days. Lastly, although the VT size was larger at 21 days in Tlr9-/- mice and correlated with decreased endothelial antigen markers, no difference in fibrosis was found. These data suggest that Tlr9 signalling in MØ is critical for later VT resolution, is associated with necrosis clearance, but does not affect later vein wall fibrosis. These findings provide insight into the Tlr9 MØ mechanisms of sterile inflammation in this disease process.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Peter K Henke
- Peter K. Henke, 1500 E. Medical Center Dr., Rm. 5463, Cardiovascular Center, Ann Arbor, MI 48109-5867, USA, Tel.: +1 734 763 0250, Fax: +1 734 647 9867, E-mail:
| |
Collapse
|
30
|
Wolberg AS, Rosendaal FR, Weitz JI, Jaffer IH, Agnelli G, Baglin T, Mackman N. Venous thrombosis. Nat Rev Dis Primers 2015; 1:15006. [PMID: 27189130 DOI: 10.1038/nrdp.2015.6] [Citation(s) in RCA: 206] [Impact Index Per Article: 22.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Venous thromboembolism (VTE) encompasses deep-vein thrombosis (DVT) and pulmonary embolism. VTE is the leading cause of lost disability-adjusted life years and the third leading cause of cardiovascular death in the world. DVT leads to post-thrombotic syndrome, whereas pulmonary embolism can cause chronic pulmonary hypertension, both of which reduce quality of life. Genetic and acquired risk factors for thrombosis include non-O blood groups, factor V Leiden mutation, oral contraceptive use, hormone replacement therapy, advanced age, surgery, hospitalization and long-haul travel. A combination of blood stasis, plasma hypercoagulability and endothelial dysfunction is thought to trigger thrombosis, which starts most often in the valve pockets of large veins. Animal studies have revealed pathogenic roles for leukocytes, platelets, tissue factor-positive microvesicles, neutrophil extracellular traps and factors XI and XII. Diagnosis of VTE requires testing and exclusion of other pathologies, and typically involves laboratory measures (such as D-dimer) and diagnostic imaging. VTE is treated with anticoagulants and occasionally with thrombolytics to prevent thrombus extension and to reduce thrombus size. Anticoagulants are also used to reduce recurrence. New therapies with improved safety profiles are needed to prevent and treat venous thrombosis. For an illustrated summary of this Primer, visit: http://go.nature.com/8ZyCuY.
Collapse
Affiliation(s)
- Alisa S Wolberg
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, 819 Brinkhous-Bullitt Building, Chapel Hill, North Carolina 27599-7525, USA.,McAllister Heart Institute, University of North Carolina at Chapel Hill, USA
| | - Frits R Rosendaal
- Department of Clinical Epidemiology and Department of Thrombosis and Hemostasis, Leiden University Medical Center, The Netherlands.,K.G. Jensen Thrombosis Research and Expertise Center, University of Tromsø, Norway
| | - Jeffrey I Weitz
- Department of Medicine and Department of Biochemistry and Biomedical Sciences, McMaster University, and Thrombosis and Atherosclerosis Research Institute, Hamilton, Ontario, Canada
| | - Iqbal H Jaffer
- Department of Medicine and Department of Biochemistry and Biomedical Sciences, McMaster University, and Thrombosis and Atherosclerosis Research Institute, Hamilton, Ontario, Canada
| | - Giancarlo Agnelli
- Division of Internal and Cardiovascular Medicine, Stroke Unit, University of Perugia, Italy
| | - Trevor Baglin
- Department of Haematology, Addenbrooke's Hospital, Cambridge University Hospitals NHS Trust, Cambridge, UK
| | - Nigel Mackman
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, 819 Brinkhous-Bullitt Building, Chapel Hill, North Carolina 27599-7525, USA.,McAllister Heart Institute, University of North Carolina at Chapel Hill, USA.,K.G. Jensen Thrombosis Research and Expertise Center, University of Tromsø, Norway.,Department of Medicine, University of North Carolina at Chapel Hill, USA
| |
Collapse
|
31
|
Kessinger CW, Kim JW, Henke PK, Thompson B, McCarthy JR, Hara T, Sillesen M, Margey RJP, Libby P, Weissleder R, Lin CP, Jaffer FA. Statins improve the resolution of established murine venous thrombosis: reductions in thrombus burden and vein wall scarring. PLoS One 2015; 10:e0116621. [PMID: 25680183 PMCID: PMC4334538 DOI: 10.1371/journal.pone.0116621] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Accepted: 12/11/2014] [Indexed: 11/18/2022] Open
Abstract
