1
|
Verhaegen J, Willems L, Wagenaar A, Spreuwers R, Dahdah N, Aversa L, Verbelen T, Delcroix M, Quarck R. Endothelial Features Along the Pulmonary Vascular Tree in Chronic Thromboembolic Pulmonary Hypertension: Distinctive or Shared Facets? Pulm Circ 2025; 15:e70096. [PMID: 40356848 PMCID: PMC12067398 DOI: 10.1002/pul2.70096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 04/25/2025] [Accepted: 04/28/2025] [Indexed: 05/15/2025] Open
Abstract
Chronic thromboembolic pulmonary hypertension (CTEPH) is a rare complication of pulmonary embolism, characterized by the presence of organized fibro-thrombotic material that partially or fully obstructs the lumen of large pulmonary arteries, microvasculopathy, and enlargement of the bronchial systemic vessels. The precise mechanisms underlying CTEPH remain unclear. However, defective angiogenesis and altered pulmonary arterial endothelial cell (PAEC) function may contribute to disease progression. Despite the observation of differences in histological features, shear stress and ischemia along the pulmonary vascular tree, the potential contribution of PAEC phenotype and function to these disparate aspects remains unexplored. Based on these observations, we postulated that angiogenic capacities and endothelial barrier function may contribute to disparities in histological features observed along the pulmonary vascular tree. We thus explored the histological characteristics of the pulmonary vascular tree using pulmonary arterial lesions obtained during pulmonary endarterectomy (PEA). We focused on the angiogenic vascular endothelial growth factor (VEGF)-A/VEGF receptor-2 (VEGFR2) axis and collagen 15A1 (COL15A1), a potential marker of endothelial cells of the systemic circulation. Concurrently, we examined In Vitro angiogenic properties and barrier function of PAECs derived from large and (sub)-segmental pulmonary arterial lesions. (Sub)-segmental pulmonary arterial lesions were abundantly recanalized by neovessels, paralleled by an enriched expression of VEGFR2. VEGF-A expression was more pronounced in large pulmonary arterial lesions. Nevertheless, no significant difference was discerned in In Vitro angiogenic capacities and barrier integrity of PAECs isolated from large and (sub)-segmental pulmonary arterial lesions. Importantly, our findings revealed the presence of endothelial cells (CD31+) expressing COL15A1, as well as CD31+ cells that did not express COL15A1. This suggests that endothelial cells from both systemic and pulmonary circulation contribute to lesion recanalization. Despite disparate in situ angiogenic cues in VEGF-A/VEGFR2 axis between large and (sub)-segmental pulmonary arterial lesions in CTEPH, In Vitro angiogenic capacities and barrier function remain unaltered.
Collapse
Affiliation(s)
- Janne Verhaegen
- Laboratory of Respiratory Diseases & Thoracic Surgery (BREATHE), Department of Chronic Diseases & Metabolism (CHROMETA)KU Leuven – University of LeuvenLeuvenBelgium
| | - Lynn Willems
- Laboratory of Respiratory Diseases & Thoracic Surgery (BREATHE), Department of Chronic Diseases & Metabolism (CHROMETA)KU Leuven – University of LeuvenLeuvenBelgium
| | - Allard Wagenaar
- Laboratory of Respiratory Diseases & Thoracic Surgery (BREATHE), Department of Chronic Diseases & Metabolism (CHROMETA)KU Leuven – University of LeuvenLeuvenBelgium
| | - Ruben Spreuwers
- Laboratory of Respiratory Diseases & Thoracic Surgery (BREATHE), Department of Chronic Diseases & Metabolism (CHROMETA)KU Leuven – University of LeuvenLeuvenBelgium
| | - Nessrine Dahdah
- Laboratory of Respiratory Diseases & Thoracic Surgery (BREATHE), Department of Chronic Diseases & Metabolism (CHROMETA)KU Leuven – University of LeuvenLeuvenBelgium
| | - Lucia Aversa
- Laboratory of Respiratory Diseases & Thoracic Surgery (BREATHE), Department of Chronic Diseases & Metabolism (CHROMETA)KU Leuven – University of LeuvenLeuvenBelgium
| | - Tom Verbelen
- Department of Cardiac SurgeryUniversity Hospitals LeuvenLeuvenBelgium
- Department of Cardiovascular SciencesKU Leuven ‐ University of LeuvenLeuvenBelgium
| | - Marion Delcroix
- Laboratory of Respiratory Diseases & Thoracic Surgery (BREATHE), Department of Chronic Diseases & Metabolism (CHROMETA)KU Leuven – University of LeuvenLeuvenBelgium
- Clinical Department of Respiratory DiseasesUniversity Hospitals LeuvenLeuvenBelgium
| | - Rozenn Quarck
- Laboratory of Respiratory Diseases & Thoracic Surgery (BREATHE), Department of Chronic Diseases & Metabolism (CHROMETA)KU Leuven – University of LeuvenLeuvenBelgium
- Clinical Department of Respiratory DiseasesUniversity Hospitals LeuvenLeuvenBelgium
| |
Collapse
|
2
|
Gendron N, Planquette B, Roche A, Chocron R, Helley D, Philippe A, Morange PE, Gaussem P, Sanchez O, Smadja DM. Circulating CD34 + Cells: A New Biomarker of Residual Pulmonary Vascular Obstruction after Pulmonary Embolism. Stem Cell Rev Rep 2025:10.1007/s12015-025-10865-0. [PMID: 40085375 DOI: 10.1007/s12015-025-10865-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/06/2025] [Indexed: 03/16/2025]
Abstract
Pulmonary embolism (PE) is a life-threatening condition with long-term complications, including residual pulmonary vascular obstruction (RPVO). RPVO is associated with an increased risk of venous thromboembolism recurrence, chronic symptoms, and reduced quality of life. We hypothesize that an endothelial activation and vascular injury play a central role in the pathophysiology of RPVO. This prospective monocentric study investigates the potential of circulating biomarkers, including CD34⁺ cells, circulating endothelial cells (CECs), and platelet-derived growth factor BB (PDGF-BB), as indicators of vascular sequelae and predictors of RPVO. We included 56 patients with a first episode of PE. Biomarker levels were measured at PE diagnosis and six months later, coinciding with RPVO assessment using ventilation-perfusion lung scans. This defined groups of patients with (RPVO ≥ 10%) and without (RPVO < 10%) perfusion defects. Associations between biomarker levels, presence of perfusion defects, and clinical parameters were analyzed. At PE diagnosis, CEC and PDGF-BB levels were significantly elevated in patients compared to healthy controls, while CD34⁺ levels showed no difference. At the six-month follow-up, patients with perfusion defects exhibited significantly lower CD34⁺ cell levels compared to those without (median 1440 cells/mL vs. 2960 cells/mL). No significant differences in CEC or PDGF-BB levels were observed at follow-up. In conclusion, low CD34⁺ cell levels at RPVO assessment suggest a decreased regenerative potential contributing to thrombus persistence. CD34⁺ cells may serve as biomarkers for perfusion defects and warrant further study for their potential role in guiding clinical management of PE complications.
Collapse
Affiliation(s)
- Nicolas Gendron
- Hematology Department, Assistance Publique - Hôpitaux de Paris-Centre Université Paris Cité (APHP-CUP), Hôpital européen Georges Pompidou, Paris, 75015, France
- Paris Cité University, INSERM UMR-S 970, Paris Cardiovascular Research Centre, Paris, France
- F-CRIN INNOVTE, Saint-Étienne, France
| | - Benjamin Planquette
- Respiratory Medicine Department, Assistance Publique - Hôpitaux de Paris-Centre Université Paris Cité (APHP-CUP), Hôpital européen Georges Pompidou, Paris, 75015, France
| | - Anne Roche
- INSERM UMR-S 999 « Pulmonary Hypertension: Pathophysiology and Novel Therapies », Hôpital Marie Lannelongue, Le Plessis-Robinson, France
- Faculté de Médecine, HPPIT, Pulmonary Hypertension: Pathophysiology and Novel Therapies, Université Paris-Saclay, Le Kremlin-Bicêtre, France
- Assistance Publique - Hôpitaux de Paris (AP-HP), Service de Pneumologie et Soins Intensifs Respiratoires, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
| | - Richard Chocron
- Paris Cité University, INSERM UMR-S 970, Paris Cardiovascular Research Centre, Paris, France
- Emergency Department, Assistance Publique - Hôpitaux de Paris-Centre Université Paris Cité (APHP-CUP), Hôpital européen Georges Pompidou, Paris, 75015, France
| | - Dominique Helley
- Hematology Department, Assistance Publique - Hôpitaux de Paris-Centre Université Paris Cité (APHP-CUP), Hôpital européen Georges Pompidou, Paris, 75015, France
- Paris Cité University, INSERM UMR-S 970, Paris Cardiovascular Research Centre, Paris, France
| | - Aurélien Philippe
- Hematology Department, Assistance Publique - Hôpitaux de Paris-Centre Université Paris Cité (APHP-CUP), Hôpital européen Georges Pompidou, Paris, 75015, France
- Paris Cité University, INSERM UMR-S 970, Paris Cardiovascular Research Centre, Paris, France
| | - Pierre-Emmanuel Morange
- F-CRIN INNOVTE, Saint-Étienne, France
- Laboratory of Hematology, La Timone Hospital, Marseille, France
- Aix Marseille Univ, INSERM, INRAE, C2VN, Marseille, France
| | - Pascale Gaussem
- Hematology Department, Assistance Publique - Hôpitaux de Paris-Centre Université Paris Cité (APHP-CUP), Hôpital européen Georges Pompidou, Paris, 75015, France
- Paris Cité University, INSERM UMR-S 970, Paris Cardiovascular Research Centre, Paris, France
| | - Olivier Sanchez
- Paris Cité University, INSERM UMR-S 970, Paris Cardiovascular Research Centre, Paris, France
- F-CRIN INNOVTE, Saint-Étienne, France
- Respiratory Medicine Department, Assistance Publique - Hôpitaux de Paris-Centre Université Paris Cité (APHP-CUP), Hôpital européen Georges Pompidou, Paris, 75015, France
| | - David M Smadja
- Hematology Department, Assistance Publique - Hôpitaux de Paris-Centre Université Paris Cité (APHP-CUP), Hôpital européen Georges Pompidou, Paris, 75015, France.
- Paris Cité University, INSERM UMR-S 970, Paris Cardiovascular Research Centre, Paris, France.
- F-CRIN INNOVTE, Saint-Étienne, France.
| |
Collapse
|
3
|
Lu MJ, Zhang JQ, Nie ZY, Yan TH, Cao YB, Zhang LC, Li L. Monocyte/macrophage-mediated venous thrombus resolution. Front Immunol 2024; 15:1429523. [PMID: 39100675 PMCID: PMC11297357 DOI: 10.3389/fimmu.2024.1429523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 07/03/2024] [Indexed: 08/06/2024] Open
Abstract
Venous thromboembolism (VTE) poses a notable risk of morbidity and mortality. The natural resolution of the venous thrombus might be a potential alternative treatment strategy for VTE. Monocytes/macrophages merge as pivotal cell types in the gradual resolution of the thrombus. In this review, the vital role of macrophages in inducing inflammatory response, augmenting neovascularization, and facilitating the degradation of fibrin and collagen during thrombus resolution was described. The two phenotypes of macrophages involved in thrombus resolution and their dual functions were discussed. Macrophages expressing various factors, including cytokines and their receptors, adhesion molecules, chemokine receptors, vascular endothelial growth factor receptors, profibrinolytic- or antifibrinolytic-related enzymes, and other elements, are explored for their potential to promote or attenuate thrombus resolution. Furthermore, this review provides a comprehensive summary of new and promising therapeutic candidate drugs associated with monocytes/macrophages that have been demonstrated to promote or impair thrombus resolution. However, further clinical trials are essential to validate their efficacy in VTE therapy.
Collapse
Affiliation(s)
- Meng-Jiao Lu
- Institute of Vascular Disease, Shanghai TCM- Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Physiology and Pharmacology, China Pharmaceutic University, Nanjing, China
| | - Jia-Qi Zhang
- Department of Pharmacy, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhou-Yu Nie
- Institute of Vascular Disease, Shanghai TCM- Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Tian-Hua Yan
- Department of Physiology and Pharmacology, China Pharmaceutic University, Nanjing, China
| | - Yong-Bing Cao
- Institute of Vascular Disease, Shanghai TCM- Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Li-Chao Zhang
- Department of Pharmacy, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ling Li
- Institute of Vascular Disease, Shanghai TCM- Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
4
|
Willems L, Kurakula K, Verhaegen J, Klok FA, Delcroix M, Goumans MJ, Quarck R. Angiogenesis in Chronic Thromboembolic Pulmonary Hypertension: A Janus-Faced Player? Arterioscler Thromb Vasc Biol 2024; 44:794-806. [PMID: 38328933 DOI: 10.1161/atvbaha.123.319852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
Chronic thromboembolic pulmonary hypertension (CTEPH) is a rare form of pulmonary hypertension characterized by the presence of organized thrombi that obstruct pulmonary arteries, ultimately leading to right heart failure and death. Among others, impaired angiogenesis and inflammatory thrombosis have been shown to contribute to the progression of CTEPH. In this review, we summarize the 2-faced nature of angiogenesis in both thrombus formation and resolution in the context of CTEPH and highlight the dual role of angiogenesis and neovascularization in resolving venous thrombi. Furthermore, we discuss relevant in vitro and in vivo models that support the benefits or drawbacks of angiogenesis in CTEPH progression. We discuss the key pathways involved in modulating angiogenesis, particularly the underexplored role of TGFβ (transforming growth factor-beta) signaling in driving fibrosis as an integral element of CTEPH pathogenesis. We finally explore innovative treatment strategies that target angiogenic pathways. These strategies have the potential to pioneer preventive, inventive, or alternative therapeutic options for patients with CTEPH who may not qualify for surgical interventions. Moreover, they could be used synergistically with established treatments such as pulmonary endarterectomy or balloon pulmonary angioplasty. In summary, this review emphasizes the crucial role of angiogenesis in the development of in fibrothrombotic tissue, a major pathological characteristic of CTEPH.
Collapse
Affiliation(s)
- Lynn Willems
- Laboratory of Respiratory Diseases and Thoracic Surgery, Department of Chronic Diseases and Metabolism, KU Leuven-University of Leuven, Belgium (L.W., J.V., M.D., R.Q.)
- Department of Cell and Chemical Biology (L.W., M.-J.G.), Leiden University Medical Centre, the Netherlands
| | - Kondababu Kurakula
- Department of Physiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Center, Free University Medical Center, the Netherlands (K.K.)
| | - Janne Verhaegen
- Laboratory of Respiratory Diseases and Thoracic Surgery, Department of Chronic Diseases and Metabolism, KU Leuven-University of Leuven, Belgium (L.W., J.V., M.D., R.Q.)
| | - Frederikus A Klok
- Department of Medicine-Thrombosis and Haemostasis (F.A.K.), Leiden University Medical Centre, the Netherlands
| | - Marion Delcroix
- Laboratory of Respiratory Diseases and Thoracic Surgery, Department of Chronic Diseases and Metabolism, KU Leuven-University of Leuven, Belgium (L.W., J.V., M.D., R.Q.)
- Clinical Department of Respiratory Diseases, University Hospital Leuven, Belgium (M.D., R.Q.)
| | - Marie-José Goumans
- Department of Cell and Chemical Biology (L.W., M.-J.G.), Leiden University Medical Centre, the Netherlands
| | - Rozenn Quarck
- Laboratory of Respiratory Diseases and Thoracic Surgery, Department of Chronic Diseases and Metabolism, KU Leuven-University of Leuven, Belgium (L.W., J.V., M.D., R.Q.)
