1
|
Zhan X, Zhong CM, Tang H, Xiao H, Guo Y, Zhang C, Qu C, Wang X, Huang C. microRNA-18a-5p promotes vascular smooth muscle cell phenotypic switch by targeting Notch2 as therapeutic targets in vein grafts restenosis. Eur J Pharmacol 2024; 985:177097. [PMID: 39522684 DOI: 10.1016/j.ejphar.2024.177097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 10/31/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024]
Abstract
Vascular smooth muscle cells (VSMCs) phenotype switching plays a crucial role in vein graft restenosis following coronary artery bypass grafting (CABG) surgery. To discover novel clinically relevant therapeutic targets for vein graft restenosis after CABG, we therefore investigated whether miRNA-18a-5p mediated phenotype switching plays a critical role in the development of vein graft restenosis. We studied miRNA-18a-5p expression in plasma samples of patients with or without vein graft restenosis at 1, 3 and 5 years after coronary artery bypass graft surgery, and in normal vs. atherosclerotic human femoral artery samples, to prove its role in VSMC phenotype switching. We found that the expression of miRNA-18a-5p significantly increased in vein grafts restenosis rat model after bypass surgery at 7, 14, 28 days and human blood specimens with vein grafts failure after grafting surgery. Through gain- and loss-of-function approaches, we determined that miRNA-18a-5p affects VSMC proliferation, migration, differentiation, and contractility. Notch2 was found to be a direct target of miRNA-18a-5p, which is critical for VSMC phenotype switching. Finally, miRNA-18a-5p knockdown used miRNA sponge via AAV6 locally delivery in vivo, miRNA-18a-5p sponge gene transfer therapy reduced the neointimal area, neointimal thickness, and intimal/media area ratio in vein grafts compared with the controls and improved vein graft hemodynamics. miRNA-18a-5p is a critical modulator of VSMC phenotypic switch during development of vein graft restenosis by downregulating Notch2, therefore targeting miRNA-18a-5p may be a helpful strategy for the treatment of vein grafts restenosis or failure after CABG surgery.
Collapse
MESH Headings
- MicroRNAs/genetics
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/cytology
- Animals
- Humans
- Receptor, Notch2/genetics
- Receptor, Notch2/metabolism
- Rats
- Male
- Phenotype
- Graft Occlusion, Vascular/genetics
- Graft Occlusion, Vascular/pathology
- Graft Occlusion, Vascular/metabolism
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Cell Proliferation/genetics
- Rats, Sprague-Dawley
- Coronary Artery Bypass/adverse effects
- Cell Movement/genetics
- Neointima/pathology
- Coronary Restenosis/genetics
- Coronary Restenosis/etiology
- Coronary Restenosis/pathology
- Coronary Restenosis/metabolism
Collapse
Affiliation(s)
- Xu Zhan
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Chang-Ming Zhong
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Hao Tang
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Hansong Xiao
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Yongzheng Guo
- Division of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Cheng Zhang
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China; Centre for Clinical Pharmacology, William Harvey Research Institute, Queen Mary University of London, London, EC1M 6BQ, United Kingdom
| | - Can Qu
- Division of Pharmacology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Xiaowen Wang
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| | - Chun Huang
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
2
|
Bradley NA, Roxburgh CSD, McMillan DC, Guthrie GJK. A systematic review of the neutrophil to lymphocyte and platelet to lymphocyte ratios in patients with lower extremity arterial disease. VASA 2024; 53:155-171. [PMID: 38563057 DOI: 10.1024/0301-1526/a001117] [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: 04/04/2024]
Abstract
Lower extremity arterial disease (LEAD) is caused by atherosclerotic plaque in the arterial supply to the lower limbs. The neutrophil to lymphocyte and platelet to lymphocyte ratios (NLR, PLR) are established markers of systemic inflammation which are related to inferior outcomes in multiple clinical conditions, though remain poorly described in patients with LEAD. This review was carried out in accordance with PRISMA guidelines. The MEDLINE database was interrogated for relevant studies. Primary outcome was the prognostic effect of NLR and PLR on clinical outcomes following treatment, and secondary outcomes were the prognostic effect of NLR and PLR on disease severity and technical success following revascularisation. There were 34 studies included in the final review reporting outcomes on a total of 19870 patients. NLR was investigated in 21 studies, PLR was investigated in two studies, and both NLR & PLR were investigated in 11 studies. Relating to increased levels of systemic inflammation, 20 studies (100%) reported inferior clinical outcomes, 13 (92.9%) studies reported increased disease severity, and seven (87.5%) studies reported inferior technical results from revascularisation. The studies included in this review support the role of elevated NLR and PLR as key components influencing the clinical outcomes, severity, and success of treatment in patients with LEAD. The use of these easily accessible, cost effective and routinely available markers is supported by the present review.