Despite anticoagulation therapy, up to one-half of patients with deep vein thrombosis (DVT) will develop the post-thrombotic syndrome (PTS). Improving the long-term outcome of DVT patients at risk for PTS will therefore require new approaches. Here we investigate the effects of statins—lipid-lowering agents with anti-thrombotic and anti-inflammatory properties—in decreasing thrombus burden and decreasing vein wall injury, mediators of PTS, in established murine stasis and non-stasis chemical-induced venous thrombosis (N = 282 mice). Treatment of mice with daily atorvastatin or rosuvastatin significantly reduced stasis venous thrombus burden by 25% without affecting lipid levels, blood coagulation parameters, or blood cell counts. Statin-driven reductions in VT burden (thrombus mass for stasis thrombi, intravital microscopy thrombus area for non-stasis thrombi) compared similarly to the therapeutic anticoagulant effects of low molecular weight heparin. Blood from statin-treated mice showed significant reductions in platelet aggregation and clot stability. Statins additionally reduced thrombus plasminogen activator inhibitor-1 (PAI-1), tissue factor, neutrophils, myeloperoxidase, neutrophil extracellular traps (NETs), and macrophages, and these effects were most notable in the earlier timepoints after DVT formation. In addition, statins reduced DVT-induced vein wall scarring by 50% durably up to day 21 in stasis VT, as shown by polarized light microscopy of picrosirius red-stained vein wall collagen. The overall results demonstrate that statins improve VT resolution via profibrinolytic, anticoagulant, antiplatelet, and anti-vein wall scarring effects. Statins may therefore offer a new pharmacotherapeutic approach to improve DVT resolution and to reduce the post-thrombotic syndrome, particularly in subjects who are ineligible for anticoagulation therapy.
Collapse
Affiliation(s)
- Chase W. Kessinger
- Cardiovascular Research Center, Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Jin Won Kim
- Cardiovascular Research Center, Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- Multimodal Imaging and Theranostic Lab, Cardiovascular Center, Korea University Guro Hospital, Seoul, Republic of Korea
| | - Peter K. Henke
- Section of Vascular Surgery, Department of Surgery, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Brian Thompson
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Jason R. McCarthy
- Center for Molecular Imaging Research, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- Center for Systems Biology, Massachusetts General Hospital, Boston, Massachusetts, United States of America
| | - Tetsuya Hara
- Cardiovascular Research Center, Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Martin Sillesen
- Division of Trauma, Emergency Surgery and Surgical Critical Care, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Ronan J. P. Margey
- Cardiovascular Research Center, Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Peter Libby
- Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Boston, Massachusetts, United States of America
| | - Ralph Weissleder
- Center for Molecular Imaging Research, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- Center for Systems Biology, Massachusetts General Hospital, Boston, Massachusetts, United States of America
| | - Charles P. Lin
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- Center for Systems Biology, Massachusetts General Hospital, Boston, Massachusetts, United States of America
| | - Farouc A. Jaffer
- Cardiovascular Research Center, Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- Center for Molecular Imaging Research, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
32
|
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: 34] [Impact Index Per Article: 3.4] [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
|
33
|
Hish GA, Diaz JA, Hawley AE, Myers DD, Lester PA. Effects of analgesic use on inflammation and hematology in a murine model of venous thrombosis. JOURNAL OF THE AMERICAN ASSOCIATION FOR LABORATORY ANIMAL SCIENCE : JAALAS 2014; 53:485-493. [PMID: 25255071 PMCID: PMC4181690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 10/25/2013] [Revised: 11/27/2013] [Accepted: 12/13/2013] [Indexed: 06/03/2023]
Abstract
Venous thrombosis (VT) is a significant cause of morbidity and mortality in humans. Surgical animal models are crucial in studies investigating the pathogenesis of this disease and evaluating VT therapies. Because inflammation is critical to both the development and resolution of VT, analgesic medications have the potential to adversely affect multiple parameters of interest in VT research. The objective of this study was to determine how several common analgesics affect key variables in a murine ligation model of deep vein thrombosis. Male C57BL/6 mice were randomly assigned to receive either local (bupivacaine) or systemic parenteral analgesia (buprenorphine, tramadol, or carprofen) or 0.9% NaCl (control). All mice underwent laparotomy and ligation of the inferior vena cava, and treatment was continued until euthanasia at 6 or 48 h after surgery. Analysis of harvested tissues and blood included: hematology, thrombus weight, serum and vein-wall cytokines (IL1β, IL6, IL10, TNFα), soluble P-selectin, and vein-wall leukocyte infiltration. Compared with 0.9% NaCl, all of the analgesics affected multiple parameters important to VT research. Carprofen and tramadol affected the most parameters and should not be used in murine models of VT. Although they affected fewer parameters, a single dose of bupivacaine increased thrombus weight at 6 h, and buprenorphine was associated with reduced vein wall macrophages at 48 h. Although we cannot recommend the use of any of the evaluated analgesic dosages in this mouse model of VT, buprenorphine merits additional investigation to ensure the highest level of laboratory animal care and welfare.