- Clinical Department of Respiratory Diseases, University Hospital Leuven, Belgium (M.D., R.Q.)
| |
Collapse
|
5
|
Bochenek ML, Saar K, Nazari-Jahantigh M, Gogiraju R, Wiedenroth CB, Münzel T, Mayer E, Fink L, Schober A, Hübner N, Guth S, Konstantinides S, Schäfer K. Endothelial Overexpression of TGF-β-Induced Protein Impairs Venous Thrombus Resolution: Possible Role in CTEPH. JACC Basic Transl Sci 2024; 9:100-116. [PMID: 38362348 PMCID: PMC10864968 DOI: 10.1016/j.jacbts.2023.08.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 08/16/2023] [Accepted: 08/16/2023] [Indexed: 02/17/2024]
Abstract
Endothelial cells play a critical role during venous thrombus remodeling, and unresolved, fibrotic thrombi with irregular vessels obstruct the pulmonary artery in patients with chronic thromboembolic pulmonary hypertension (CTEPH). This study sought to identify endothelial mediators of impaired venous thrombus resolution and to determine their role in the pathogenesis of the vascular obstructions in patients with CTEPH. Endothelial cells outgrown from pulmonary endarterectomy specimens (PEA) were processed for mRNA profiling, and nCounter gene expression and immunohistochemistry analysis of PEA tissue microarrays and immunoassays of plasma were used to validate the expression in CTEPH. Lentiviral overexpression in human pulmonary artery endothelial cells (HPAECs) and exogenous administration of the recombinant protein into C57BL/6J mice after inferior Vena cava ligation were employed to assess their role for venous thrombus resolution. RT2 PCR profiler analysis demonstrated the significant overexpression of factors downstream of transforming growth factor beta (TGFβ), that is TGFβ-Induced Protein (TGFBI or BIGH3) and transgelin (TAGLN), or involved in TGFβ signaling, that is follistatin-like 3 (FSTL3) and stanniocalcin-2 (STC2). Gene expression and immunohistochemistry analysis of tissue microarrays localized potential disease candidates to vessel-rich regions. Lentiviral overexpression of TGFBI in HPAECs increased fibrotic remodeling of human blood clots in vitro, and exogenous administration of recombinant TGFBI in mice delayed venous thrombus resolution. Significantly elevated plasma TGFBI levels were observed in patients with CTEPH and decreased after PEA. Our findings suggest that overexpression of TGFBI in endothelial promotes venous thrombus non-resolution and fibrosis and is causally involved in the pathophysiology of CTEPH.
Collapse
Affiliation(s)
- Magdalena L. Bochenek
- Department of Cardiology, Cardiology I, University Medical Center Mainz, Germany
- Center for Thrombosis and Hemostasis, University Medical Center Mainz, Germany
- Deutsches Zentrum für Herz-Kreislauf-Forschung, RheinMain, Germany
| | - Kathrin Saar
- Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
- Deutsches Zentrum für Herz-Kreislauf-Forschung, Berlin, Germany
| | - Maliheh Nazari-Jahantigh
- Institute for Prophylaxis and Epidemiology of Cardiovascular Diseases, Clinic of the University of Munich, Germany
- Deutsches Zentrum für Herz-Kreislauf-Forschung, Munich, Germany
| | - Rajinikanth Gogiraju
- Department of Cardiology, Cardiology I, University Medical Center Mainz, Germany
- Deutsches Zentrum für Herz-Kreislauf-Forschung, RheinMain, Germany
| | | | - Thomas Münzel
- Department of Cardiology, Cardiology I, University Medical Center Mainz, Germany
- Deutsches Zentrum für Herz-Kreislauf-Forschung, RheinMain, Germany
| | - Eckhard Mayer
- Department of Thoracic Surgery, Kerckhoff Heart and Thorax Center, Bad Nauheim, Germany
| | - Ludger Fink
- Institute for Pathology, Cytology and Molecular Pathology, MVZ, Wetzlar, Germany
| | - Andreas Schober
- Institute for Prophylaxis and Epidemiology of Cardiovascular Diseases, Clinic of the University of Munich, Germany
- Deutsches Zentrum für Herz-Kreislauf-Forschung, Munich, Germany
| | - Norbert Hübner
- Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
- Deutsches Zentrum für Herz-Kreislauf-Forschung, Berlin, Germany
| | - Stefan Guth
- Department of Thoracic Surgery, Kerckhoff Heart and Thorax Center, Bad Nauheim, Germany
| | | | - Katrin Schäfer
- Department of Cardiology, Cardiology I, University Medical Center Mainz, Germany
- Deutsches Zentrum für Herz-Kreislauf-Forschung, RheinMain, Germany
| |
Collapse
|
6
|
Delcroix M, de Perrot M, Jaïs X, Jenkins DP, Lang IM, Matsubara H, Meijboom LJ, Quarck R, Simonneau G, Wiedenroth CB, Kim NH. Chronic thromboembolic pulmonary hypertension: realising the potential of multimodal management. THE LANCET. RESPIRATORY MEDICINE 2023; 11:836-850. [PMID: 37591299 DOI: 10.1016/s2213-2600(23)00292-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 07/13/2023] [Accepted: 07/24/2023] [Indexed: 08/19/2023]
Abstract
Chronic thromboembolic pulmonary hypertension (CTEPH) is a rare complication of acute pulmonary embolism. Important advances have enabled better understanding, characterisation, and treatment of this condition. Guidelines recommending systematic follow-up after acute pulmonary embolism, and the insight that CTEPH can mimic acute pulmonary embolism on initial presentation, have led to the definition of CTEPH imaging characteristics, the introduction of artificial intelligence diagnosis pathways, and thus the prospect of easier and earlier CTEPH diagnosis. In this Series paper, we show how the understanding of CTEPH as a sequela of inflammatory thrombosis has driven successful multidisciplinary management that integrates surgical, interventional, and medical treatments. We provide imaging examples of classical major vessel targets, describe microvascular targets, define available tools, and depict an algorithm facilitating the initial treatment strategy in people with newly diagnosed CTEPH based on a multidisciplinary team discussion at a CTEPH centre. Further work is needed to optimise the use and combination of multimodal therapeutic options in CTEPH to improve long-term outcomes for patients.
Collapse
Affiliation(s)
- Marion Delcroix
- Clinical Department of Respiratory Diseases, University Hospitals of Leuven and Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven-University of Leuven, Leuven, Belgium.
| | - Marc de Perrot
- Division of Thoracic Surgery, Toronto General Hospital, Toronto, ON, Canada
| | - Xavier Jaïs
- Assistance Publique-Hôpitaux de Paris (AP-HP), Service de Pneumologie, Hôpital Bicêtre, Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | - David P Jenkins
- Department of Cardiothoracic Surgery, Royal Papworth Hospital, Cambridge, UK
| | - Irene M Lang
- Division of Cardiology, Department of Internal Medicine II, Vienna General Hospital, Centre for CardioVascular Medicine, Medical University of Vienna, Vienna, Austria
| | - Hiromi Matsubara
- National Hospital Organization Okayama Medical Center, Okayama, Japan
| | - Lilian J Meijboom
- Department of Radiology and Nuclear Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Rozenn Quarck
- Clinical Department of Respiratory Diseases, University Hospitals of Leuven and Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven-University of Leuven, Leuven, Belgium
| | - Gérald Simonneau
- Assistance Publique-Hôpitaux de Paris (AP-HP), Service de Pneumologie, Hôpital Bicêtre, Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | | | - Nick H Kim
- Division of Pulmonary, Critical Care and Sleep Medicine, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
7
|
Mosquera MS, Diaz JA. Back To Basics: Theory of Thrombus Formation and Potential Implications for Therapies? Tech Vasc Interv Radiol 2023; 26:100894. [PMID: 37865449 DOI: 10.1016/j.tvir.2023.100894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2023]
Abstract
Venous thromboembolism (VTE), including deep vein thrombosis (DVT) and pulmonary embolism (PE), is a common and potentially fatal condition. Despite existing treatments, recurrence rates and complications remain high. Understanding the pathophysiology of thrombus formation is crucial for developing effective therapies. This narrative review provides an overview of the critical elements of acute and chronic DVT, presents a theoretical framework for understanding thrombus formation, and discusses potential implications for therapeutic interventions. In addition, a hypothesis of thrombus formation is formulated, encompassing all elements described in this work.
Collapse
Affiliation(s)
- M Silena Mosquera
- Department of Surgery, Center for Fetal Research, The Children's Hospital of Philadelphia Research Institute, Philadelphia, PA
| | - Jose A Diaz
- Division of Surgical Research, Surgical Sciences, Vanderbilt University Medical Center, Nashville, TN.
| |
Collapse
|
8
|
Wu C, Li X, Zhao H, Ling Y, Ying Y, He Y, Zhang S, Liang S, Wei J, Gan X. Resistance exercise promotes the resolution and recanalization of deep venous thrombosis in a mouse model via SIRT1 upregulation. BMC Cardiovasc Disord 2023; 23:18. [PMID: 36639616 PMCID: PMC9837998 DOI: 10.1186/s12872-022-02908-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 10/19/2022] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Early exercise for acute deep venous thrombosis (DVT) improves the patient's symptoms and does not increase the risk of pulmonary embolism. However, information about its effect on thrombus resolution is limited. The aim of this study was to investigate the role of resistance exercise (RE) in thrombus resolution and recanalization and determine its underlying mechanisms. METHODS: Ninety-six C57BL/6 J mice were randomly divided into four groups: Control group (C, n = 24); DVT group (D, n = 24); RE + DVT group (ED, n = 24); and inhibitor + RE + DVT group (IED, n = 24). A DVT model was induced by stenosis of the inferior vena cava (IVC). After undergoing IVC ultrasound within 24 h post-operation to confirm DVT formation, mice without thrombosis were excluded. Other mice were sacrificed and specimens were obtained 14 or 28 days after operation. Thrombus-containing IVC was weighed, and the thrombus area and recanalization rate were calculated using HE staining. Masson's trichrome staining was used to analyze the collagen content. RT-PCR and ELISA were performed to examine IL-6, TNF-α, IL-10, and VEGF expression levels. SIRT1 expression was assessed using immunohistochemistry staining and RT-PCR. VEGF-A protein expression and CD-31-positive microvascular density (MVD) in the thrombus were observed using immunohistochemistry. RESULTS: RE did not increase the incidence of pulmonary embolism. It reduced the weight and size of the thrombus and the collagen content. Conversely, it increased the recanalization rate. It also decreased the levels of the pro-inflammatory factors IL-6 and TNF-α and increased the expression levels of the anti-inflammatory factor IL-10. RE enhanced VEGF and SIRT1 expression levels and increased the MVD in the thrombosis area. After EX527 (SIRT1 inhibitor) was applied, the positive effects of exercise were suppressed. CONCLUSIONS RE can inhibit inflammatory responses, reduce collagen deposition, and increase angiogenesis in DVT mice, thereby promoting thrombus resolution and recanalization. Its underlying mechanism may be associated with the upregulation of SIRT1 expression.
Collapse
Affiliation(s)
- Caijiao Wu
- grid.412594.f0000 0004 1757 2961Department of Nursing, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021 Guangxi China
| | - Xiaorong Li
- grid.412594.f0000 0004 1757 2961Department of Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi China
| | - Huihan Zhao
- grid.412594.f0000 0004 1757 2961Department of Nursing, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021 Guangxi China
| | - Ying Ling
- grid.412594.f0000 0004 1757 2961Department of Nursing, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021 Guangxi China
| | - Yanping Ying
- grid.412594.f0000 0004 1757 2961Department of Nursing, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021 Guangxi China
| | - Yu He
- grid.412594.f0000 0004 1757 2961Medical Lab, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021 Guangxi China
| | - Shaohan Zhang
- grid.412594.f0000 0004 1757 2961Department of Nursing, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021 Guangxi China
| | - Shijing Liang
- grid.412594.f0000 0004 1757 2961Department of Nursing, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021 Guangxi China
| | - Jiani Wei
- grid.412594.f0000 0004 1757 2961Department of Nursing, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021 Guangxi China
| | - Xiao Gan
- grid.412594.f0000 0004 1757 2961Department of Nursing, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021 Guangxi China
| |
Collapse
|
9
|
Jordan KR, Wyatt CR, Fallon ME, Woltjer R, Neuwelt EA, Cheng Q, Gailani D, Lorentz C, Tucker EI, McCarty OJ, Hinds MT, Nguyen KP. Pharmacological reduction of coagulation factor XI reduces macrophage accumulation and accelerates deep vein thrombosis resolution in a mouse model of venous thrombosis. J Thromb Haemost 2022; 20:2035-2045. [PMID: 35638310 PMCID: PMC9580566 DOI: 10.1111/jth.15777] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 05/10/2022] [Accepted: 05/25/2022] [Indexed: 11/27/2022]
Abstract
BACKGROUND Deep vein thrombosis (DVT) and post-thrombotic syndrome (PTS) remain highly prevalent despite modern medical therapy. Contact activation is a promising target for safe antithrombotic anticoagulation. The anti-factor XI (FXI) monoclonal antibody 14E11 reduces circulating levels of FXI without compromising hemostasis. The human recombinant analog, AB023, is in clinical development. The role of FXI in mediation of inflammation during DVT resolution is unknown. OBJECTIVES Investigate the effects of pharmacological targeting of FXI with 14E11 in an experimental model of venous thrombosis. METHODS Adult wild-type CD1 mice were treated with subcutaneous anti-FXI antibody (14E11, 5 mg/kg) versus saline prior to undergoing surgical constriction of the inferior vena cava (IVC). Mice were evaluated at various time points to assess thrombus weight and volume, as well as histology analysis, ferumoxytol enhanced magnetic resonance imaging (Fe-MRI), and whole blood flow cytometry. RESULTS 14E11-treated mice had reduced thrombus weights and volumes after IVC constriction on day 7 compared to saline-treated mice. 14E11 treatment reduced circulating monocytes by flow cytometry and macrophage content within thrombi as evaluated by histologic staining and Fe-MRI. Collagen deposition was increased at day 3 while CD31 and smooth muscle cell actin expression was increased at day 7 in the thrombi of 14E11-treated mice compared to saline-treated mice. CONCLUSION Pharmacologic targeting of FXI enhances the early stages of experimental venous thrombus resolution in wild-type CD1 mice, and may be of interest for future clinical evaluation of the antibody in DVT and PTS.
Collapse
Affiliation(s)
- Kelley R. Jordan
- Department of Biomedical Engineering, School of Medicine, Oregon Health & Science University, Portland, Oregon, USA
| | - Cory R. Wyatt
- Advanced Imaging Research Center, Oregon Health & Science University, Portland, Oregon, USA
| | - Meghan E. Fallon
- Department of Biomedical Engineering, School of Medicine, Oregon Health & Science University, Portland, Oregon, USA
| | - Randy Woltjer
- Department of Pathology, Oregon Health & Science University, Portland, Oregon, USA
| | - Edward A. Neuwelt
- Department of Neurology, Oregon Health & Science University, Portland, Oregon, USA
| | - Quifang Cheng
- Department of Medicine, Vanderbilt University, Nashville, TN, USA
- Department of Pathology, Microbiology and Immunology, Vanderbilt University, Nashville, TN, USA
| | - David Gailani
- Department of Medicine, Vanderbilt University, Nashville, TN, USA
- Department of Pathology, Microbiology and Immunology, Vanderbilt University, Nashville, TN, USA
| | - Christina Lorentz
- Department of Biomedical Engineering, School of Medicine, Oregon Health & Science University, Portland, Oregon, USA
- Aronora Inc., Portland, OR, USA
| | - Erik I. Tucker
- Department of Biomedical Engineering, School of Medicine, Oregon Health & Science University, Portland, Oregon, USA
- Aronora Inc., Portland, OR, USA
| | - Owen J.T. McCarty
- Department of Biomedical Engineering, School of Medicine, Oregon Health & Science University, Portland, Oregon, USA
| | - Monica T. Hinds
- Department of Biomedical Engineering, School of Medicine, Oregon Health & Science University, Portland, Oregon, USA
| | - Khanh P. Nguyen
- Department of Biomedical Engineering, School of Medicine, Oregon Health & Science University, Portland, Oregon, USA
- VA Portland Health Care System, Portland, Oregon, USA
| |
Collapse
|
10
|
de Perrot M, Gopalan D, Jenkins D, Lang IM, Fadel E, Delcroix M, Benza R, Heresi GA, Kanwar M, Granton JT, McInnis M, Klok FA, Kerr KM, Pepke-Zaba J, Toshner M, Bykova A, Armini AMD, Robbins IM, Madani M, McGiffin D, Wiedenroth CB, Mafeld S, Opitz I, Mercier O, Uber PA, Frantz RP, Auger WR. Evaluation and management of patients with chronic thromboembolic pulmonary hypertension - consensus statement from the ISHLT. J Heart Lung Transplant 2021; 40:1301-1326. [PMID: 34420851 DOI: 10.1016/j.healun.2021.07.020] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Accepted: 07/22/2021] [Indexed: 02/08/2023] Open
Abstract
ISHLT members have recognized the importance of a consensus statement on the evaluation and management of patients with chronic thromboembolic pulmonary hypertension. The creation of this document required multiple steps, including the engagement of the ISHLT councils, approval by the Standards and Guidelines Committee, identification and selection of experts in the field, and the development of 6 working groups. Each working group provided a separate section based on an extensive literature search. These sections were then coalesced into a single document that was circulated to all members of the working groups. Key points were summarized at the end of each section. Due to the limited number of comparative trials in this field, the document was written as a literature review with expert opinion rather than based on level of evidence.