Collapse
|
3
|
Emmert MY, Bonatti J, Caliskan E, Gaudino M, Grabenwöger M, Grapow MT, Heinisch PP, Kieser-Prieur T, Kim KB, Kiss A, Mouriquhe F, Mach M, Margariti A, Pepper J, Perrault LP, Podesser BK, Puskas J, Taggart DP, Yadava OP, Winkler B. Consensus statement-graft treatment in cardiovascular bypass graft surgery. Front Cardiovasc Med 2024; 11:1285685. [PMID: 38476377 PMCID: PMC10927966 DOI: 10.3389/fcvm.2024.1285685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 01/15/2024] [Indexed: 03/14/2024] Open
Abstract
Coronary artery bypass grafting (CABG) is and continues to be the preferred revascularization strategy in patients with multivessel disease. Graft selection has been shown to influence the outcomes following CABG. During the last almost 60 years saphenous vein grafts (SVG) together with the internal mammary artery have become the standard of care for patients undergoing CABG surgery. While there is little doubt about the benefits, the patency rates are constantly under debate. Despite its acknowledged limitations in terms of long-term patency due to intimal hyperplasia, the saphenous vein is still the most often used graft. Although reendothelialization occurs early postoperatively, the process of intimal hyperplasia remains irreversible. This is due in part to the persistence of high shear forces, the chronic localized inflammatory response, and the partial dysfunctionality of the regenerated endothelium. "No-Touch" harvesting techniques, specific storage solutions, pressure controlled graft flushing and external stenting are important and established methods aiming to overcome the process of intimal hyperplasia at different time levels. Still despite the known evidence these methods are not standard everywhere. The use of arterial grafts is another strategy to address the inferior SVG patency rates and to perform CABG with total arterial revascularization. Composite grafting, pharmacological agents as well as latest minimal invasive techniques aim in the same direction. To give guide and set standards all graft related topics for CABG are presented in this expert opinion document on graft treatment.
Collapse
Affiliation(s)
- Maximilian Y. Emmert
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charite (DHZC), Berlin, Germany
- Institute for Regenerative Medicine (IREM), University of Zurich, Zurich, Switzerland
| | - Johannes Bonatti
- Department of Cardiothoracic Surgery, UPMC Heart and Vascular Institute, University of Pittsburgh, Pittsburgh, PA, United States
| | - Etem Caliskan
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charite (DHZC), Berlin, Germany
| | - Mario Gaudino
- Department of Cardiothoracic Surgery, Weill Cornell Medicine, New York, NY, United States
| | - Martin Grabenwöger
- Sigmund Freud Private University, Vienna, Austria
- Department of Cardiovascular Surgery KFL, Vienna Health Network, Vienna, Austria
| | | | - Paul Phillip Heinisch
- German Heart Center Munich, Technical University of Munich, School of Medicine, Munich, Germany
| | - Teresa Kieser-Prieur
- LIBIN Cardiovascular Institute of Alberta, University of Calgary, Calgary, AB, Canada
| | - Ki-Bong Kim
- Department of Thoracic and Cardiovascular Surgery, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Attila Kiss
- Ludwig Boltzmann Institute at the Center for Biomedical Research, Medical University of Vienna, Vienna, Austria
| | | | - Markus Mach
- Department of Cardiac Surgery, Medical University Vienna, Vienna, Austria
| | - Adrianna Margariti
- The Wellcome-Wolfson Institute of Experimental Medicine, Belfast, United Kingdom
| | - John Pepper
- Cardiology and Aortic Centre, Royal Brompton Hospital, Royal Brompton and Harefield NHS Foundation Trust, London, United Kingdom
| | | | - Bruno K. Podesser
- Ludwig Boltzmann Institute at the Center for Biomedical Research, Medical University of Vienna, Vienna, Austria
| | - John Puskas
- Department of Cardiovascular Surgery, Mount Sinai Morningside, New York, NY, United States
| | - David P. Taggart
- Nuffield Dept Surgical Sciences, Oxford University, Oxford, United Kingdom
| | | | - Bernhard Winkler
- Department of Cardiovascular Surgery KFL, Vienna Health Network, Vienna, Austria