Collapse
Affiliation(s)
- Gerald A Hish
- Department of Comparative Medicine, University of Washington, Seattle, Washington, USA
| | - Jose A Diaz
- Conrad Jobst Vascular Research Laboratories, University of Michigan, Ann Arbor, Michigan, USA
| | - Angela E Hawley
- Conrad Jobst Vascular Research Laboratories, University of Michigan, Ann Arbor, Michigan, USA
| | - Daniel D Myers
- Conrad Jobst Vascular Research Laboratories, University of Michigan, Ann Arbor, Michigan, USA; Unit for Laboratory Animal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Patrick A Lester
- Unit for Laboratory Animal Medicine, University of Michigan, Ann Arbor, Michigan, USA.
| |
Collapse
|
34
|
Savchenko AS, Martinod K, Seidman MA, Wong SL, Borissoff JI, Piazza G, Libby P, Goldhaber SZ, Mitchell RN, Wagner DD. Neutrophil extracellular traps form predominantly during the organizing stage of human venous thromboembolism development. J Thromb Haemost 2014; 12:860-70. [PMID: 24674135 PMCID: PMC4055516 DOI: 10.1111/jth.12571] [Citation(s) in RCA: 182] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Accepted: 03/21/2014] [Indexed: 12/12/2022]
Abstract
BACKGROUND A growing health problem, venous thromboembolism (VTE), including pulmonary embolism (PE) and deep vein thrombosis (DVT), requires refined diagnostic and therapeutic approaches. Neutrophils contribute to thrombus initiation and development in experimental DVT. Recent animal studies recognized neutrophil extracellular traps (NETs) as an important scaffold supporting thrombus stability. However, the hypothesis that human venous thrombi involve NETs has not undergone rigorous testing. OBJECTIVE To explore the cellular composition and the presence of NETs within human venous thrombi at different stages of development. PATIENTS AND METHODS We examined 16 thrombi obtained from 11 patients during surgery or at autopsy using histomorphological, immunohistochemical and immunofluorescence analyses. RESULTS We classified thrombus regions as unorganized, organizing and organized according to their morphological characteristics. We then evaluated them, focusing on neutrophil and platelet deposition as well as micro-vascularization of the thrombus body. We observed evidence of NET accumulation, including the presence of citrullinated histone H3 (H3Cit)-positive cells. NETs, defined as extracellular diffuse H3Cit areas associated with myeloperoxidase and DNA, localized predominantly during the phase of organization in human venous thrombi. CONCLUSIONS NETs are present in organizing thrombi in patients with VTE. They are associated with thrombus maturation in humans. Dissolution of NETs might thus facilitate thrombolysis. This finding provides new insights into the clinical development and pathology of thrombosis and provides new perspectives for therapeutic advances.