Collapse
Affiliation(s)
- Marc de Perrot
- Division of Thoracic Surgery, Toronto General Hospital, Toronto, Ontario, Canada.
| | - Deepa Gopalan
- Department of Radiology, Imperial College Healthcare NHS Trust, London & Cambridge University Hospital, Cambridge, UK
| | - David Jenkins
- National Pulmonary Endarterectomy Service, Department of Cardiothoracic Surgery, Papworth Hospital, Cambridge, UK
| | - Irene M Lang
- Department of Cardiology, Pulmonary Hypertension Unit, Medical University of Vienna, Vienna, Austria
| | - Elie Fadel
- Department of Thoracic and Vascular Surgery and Heart Lung Transplantation, Marie-Lannelongue Hospital, Paris Saclay University, Le Plessis-Robinson, France
| | - Marion Delcroix
- Clinical Department of Respiratory Diseases, Pulmonary Hypertension Centre, UZ Leuven, Leuven, Belgium; Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of Chronic Diseases and Metabolism (CHROMETA), KU, Leuven, Belgium
| | - Raymond Benza
- Division of Cardiovascular Medicine, The Ohio State University, Columbus, Ohio
| | - Gustavo A Heresi
- Department of Pulmonary and Critical Care Medicine, Respiratory Institute, Cleveland Clinic, Cleveland, Ohio
| | - Manreet Kanwar
- Cardiovascular Institute, Allegheny Health Network, Pittsburgh, Pennsylvania
| | - John T Granton
- Division of Respirology, University Health Network, Toronto, Ontario, Canada
| | - Micheal McInnis
- Joint Department of Medical Imaging, University Health Network, Toronto, Ontario, Canada
| | - Frederikus A Klok
- Department of Medicine, Thrombosis and Hemostasis, Leiden University Medical Center, Leiden, The Netherlands
| | - Kim M Kerr
- University of California San Diego Medical Health, Division of Pulmonary Critical Care and Sleep Medicine, San Diego, California
| | - Joanna Pepke-Zaba
- Pulmonary Vascular Disease Unit, Royal Papworth Hospital NHS foundation Trust, Cambridge, Cambridgeshire, UK
| | - Mark Toshner
- Pulmonary Vascular Disease Unit, Royal Papworth Hospital NHS foundation Trust, Cambridge, Cambridgeshire, UK; Heart Lung Research Institute, University of Cambridge, Cambridge, UK
| | - Anastasia Bykova
- Division of Thoracic Surgery, Toronto General Hospital, Toronto, Ontario, Canada
| | - Andrea M D' Armini
- Unit of Cardiac Surgery, Intrathoracic-Trasplantation and Pulmonary Hypertension, University of Pavia, Foundation I.R.C.C.S. Policlinico San Matteo, Pavia, Italy
| | - Ivan M Robbins
- Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Michael Madani
- Department of Cardiovascular and Thoracic Surgery, University of California San Diego, La Jolla, California
| | - David McGiffin
- Department of Cardiothoracic Surgery, The Alfred Hospital and Monash University, Melbourne, VIC, Australia
| | - Christoph B Wiedenroth
- Department of Thoracic Surgery, Campus Kerckhoff of the University of Giessen, Kerckhoff Heart and Thorax Center, Bad Nauheim, Germany
| | - Sebastian Mafeld
- Division of Vascular and Interventional Radiology, Joint Department of Medical Imaging, University Health Network, Toronto, Ontario, Canada
| | - Isabelle Opitz
- Department of Thoracic Surgery, University Hospital Zurich, Zurich, Switzerland
| | - Olaf Mercier
- Department of Thoracic and Vascular Surgery and Heart Lung Transplantation, Marie-Lannelongue Hospital, Paris Saclay University, Le Plessis-Robinson, France
| | - Patricia A Uber
- Pauley Heart Center, Virginia Commonwealth University Health System, Richmond, Virginia
| | - Robert P Frantz
- Department of Cardiovascular Disease, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - William R Auger
- Pulmonary Hypertension and CTEPH Research Program, Temple Heart and Vascular Institute, Temple University, Lewis Katz School of Medicine, Philadelphia, Pennsylvania
| |
Collapse
|
11
|
Alba GA, Atri D, Darbha S, Singh I, Tapson VF, Lewis MI, Chun HJ, Yu YR, Maron BA, Rajagopal S. Chronic Thromboembolic Pulmonary Hypertension: the Bench. Curr Cardiol Rep 2021; 23:141. [PMID: 34410515 DOI: 10.1007/s11886-021-01572-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/27/2021] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW Chronic thromboembolic pulmonary hypertension (CTEPH) is an uncommon complication of acute pulmonary embolism (PE), in which the red, platelet-rich thrombus does not resolve but forms into an organized yellow, fibrotic scar-like obstruction in the pulmonary vasculature. Here we review the pathobiology of CTEPH. RECENT FINDINGS Our current knowledge has predominantly been informed by studies of human samples and animal models that are inherently limited in their ability to recapitulate all aspects of the disease. These studies have identified alterations in platelet biology and inflammation in the formation of a scar-like thrombus that comprised endothelial cells, myofibroblasts, and immune cells, along with a small vessel pulmonary arterial hypertension-like vasculopathy. The development of CTEPH-specific therapies is currently hindered by a limited knowledge of its pathobiology. The development of new CTEPH medical therapies will require new insights into its pathobiology that bridge the gap from bench to bedside.
Collapse
Affiliation(s)
- George A Alba
- Division of Pulmonary and Critical Care, Massachusetts General Hospital, Boston, MA, USA
| | - Deepak Atri
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Sriranjani Darbha
- College of Natural Sciences, The University of Texas, Austin, TX, USA
| | - Inderjit Singh
- Division of Pulmonary, Critical Care, and Sleep Medicine, Yale New Haven Hospital and Yale School of Medicine, New Haven, CT, USA
| | - Victor F Tapson
- Division of Pulmonary and Critical Care Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Michael I Lewis
- Division of Pulmonary and Critical Care Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Hyung J Chun
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale Cardiovascular Research Center, Yale School of Medicine, New Haven, CT, USA
| | - Yen-Rei Yu
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - Bradley A Maron
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Section of Cardiology, Veterans Affairs Boston Healthcare System, Boston, MA, USA
| | - Sudarshan Rajagopal
- Division of Cardiology, Department of Medicine, Duke University Medical Center, Durham, NC, USA.
| |
Collapse
|
12
|
Sun Y, Wang T, Wen QT, Yu DH, Chen JX. VEGF gene transfection restores the angiogenesis of oral submucous fibrosis in mice. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:930. [PMID: 34350245 PMCID: PMC8263869 DOI: 10.21037/atm-21-2213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 06/02/2021] [Indexed: 11/16/2022]
Abstract
BACKGROUND To explore the effectiveness of adenovirus-enhanced green fluorescent protein-vascular endothelial growth factor165 (AD-EGFP-VEGF165) transfection on fibroblasts from mice, and we assessed whether VEGF165 restores the angiogenesis of oral submucous fibrosis (OSF) in mice. METHODS AD-EGFP-VEGF165 and AD-EGFP were transfected into fibroblasts from mouse buccal tissues in vitro. The expression of VEGF before and after transfection was detected by RT-qPCR and ELISA in each group of fibroblasts. Fifteen OSF mice (pre-experimental construction) were randomly divided into 3 groups, and equal amounts of AD-EGFP-VEGF165 virus, AD-EGFP virus, and saline were injected into the buccal submucosal tissue of OSF mice. The expression of VEGF and local tissue angiogenesis were observed and measured in each group of animals. RESULTS The Ad-EGFP-VEGF165-transfected fibroblasts increased human and mouse VEGF expression compared to the Ad-EGFP group and control group (P<0.05). The buccal submucosal tissue of mice was injected with Ad-EGFP-VEGF165 after the 6th day, and the expression of VEGF was effectively expressed in AD-EGFP-VEGF165 group (P<0.05), while no positive expression observed in other groups. and the number of microvessels in the AD-EGFP-VEGF165 group increased significantly compared to the other groups (P<0.05). CONCLUSIONS Ad-EGFP-VEGF165 can be successfully transfected into fibroblasts from mice, and restored the angiogenesis of OSF in mice.
Collapse
Affiliation(s)
- Ying Sun
- Dental Medical Center, Hainan General Hospital (Hainan Affiliated Hospital of Hainan medical University), Haikou, China
| | - Tao Wang
- Dental Medical Center, Hainan General Hospital (Hainan Affiliated Hospital of Hainan medical University), Haikou, China
| | - Qi-Tao Wen
- Dental Medical Center, Hainan General Hospital (Hainan Affiliated Hospital of Hainan medical University), Haikou, China
| | - Da-Hai Yu
- Department of Stomatology, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jing-Xin Chen
- Dental Medical Center, Hainan General Hospital (Hainan Affiliated Hospital of Hainan medical University), Haikou, China
| |
Collapse
|
13
|
Hobohm L, Kölmel S, Niemann C, Kümpers P, Krieg VJ, Bochenek ML, Lukasz AH, Reiss Y, Plate KH, Liebetrau C, Wiedenroth CB, Guth S, Münzel T, Hasenfuß G, Wenzel P, Mayer E, Konstantinides SV, Schäfer K, Lankeit M. Role of angiopoietin-2 in venous thrombus resolution and chronic thromboembolic disease. Eur Respir J 2021; 58:13993003.04196-2020. [PMID: 33986029 DOI: 10.1183/13993003.04196-2020] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Accepted: 04/10/2021] [Indexed: 11/05/2022]
Abstract
Defective angiogenesis, incomplete thrombus revascularisation and fibrosis are considered critical pathomechanisms of chronic thromboembolic pulmonary hypertension (CTEPH) after pulmonary embolism (PE). Angiopoietin-2 (ANGPT2) has been shown to regulate angiogenesis, but its importance for thrombus resolution and remodelling is unknown.ANGPT2 plasma concentrations were measured in patients with CTEPH (n=68) and acute PE (n=84). Tissue removed during pulmonary endarterectomy (PEA) for CTEPH was analysed (immuno)histologically. A mouse model of inferior vena cava ligation was used to study the kinetics of venous thrombus resolution in wild-type mice receiving recombinant ANGPT2 via osmotic pumps, and in transgenic mice overexpressing ANGPT2 in endothelial cells.Circulating ANGPT2 levels were higher in CTEPH patients compared to patients with idiopathic pulmonary arterial hypertension and healthy controls, and decreased after PEA. Plasma ANGPT2 levels were also elevated in patients with PE and diagnosis of CTEPH during follow-up. Histological analysis of PEA specimens confirmed increased ANGPT2 expression, and low levels of phosphorylated TIE2 were observed in regions with early-organised pulmonary thrombi, myofibroblasts and fibrosis. Microarray and high-resolution microscopy analysis could localise ANGPT2 overexpression to endothelial cells, and hypoxia and TGF-β1 were identified as potential stimuli. Gain-of-function experiments in mice demonstrated that exogenous ANGPT2 administration and transgenic endothelial ANGPT2 overexpression resulted in delayed venous thrombus resolution, and thrombi were characterised by lower TIE2 phosphorylation and fewer microvessels.Our findings suggest that ANGPT2 delays venous thrombus resolution and that overexpression of ANGPT2 contributes to thrombofibrosis and may thus support the transition from PE to CTEPH.
Collapse
Affiliation(s)
- Lukas Hobohm
- Center for Thrombosis and Hemostasis (CTH), University Medical Center, Mainz, Germany.,Department of Cardiology, Cardiology I, University Medical Center, Mainz, Germany
| | - Sebastian Kölmel
- Internal Medicine & Endocrinology/Diabetes, Kantonsspital St.Gallen, Sankt Gallen, Switzerland
| | - Caroline Niemann
- Clinic of Gynaecology, St. Franziskus Hospital Münster, Münster, Germany
| | - Philipp Kümpers
- Department of Medicine D, Division of General Internal Medicine, Nephrology and Rheumatology, University Hospital Münster, Münster, Germany
| | - Valentin J Krieg
- Division of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Magdalena L Bochenek
- Center for Thrombosis and Hemostasis (CTH), University Medical Center, Mainz, Germany.,Department of Cardiology, Cardiology I, University Medical Center, Mainz, Germany.,German Cardiovascular Research Centre, partner site Rhine-Main, Germany
| | - Alexander H Lukasz
- Department of Medicine D, Division of General Internal Medicine, Nephrology and Rheumatology, University Hospital Münster, Münster, Germany
| | - Yvonne Reiss
- German Cardiovascular Research Centre, partner site Rhine-Main, Germany.,Institute of Neurology (Edinger Institute), University Hospital, Goethe University, Frankfurt, Germany
| | - Karl-Heinz Plate
- German Cardiovascular Research Centre, partner site Rhine-Main, Germany.,Institute of Neurology (Edinger Institute), University Hospital, Goethe University, Frankfurt, Germany
| | - Christoph Liebetrau
- German Cardiovascular Research Centre, partner site Rhine-Main, Germany.,Department of Cardiology, Kerckhoff Clinic, Bad Nauheim, Germany.,Department of Cardiology, Justus-Liebig University of Giessen, Giessen, Germany
| | | | - Stefan Guth
- Department of Thoracic Surgery, Kerckhoff Clinic, Bad Nauheim, Germany
| | - Thomas Münzel
- Department of Cardiology, Cardiology I, University Medical Center, Mainz, Germany.,German Cardiovascular Research Centre, partner site Rhine-Main, Germany
| | - Gerd Hasenfuß
- Clinic of Cardiology and Pneumology, Heart Center, University Medical Center Göttingen, Goettingen, Germany.,German Cardiovascular Research Centre, partner site Goettingen, Germany
| | - Philip Wenzel
- Center for Thrombosis and Hemostasis (CTH), University Medical Center, Mainz, Germany.,Department of Cardiology, Cardiology I, University Medical Center, Mainz, Germany.,German Cardiovascular Research Centre, partner site Rhine-Main, Germany
| | - Eckhard Mayer
- Department of Thoracic Surgery, Kerckhoff Clinic, Bad Nauheim, Germany
| | - Stavros V Konstantinides
- Center for Thrombosis and Hemostasis (CTH), University Medical Center, Mainz, Germany.,Department of Cardiology, Democritus University of Thrace, Alexandroupolis, Greece
| | - Katrin Schäfer
- Department of Cardiology, Cardiology I, University Medical Center, Mainz, Germany.,German Cardiovascular Research Centre, partner site Rhine-Main, Germany
| | - Mareike Lankeit
- Center for Thrombosis and Hemostasis (CTH), University Medical Center, Mainz, Germany .,Clinic of Cardiology and Pneumology, Heart Center, University Medical Center Göttingen, Goettingen, Germany.,Department of Internal Medicine and Cardiology, Campus Virchow Klinikum, Charité - University Medicine, Berlin, Germany.,German Cardiovascular Research Centre, partner site Berlin, Germany
| |
Collapse
|
14
|
Adventitial Progenitor Cells of Human Great Saphenous Vein Enhance the Resolution of Venous Thrombosis via Neovascularization. Stem Cells Int 2021; 2021:8816763. [PMID: 33679991 PMCID: PMC7926266 DOI: 10.1155/2021/8816763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 01/20/2021] [Accepted: 02/06/2021] [Indexed: 11/24/2022] Open
Abstract
Background Vascular adventitia contains progenitor cells and is shown to participate in vascular remolding. Progenitor cells are recruited into the venous thrombi in mice to promote neovascularization. We hypothesized that the adventitial progenitor cells of human great saphenous vein (HGSV-AdPC) enhance the resolution of venous thrombosis via neovascularization. Methods Human great saphenous vein (HGSV) was harvested from the patients with great saphenous vein varicose and sectioned for immunohistochemistry, or minced for progenitor cell primary culture, or placed in sodium dodecyl sulfate solution for decellularization. Human venous thrombi were collected from patients with great saphenous vein varicose and superficial thrombophlebitis. Infrarenal abdominal aorta of New Zealand white rabbits was replaced with interposing decellularized vessel, and the patency of the grafts was confirmed by ultrasonic examination. Animal venous thrombi in the left infrarenal vena cava of mice were produced with Prolene suture ligation and ophthalmic force clipping of this portion. After HGSVs were digested by collagenase, the CD34+CD117+ HGSV-AdPC were isolated on FACS system, labelled with CM-Dil, and transplanted into the adventitia of infrarenal vena cava of nude mice. The percentage of thrombus organization area to the thrombus area was calculated as the organization rate. The thrombus cell, endothelial cells, and macrophages in the thrombi were counted in sections. Cell smears and frozen sections of human saphenous veins and venous thrombi were labeled with Sca1, CD34, CD117, Flk1, CD31, and F4/80 antibodies. The CD34+CD117+ HGSV-AdPC were cultured in endothelial growth medium with vascular endothelial growth factor (VEGF) to induce endothelial cell differentiation and analyzed with real time-PCR, Western blotting, and tube formation assays. Results Immunohistochemical staining showed that the CD34+CD117+ cells were located within the adventitia of HGSVs, and many CD34+ and CD117+ cells have emerged in the human venous thrombi. The number of progenitor cells within the marginal area of 7 days mice thrombi was shown to be Sca1+ ≈21%, CD34+ ≈12%, CD117+ ≈9%, and Flk1+ ≈5%. Many CD34+adventitial progenitor cells have migrated into the decellularized vessels. FACS showed that the number of CD34+CD117+ HGSV-AdPC in primary cultured cells as 1.2 ± 0.07%. After CD34+CD117+HGSV-AdPC were transplanted into the adventitia of nude mice vena cava with venous thrombi, the organization rate, nucleate cell count, endothelial cells, and macrophage cells of thrombi were shown to be significantly increased. The transplanted CD34+CD117+ HGSV-AdPC at the adventitia have crossed the vein wall, entered the venous thrombi, and differentiated into endothelial cells. The CD34+CD117+ HGSV-AdPC in the culture medium in the presence of VEGF-promoted gene and protein expression of endothelial cell markers in vitro and induced tube formation. Conclusions HGSV-AdPC could cross the vein wall and migrate from the adventitia into the venous thrombi. Increased HGSV-AdPC in the adventitia has enhanced the resolution of venous thrombi via differentiating into endothelial cells of neovascularization.