- Ludwig Boltzmann Institute at the Center for Biomedical Research, Medical University of Vienna, Vienna, Austria
- Karld Landsteiner Institute for Cardiovascular Research Clinic Floridsdorf, Vienna, Austria
| |
Collapse
|
4
|
Gemelli M, Addonizio M, Geatti V, Gallo M, Dixon LK, Slaughter MS, Gerosa G. Techniques and Technologies to Improve Vein Graft Patency in Coronary Surgery. Med Sci (Basel) 2024; 12:6. [PMID: 38249082 PMCID: PMC10801616 DOI: 10.3390/medsci12010006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 12/22/2023] [Accepted: 01/05/2024] [Indexed: 01/23/2024] Open
Abstract
Vein grafts are the most used conduits in coronary artery bypass grafting (CABG), even though many studies have suggested their lower patency compared to arterial alternatives. We have reviewed the techniques and technologies that have been investigated over the years with the aim of improving the quality of these conduits. We found that preoperative and postoperative optimal medical therapy and no-touch harvesting techniques have the strongest evidence for optimizing vein graft patency. On the other hand, the use of venous external support, endoscopic harvesting, vein preservation solution and anastomosis, and graft configuration need further investigation. We have also analyzed strategies to treat vein graft failure: when feasible, re-doing the CABG and native vessel primary coronary intervention (PCI) are the best options, followed by percutaneous procedures targeting the failed grafts.
Collapse
Affiliation(s)
- Marco Gemelli
- Cardiac Surgery Unit, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padua, 35128 Padova, Italy; (M.A.); (V.G.); (G.G.)
| | - Mariangela Addonizio
- Cardiac Surgery Unit, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padua, 35128 Padova, Italy; (M.A.); (V.G.); (G.G.)
| | - Veronica Geatti
- Cardiac Surgery Unit, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padua, 35128 Padova, Italy; (M.A.); (V.G.); (G.G.)
| | - Michele Gallo
- Department of Cardiothoracic Surgery, University of Louisville, Louisville, KY 40292, USA; (M.G.); (M.S.S.)
| | - Lauren K. Dixon
- Clinical Effectiveness Unit, The Royal College of Surgeons of England, London WC2A 3PE, UK;
| | - Mark S. Slaughter
- Department of Cardiothoracic Surgery, University of Louisville, Louisville, KY 40292, USA; (M.G.); (M.S.S.)
| | - Gino Gerosa
- Cardiac Surgery Unit, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padua, 35128 Padova, Italy; (M.A.); (V.G.); (G.G.)
| |
Collapse
|
5
|
Raval AJ, Parikh JK, Desai MA. A review on the treatment of intimal hyperplasia with perivascular medical devices: role of mechanical factors and drug release kinetics. Expert Rev Med Devices 2023; 20:805-819. [PMID: 37559556 DOI: 10.1080/17434440.2023.2244875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Accepted: 08/02/2023] [Indexed: 08/11/2023]
Abstract
INTRODUCTION Intimal hyperplasia (IH) is a significant factor limiting the success of revascularization surgery for blood flow restoration. IH results from a foreign body response and mechanical disparity that involves complex biochemical reactions resulting in graft failure. The available treatment option utilizes either different pharmacological interventions or mechanical support to the vascular grafts with limited success. AREAS COVERED This review explains the pathophysiology of IH, responsible mechanical and biological factors, and treatment options, emphasizing perivascular devices. They are designed to provide mechanical support and pharmacology actions. The perivascular drug delivery concept has successfully demonstrated efficacy in various animal studies. Accurate projections of drug release mechanisms using mathematical modeling could be used to formulate prolonged drug elution devices. Numerical modeling aspects for the prediction of design outcomes have been given due importance that fulfills the unmet clinical need for better patient care. EXPERT OPINION IH could be effectively prevented by simultaneous mechanical scaffolding and sustained local drug delivery. Future perivascular medical devices could be designed to integrate these essential features. Numerical modeling for device performance prediction should be utilized in the development of next-generation perivascular devices.