Collapse
Affiliation(s)
- A S Savchenko
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA; Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Abstract
The contributions by blood cells to pathological venous thrombosis were only recently appreciated. Both platelets and neutrophils are now recognized as crucial for thrombus initiation and progression. Here we review the most recent findings regarding the role of neutrophil extracellular traps (NETs) in thrombosis. We describe the biological process of NET formation (NETosis) and how the extracellular release of DNA and protein components of NETs, such as histones and serine proteases, contributes to coagulation and platelet aggregation. Animal models have unveiled conditions in which NETs form and their relation to thrombogenesis. Genetically engineered mice enable further elucidation of the pathways contributing to NETosis at the molecular level. Peptidylarginine deiminase 4, an enzyme that mediates chromatin decondensation, was identified to regulate both NETosis and pathological thrombosis. A growing body of evidence reveals that NETs also form in human thrombosis and that NET biomarkers in plasma reflect disease activity. The cell biology of NETosis is still being actively characterized and may provide novel insights for the design of specific inhibitory therapeutics. After a review of the relevant literature, we propose new ways to approach thrombolysis and suggest potential prophylactic and therapeutic agents for thrombosis.
Collapse
|
36
|
Laser A, Elfline M, Luke C, Slack D, Shah A, Sood V, Deatrick B, McEvoy B, Ostra C, Comerota A, Kunkel S, Hogaboam C, Henke PK. Deletion of cysteine-cysteine receptor 7 promotes fibrotic injury in experimental post-thrombotic vein wall remodeling. Arterioscler Thromb Vasc Biol 2013; 34:377-85. [PMID: 24311382 DOI: 10.1161/atvbaha.113.302428] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
OBJECTIVE Deep vein thrombosis (VT) can result in vein wall injury, which clinically manifests as post-thrombotic syndrome. Postinjury fibrosis may be modulated in part through cellular cysteine-cysteine receptor 7 (CCR7)-mediated events. We tested the hypothesis that late vein wall fibrotic remodeling is dependent on CCR7. APPROACH AND RESULTS CCR7(-/-) and C57BL/6 wild-type mice had inferior vena cava VT induced by nonstasis or stasis mechanisms. In both models, VT size was largest at day 1 and trended down by day 21, and CCR7(+) cells peaked at day 8 in wild-type mice. No significant differences in VT resolution were found in CCR7(-/-) as compared with wild type in either model. In the nonstasis VT model, vein wall changes consistent with fibrotic injury were evidenced by significant increases in collagen I, III, matrix metalloproteinase 2, and transforming growth factor-β gene expression, increases in α-smooth muscle actin and fibroblast specific protein-1 antigen, and total collagen at 8 days. Correspondingly, SM22α and fibroblast specific protein-1, but not DDR2(+) cells, were increased at 8 days. Early wild-type thrombus exposure inhibited profibrotic gene expression in CCR7(-/-) in ex vivo vein wall culture. Bone marrow chimera experiments further showed that circulating CCR7(+) leukocytes partially rescued midterm profibrotic changes in CCR7(-/-) mice. In human histological sections of chronic thrombosed femoral veins, CCR7(+) cells were present in the fibrotic areas. CONCLUSIONS Post-thrombotic vein wall remodeling is impaired in CCR7(-/-) mice, with a profibrotic phenotype, is dependent on the thrombotic mechanism, and is mediated by circulating CCR7(+) cells. Unlike other postinjury fibrotic responses, CCR7(+) signaling may be important for positive vein wall remodeling after VT.
Collapse
Affiliation(s)
- Adriana Laser
- From the Jobst Vascular Surgery Laboratory, Section of Vascular Surgery, Department of Surgery (A.L., M.E., C.L., D.S., A.S., V.S., B.D., B.M., S.K., C.H., P.K.H.) and Department of Pathology (A.L., M.E., C.L., D.S., A.S., V.S., B.D., B.M., S.K., C.H., P.K.H.), University of Michigan Medical School, Ann Arbor; and Jobst Vascular Center, Toledo, OH (C.O, A.C.)