Collapse
|
15
|
DeRoo E, Martinod K, Cherpokova D, Fuchs T, Cifuni S, Chu L, Staudinger C, Wagner DD. The role of platelets in thrombus fibrosis and vessel wall remodeling after venous thrombosis. J Thromb Haemost 2021; 19:387-399. [PMID: 33058430 PMCID: PMC8530247 DOI: 10.1111/jth.15134] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 09/09/2020] [Accepted: 10/06/2020] [Indexed: 01/20/2023]
Abstract
PURPOSE Platelets are known to play an important role in venous thrombogenesis, but their role in thrombus maturation, resolution, and postthrombotic vein wall remodeling is unclear. The purpose of this study was to determine the role that circulating platelets play in the later phases of venous thrombosis. METHODS We used a murine inferior vena cava (IVC) stenosis model. Baseline studies in untreated mice were performed to determine an optimal postthrombotic time point for tissue harvest that would capture both thrombus maturation/resolution and postthrombotic vein wall remodeling. This time point was found to be postoperative day 10. After undergoing IVC ultrasound on day 2 to confirm venous thrombus formation, mice were treated with a daily injection of platelet-depleting antibody (anti-GP1bα) to maintain thrombocytopenia or with control IgG until postoperative day 10, at which time IVC and thrombi were harvested and thrombus length, volume, fibrosis, neovascularization, and smooth muscle cell invasion analyzed. Vein wall fibrosis and intimal thickening were also determined. RESULTS Mice that were made thrombocytopenic after venous thrombogenesis had thrombi that were less fibrotic, with fewer invading smooth muscle cells. Furthermore, thrombocytopenia in the setting of venous thrombosis resulted in less postthrombotic vein wall intimal thickening. Thrombus volume did not differ between thrombocytopenic mice and their control peers. CONCLUSIONS This work suggests that circulating platelets contribute to venous thrombus maturation, fibrosis, and adverse vein wall remodeling, and that that inhibition of platelet recruitment may decrease thrombus and vein wall fibrosis, thus helping thrombolysis and preventing postthrombotic syndrome.
Collapse
Affiliation(s)
- Elise DeRoo
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Kimberly Martinod
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Deya Cherpokova
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Tobias Fuchs
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Stephen Cifuni
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Long Chu
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Caleb Staudinger
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Denisa D. Wagner
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
- Division of Hematology/Oncology, Boston Children’s Hospital, Boston, MA, USA
| |
Collapse
|
16
|
Gutmann C, Siow R, Gwozdz AM, Saha P, Smith A. Reactive Oxygen Species in Venous Thrombosis. Int J Mol Sci 2020; 21:E1918. [PMID: 32168908 PMCID: PMC7139897 DOI: 10.3390/ijms21061918] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 03/02/2020] [Accepted: 03/04/2020] [Indexed: 01/03/2023] Open
Abstract
Reactive oxygen species (ROS) have physiological roles as second messengers, but can also exert detrimental modifications on DNA, proteins and lipids if resulting from enhanced generation or reduced antioxidant defense (oxidative stress). Venous thrombus (DVT) formation and resolution are influenced by ROS through modulation of the coagulation, fibrinolysis, proteolysis and the complement system, as well as the regulation of effector cells such as platelets, endothelial cells, erythrocytes, neutrophils, mast cells, monocytes and fibroblasts. Many conditions that carry an elevated risk of venous thrombosis, such as the Antiphospholipid Syndrome, have alterations in their redox homeostasis. Dietary and pharmacological antioxidants can modulate several important processes involved in DVT formation, but their overall effect is unknown and there are no recommendations regarding their use. The development of novel antioxidant treatments that aim to abrogate the formation of DVT or promote its resolution will depend on the identification of targets that enable ROS modulation confined to their site of interest in order to prevent off-target effects on physiological redox mechanisms. Subgroups of patients with increased systemic oxidative stress might benefit from unspecific antioxidant treatment, but more clinical studies are needed to bring clarity to this issue.
Collapse
Affiliation(s)
- Clemens Gutmann
- King’s British Heart Foundation Centre, King’s College London, 125 Coldharbour Lane, London SE5 9NU, UK;
| | - Richard Siow
- Vascular Biology & Inflammation Section, School of Cardiovascular Medicine & Sciences, British Heart Foundation of Research Excellence, King’s College London, SE1 9NH, UK;
| | - Adam M. Gwozdz
- Academic Department of Surgery, School of Cardiovascular Medicine & Sciences, British Heart Foundation of Research Excellence, King’s College London, London SE1 7EH, UK; (A.M.G.); (P.S.)
| | - Prakash Saha
- Academic Department of Surgery, School of Cardiovascular Medicine & Sciences, British Heart Foundation of Research Excellence, King’s College London, London SE1 7EH, UK; (A.M.G.); (P.S.)
| | - Alberto Smith
- Academic Department of Surgery, School of Cardiovascular Medicine & Sciences, British Heart Foundation of Research Excellence, King’s College London, London SE1 7EH, UK; (A.M.G.); (P.S.)
| |
Collapse
|
17
|
Diaz JA, Saha P, Cooley B, Palmer OR, Grover SP, Mackman N, Wakefield TW, Henke PK, Smith A, Lal BK. Choosing a Mouse Model of Venous Thrombosis. Arterioscler Thromb Vasc Biol 2020; 39:311-318. [PMID: 30786739 DOI: 10.1161/atvbaha.118.311818] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Murine models are widely used valuable tools to study deep vein thrombosis. Leading experts in venous thrombosis research came together through the American Venous Forum to develop a consensus on maximizing the utility and application of available mouse models of venous thrombosis. In this work, we provide an algorithm for model selection, with discussion of the advantages, disadvantages, and applications of the main mouse models of venous thrombosis. Additionally, we provide a detailed surgical description of the models with guidelines to validate surgical technique.
Collapse
Affiliation(s)
- Jose A Diaz
- From the Department of Surgery, Vascular Surgery, University of Michigan, Ann Arbor (J.A.D., O.R.P., T.W.W., P.K.H.)
| | - Prakash Saha
- Academic Department of Vascular Surgery, King's College London, UK (P.S., A.S.)
| | - Brian Cooley
- Department of Medicine, Division of Hematology/Oncology, University of North Carolina at Chapel Hill (B.C., S.P.G., N.M.)
| | - Olivia R Palmer
- From the Department of Surgery, Vascular Surgery, University of Michigan, Ann Arbor (J.A.D., O.R.P., T.W.W., P.K.H.)
| | - Steven P Grover
- Department of Medicine, Division of Hematology/Oncology, University of North Carolina at Chapel Hill (B.C., S.P.G., N.M.)
| | - Nigel Mackman
- Department of Medicine, Division of Hematology/Oncology, University of North Carolina at Chapel Hill (B.C., S.P.G., N.M.)
| | - Thomas W Wakefield
- From the Department of Surgery, Vascular Surgery, University of Michigan, Ann Arbor (J.A.D., O.R.P., T.W.W., P.K.H.)
| | - Peter K Henke
- From the Department of Surgery, Vascular Surgery, University of Michigan, Ann Arbor (J.A.D., O.R.P., T.W.W., P.K.H.)
| | - Alberto Smith
- Academic Department of Vascular Surgery, King's College London, UK (P.S., A.S.)
| | - Brajesh K Lal
- Department of Surgery, University of Maryland, College Park (B.K.L.)
| |
Collapse
|
18
|
Bochenek ML, Leidinger C, Rosinus NS, Gogiraju R, Guth S, Hobohm L, Jurk K, Mayer E, Münzel T, Lankeit M, Bosmann M, Konstantinides S, Schäfer K. Activated Endothelial TGFβ1 Signaling Promotes Venous Thrombus Nonresolution in Mice Via Endothelin-1: Potential Role for Chronic Thromboembolic Pulmonary Hypertension. Circ Res 2019; 126:162-181. [PMID: 31747868 DOI: 10.1161/circresaha.119.315259] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
RATIONALE Chronic thromboembolic pulmonary hypertension (CTEPH) is characterized by defective thrombus resolution, pulmonary artery obstruction, and vasculopathy. TGFβ (transforming growth factor-β) signaling mutations have been implicated in pulmonary arterial hypertension, whereas the role of TGFβ in the pathophysiology of CTEPH is unknown. OBJECTIVE To determine whether defective TGFβ signaling in endothelial cells contributes to thrombus nonresolution and fibrosis. METHODS AND RESULTS Venous thrombosis was induced by inferior vena cava ligation in mice with genetic deletion of TGFβ1 in platelets (Plt.TGFβ-KO) or TGFβ type II receptors in endothelial cells (End.TGFβRII-KO). Pulmonary endarterectomy specimens from CTEPH patients were analyzed using immunohistochemistry. Primary human and mouse endothelial cells were studied using confocal microscopy, quantitative polymerase chain reaction, and Western blot. Absence of TGFβ1 in platelets did not alter platelet number or function but was associated with faster venous thrombus resolution, whereas endothelial TGFβRII deletion resulted in larger, more fibrotic and higher vascularized venous thrombi. Increased circulating active TGFβ1 levels, endothelial TGFβRI/ALK1 (activin receptor-like kinase), and TGFβRI/ALK5 expression were detected in End.TGFβRII-KO mice, and activated TGFβ signaling was present in vessel-rich areas of CTEPH specimens. CTEPH-endothelial cells and murine endothelial cells lacking TGFβRII simultaneously expressed endothelial and mesenchymal markers and transcription factors regulating endothelial-to-mesenchymal transition, similar to TGFβ1-stimulated endothelial cells. Mechanistically, increased endothelin-1 levels were detected in TGFβRII-KO endothelial cells, murine venous thrombi, or endarterectomy specimens and plasma of CTEPH patients, and endothelin-1 overexpression was prevented by inhibition of ALK5, and to a lesser extent of ALK1. ALK5 inhibition and endothelin receptor antagonization inhibited mesenchymal lineage conversion in TGFβ1-exposed human and murine endothelial cells and improved venous thrombus resolution and pulmonary vaso-occlusions in End.TGFβRII-KO mice. CONCLUSIONS Endothelial TGFβ1 signaling via type I receptors and endothelin-1 contribute to mesenchymal lineage transition and thrombofibrosis, which were prevented by blocking endothelin receptors. Our findings may have relevant implications for the prevention and management of CTEPH.
Collapse
Affiliation(s)
- Magdalena L Bochenek
- From the Center for Cardiology, Cardiology I (M.L.B., C.L., N.S.R., R.G., L.H., T.M., K.S.), University Medical Center Mainz, Germany.,Center for Thrombosis and Hemostasis (M.L.B., L.H., K.J., M.L., M.B., S.K.), University Medical Center Mainz, Germany.,German Center for Cardiovascular Research (DZHK e.V.; RheinMain) (M.L.B., N.S.R., R.G., E.M., T.M., K.S.)
| | - Christiane Leidinger
- From the Center for Cardiology, Cardiology I (M.L.B., C.L., N.S.R., R.G., L.H., T.M., K.S.), University Medical Center Mainz, Germany
| | - Nico S Rosinus
- From the Center for Cardiology, Cardiology I (M.L.B., C.L., N.S.R., R.G., L.H., T.M., K.S.), University Medical Center Mainz, Germany.,German Center for Cardiovascular Research (DZHK e.V.; RheinMain) (M.L.B., N.S.R., R.G., E.M., T.M., K.S.)
| | - Rajinikanth Gogiraju
- From the Center for Cardiology, Cardiology I (M.L.B., C.L., N.S.R., R.G., L.H., T.M., K.S.), University Medical Center Mainz, Germany.,German Center for Cardiovascular Research (DZHK e.V.; RheinMain) (M.L.B., N.S.R., R.G., E.M., T.M., K.S.)
| | - Stefan Guth
- Thoracic Surgery, Kerckhoff Clinic, Bad Nauheim, Germany (S.G., E.M.)
| | - Lukas Hobohm
- From the Center for Cardiology, Cardiology I (M.L.B., C.L., N.S.R., R.G., L.H., T.M., K.S.), University Medical Center Mainz, Germany.,Center for Thrombosis and Hemostasis (M.L.B., L.H., K.J., M.L., M.B., S.K.), University Medical Center Mainz, Germany
| | - Kerstin Jurk
- Center for Thrombosis and Hemostasis (M.L.B., L.H., K.J., M.L., M.B., S.K.), University Medical Center Mainz, Germany
| | - Eckhard Mayer
- Thoracic Surgery, Kerckhoff Clinic, Bad Nauheim, Germany (S.G., E.M.).,German Center for Cardiovascular Research (DZHK e.V.; RheinMain) (M.L.B., N.S.R., R.G., E.M., T.M., K.S.)
| | - Thomas Münzel
- From the Center for Cardiology, Cardiology I (M.L.B., C.L., N.S.R., R.G., L.H., T.M., K.S.), University Medical Center Mainz, Germany.,German Center for Cardiovascular Research (DZHK e.V.; RheinMain) (M.L.B., N.S.R., R.G., E.M., T.M., K.S.)
| | - Mareike Lankeit
- Center for Thrombosis and Hemostasis (M.L.B., L.H., K.J., M.L., M.B., S.K.), University Medical Center Mainz, Germany.,Department of Internal Medicine and Cardiology, Campus Virchow Klinikum, Charité -University Medicine, Berlin, Germany (M.L.)
| | - Markus Bosmann
- Center for Thrombosis and Hemostasis (M.L.B., L.H., K.J., M.L., M.B., S.K.), University Medical Center Mainz, Germany.,Department of Medicine, Boston University School of Medicine, MA (M.B.)
| | - Stavros Konstantinides
- Center for Thrombosis and Hemostasis (M.L.B., L.H., K.J., M.L., M.B., S.K.), University Medical Center Mainz, Germany.,Department of Cardiology, Democritus University of Thrace, Alexandroupolis, Greece (S.K.)
| | - Katrin Schäfer
- From the Center for Cardiology, Cardiology I (M.L.B., C.L., N.S.R., R.G., L.H., T.M., K.S.), University Medical Center Mainz, Germany.,German Center for Cardiovascular Research (DZHK e.V.; RheinMain) (M.L.B., N.S.R., R.G., E.M., T.M., K.S.)
| |
Collapse
|
19
|
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
|
20
|
Diaz JA, Saha P, Cooley B, Palmer OR, Grover SP, Mackman N, Wakefield TW, Henke PK, Smith A, Lal BK. Choosing a mouse model of venous thrombosis: a consensus assessment of utility and application. J Thromb Haemost 2019; 17:699-707. [PMID: 30927321 DOI: 10.1111/jth.14413] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Murine models are widely used valuable tools to study deep vein thrombosis (VT). Leading experts in VT research came together through the American Venous Forum to develop a consensus on maximizing the utility and application of available mouse models of VT. In this work, we provide an algorithm for model selection, with discussion of the advantages, disadvantages, and applications of the main mouse models of VT. Additionally, we provide a detailed surgical description of the models with guidelines to validate surgical technique.