Collapse
Affiliation(s)
- Ankur J Raval
- Department of Chemical Engineering, Sardar Vallabhbhai National of Technology, Surat, Gujarat, India
- Research and Development Department, Sahajanand Medical Technologies Ltd, Surat, Gujarat, India
| | - Jigisha K Parikh
- Department of Chemical Engineering, Sardar Vallabhbhai National of Technology, Surat, Gujarat, India
| | - Meghal A Desai
- Department of Chemical Engineering, Sardar Vallabhbhai National of Technology, Surat, Gujarat, India
| |
Collapse
|
6
|
Reolizo LM, Williams H, Wadey K, Frankow A, Li Z, Gaston K, Jayaraman PS, Johnson JL, George SJ. Inhibition of Intimal Thickening By PRH (Proline-Rich Homeodomain) in Mice. Arterioscler Thromb Vasc Biol 2023; 43:456-473. [PMID: 36700427 PMCID: PMC9944393 DOI: 10.1161/atvbaha.122.318367] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 01/05/2023] [Indexed: 01/27/2023]
Abstract
BACKGROUND Late vein graft failure is caused by intimal thickening resulting from endothelial cell (EC) damage and inflammation which promotes vascular smooth muscle cell (VSMC) dedifferentiation, migration, and proliferation. Nonphosphorylatable PRH (proline-rich homeodomain) S163C:S177C offers enhanced stability and sustained antimitotic effect. Therefore, we investigated whether adenovirus-delivered PRH S163C:S177C protein attenuates intimal thickening via VSMC phenotype modification without detrimental effects on ECs. METHODS PRH S163C:S177C was expressed in vitro (human saphenous vein-VSMCs and human saphenous vein-ECs) and in vivo (ligated mouse carotid arteries) by adenoviruses. Proliferation, migration, and apoptosis were quantified and phenotype was assessed using Western blotting for contractile filament proteins and collagen gel contraction. EC inflammation was quantified using VCAM (vascular cell adhesion protein)-1, ICAM (intercellular adhesion molecule)-1, interleukin-6, and monocyte chemotactic factor-1 measurement and monocyte adhesion. Next Generation Sequencing was utilized to identify novel downstream mediators of PRH action and these and intimal thickening were investigated in vivo. RESULTS PRH S163C:S177C inhibited proliferation, migration, and apoptosis and promoted contractile phenotype (enhanced contractile filament proteins and collagen gel contraction) compared with virus control in human saphenous vein-VSMCs. PRH S163C:S177C expression in human saphenous vein-ECs significantly reduced apoptosis, without affecting cell proliferation and migration, while reducing TNF (tumor necrosis factor)-α-induced VCAM-1 and ICAM-1 and monocyte adhesion and suppressing interleukin-6 and monocyte chemotactic factor-1 protein levels. PRH S163C:S177C expression in ligated murine carotid arteries significantly impaired carotid artery ligation-induced neointimal proliferation and thickening without reducing endothelial coverage. Next Generation Sequencing revealed STAT-1 (signal transducer and activator of transcription 1) and HDAC-9 (histone deacetylase 9) as mediators of PRH action and was supported by in vitro and in vivo analyses. CONCLUSIONS We observed PRH S163C:S177C attenuated VSMC proliferation, and migration and enhanced VSMC differentiation at least in part via STAT-1 and HDAC-9 signaling while promoting endothelial repair and anti-inflammatory properties. These findings highlight the potential for PRH S163C:S177C to preserve endothelial function whilst suppressing intimal thickening, and reducing late vein graft failure.
Collapse
Affiliation(s)
- Lien M. Reolizo
- Bristol Heart Institute, University of Bristol, UK (L.M.R., H.W., K.W., A.F., Z.L., J.L.J., S.J.G.)
| | - Helen Williams
- Bristol Heart Institute, University of Bristol, UK (L.M.R., H.W., K.W., A.F., Z.L., J.L.J., S.J.G.)
| | - Kerry Wadey
- Bristol Heart Institute, University of Bristol, UK (L.M.R., H.W., K.W., A.F., Z.L., J.L.J., S.J.G.)
| | - Aleksandra Frankow
- Bristol Heart Institute, University of Bristol, UK (L.M.R., H.W., K.W., A.F., Z.L., J.L.J., S.J.G.)
| | - Ze Li
- Bristol Heart Institute, University of Bristol, UK (L.M.R., H.W., K.W., A.F., Z.L., J.L.J., S.J.G.)