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Sugita C, Yamashita A, Matsuura Y, Iwakiri T, Okuyama N, Matsuda S, Matsumoto T, Inoue O, Harada A, Kitazawa T, Hattori K, Shima M, Asada Y. Elevated plasma factor VIII enhances venous thrombus formation in rabbits: contribution of factor XI, von Willebrand factor and tissue factor. Thromb Haemost 2013; 110:62-75. [PMID: 23636277 DOI: 10.1160/th13-01-0069] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2013] [Accepted: 04/12/2013] [Indexed: 01/13/2023]
Abstract
Elevated plasma levels of factor VIII (FVIII) are associated with increased risk of deep venous thrombosis. The aim of this study is to elucidate how elevated FVIII levels affect venous thrombus formation and propagation in vivo. We examined rabbit plasma FVIII activity, plasma thrombin generation, whole blood coagulation, platelet aggregation and venous wall thrombogenicity before and one hour after an intravenous infusion of recombinant human FVIII (rFVIII). Venous thrombus induced by the endothelial denudation of rabbit jugular veins was histologically assessed. Thrombus propagation was evaluated as indocyanine green fluorescence intensity. Argatroban, a thrombin inhibitor, and neutralised antibodies for tissue factor (TF), factor XI (FXI), and von Willebrand factor (VWF) were infused before or after thrombus induction to investigate their effects on venous thrombus formation or propagation. Recombinant FVIII (100 IU/kg) increased rabbit plasma FVIII activity two-fold and significantly enhanced whole blood coagulation and total plasma thrombin generation, but did not affect initial thrombin generation time, platelet aggregation and venous wall thrombogenicity. The rFVIII infusion also increased the size of venous thrombus 1 hour after thrombus induction. Argatroban and the antibodies for TF, FXI or VWF inhibited such enhanced thrombus formation and all except TF suppressed thrombus propagation. In conclusion, elevated plasma FVIII levels enhance venous thrombus formation and propagation. Excess thrombin generation by FXI and VWF-mediated FVIII recruitment appear to contribute to the growth of FVIII-driven venous thrombus.
Collapse
Affiliation(s)
- Chihiro Sugita
- Department of Pathology, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki 889-1692, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
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: 2.6] [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
|
39
|
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: 3.6] [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
|
40
|
Cooley BC. Collagen-induced thrombosis in murine arteries and veins. Thromb Res 2013; 131:49-54. [DOI: 10.1016/j.thromres.2012.09.019] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2012] [Revised: 09/20/2012] [Accepted: 09/25/2012] [Indexed: 11/26/2022]
|
41
|
Culmer DL, Diaz JA, Hawley AE, Jackson TO, Shuster KA, Sigler RE, Wakefield TW, Myers DD. Circulating and vein wall P-selectin promote venous thrombogenesis during aging in a rodent model. Thromb Res 2012; 131:42-8. [PMID: 23174624 DOI: 10.1016/j.thromres.2012.10.013] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2012] [Revised: 10/24/2012] [Accepted: 10/27/2012] [Indexed: 01/24/2023]
Abstract
INTRODUCTION The objective of this study was to identify the direct relationship between aging and selectin activation during acute venous thrombosis in mice of varying ages. We hypothesized that older animals would have increased venous thrombus formation as a result of age associated-increases of pro-inflammatory molecules within the vein wall when compared to younger animals. MATERIALS AND METHODS Deep venous thrombosis (DVT) was induced in 4 and 18month old C57BL/6 mice using the electrolytic inferior vena cava model (EIM) of DVT. Blood and tissue samples were collected at baseline (TC), 6hours, and 2days post-thrombosis induction. RESULTS Older mice had significantly larger thrombi versus younger mice at 6H (18.4±6.21 vs. 13.0±4.29×10(-3) grams, p=0.0033) and 2D (18.4±4.27 vs. 13.0±5.01×10(-3) grams, p=0.0005), higher soluble P-selectin levels at 6H (13±2.5 vs. 8.4±2.7ng/mg p=0.0010) and 2D (12.7±5.0 vs. 5.9±1.3ng/mg p=0.0020), and higher vein wall P-selectin levels at 6H (1.94×10(5)±3.56×10(4) vs. 4.81±2.29×10(4) pg/mg p=0.0001) and 2D (1.38×10(5)±5.65×10(4) vs. 3.73±1.66×10(4) pg/mg p=0.0177). Older animals also had significantly higher platelet numbers at 6H (841±203.8 vs. 564±164.8K/μL p=0.0001), and 2D (1002±342.9 vs. 690±186.1K/μL p=0.0003), with corresponding increases in mean platelet volume versus younger mice post thrombosis (p≤0.01). CONCLUSIONS Older animals had significantly larger venous thrombi versus younger animals post-thombosis, as a result of high levels of P-selectin both in the circulation and locally at the level of the vein wall. Expression of local and soluble P-selectin increased with age, resulting in a pro-thrombotic environment not represented in younger mice.