Collapse
|
21
|
Naito A, Sakao S, Lang IM, Voelkel NF, Jujo T, Ishida K, Sugiura T, Matsumiya G, Yoshino I, Tanabe N, Tatsumi K. Endothelial cells from pulmonary endarterectomy specimens possess a high angiogenic potential and express high levels of hepatocyte growth factor. BMC Pulm Med 2018; 18:197. [PMID: 30594174 PMCID: PMC6310963 DOI: 10.1186/s12890-018-0769-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Accepted: 12/18/2018] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Impaired angiogenesis is assumed to be an important factor in the development of chronic thromboembolic pulmonary hypertension (CTEPH). However, the role of endothelial cells (ECs) in CTEPH remains unclear. The aim of this study was to investigate the angiogenic potential of ECs from pulmonary endarterectomy (PEA) specimens. METHODS We isolated ECs from PEA specimens (CTEPH-ECs) and control EC lines from the intact pulmonary arteries of patients with peripheral lung cancers, using a MACS system. These cells were analyzed in vitro including PCR-array analysis, and the PEA specimens were analyzed with immunohistochemistry. Additionally, the serum HGF levels were determined in CTEPH patients. RESULTS A three-dimensional culture assay revealed that CTEPH-ECs were highly angiogenic. An angiogenesis-focused gene PCR array revealed a high expression of hepatocyte growth factor (HGF) in CTEPH-ECs. The high expression of HGF was also confirmed in the supernatant extracted from PEA specimens. The immunohistochemical analysis showed expression of HGF on the surface of the thrombus vessels. The serum HGF levels in CTEPH patients were higher than those in pulmonary thromboembolism survivors. CONCLUSION Our study suggests that there are ECs with pro-angiogenetic character and high expression of HGF in PEA specimens. It remains unknown how these results are attributable to the etiology. However, further investigation focused on the HGF pathway may provide novel diagnostic and therapeutic tools for patients with CTEPH.
Collapse
Affiliation(s)
- Akira Naito
- Department of Respirology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-Ku, Chiba City, 260-8670, Japan.,Department of Advancing Research on Treatment Strategies for respiratory disease, Graduate School of Medicine, Chiba University, 1-8-1, Inohana, Chuo-Ku, Chiba City, 260-8670, Japan
| | - Seiichiro Sakao
- Department of Respirology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-Ku, Chiba City, 260-8670, Japan.
| | - Irene M Lang
- Department of Internal Medicine II, Cardiology, Medical University of Vienna, Spitalgasse 23, 1090, Vienna, Austria
| | - Norbert F Voelkel
- Victoria Johnson Center for Obstructive Lung Disease, Virginia Commonwealth University, 1101 East Marshall Street, Sanger Hall, Richmond, VA, 23298-0565, USA
| | - Takayuki Jujo
- Department of Respirology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-Ku, Chiba City, 260-8670, Japan.,Department of Advanced Medicine in Pulmonary Hypertension, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-Ku, Chiba City, 260-8670, Japan
| | - Keiichi Ishida
- Department of Cardiovascular Surgery, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-Ku, Chiba City, 260-8670, Japan
| | - Toshihiko Sugiura
- Department of Respirology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-Ku, Chiba City, 260-8670, Japan
| | - Goro Matsumiya
- Department of Cardiovascular Surgery, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-Ku, Chiba City, 260-8670, Japan
| | - Ichiro Yoshino
- Department of Thoracic Surgery, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-Ku, Chiba City, 260-8670, Japan
| | - Nobuhiro Tanabe
- Department of Respirology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-Ku, Chiba City, 260-8670, Japan
| | - Koichiro Tatsumi
- Department of Respirology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-Ku, Chiba City, 260-8670, Japan
| |
Collapse
|
22
|
Zhang J, Zhang H, Chen Y, Fu J, Lei Y, Sun J, Tang B. Platelet‑derived growth factor D promotes the angiogenic capacity of endothelial progenitor cells. Mol Med Rep 2018; 19:125-132. [PMID: 30483778 PMCID: PMC6297765 DOI: 10.3892/mmr.2018.9692] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Accepted: 10/09/2018] [Indexed: 01/04/2023] Open
Abstract
Neovascularization and re-endothelialization rely on endothelial progenitor cells (EPCs). However, the recruitment and angiogenic roles of EPCs are subject to regulation through the vascular microenvironment, which remains largely unknown. Platelet-derived growth factor D (PDGF-D) is a new member of the PDGF family that binds the PDGFR-β homodimer. However, it remains unknown whether and how it affects the angiogenic capacity of EPCs and participates in tube-like formation. EPCs were derived from rat bone marrow cells, and the gain-of-function approach was used to study the effects of PDGF-D on the biological activities of EPCs. EPCs that stably express PDGF-D were generated by lentiviral-mediated transduction, and the expression levels were evaluated by western blotting and reverse transcription, followed by real-time quantitative polymerase chain reaction (RT-qPCR). The biological activities of EPCs evaluated in the present study included proliferation, adhesion, migration, tube formation and senescence. Furthermore, the downstream signaling of PDGF-D was explored by western blot analysis. The results revealed that the lentiviral-mediated expression of PDGF-D in the microenvironment promoted the migration, proliferation, adhesion and tube formation of EPCs. In addition, PDGF-D suppressed the senescence of EPCs. Mechanistically, PDGF-D induced the phosphorylation of several signaling molecules, including STAT3, AKT, ERK1/2, mTOR and GSK-3β, suggesting that PDGF-D enhanced the angiogenic function of EPCs through PDGF receptor-dependent and -independent signaling pathways. In conclusion, PDGF-D promotes the angiogenic capacity of EPCs, including proliferation, migration, adhesion and tube formation, and thereby contributes to angiogenesis.
Collapse
Affiliation(s)
- Jianbo Zhang
- Department of Vascular Surgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, P.R. China
| | - Haolong Zhang
- Department of Vascular Surgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, P.R. China
| | - Yikuan Chen
- Department of Vascular Surgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, P.R. China
| | - Jian Fu
- Department of Vascular Surgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, P.R. China
| | - Yu Lei
- Department of Vascular Surgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, P.R. China
| | - Jianming Sun
- Department of Vascular Surgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, P.R. China
| | - Bo Tang
- Department of Vascular Surgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, P.R. China
| |
Collapse
|
23
|
Inhibition of prolyl hydroxylase domain proteins selectively enhances venous thrombus neovascularisation. Thromb Res 2018; 169:105-112. [PMID: 30031289 DOI: 10.1016/j.thromres.2018.07.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 06/30/2018] [Accepted: 07/09/2018] [Indexed: 11/21/2022]
Abstract
BACKGROUND Hypoxia within acute venous thrombi is thought to drive resolution through stabilisation of hypoxia inducible factor 1 alpha (HIF1α). Prolyl hydroxylase domain (PHD) isoforms are critical regulators of HIF1α stability. Non-selective inhibition of PHD isoforms with l-mimosine has been shown to increase HIF1α stabilisation and promote thrombus resolution. OBJECTIVE The aim of this study was to investigate the therapeutic potential of PHD inhibition in venous thrombus resolution. METHODS Thrombosis was induced in the inferior vena cava of mice using a combination of flow restriction and endothelial activation. Gene and protein expression of PHD isoforms in the resolving thrombus was measured by RT-PCR and immunohistochemistry. Thrombus resolution was quantified in mice treated with pan PHD inhibitors AKB-4924 and JNJ-42041935 or inducible all-cell Phd2 knockouts by micro-computed tomography, 3D high frequency ultrasound or endpoint histology. RESULTS Resolving venous thrombi demonstrated significant temporal gene expression profiles for PHD2 and PHD3 (P < 0.05), but not for PHD1. PHD isoform protein expression was localised to early and late inflammatory cell infiltrates. Treatment with selective pan PHD inhibitors, AKB-4924 and JNJ-42041935, enhanced thrombus neovascularisation (P < 0.05), but had no significant effect on overall thrombus resolution. Thrombus resolution or its markers, macrophage accumulation and neovascularisation, did not differ significantly in inducible all-cell homozygous Phd2 knockouts compared with littermate controls (P > 0.05). CONCLUSIONS This data suggests that PHD-mediated thrombus neovascularisation has a limited role in the resolution of venous thrombi. Directly targeting angiogenesis alone may not be a viable therapeutic strategy to enhance venous thrombus resolution.
Collapse
|
24
|
Abstract
After achievement of adequate anticoagulation, the natural history of acute pulmonary emboli ranges from near total resolution of vascular perfusion to long-term persistence of hemodynamically consequential residual perfusion defects. The persistence of perfusion defects is necessary, but not sufficient, for the development of chronic thromboembolic pulmonary hypertension (CTEPH). Approximately 30% of patients have persistent defects after 6 months of anticoagulation, but only 10% of those with persistent defects subsequently develop CTEPH. A number of clinical risk factors including increasing age, delay in anticoagulation from symptom onset, and the size of the initial thrombus have been associated with the persistence of perfusion defects. Likewise, a number of cellular and molecular pathways have been implicated in the failure of thrombus resolution, including impaired fibrinolysis, altered fibrinogen structure and function, increased local or systemic inflammation, and remodeling of the embolic material by neovascularization. Treatment with fibrinolytic agents at the time of initial presentation has not clearly improved the frequency or degree of recovery of pulmonary vascular perfusion. A better understanding of the interplay between clinical risk factors and pathogenic mechanisms may enhance the ability to prevent and treat CTEPH in the future.
Collapse
|
25
|
Sharma S, Lang IM. Current understanding of the pathophysiology of chronic thromboembolic pulmonary hypertension. Thromb Res 2017. [PMID: 28624155 DOI: 10.1016/j.thromres.2017.06.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Chronic thromboembolic pulmonary hypertension (CTEPH) is a unique form of pulmonary hypertension arising from fibrotic obliteration of major pulmonary arteries. Pro-thrombotic states, large clot burden and impaired dissolution are believed to contribute to the occurrence and progression of thrombosis after an acute pulmonary embolic event. Recent data utilizing several models have facilitated the understanding of clot resolution. This review summarizes current knowledge on pathophysiological mechanisms of major vessel occlusion in CTEPH.
Collapse
Affiliation(s)
- Smriti Sharma
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Irene M Lang
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
26
|
Prodger A, Saha P, Smith A, Evans CE. Cancer-Associated Thrombosis: Regulatory Mechanisms and Emerging Directions. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 906:115-122. [PMID: 27638621 DOI: 10.1007/5584_2016_110] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Venous thrombosis is a common complication in cancer patients, and some cancer chemotherapies are associated with an increased risk of venous thromboembolism. The regulatory mechanisms that control thrombus formation and subsequent resolution in patients with cancer, however, are incompletely understood, and novel treatments for cancer-associated thrombosis may arise from a better understanding of such mechanisms. In this chapter, pathways that regulate cancer-associated thrombus formation are outlined, and the effects of anti-angiogenic cancer chemotherapies on venous thrombus resolution are highlighted. Potentially pro-thrombotic effects of anti-angiogenic agents are important considerations when managing the complications of venous thrombosis in cancer patients.
Collapse
Affiliation(s)
- Alice Prodger
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Prakash Saha
- Academic Department of Vascular Surgery, King's College London, London, UK
| | - Alberto Smith
- Academic Department of Vascular Surgery, King's College London, London, UK
| | - Colin E Evans
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK.
| |
Collapse
|
27
|
Grover SP, Evans CE, Patel AS, Modarai B, Saha P, Smith A. Assessment of Venous Thrombosis in Animal Models. Arterioscler Thromb Vasc Biol 2015; 36:245-52. [PMID: 26681755 DOI: 10.1161/atvbaha.115.306255] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Accepted: 11/30/2015] [Indexed: 12/19/2022]
Abstract
Deep vein thrombosis and common complications, including pulmonary embolism and post-thrombotic syndrome, represent a major source of morbidity and mortality worldwide. Experimental models of venous thrombosis have provided considerable insight into the cellular and molecular mechanisms that regulate thrombus formation and subsequent resolution. Here, we critically appraise the ex vivo and in vivo techniques used to assess venous thrombosis in these models. Particular attention is paid to imaging modalities, including magnetic resonance imaging, micro-computed tomography, and high-frequency ultrasound that facilitate longitudinal assessment of thrombus size and composition.
Collapse
Affiliation(s)
- Steven P Grover
- From the Cardiovascular Division, Academic Department of Vascular Surgery, Kings College London, BHF Centre of Research Excellence and NIHR Biomedical Research Centre at Kings Health Partners, St Thomas' Hospital, London, United Kingdom
| | - Colin E Evans
- From the Cardiovascular Division, Academic Department of Vascular Surgery, Kings College London, BHF Centre of Research Excellence and NIHR Biomedical Research Centre at Kings Health Partners, St Thomas' Hospital, London, United Kingdom
| | - Ashish S Patel
- From the Cardiovascular Division, Academic Department of Vascular Surgery, Kings College London, BHF Centre of Research Excellence and NIHR Biomedical Research Centre at Kings Health Partners, St Thomas' Hospital, London, United Kingdom
| | - Bijan Modarai
- From the Cardiovascular Division, Academic Department of Vascular Surgery, Kings College London, BHF Centre of Research Excellence and NIHR Biomedical Research Centre at Kings Health Partners, St Thomas' Hospital, London, United Kingdom
| | - Prakash Saha
- From the Cardiovascular Division, Academic Department of Vascular Surgery, Kings College London, BHF Centre of Research Excellence and NIHR Biomedical Research Centre at Kings Health Partners, St Thomas' Hospital, London, United Kingdom.
| | - Alberto Smith
- From the Cardiovascular Division, Academic Department of Vascular Surgery, Kings College London, BHF Centre of Research Excellence and NIHR Biomedical Research Centre at Kings Health Partners, St Thomas' Hospital, London, United Kingdom
| |
Collapse
|
28
|
Altmann J, Sharma S, Lang IM. Advances in our understanding of mechanisms of venous thrombus resolution. Expert Rev Hematol 2015; 9:69-78. [PMID: 26629617 DOI: 10.1586/17474086.2016.1112264] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Traditionally, venous thrombosis has been seen as the consequence of a regulated cascade of proteolytic steps leading to the polymerization of fibrinogen and fibrin crosslinking that is facilitated by platelets. A new view of thrombosis is providing a more integrated concept, with components of the vascular wall contributing to the vascular remodeling of thrombosis. Angiogenesis and inflammation are two key mechanisms that safeguard venous thrombus resolution and restitution of vascular patency after thrombosis. Disturbance of these processes leads to thrombus persistence and has potentially severe consequences for affected patients. Examples for clinical conditions associated with recurrent or persisting venous thrombosis are post-thrombotic syndrome or chronic thromboembolic pulmonary hypertension. Recently, studies using animal models of venous thrombosis have contributed to a better understanding of thrombus non-resolution that will eventually lead to modification of current treatment concepts. For example, recent data suggest that innate immunity is involved in the modification of thrombosis.