| | - Kevin Gaston
- School of Medicine and Biodiscovery Institute, Faculty of Medicine & Health Sciences, University of Nottingham, UK (K.G., P.-S.J.)
| | - Padma-Sheela Jayaraman
- School of Medicine and Biodiscovery Institute, Faculty of Medicine & Health Sciences, University of Nottingham, UK (K.G., P.-S.J.)
| | - Jason L. Johnson
- Bristol Heart Institute, University of Bristol, UK (L.M.R., H.W., K.W., A.F., Z.L., J.L.J., S.J.G.)
| | - Sarah J. George
- Bristol Heart Institute, University of Bristol, UK (L.M.R., H.W., K.W., A.F., Z.L., J.L.J., S.J.G.)
| |
Collapse
|
7
|
Systemic Delivery of Clopidogrel Inhibits Neointimal Formation in a Mouse Vein Graft Model. J Cardiovasc Pharmacol 2022; 80:832-841. [PMID: 36027583 DOI: 10.1097/fjc.0000000000001361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 07/30/2022] [Indexed: 12/13/2022]
Abstract
ABSTRACT Clopidogrel inhibits platelet aggregation and has beneficial effects on patients undergoing coronary artery bypass grafting surgery, but it is unknown whether clopidogrel inhibits the neointima formation of grafted veins. In this study, we used a murine vein graft model to study the effect of clopidogrel on intima hyperplasia of the vein graft. Vein grafting was performed among C57BL/6J mice, immediately after surgery; 1 mg/kg clopidogrel and vehicle control were used to inject mice peritoneally daily for 2 weeks. As compared with the vehicle, clopidogrel significantly inhibited the neointima formation of vein grafts at 4 weeks after surgeries. The immunohistochemistry study showed that as compared with the vehicle, clopidogrel significantly decreased the rate of proliferating cell nuclear antigen-positive cells in the wall of vein grafts and significantly increased the expression of vascular smooth muscle cell (VSMC) contractile protein markers (α-smooth muscle actin, calponin, and SM22) within the neointima area of vein grafts. Clopidogrel significantly decreased the plasma interleukin 6 (IL-6) level at 1 week after surgery as compared with the vehicle. We isolated VSMCs from mouse aortic arteries. As compared with the vehicle, clopidogrel significantly inhibited thrombin-induced VSMC proliferation and migration, significantly decreased IL-6 mRNA expression and protein secretion, and increased intracellular cyclic adenosine monophosphate generation in a dose-dependent manner. In conclusion, systemic delivery of clopidogrel inhibits neointima formation of the mouse vein graft, the mechanisms of which are associated with its inhibitory effects on VSMC proliferation, migration, and the tendency to synthetic phenotype after vein graft surgery, reducing the expression of IL-6 and increasing the intracellular cyclic adenosine monophosphate level.
Collapse
|
8
|
Obed D, Dastagir N, Liebsch C, Bingoel AS, Strauss S, Vogt PM, Dastagir K. In Vitro Differentiation of Myoblast Cell Lines on Spider Silk Scaffolds in a Rotating Bioreactor for Vascular Tissue Engineering. J Pers Med 2022; 12:jpm12121986. [PMID: 36556206 PMCID: PMC9783533 DOI: 10.3390/jpm12121986] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Revised: 11/28/2022] [Accepted: 11/29/2022] [Indexed: 12/02/2022] Open
Abstract
Functional construction of tissue-engineered vessels as an alternative to autologous vascular grafts has been shown to be feasible, however the proliferation of seeded smooth-muscle cells remains a limiting factor. We employed a rotating bioreactor system to improve myoblast cell differentiation on a spider silk scaffold for tissue-engineered vessel construction. C2C12 myofibroblast cells were seeded on the surface of spider silk scaffold constructs and cultivated in a rotating bioreactor system with a continuous rotation speed (1 rpm). Cell function, cell growth and morphological structure and expression of biomarkers were analyzed using scanning electron microscopy, the LIVE/DEAD® assay, Western blot and quantitative real-time PCR analyses. A dense myofibroblast cell sheet could be developed which resembled native blood vessel muscular tissue in morphological structure and in function. Bioreactor perfusion positively affected cell morphology, and increased cell viability and cell differentiation. The expression of desmin, MYF5 and MEF2D surged as an indication of myoblast differentiation. Cell-seeded scaffolds showed a tear-down at 18 N when strained at a set speed (20 mm min-1). Spider silk scaffolds appear to offer a reliable basis for engineered vascular constructs and rotating bioreactor cultivation may be considered an effective alternative to complex bioreactor setups to improve cell viability and biology.