Collapse
Affiliation(s)
- Dorian Laird Culmer
- Unit for Laboratory Animal Medicine, University of Michigan, Ann Arbor, Michigan, United States.
| | | | | | | | | | | | | | | |
Collapse
|
42
|
|
43
|
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: 191] [Impact Index Per Article: 15.9] [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
|
44
|
Emmerechts J, Vanassche T, Loyen S, Van Linthout I, Cludts K, Kauskot A, Long C, Jacquemin M, Hoylaerts M, Verhamme P. Partial versus complete factor VIII inhibition in a mouse model of venous thrombosis. Thromb Res 2012; 129:514-9. [DOI: 10.1016/j.thromres.2011.06.027] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2011] [Revised: 06/20/2011] [Accepted: 06/29/2011] [Indexed: 10/17/2022]
|
45
|
von Brühl ML, Stark K, Steinhart A, Chandraratne S, Konrad I, Lorenz M, Khandoga A, Tirniceriu A, Coletti R, Köllnberger M, Byrne RA, Laitinen I, Walch A, Brill A, Pfeiler S, Manukyan D, Braun S, Lange P, Riegger J, Ware J, Eckart A, Haidari S, Rudelius M, Schulz C, Echtler K, Brinkmann V, Schwaiger M, Preissner KT, Wagner DD, Mackman N, Engelmann B, Massberg S. Monocytes, neutrophils, and platelets cooperate to initiate and propagate venous thrombosis in mice in vivo. ACTA ACUST UNITED AC 2012; 209:819-35. [PMID: 22451716 PMCID: PMC3328366 DOI: 10.1084/jem.20112322] [Citation(s) in RCA: 1284] [Impact Index Per Article: 107.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Deep vein thrombosis (DVT) is a major cause of cardiovascular death. The sequence of events that promote DVT remains obscure, largely as a result of the lack of an appropriate rodent model. We describe a novel mouse model of DVT which reproduces a frequent trigger and resembles the time course, histological features, and clinical presentation of DVT in humans. We demonstrate by intravital two-photon and epifluorescence microscopy that blood monocytes and neutrophils crawling along and adhering to the venous endothelium provide the initiating stimulus for DVT development. Using conditional mutants and bone marrow chimeras, we show that intravascular activation of the extrinsic pathway of coagulation via tissue factor (TF) derived from myeloid leukocytes causes the extensive intraluminal fibrin formation characteristic of DVT. We demonstrate that thrombus-resident neutrophils are indispensable for subsequent DVT propagation by binding factor XII (FXII) and by supporting its activation through the release of neutrophil extracellular traps (NETs). Correspondingly, neutropenia, genetic ablation of FXII, or disintegration of NETs each confers protection against DVT amplification. Platelets associate with innate immune cells via glycoprotein Ibα and contribute to DVT progression by promoting leukocyte recruitment and stimulating neutrophil-dependent coagulation. Hence, we identified a cross talk between monocytes, neutrophils, and platelets responsible for the initiation and amplification of DVT and for inducing its unique clinical features.
Collapse
Affiliation(s)
- Marie-Luise von Brühl
- Deutsches Herzzentrum and I. Medizinische Klinik, Technische Universität München (TUM), 80333 Munich, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Abstract
BACKGROUND Venous thrombosis (VT) is an important cause of morbidity and mortality in clinical medicine. Animal models studying venous thrombosis are scarce and, in most cases, very crude and rely on sacrificing the animals to excise formed thrombi. Developing an in vivo murine model of venous thrombosis can be a powerful tool for studying venous thrombosis. OBJECTIVES We sought to use a high-frequency ultrasound system (HFUS) to dynamically and non-invasively monitor thrombus formation in the inferior vena cava (IVC) of mice. METHODS We developed a murine model of venous thrombosis using, for detection, the Vevo 770(®), a micro-imaging HFUS. Two different thrombosis models were used to generate thrombi in the IVC of C57Bl/6NCr mice: (i) ligation and (ii) application of ferric chloride (FeCl(3)). We then assessed venous thrombosis by HFUS. RESULTS In both models, measurements of the clot pathologically correlated favorably with measurements acquired with HFUS. Thrombus develops less than an hour after ligation or FeCl(3) -induced injury of the IVC and the size of the clot increases over time for up to 24 h. Importantly, we demonstrate that HFUS can be used to monitor the effect of an anticoagulant such as dalteparin until complete resolution of the thrombus. CONCLUSIONS These data show that HFUS assesses venous thrombosis in mice reliably and non-invasively. Developing a murine model of thrombosis using more accurate, and clinically more relevant, techniques such as ultrasonography, is a step towards a better understanding of the pathophysiology of venous thromboembolism.