Collapse
Affiliation(s)
- Johanna Altmann
- a Division of Cardiology, Department of Internal Medicine II , Vienna General Hospital, Medical University of Vienna , Vienna , Austria
| | - Smriti Sharma
- a Division of Cardiology, Department of Internal Medicine II , Vienna General Hospital, Medical University of Vienna , Vienna , Austria
| | - Irene M Lang
- a Division of Cardiology, Department of Internal Medicine II , Vienna General Hospital, Medical University of Vienna , Vienna , Austria
| |
Collapse
|
29
|
Wang X, Wang HQ, Jian T, Qin WW, Xu F, Xin ZL, Wang YH, Zhang M, Lu HJ. VEGFR2 Gene Polymorphism Correlates with Deep Venous Thrombosis Risk in Chinese Han Population. Genet Test Mol Biomarkers 2015; 19:673-8. [PMID: 26600200 DOI: 10.1089/gtmb.2015.0129] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
OBJECTIVE To investigate the correlations between three vascular endothelial growth factor 2 (VEGFR2) gene polymorphisms, +1192C>T, +1719T>A, and -604T>C, and deep venous thrombosis (DVT) in Chinese Han population. METHODS We conducted a case-control study, between September 2009 and September 2012, in a Chinese Han population with onset of lower extremity DVT. A total of 135 patients were enrolled in the case group and 156 healthy individuals in the control group. Polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) was used to detect the genotype and allele frequencies of the VEGFR2 gene polymorphisms +1192C>T, +1719T>A, and -604T>C. Haplotype analyses were conducted with SHEsis program. Logistic regression was used to detect the risk factors of DVT. Outpatient review and telephone follow-up were conducted to analyze the long-term treatment of DVT patients. RESULTS The allele and genotype frequencies of -604T>C VEGFR2 polymorphism exhibited significant differences between the case and control groups (both p < 0.05). Haplotype analyses showed remarkable differences between the case and control groups in the distribution frequency of TAC and CTT haplotypes in the VEGFR2 gene (both p < 0.05). Logistic regression analysis showed independent correlation between the incidence of DVT and TAC haplotype in the VEGFR2 gene (p < 0.05). In addition, the TAC haplotype may be a risk factor for DVT treatment failure. CONCLUSION Our findings suggest that the VEGFR2 gene -604T>C polymorphism and TAC haplotype are associated with DVT, and the TAC haplotype might affect the efficacy of long-term treatment of DVT patients.
Collapse
Affiliation(s)
- Xu Wang
- 1 Department of Vascular Surgery, Jining No. 1 People's Hospital , Jining, People's Republic of China
| | - Hai-Qing Wang
- 1 Department of Vascular Surgery, Jining No. 1 People's Hospital , Jining, People's Republic of China
| | - Tao Jian
- 1 Department of Vascular Surgery, Jining No. 1 People's Hospital , Jining, People's Republic of China
| | - Wei-Wei Qin
- 1 Department of Vascular Surgery, Jining No. 1 People's Hospital , Jining, People's Republic of China
| | - Fei Xu
- 1 Department of Vascular Surgery, Jining No. 1 People's Hospital , Jining, People's Republic of China
| | - Zhen-Lei Xin
- 1 Department of Vascular Surgery, Jining No. 1 People's Hospital , Jining, People's Republic of China
| | - Yan-Hua Wang
- 2 Operation Room, Jining No. 1 People's Hospital , Jining, People's Republic of China
| | - Mi Zhang
- 1 Department of Vascular Surgery, Jining No. 1 People's Hospital , Jining, People's Republic of China
| | - Hui-Jun Lu
- 2 Operation Room, Jining No. 1 People's Hospital , Jining, People's Republic of China
| |
Collapse
|
30
|
Maiborodin IV, Morozov VV, Markevich YV, Matveeva VA, Artem'eva LV, Matveev AL, Chastikin GA, Seryapina YV. Acceleration of Angiogenesis after Paravasal Injection of Mesenchymal Stem Cells at the Site of Modeled Venous Thrombosis. Bull Exp Biol Med 2015; 159:128-33. [PMID: 26033602 DOI: 10.1007/s10517-015-2907-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Indexed: 11/26/2022]
Abstract
The results of transplantation of autologous bone marrow multipotent mesenchymal stem cells carrying GFP gene and labeled with cell nucleus-specific dye DAPI near the thrombosed vein in rat hind limb were studied by methods of luminescent microscopy. It was demonstrated that autologous multipotent mesenchymal stem cells participate in the formation of granulations at the site of surgery. The blood fl ow in the thrombosed great vein was always restored through thrombolysis. We observed no signs of incorporation of the transplanted cells into the wall of the great vessel, clot recanalization, or formation of collaterals. Small branches of the great vein in the affected region were also thrombosed. The blood fl ow in these branches was always restored with participation of the transplanted cells or through clot recanalization or through obliteration of the thrombosed vessels and formation of new vessels. The transplanted cells and structures formed by them were gradually replaced by the recipient cells.
Collapse
Affiliation(s)
- I V Maiborodin
- Center of New Medical Technologies, Institute of Chemical Biology and Fundamental Medicine, Siberian Division of the Russian Academy of Sciences, Novosibirsk, Russia,
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Abstract
Chronic thromboembolic pulmonary hypertension (CTEPH) is a distinct subtype of pulmonary hypertension (PH). One disease hypothesis is that CTEPH results from the non-resolution of venous thromboembolism. CTEPH is characterised by the presence of obstructive fibrotic thromboembolic material in the major pulmonary vessels, with concomitant microvascular arteriopathy, resulting in progressive PH. The clinical presentation of CTEPH is similar to pulmonary arterial hypertension with nonspecific symptoms, but it is distinguished from pulmonary arterial hypertension by the presence of mismatched segmental defects on the ventilation/perfusion scan. The exact prevalence and incidence of CTEPH are unknown, but are thought to have been underestimated in the past. CTEPH is unique among the subgroups of PH in that it is potentially curable with pulmonary endarterectomy, a surgical intervention intended to remove the occlusive material from the pulmonary vasculature. However, in some patients the obstructions are technically inaccessible or the risk/benefit ratios are unfavourable, making the condition inoperable. It is thought that the involvement of the smaller, more distal vessels is a target for medical treatment. Untreated, CTEPH may result in right heart failure and death. The pathophysiological mechanisms which cause CTEPH are complex and have not yet been fully elucidated. CTEPH is distinct from other types of pulmonary hypertension, both in terms of its pathophysiology and treatmenthttp://ow.ly/L54ag
Collapse
|
32
|
Chabasse C, Siefert SA, Chaudry M, Hoofnagle MH, Lal BK, Sarkar R. Recanalization and flow regulate venous thrombus resolution and matrix metalloproteinase expression in vivo. J Vasc Surg Venous Lymphat Disord 2015; 3:64-74. [PMID: 26993683 PMCID: PMC4892699 DOI: 10.1016/j.jvsv.2014.03.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Accepted: 03/11/2014] [Indexed: 10/25/2022]
Abstract
OBJECTIVE We examined the role of thrombus recanalization and ongoing blood flow in the process of thrombus resolution by comparing two murine in vivo models of deep venous thrombosis. METHODS In CD1 mice, we performed surgical inferior vena cava ligation (stasis thrombosis), stenosis (thrombosis with recanalization), or sham procedure. We analyzed thrombus weight over time as a measure of thrombus resolution and quantified the messenger RNA and protein levels of membrane-type matrix metalloproteinases (MT-MMPs) as well as effectors of the plasmin complex at days 4, 8, and 12 after surgery. RESULTS Despite similar initial thrombus size, the presence of ongoing blood flow (stenosis model) was associated with a 45.91% subsequent improvement in thrombus resolution at day 8 and 12.57% at day 12 compared with stasis thrombosis (ligation model). Immunoblot and real-time polymerase chain reaction analysis demonstrated a difference in MMP-2 and MMP-9 activity at day 8 between the two models (P = .03 and P = .006, respectively) as well as a difference in MT2-MMP gene expression at day 8 (P = .044) and day 12 (P = .03) and MT1-MMP protein expression at day 4 (P = .021). Histologic analyses revealed distinct areas of recanalization in the thrombi of the stenosis model compared with the ligation model as well as the recruitment of inflammatory cells, especially macrophages, and a focal pattern of localized expression of MT1-MMP and MT3-MMP proteins surrounding the areas of recanalization in the stenosis model. CONCLUSIONS Recanalization and ongoing blood flow accelerate deep venous thrombus resolution in vivo and are associated with distinct patterns of MT1-MMP and MT3-MMP expression and macrophage localization in areas of intrathrombus recanalization.
Collapse
Affiliation(s)
- Christine Chabasse
- Center for Vascular and Inflammatory Diseases, School of Medicine, University of Maryland, Baltimore, Md; Department of Surgery, School of Medicine, University of Maryland, Baltimore, Md
| | - Suzanne A Siefert
- Center for Vascular and Inflammatory Diseases, School of Medicine, University of Maryland, Baltimore, Md; Department of Surgery, School of Medicine, University of Maryland, Baltimore, Md
| | - Mohammed Chaudry
- Center for Vascular and Inflammatory Diseases, School of Medicine, University of Maryland, Baltimore, Md; Department of Surgery, School of Medicine, University of Maryland, Baltimore, Md
| | - Mark H Hoofnagle
- Center for Vascular and Inflammatory Diseases, School of Medicine, University of Maryland, Baltimore, Md; Department of Surgery, School of Medicine, University of Maryland, Baltimore, Md
| | - Brajesh K Lal
- Department of Surgery, School of Medicine, University of Maryland, Baltimore, Md
| | - Rajabrata Sarkar
- Center for Vascular and Inflammatory Diseases, School of Medicine, University of Maryland, Baltimore, Md; Department of Surgery, School of Medicine, University of Maryland, Baltimore, Md.
| |
Collapse
|
33
|
Alias S, Lang IM. Coagulation and the vessel wall in pulmonary embolism. Pulm Circ 2014; 3:728-38. [PMID: 25006391 DOI: 10.1086/674768] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2012] [Accepted: 08/05/2013] [Indexed: 01/28/2023] Open
Abstract
Venous thromboembolism comprises deep-vein thrombosis, thrombus in transit, acute pulmonary embolism, and chronic thromboembolic pulmonary hypertension (CTEPH). Pulmonary thromboemboli commonly resolve, with restoration of normal pulmonary hemodynamics. When they fail to resorb, permanent occlusion of the deep veins and/or CTEPH are the consequences. Apart from endogenous fibrinolysis, venous thrombi resolve by a process of mechanical fragmentation, through organization of the thromboembolus by invasion of endothelial cells, leukocytes, and fibroblasts leading to recanalization. Recent data utilizing various models have contributed to a better understanding of venous thrombosis and the resolution process that is directed at maintaining vascular patency. This review summarizes the plasmatic and cellular components of venous thrombus formation and resolution.
Collapse
Affiliation(s)
- Sherin Alias
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Irene M Lang
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
34
|
Hoeper MM, Madani MM, Nakanishi N, Meyer B, Cebotari S, Rubin LJ. Chronic thromboembolic pulmonary hypertension. THE LANCET RESPIRATORY MEDICINE 2014; 2:573-82. [PMID: 24898750 DOI: 10.1016/s2213-2600(14)70089-x] [Citation(s) in RCA: 118] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
Chronic thromboembolic pulmonary hypertension (CTEPH) is a rare but debilitating and life-threatening complication of acute pulmonary embolism. CTEPH results from persistent obstruction of pulmonary arteries and progressive vascular remodelling. Not all patients presenting with CTEPH have a history of clinically overt pulmonary embolism. The diagnostic work-up to detect or rule out CTEPH should include ventilation-perfusion scintigraphy, which has high sensitivity and a negative predictive value of nearly 100%. CT angiography usually reveals typical features of CTEPH, including mosaic perfusion, part or complete occlusion of pulmonary arteries, and intraluminal bands and webs. Patients with suspected CTEPH should be referred to a specialist centre for right-heart catheterisation and pulmonary angiography. Surgical pulmonary endarterectomy remains the treatment of choice for CTEPH and is associated with excellent long-term results and a high probability of cure. For patients with inoperable CTEPH, various medical and interventional therapies are being developed.
Collapse
Affiliation(s)
- Marius M Hoeper
- Department of Respiratory Medicine, Hannover Medical School and German Centre for Lung Research (DZL), Hannover, Germany.
| | - Michael M Madani
- Department of Cardiothoracic Surgery, University of California, San Diego, CA, USA
| | - Norifumi Nakanishi
- Department of Cardiovascular Medicine, National Cardiovascular Centre, Osaka, Japan
| | - Bernhard Meyer
- Department of Radiology, Hannover Medical School, Hannover, Germany
| | - Serghei Cebotari
- Department of Cardiovascular, Thoracic and Transplantation Surgery, Hannover Medical School, Hannover, Germany
| | - Lewis J Rubin
- Department of Respiratory Medicine, University of California, San Diego, CA, USA
| |
Collapse
|
35
|
Candan F, Yildiz G, Kayataş M. Role of the VEGF 936 gene polymorphism and VEGF-A levels in the late-term arteriovenous fistula thrombosis in patients undergoing hemodialysis. Int Urol Nephrol 2014; 46:1815-23. [PMID: 24748065 DOI: 10.1007/s11255-014-0711-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2014] [Accepted: 03/31/2014] [Indexed: 12/16/2022]
Abstract
PURPOSE Vascular access is vital for hemodialysis patients. A major factor that facilitates arteriovenous (AV) fistula failure is stenosis and thrombosis due to intimal hyperplasia developing in the venous segment of AV fistula. It has been reported that VEGF accelerated re-endothelialization, reduction in intimal thickening, and/or mural thrombus formed in the injured vascular structures. In this study, we aimed to identify the effect of the VEGF 936 gene polymorphism and vascular endothelial growth factor-A (VEGF-A) levels in the late period of AV fistula loss in hemodialysis patients. METHODS The study was carried out with a patient group of 42 individuals who experienced two or more fistula thrombosis in the late period after the AV fistula operation and also a control group of 38 patients who have not had any AV fistula thrombosis history for 3 years or more. All participants were assessed for VEGF-936C/T gene polymorphism and VEGF-A levels. RESULTS VEGF-936C/T genotypes were determined in the large proportion in the control group (31.6 %), while VEGF-936C/C genotypes were determined in a large proportion in the patient group (90.5 %). Individuals carrying the VEGF-936C/C genotype had an increased risk of 5.54 for getting AV fistula thrombosis. The VEGF-A levels of patient group (27.3 ± 43.5 pg/ml) were significantly lower than those of the control group (70.7 ± 53.1 pg/ml). CONCLUSION There is an increased risk of AV fistula thrombosis in individuals carrying the VEGF-936C/C genotype. The other renal replacement modalities should be considered in patients with this genotype. As a result, it will be possible to prevent the morbidity and mortality due to fistula failure.
Collapse
Affiliation(s)
- Ferhan Candan
- Division of Nephrology, Department of Internal Medicine, Faculty of Medicine, Cumhuriyet University, 58140, Sivas, Turkey
| | | | | |
Collapse
|
36
|
Why acute pulmonary embolism becomes chronic thromboembolic pulmonary hypertension: clinical and genetic insights. Curr Opin Pulm Med 2014; 19:422-9. [PMID: 23907454 DOI: 10.1097/mcp.0b013e328364379f] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
PURPOSE OF REVIEW Chronic thromboembolic pulmonary hypertension (CTEPH) is a life-threatening complication that affects a small but appreciable percentage of patients after acute pulmonary embolism. The cause of CTEPH is under investigation, but no single causative mechanism has yet been identified. RECENT FINDINGS CTEPH is likely a complication of residual thrombotic material in the pulmonary arteries that becomes transformed into intravascular scars. Pulmonary artery residua are relatively common after acute pulmonary embolism, and CTEPH may be an extreme manifestation of this phenomenon. Several intriguing observations have been made in patients with CTEPH that give insights into the mechanisms responsible for its formation. Two general pathways have been investigated: resistance of thromboemboli to lysis and attenuation of cellular processes involved in thrombus resolution. This review discusses the evidence supporting each pathway as a mechanism for CTEPH formation, as well as the interaction between the two. SUMMARY CTEPH may be due to a complex interaction between thrombotic/thrombolytic processes and angiogenic cellular remodeling of organized thrombi. The factors involved may, in fact, vary among CTEPH patients. An understanding of the interplay between the factors that cause CTEPH may help quantify the risk of its occurrence and provide insights into how it can be prevented.
Collapse
|
37
|
Evans CE, Grover SP, Humphries J, Saha P, Patel AP, Patel AS, Lyons OT, Waltham M, Modarai B, Smith A. Antiangiogenic Therapy Inhibits Venous Thrombus Resolution. Arterioscler Thromb Vasc Biol 2014; 34:565-70. [DOI: 10.1161/atvbaha.113.302998] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Colin E. Evans
- From the Academic Department of Vascular Surgery, Cardiovascular Division, Kings College London, BHF Centre of Research Excellence & NIHR Biomedical Research Centre at Kings Health Partners, St Thomas’ Hospital, London, United Kingdom
| | - Steven P. Grover
- From the Academic Department of Vascular Surgery, Cardiovascular Division, Kings College London, BHF Centre of Research Excellence & NIHR Biomedical Research Centre at Kings Health Partners, St Thomas’ Hospital, London, United Kingdom
| | - Julia Humphries
- From the Academic Department of Vascular Surgery, Cardiovascular Division, Kings College London, BHF Centre of Research Excellence & NIHR Biomedical Research Centre at Kings Health Partners, St Thomas’ Hospital, London, United Kingdom
| | - Prakash Saha
- From the Academic Department of Vascular Surgery, Cardiovascular Division, Kings College London, BHF Centre of Research Excellence & NIHR Biomedical Research Centre at Kings Health Partners, St Thomas’ Hospital, London, United Kingdom
| | - Anant P. Patel
- From the Academic Department of Vascular Surgery, Cardiovascular Division, Kings College London, BHF Centre of Research Excellence & NIHR Biomedical Research Centre at Kings Health Partners, St Thomas’ Hospital, London, United Kingdom
| | - Ashish S. Patel
- From the Academic Department of Vascular Surgery, Cardiovascular Division, Kings College London, BHF Centre of Research Excellence & NIHR Biomedical Research Centre at Kings Health Partners, St Thomas’ Hospital, London, United Kingdom
| | - Oliver T. Lyons
- From the Academic Department of Vascular Surgery, Cardiovascular Division, Kings College London, BHF Centre of Research Excellence & NIHR Biomedical Research Centre at Kings Health Partners, St Thomas’ Hospital, London, United Kingdom
| | - Matt Waltham
- From the Academic Department of Vascular Surgery, Cardiovascular Division, Kings College London, BHF Centre of Research Excellence & NIHR Biomedical Research Centre at Kings Health Partners, St Thomas’ Hospital, London, United Kingdom
| | - Bijan Modarai
- From the Academic Department of Vascular Surgery, Cardiovascular Division, Kings College London, BHF Centre of Research Excellence & NIHR Biomedical Research Centre at Kings Health Partners, St Thomas’ Hospital, London, United Kingdom
| | - Alberto Smith
- From the Academic Department of Vascular Surgery, Cardiovascular Division, Kings College London, BHF Centre of Research Excellence & NIHR Biomedical Research Centre at Kings Health Partners, St Thomas’ Hospital, London, United Kingdom
| |
Collapse
|
38
|
Frey MK, Alias S, Winter MP, Redwan B, Stübiger G, Panzenboeck A, Alimohammadi A, Bonderman D, Jakowitsch J, Bergmeister H, Bochkov V, Preissner KT, Lang IM. Splenectomy is modifying the vascular remodeling of thrombosis. J Am Heart Assoc 2014; 3:e000772. [PMID: 24584745 PMCID: PMC3959675 DOI: 10.1161/jaha.113.000772] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Background Splenectomy is a clinical risk factor for complicated thrombosis. We hypothesized that the loss of the mechanical filtering function of the spleen may enrich for thrombogenic phospholipids in the circulation, thereby affecting the vascular remodeling of thrombosis. Methods and Results We investigated the effects of splenectomy both in chronic thromboembolic pulmonary hypertension (CTEPH), a human model disease for thrombus nonresolution, and in a mouse model of stagnant flow venous thrombosis mimicking deep vein thrombosis. Surgically excised thrombi from rare cases of CTEPH patients who had undergone previous splenectomy were enriched for anionic phospholipids like phosphatidylserine. Similar to human thrombi, phosphatidylserine accumulated in thrombi after splenectomy in the mouse model. A postsplenectomy state was associated with larger and more persistent thrombi. Higher counts of procoagulant platelet microparticles and increased leukocyte–platelet aggregates were observed in mice after splenectomy. Histological inspection revealed a decreased number of thrombus vessels. Phosphatidylserine‐enriched phospholipids specifically inhibited endothelial proliferation and sprouting. Conclusions After splenectomy, an increase in circulating microparticles and negatively charged phospholipids is enhanced by experimental thrombus induction. The initial increase in thrombus volume after splenectomy is due to platelet activation, and the subsequent delay of thrombus resolution is due to inhibition of thrombus angiogenesis. The data illustrate a potential mechanism of disease in CTEPH.
Collapse
Affiliation(s)
- Maria K Frey
- Department of Cardiology, Medical University Vienna, Vienna, Austria
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Alias S, Redwan B, Panzenboeck A, Winter MP, Schubert U, Voswinckel R, Frey MK, Jakowitsch J, Alimohammadi A, Hobohm L, Mangold A, Bergmeister H, Sibilia M, Wagner EF, Mayer E, Klepetko W, Hoelzenbein TJ, Preissner KT, Lang IM. Defective angiogenesis delays thrombus resolution: a potential pathogenetic mechanism underlying chronic thromboembolic pulmonary hypertension. Arterioscler Thromb Vasc Biol 2014; 34:810-819. [PMID: 24526692 DOI: 10.1161/atvbaha.113.302991] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Restoration of patency is a natural target of vascular remodeling after venous thrombosis that involves vascular endothelial cells and smooth muscle cells, as well as leukocytes. Acute pulmonary emboli usually resolve <6 months. However, in some instances, thrombi transform into fibrous vascular obstructions, resulting in occlusion of the deep veins, or in chronic thromboembolic pulmonary hypertension (CTEPH). We proposed that dysregulated thrombus angiogenesis may contribute to thrombus persistence. APPROACH AND RESULTS Mice with an endothelial cell-specific conditional deletion of vascular endothelial growth factor receptor 2/kinase insert domain protein receptor were used in a model of stagnant flow venous thrombosis closely resembling human deep vein thrombosis. Biochemical and functional analyses were performed on pulmonary endarterectomy specimens from patients with CTEPH, a human model of nonresolving venous thromboembolism. Endothelial cell-specific deletion of kinase insert domain protein receptor and subsequent ablation of thrombus vascularization delayed thrombus resolution. In accordance with these findings, organized human CTEPH thrombi were largely devoid of vascular structures. Several vessel-specific genes, such as kinase insert domain protein receptor, vascular endothelial cadherin, and podoplanin, were expressed at lower levels in white CTEPH thrombi than in organizing deep vein thrombi and organizing thrombi from aortic aneurysms. In addition, red CTEPH thrombi attenuated the angiogenic response induced by vascular endothelial growth factor. CONCLUSIONS In the present work, we propose a mechanism of thrombus nonresolution demonstrating that endothelial cell-specific deletion of kinase insert domain protein receptor abates thrombus vessel formation, misguiding thrombus resolution. Medical conditions associated with the development of CTEPH may be compromising early thrombus angiogenesis.
Collapse
Affiliation(s)
- Sherin Alias
- Department of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Bassam Redwan
- Department of Cardiothoracic Surgery, University Hospital Muenster, Muenster, Germany
| | | | - Max P Winter
- Department of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Uwe Schubert
- Institute for Biochemistry, Medical School, Justus-Liebig-University, Giessen, Germany
| | - Robert Voswinckel
- Department of Lung Development and Remodeling, Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Maria K Frey
- Department of Cardiology, Medical University of Vienna, Vienna, Austria
| | | | | | - Lukas Hobohm
- Department of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Andreas Mangold
- Department of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Helga Bergmeister
- Department of Biomedical Research, Medical University of Vienna, Vienna, Austria
| | - Maria Sibilia
- Institute for Cancer Research, Department of Medicine I, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Erwin F Wagner
- Centro Nacional de Investigaciones Oncológicas, Madrid, Spain
| | - Eckhard Mayer
- Department of Thoracic Surgery, Kerckhoff Clinic Heart and Lung Centre, Bad Nauheim, Germany
| | - Walter Klepetko
- Department of Cardiothoracic Surgery, Medical University of Vienna, Vienna, Austria
| | - Thomas J Hoelzenbein
- Department of Vascular and Endovascular Surgery, Paracelsus Medical University, Salzburg, Austria
| | - Klaus T Preissner
- Institute for Biochemistry, Medical School, Justus-Liebig-University, Giessen, Germany
| | - Irene M Lang
- Department of Cardiology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
40
|
Qin C, Lan X, He J, Xia X, Tian Y, Pei Z, Yuan H, Zhang Y. An in vitro and in vivo evaluation of a reporter gene/probe system hERL/(18)F-FES. PLoS One 2013; 8:e61911. [PMID: 23593502 PMCID: PMC3625158 DOI: 10.1371/journal.pone.0061911] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2012] [Accepted: 03/14/2013] [Indexed: 11/18/2022] Open
Abstract
Purpose To evaluate the feasibility of a reporter gene/probe system, namely the human estrogen receptor ligand binding domain (hERL)/16α-[18F] fluoro-17β-estradiol (18F-FES), for monitoring gene and cell therapy. Methods The recombinant adenovirus vector Ad5-hERL-IRES-VEGF (Ad-EIV), carrying a reporter gene (hERL) and a therapeutic gene (vascular endothelial growth factor, VEGF165) through an internal ribosome entry site (IRES), was constructed. After transfection of Ad-EIV into bone marrow mesenchymal stem cells (Ad-EIV-MSCs), hERL and VEGF165 mRNA and protein expressions were identified using Real-Time qRT-PCR and immunofluorescence. The uptake of 18F-FES was measured in both Ad-EIV-MSCs and nontransfected MSCs after different incubation time. Micro-PET/CT images were obtained at 1 day after injection of Ad-EIV-MSCs into the left foreleg of the rat. The right foreleg was injected with nontransfected MSCs, which served as self-control. Results After transfection with Ad-EIV, the mRNA and protein expression of hERL and VEGF165 were successfully detected in MSCs, and correlated well with each other (R2 = 0.9840, P<0.05). This indicated the reporter gene could reflect the therapeutic gene indirectly. Ad-EIV-MSCs uptake of 18F-FES increased with incubation time with a peak value of 9.13%±0.33% at 150 min, which was significantly higher than that of the control group. A far higher level of radioactivity could be seen in the left foreleg on the micro-PET/CT image than in the opposite foreleg. Conclusion These preliminary in vitro and in vivo studies confirmed that hERL/18F-FES might be used as a novel reporter gene/probe system for monitoring gene and cell therapy. This imaging platform may have broad applications for basic research and clinical studies.
Collapse
Affiliation(s)
- Chunxia Qin
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoli Lan
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- * E-mail:
| | - Jiang He
- Department of Radiology and Medical Imaging, University of Virginia, School of Medicine, Charlottesville, Virginia, United States of America
| | - Xiaotian Xia
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yueli Tian
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhijun Pei
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hui Yuan
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yongxue Zhang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
41
|
Bäck M, Gasser TC, Michel JB, Caligiuri G. Biomechanical factors in the biology of aortic wall and aortic valve diseases. Cardiovasc Res 2013; 99:232-41. [PMID: 23459103 PMCID: PMC3695745 DOI: 10.1093/cvr/cvt040] [Citation(s) in RCA: 153] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
The biomechanical factors that result from the haemodynamic load on the cardiovascular system are a common denominator of several vascular pathologies. Thickening and calcification of the aortic valve will lead to reduced opening and the development of left ventricular outflow obstruction, referred to as aortic valve stenosis. The most common pathology of the aorta is the formation of an aneurysm, morphologically defined as a progressive dilatation of a vessel segment by more than 50% of its normal diameter. The aortic valve is exposed to both haemodynamic forces and structural leaflet deformation as it opens and closes with each heartbeat to assure unidirectional flow from the left ventricle to the aorta. The arterial pressure is translated into tension-dominated mechanical wall stress in the aorta. In addition, stress and strain are related through the aortic stiffness. Furthermore, blood flow over the valvular and vascular endothelial layer induces wall shear stress. Several pathophysiological processes of aortic valve stenosis and aortic aneurysms, such as macromolecule transport, gene expression alterations, cell death pathways, calcification, inflammation, and neoangiogenesis directly depend on biomechanical factors.
Collapse
Affiliation(s)
- Magnus Bäck
- Center for Molecular Medicine, Department of Medicine, Karolinska Institutet, Stockholm, Sweden.
| | | | | | | |
Collapse
|
42
|
Ripplinger CM, Kessinger CW, Li C, Kim JW, McCarthy JR, Weissleder R, Henke PK, Lin CP, Jaffer FA. Inflammation modulates murine venous thrombosis resolution in vivo: assessment by multimodal fluorescence molecular imaging. Arterioscler Thromb Vasc Biol 2012; 32:2616-24. [PMID: 22995524 PMCID: PMC3516622 DOI: 10.1161/atvbaha.112.251983] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2012] [Accepted: 09/06/2012] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Assessment of thrombus inflammation in vivo could provide new insights into deep vein thrombosis (DVT) resolution. Here, we develop and evaluate 2 integrated fluorescence molecular-structural imaging strategies to quantify DVT-related inflammation and architecture and to assess the effect of thrombus inflammation on subsequent DVT resolution in vivo. METHODS AND RESULTS Murine DVT were created with topical 5% FeCl(3) application to thigh or jugular veins (n=35). On day 3, mice received macrophage and matrix metalloproteinase activity fluorescence imaging agents. On day 4, integrated assessment of DVT inflammation and architecture was performed using confocal fluorescence intravital microscopy. Day 4 analyses showed robust relationships among in vivo thrombus macrophages, matrix metalloproteinase activity, and fluorescein isothiocyanate-dextran deposition (r>0.70; P<0.01). In a serial 2-time point study, mice with DVT underwent intravital microscopy at day 4 and day 6. Analyses revealed that the intensity of thrombus inflammation at day 4 predicted the magnitude of DVT resolution at day 6 (P<0.05). In a second approach, noninvasive fluorescence molecular tomography-computed tomography was used and detected macrophages within jugular DVT (P<0.05 versus sham controls). CONCLUSIONS Integrated fluorescence molecular-structural imaging demonstrates that the DVT-induced inflammatory response can be readily assessed in vivo and can inform the magnitude of thrombus resolution.