Collapse
Affiliation(s)
- M N Aghourian
- Lady Davis Institute for Medical Research, Sir Mortimer B. Davis-Jewish General Hospital, Montreal, Quebec, Canada
| | | | | |
Collapse
|
47
|
Hampton AL, Diaz JA, Hawley AE, Wrobleski SK, Wang JG, Lee RD, Kirchhofer D, Sigler RE, Wakefield TW, Mackman N, Myers DD. Myeloid cell tissue factor does not contribute to venous thrombogenesis in an electrolytic injury model. Thromb Res 2011; 130:640-5. [PMID: 22192154 DOI: 10.1016/j.thromres.2011.11.027] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2011] [Revised: 10/10/2011] [Accepted: 11/15/2011] [Indexed: 11/28/2022]
Abstract
INTRODUCTION Tissue factor (TF) is a potent initiator of the extrinsic coagulation cascade. The role and source of TF in venous thrombotic disease is not clearly defined. Our study objective was to identify the contribution of myeloid cell TF to venous thrombogenesis in mice. MATERIALS AND METHODS The mouse electrolytic inferior vena cava model was used to induce thrombosis. The following groups of mice were used (1) TF(flox/flox)LysMCre(+) mice that have reduced TF expression in myeloid cells, (2) TF(flox/flox)LysMCre(-) littermate controls, (3) Wild type mice given a monoclonal anti-mouse TF antibody (1H1) to inhibit TF activity, and (4) Wild type mice given rat IgG. Evaluations at baseline, day 2, and day 6 post thrombosis included thrombus weight, vein wall inflammatory cell migration, vein wall TF mRNA, and plasma D-dimer levels. RESULTS Inhibition of TF significantly decreased thrombus weight 2days post venous thrombosis. In contrast, TF(flox/flox)LysMCre(+) had no change in thrombus weight when compared to littermate controls. The absence of myeloid cell TF did not affect infiltration of neutrophils or monocytes into the vein wall. TF mRNA expression in the vein wall decreased at 2days but then returned to baseline levels by 6days post thrombosis. D-dimer levels peaked at 2days post thrombosis in mice with or without myeloid cell TF. CONCLUSIONS TF is important in the formation of venous thrombi in the macrovasculature. However, TF expression by myeloid cells does not significantly contribute to venous thrombogenesis in this model.
Collapse
Affiliation(s)
- Anna L Hampton
- Department of Surgery, Section of Vascular Surgery, University of Michigan, Ann Arbor, Michigan, USA; Unit for Laboratory Animal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Abstract
Blood contains microparticles (MPs) derived from a variety of cell types, including platelets, monocytes, and endothelial cells. In addition, tumors release MPs into the circulation. MPs are formed from membrane blebs that are released from the cell surface by proteolytic cleavage of the cytoskeleton. All MPs are procoagulant because they provide a membrane surface for the assembly of components of the coagulation protease cascade. Importantly, procoagulant activity is increased by the presence of anionic phospholipids, particularly phosphatidylserine (PS), and the procoagulant protein tissue factor (TF), which is the major cellular activator of the clotting cascade. High levels of platelet-derived PS(+) MPs are present in healthy individuals, whereas the number of TF(+), PS(+) MPs is undetectable or very low. However, levels of PS(+), TF(+) MPs are readily detected in a variety of diseases, and monocytes appear to be the primary cellular source. In cancer, PS(+), TF(+) MPs are derived from tumors and may serve as a useful biomarker to identify patients at risk for venous thrombosis. This review will summarize our current knowledge of the role of procoagulant MPs in hemostasis and thrombosis.