Collapse
Affiliation(s)
- Crystal M. Ripplinger
- Cardiovascular Research Center, Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, MA
- Department of Pharmacology, UC Davis School of Medicine, Davis, CA
| | - Chase W. Kessinger
- Cardiovascular Research Center, Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Chunqiang Li
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Jin Won Kim
- Cardiovascular Research Center, Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, MA
- Cardiovascular Center, Korea University, Guro Hospital, Seoul, Republic of Korea
| | - Jason R. McCarthy
- Center for Molecular Imaging Research, Massachusetts General Hospital, Harvard Medical School, Boston, MA
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Ralph Weissleder
- Center for Molecular Imaging Research, Massachusetts General Hospital, Harvard Medical School, Boston, MA
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Peter K. Henke
- Section of Vascular Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI
| | - Charles P. Lin
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Farouc A. Jaffer
- Cardiovascular Research Center, Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, MA
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA
- Center for Molecular Imaging Research, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
43
|
Evans CE, Humphries J, Mattock K, Saha P, Smith A. HIF1 signalling regulates venous thrombus resolution. Thromb Res 2012; 130:971-3. [PMID: 22909829 DOI: 10.1016/j.thromres.2012.08.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2012] [Revised: 08/02/2012] [Accepted: 08/02/2012] [Indexed: 10/28/2022]
|
44
|
Evans CE, Humphries J, Waltham M, Saha P, Mattock K, Patel A, Ahmad A, Wadoodi A, Modarai B, Burnand K, Smith A. Adenoviral delivery of constitutively active HIF1alpha into venous thrombus. Thromb Res 2012; 129:812-4. [PMID: 22425317 DOI: 10.1016/j.thromres.2012.02.043] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2012] [Revised: 02/21/2012] [Accepted: 02/22/2012] [Indexed: 10/28/2022]
|
45
|
Nosaka M, Ishida Y, Kimura A, Kuninaka Y, Inui M, Mukaida N, Kondo T. Absence of IFN-γ accelerates thrombus resolution through enhanced MMP-9 and VEGF expression in mice. J Clin Invest 2011; 121:2911-2920. [PMID: 21646723 PMCID: PMC3223815 DOI: 10.1172/jci40782] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2011] [Accepted: 04/20/2011] [Indexed: 11/17/2022] Open
Abstract
Deep vein thrombosis (DVT) is a major cause of pulmonary thromboembolism, a leading cause of death in individuals with DVT. Several lines of evidence indicate proinflammatory cytokines such as TNF-α are involved in thrombus formation and resolution, but the roles of IFN-γ remain unclear. To address this issue, we performed ligation of the inferior vena cava to induce DVT in WT and IFN-γ-deficient (Ifng-/-) mice. In WT mice, intrathrombotic IFN-γ levels were elevated progressively as the postligation interval was extended. Thrombus size was substantially smaller at 10 and 14 days in Ifng-/- mice than in WT mice. Intrathrombotic collagen content was remarkably reduced at more than 10 days after the ligation in Ifng-/- mice compared with WT mice. The expression and activity of MMP-9, but not MMP-2, was higher at the late phase in Ifng-/- mice than in WT mice. Moreover, intrathrombotic recanalization was increased in Ifng-/- mice, with enhanced Vegf gene expression, compared with that in WT mice. Activation of the IFN-γ/Stat1 signal pathway suppressed PMA-induced Mmp9 and Vegf gene expression in peritoneal macrophages. Furthermore, administration of anti-IFN-γ mAbs accelerated thrombus resolution in WT mice. Collectively, these findings indicate that IFN-γ can have detrimental roles in thrombus resolution and may be a good molecular target for the acceleration of thrombus resolution in individuals with DVT.
Collapse
Affiliation(s)
- Mizuho Nosaka
- Department of Forensic Medicine, and
Department of Immunology, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, Japan.
Division of Molecular Bioregulation, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | - Yuko Ishida
- Department of Forensic Medicine, and
Department of Immunology, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, Japan.
Division of Molecular Bioregulation, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | - Akihiko Kimura
- Department of Forensic Medicine, and
Department of Immunology, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, Japan.
Division of Molecular Bioregulation, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | - Yumi Kuninaka
- Department of Forensic Medicine, and
Department of Immunology, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, Japan.
Division of Molecular Bioregulation, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | - Masanori Inui
- Department of Forensic Medicine, and
Department of Immunology, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, Japan.
Division of Molecular Bioregulation, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | - Naofumi Mukaida
- Department of Forensic Medicine, and
Department of Immunology, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, Japan.
Division of Molecular Bioregulation, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | - Toshikazu Kondo
- Department of Forensic Medicine, and
Department of Immunology, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, Japan.
Division of Molecular Bioregulation, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| |
Collapse
|
46
|
Upregulation of hypoxia-inducible factor 1 alpha in local vein wall is associated with enhanced venous thrombus resolution. Thromb Res 2011; 128:346-51. [PMID: 21621825 PMCID: PMC3189511 DOI: 10.1016/j.thromres.2011.05.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2011] [Revised: 05/02/2011] [Accepted: 05/03/2011] [Indexed: 01/18/2023]
Abstract
Introduction Venous thrombus resolution may be regulated by an angiogenic process that involves the surrounding vein wall. The aims of this study were to determine whether: (i) thrombosis stimulates activation of the angiogenic transcription factor, hypoxia-inducible factor (HIF) 1α, and downstream expression of growth factors in vein wall; and (ii) upregulation of HIF1α in vein wall leads to increased growth factor expression and enhanced thrombus resolution. Materials and methods HIF1α, vascular endothelial growth factor (VEGF), and placental growth factor (PLGF) were quantified in mouse inferior vena cava (IVC) at days 1, 3, 7, and 14 after thrombus formation (n = 10-13 per group). An additional group of thrombosed mice were treated with the prolyl-hydroxylase domain (PHD) inhibitor, L-mimosine (L-mim) or vehicle control. HIF1α, VEGF, and PLGF in IVC were measured at days 1 and 7; and vein recanalisation and thrombus resolution were measured at days 7 and 10 (n = 6-7 per group). Results HIF1α was expressed in thrombosed IVC and its levels remained relatively constant throughout natural resolution. The levels of VEGF in thrombosed IVC were elevated at days 1 (P < 0.0001) and 3 (P < 0.05); and PLGF at days 1 (P < 0.0001), 3 (P < 0.0001), and 7 (P < 0.0001). Treatment with L-mim led to: increased HIF1α (P < 0.05), VEGF (P < 0.005), and PLGF (P < 0.001) levels in the IVC; decreased thrombus size (P < 0.01); and increased vein recanalisation (P < 0.001). Conclusions HIF1α levels in vein wall are not affected by thrombosis and it appears that the angiogenic drive in the vein surrounding resolving thrombus is regulated independently of HIF1α. Stimulating HIF1α levels in the vein wall leads to an increased angiogenic drive and promotes vein recanalisation and thrombus resolution.
Collapse
|
47
|
Hong L, Li S, Han Y, Du J, Zhang H, Li J, Zhao Q, Wu K, Fan D. Angiogenesis-related molecular targets in esophageal cancer. Expert Opin Investig Drugs 2011; 20:637-44. [DOI: 10.1517/13543784.2011.571203] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
48
|
Saha P, Humphries J, Modarai B, Mattock K, Waltham M, Evans CE, Ahmad A, Patel AS, Premaratne S, Lyons OTA, Smith A. Leukocytes and the natural history of deep vein thrombosis: current concepts and future directions. Arterioscler Thromb Vasc Biol 2011; 31:506-12. [PMID: 21325673 PMCID: PMC3079895 DOI: 10.1161/atvbaha.110.213405] [Citation(s) in RCA: 142] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2010] [Accepted: 12/21/2010] [Indexed: 12/24/2022]
Abstract
Observational studies have shown that inflammatory cells accumulate within the thrombus and surrounding vein wall during the natural history of venous thrombosis. More recent studies have begun to unravel the mechanisms that regulate this interaction and have confirmed that thrombosis and inflammation are intimately linked. This review outlines our current knowledge of the complex relationship between inflammatory cell activity and venous thrombosis and highlights new areas of research in this field. A better understanding of this relationship could lead to the development of novel therapeutic targets that inhibit thrombus formation or promote its resolution.
Collapse
Affiliation(s)
- Prakash Saha
- Kings College London, British Heart Foundation Centre of Research Excellence and National Institute for Health Research Biomedical Research Centre at Kings Health Partners, Academic Department of Surgery, St Thomas' Hospital, London, United Kingdom
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Evans CE, Humphries J, Mattock K, Waltham M, Wadoodi A, Saha P, Modarai B, Maxwell PJ, Smith A. Hypoxia and Upregulation of Hypoxia-Inducible Factor 1α Stimulate Venous Thrombus Recanalization. Arterioscler Thromb Vasc Biol 2010; 30:2443-51. [DOI: 10.1161/atvbaha.110.215038] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Affiliation(s)
- Colin Edward Evans
- From Kings College London (C.E.E., J.H., K.M., M.W., A.W., P.S., B.M., and A.S.), BHF Centre of Research Excellence & NIHR Biomedical Research Centre at Kings Health Partners, Academic Department of Surgery, London, United Kingdom; and Centre for Cell Signalling and Molecular Genetics (P.H.M.), Rayne Institute, University College London, United Kingdom
| | - Julia Humphries
- From Kings College London (C.E.E., J.H., K.M., M.W., A.W., P.S., B.M., and A.S.), BHF Centre of Research Excellence & NIHR Biomedical Research Centre at Kings Health Partners, Academic Department of Surgery, London, United Kingdom; and Centre for Cell Signalling and Molecular Genetics (P.H.M.), Rayne Institute, University College London, United Kingdom
| | - Katherine Mattock
- From Kings College London (C.E.E., J.H., K.M., M.W., A.W., P.S., B.M., and A.S.), BHF Centre of Research Excellence & NIHR Biomedical Research Centre at Kings Health Partners, Academic Department of Surgery, London, United Kingdom; and Centre for Cell Signalling and Molecular Genetics (P.H.M.), Rayne Institute, University College London, United Kingdom
| | - Matthew Waltham
- From Kings College London (C.E.E., J.H., K.M., M.W., A.W., P.S., B.M., and A.S.), BHF Centre of Research Excellence & NIHR Biomedical Research Centre at Kings Health Partners, Academic Department of Surgery, London, United Kingdom; and Centre for Cell Signalling and Molecular Genetics (P.H.M.), Rayne Institute, University College London, United Kingdom
| | - Ashar Wadoodi
- From Kings College London (C.E.E., J.H., K.M., M.W., A.W., P.S., B.M., and A.S.), BHF Centre of Research Excellence & NIHR Biomedical Research Centre at Kings Health Partners, Academic Department of Surgery, London, United Kingdom; and Centre for Cell Signalling and Molecular Genetics (P.H.M.), Rayne Institute, University College London, United Kingdom
| | - Prakash Saha
- From Kings College London (C.E.E., J.H., K.M., M.W., A.W., P.S., B.M., and A.S.), BHF Centre of Research Excellence & NIHR Biomedical Research Centre at Kings Health Partners, Academic Department of Surgery, London, United Kingdom; and Centre for Cell Signalling and Molecular Genetics (P.H.M.), Rayne Institute, University College London, United Kingdom
| | - Bijan Modarai
- From Kings College London (C.E.E., J.H., K.M., M.W., A.W., P.S., B.M., and A.S.), BHF Centre of Research Excellence & NIHR Biomedical Research Centre at Kings Health Partners, Academic Department of Surgery, London, United Kingdom; and Centre for Cell Signalling and Molecular Genetics (P.H.M.), Rayne Institute, University College London, United Kingdom
| | - Patrick J. Maxwell
- From Kings College London (C.E.E., J.H., K.M., M.W., A.W., P.S., B.M., and A.S.), BHF Centre of Research Excellence & NIHR Biomedical Research Centre at Kings Health Partners, Academic Department of Surgery, London, United Kingdom; and Centre for Cell Signalling and Molecular Genetics (P.H.M.), Rayne Institute, University College London, United Kingdom
| | - Alberto Smith
- From Kings College London (C.E.E., J.H., K.M., M.W., A.W., P.S., B.M., and A.S.), BHF Centre of Research Excellence & NIHR Biomedical Research Centre at Kings Health Partners, Academic Department of Surgery, London, United Kingdom; and Centre for Cell Signalling and Molecular Genetics (P.H.M.), Rayne Institute, University College London, United Kingdom
| |
Collapse
|
50
|
Humphries J, Gossage JA, Modarai B, Burnand KG, Sisson TH, Murdoch C, Smith A. Monocyte urokinase-type plasminogen activator up-regulation reduces thrombus size in a model of venous thrombosis. J Vasc Surg 2009; 50:1127-34. [PMID: 19703758 PMCID: PMC2778796 DOI: 10.1016/j.jvs.2009.06.047] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2009] [Revised: 06/08/2009] [Accepted: 06/21/2009] [Indexed: 11/25/2022]
Abstract
Background Our previous studies showed that the direct injection of an adenovirus construct expressing urokinase-type plasminogen activator (uPA) into experimental venous thrombi significantly reduces thrombus weight. The systemic use of adenovirus vectors is limited by inherent hepatic tropism and inflammatory response. As macrophages are recruited into venous thrombi, it is reasonable to speculate that these cells could be used to target the adenovirus uPA (ad-uPA) gene construct to the thrombus. The aims of this study were to determine whether macrophages transduced with ad-uPA have increased fibrinolytic activity and whether systemic injection of transduced cells could be used to target uPA expression to the thrombus and reduce its size. Methods The effect of up-regulating uPA was examined in an immortalized macrophage cell line (MM6) and macrophages differentiated from human blood monocyte-derived macrophages (HBMMs). Cells were infected with ad-uPA or blank control virus (ad-blank). Fibrinolytic mediator expression, cell viability, and cytokine expression were measured by activity assays and enzyme-linked immunosorbent assays. Monocyte migration was measured using a modified Boyden chamber assay. A model of venous thrombosis was developed and characterized in mice with severe combined immunodeficiency (SCID). This model was used to study whether systemically administered macrophages over-expressing uPA reduced thrombus size. Uptake of HBMMs into the thrombus induced in these mice was confirmed by a combination of PKH2-labeled cell tracking and colocalization with human leukocyte antigen (HLA) by immunohistology. Results Compared with ad-blank, treated HBMMs transduction with ad-uPA increased uPA production by >1000-fold (P = .003), uPA activity by 150-fold (P = .0001), and soluble uPA receptor (uPAR) by almost twofold (P = .043). Expression of plasminogen activator inhibitor (PAI-1) and PAI-2 was decreased by about twofold (P = .011) and threefold (P = .005), respectively. Up-regulation of uPA had no effect on cell viability or inflammatory cytokine production compared with ad-blank or untreated cells. Ad-uPA transduction increased the migration rate of HBMMs (about 20%, P = .03) and MM6 cells (>twofold, P = .005) compared with ad-blank treated controls. Human macrophage recruitment into the mouse thrombus was confirmed by the colocalization of HLA with the PKH2-marked cells. Systemic injection of uPA-up-regulated HBMMs reduced thrombus weight by approximately 20% compared with ad-blank (P = .038) or sham-treated controls (P = .0028). Conclusion Transduction of HBBM with ad-uPA increases their fibrinolytic activity. Systemic administration of uPA up-regulated HBBMs reduced thrombus size in an experimental model of venous thrombosis. Alternative methods of delivering fibrinolytic agents are worth exploring. The use of thrombolysis in the treatment of acute iliofemoral deep vein thrombosis is not suitable for all patients. Our previous studies have shown that direct injection of an adenovirus construct expressing urokinase plasminogen activator (uPA) into experimental venous thrombi significantly reduced thrombus weight. The systemic use of adenovirus vectors is, however, limited by both their inherent hepatic tropism, which precludes targeted delivery to disease sites, and by the associated host inflammatory response. As macrophages are recruited into venous thrombi, these cells could be used to target uPA gene constructs to the thrombus after systemic administration.
Collapse
Affiliation(s)
- Julia Humphries
- King's College London British Heart Foundation Centre, Academic Department of Surgery, Cardiovascular Division, National Institute for Health Research Biomedical Research Centre at Guy's and St Thomas' National Health Service Foundation Trust, London, United Kingdom
| | | | | | | | | | | | | |
Collapse
|