Collapse
Affiliation(s)
- A Phillip Owens
- Division of Hematology/Oncology, Department of Medicine, McAllister Heart Institute, University of North Carolina at Chapel Hill, North Carolina, USA
| | | |
Collapse
|
49
|
Diaz JA, Wrobleski SK, Hawley AE, Lucchesi BR, Wakefield TW, Myers DD. Electrolytic inferior vena cava model (EIM) of venous thrombosis. J Vis Exp 2011:e2737. [PMID: 21775963 PMCID: PMC3196180 DOI: 10.3791/2737] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Animal models serve a vital role in deep venous thrombosis (DVT) research in order to study thrombus formation, thrombus resolution and to test potential therapeutic compounds (1). New compounds to be utilized in the treatment and prevention of DVT are currently being developed. The delivery of potential therapeutic antagonist compounds to an affected thrombosed vein has been problematic. In the context of therapeutic applications, a model that uses partial stasis and consistently generates thrombi within a major vein has been recently established. The Electrolytic Inferior vena cava Model (EIM) is mouse model of DVT that permits thrombus formation in the presence of continuous blood flow. This model allows therapeutic agents to be in contact with the thrombus in a dynamic fashion, and is more sensitive than other models of DVT (1). In addition, this thrombosis model closely simulates clinical situations of thrombus formation and is ideal to study venous endothelial cell activation, leukocyte migration, venous thrombogenesis, and to test therapeutic applications (1). The EIM model is technically simple, easily reproducible, creates consistent thrombi sizes and allows for a large sample (i.e. thrombus and vein wall) which is required for analytical purposes.
Collapse
Affiliation(s)
- Jose A Diaz
- Conrad Jobst Vascular Research Laboratories, Section of Vascular Surgery, University of Michigan, USA.
| | | | | | | | | | | |
Collapse
|
50
|
Alvarado CM, Diaz JA, Hawley AE, Wrobleski SK, Sigler RE, Myers DD. Male mice have increased thrombotic potential: sex differences in a mouse model of venous thrombosis. Thromb Res 2011; 127:478-86. [PMID: 21296387 DOI: 10.1016/j.thromres.2011.01.004] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2010] [Revised: 12/08/2010] [Accepted: 01/05/2011] [Indexed: 11/20/2022]
Abstract
INTRODUCTION Our objectives were to characterize sex differences during venous thrombosis, using the electrolytic inferior vena cava model of the disease. MATERIALS AND METHODS Male and female C57BL/6 mice (6-8 weeks) underwent inferior vena cava thrombosis. Time points included 6 hours, day 2, day 6, and day 14 post surgery, along with surgically naïve true controls and surgical shams. Analyses included thrombus weight, vein wall morphometrics, vein wall protein and gene expression for P-selectin, interleukin-1β, and tumor necrosis factor-α; hematology, soluble P-selectin, and plasma microparticle tissue factor activity assays. RESULTS Male venous thrombi were significantly larger than females at days 2 (13.1 ± 1.0 vs. 6.8 ± 0.5 × 10(-3) grams, p < 0.01), 6 (10.4 ± 0.8 vs. 5.4 ± 0.5 × 10(-3) grams, p < 0.01) and 14 (6.3 ± 0.5 vs. 4.1 ± 0.3 × 10(-3) grams, p < 0.01). Both male and female mice exhibited significantly increased vein wall P-selectin at 6 hours, vs. true controls (p < 0.05). Males had increased vein wall interleukin-1β, versus females, at 6 hours (180.926 ± 24.596 vs. 60.417 ± 10.478 pg/mL, p < 0.05) and day 6 (76.966 ± 13.081 vs. 33.834 ± 4.198 pg/mL, p < 0.01). Males showed decreased tumor necrosis factor-α expression (-66 %) at 6 hours. Females had increased tumor necrosis factor-α expression at 6 hours (+541%) and day 6 (+539%). Both sexes demonstrated decreased peripheral platelets at 6 hours (p < 0.05), coinciding with thrombogenesis. Plasma P-selectin increased in both sexes, versus controls, through day 6 (p < 0.05). CONCLUSIONS Males had significantly larger venous thrombi than females. Sex differences in vascular anatomy and response to inflammation may influence thrombus formation in our mouse thrombosis model.
Collapse
Affiliation(s)
- Christine M Alvarado
- Department of Surgery, Section of Vascular Surgery, University of Michigan, Ann Arbor, MI 48109-0654, USA
| | | | | | | | | | | |
Collapse
|