Collapse
Affiliation(s)
- Doha Obed
- Correspondence: ; Tel.: +49-511-532-8894; Fax: +49-511-532-8864
| | | | | | | | | | | | | |
Collapse
|
9
|
Ladak SS, McQueen LW, Layton GR, Aujla H, Adebayo A, Zakkar M. The Role of Endothelial Cells in the Onset, Development and Modulation of Vein Graft Disease. Cells 2022; 11:3066. [PMID: 36231026 PMCID: PMC9561968 DOI: 10.3390/cells11193066] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 09/22/2022] [Accepted: 09/26/2022] [Indexed: 01/23/2023] Open
Abstract
Endothelial cells comprise the intimal layer of the vasculature, playing a crucial role in facilitating and regulating aspects such nutrient transport, vascular homeostasis, and inflammatory response. Given the importance of these cells in maintaining a healthy haemodynamic environment, dysfunction of the endothelium is central to a host of vascular diseases and is a key predictor of cardiovascular risk. Of note, endothelial dysfunction is believed to be a key driver for vein graft disease-a pathology in which vein grafts utilised in coronary artery bypass graft surgery develop intimal hyperplasia and accelerated atherosclerosis, resulting in poor long-term patency rates. Activation and denudation of the endothelium following surgical trauma and implantation of the graft encourage a host of immune, inflammatory, and cellular differentiation responses that risk driving the graft to failure. This review aims to provide an overview of the current working knowledge regarding the role of endothelial cells in the onset, development, and modulation of vein graft disease, as well as addressing current surgical and medical management approaches which aim to beneficially modulate endothelial function and improve patient outcomes.
Collapse
Affiliation(s)
| | | | | | | | | | - Mustafa Zakkar
- Department of Cardiovascular Sciences, Clinical Science Wing, University of Leicester, Glenfield Hospital, Leicester LE3 9QP, UK
| |
Collapse
|
10
|
Pan RL, Martyniak K, Karimzadeh M, Gelikman DG, DeVries J, Sutter K, Coathup M, Razavi M, Sawh-Martinez R, Kean TJ. Systematic review on the application of 3D-bioprinting technology in orthoregeneration: current achievements and open challenges. J Exp Orthop 2022; 9:95. [PMID: 36121526 PMCID: PMC9485345 DOI: 10.1186/s40634-022-00518-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 08/08/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Joint degeneration and large or complex bone defects are a significant source of morbidity and diminished quality of life worldwide. There is an unmet need for a functional implant with near-native biomechanical properties. The potential for their generation using 3D bioprinting (3DBP)-based tissue engineering methods was assessed. We systematically reviewed the current state of 3DBP in orthoregeneration. METHODS This review was performed using PubMed and Web of Science. Primary research articles reporting 3DBP of cartilage, bone, vasculature, and their osteochondral and vascular bone composites were considered. Full text English articles were analyzed. RESULTS Over 1300 studies were retrieved, after removing duplicates, 1046 studies remained. After inclusion and exclusion criteria were applied, 114 articles were analyzed fully. Bioink material types and combinations were tallied. Cell types and testing methods were also analyzed. Nearly all papers determined the effect of 3DBP on cell survival. Bioink material physical characterization using gelation and rheology, and construct biomechanics were performed. In vitro testing methods assessed biochemistry, markers of extracellular matrix production and/or cell differentiation into respective lineages. In vivo proof-of-concept studies included full-thickness bone and joint defects as well as subcutaneous implantation in rodents followed by histological and µCT analyses to demonstrate implant growth and integration into surrounding native tissues. CONCLUSIONS Despite its relative infancy, 3DBP is making an impact in joint and bone engineering. Several groups have demonstrated preclinical efficacy of mechanically robust constructs which integrate into articular joint defects in small animals. However, notable obstacles remain. Notably, researchers encountered pitfalls in scaling up constructs and establishing implant function and viability in long term animal models. Further, to translate from the laboratory to the clinic, standardized quality control metrics such as construct stiffness and graft integration metrics should be established with investigator consensus. While there is much work to be done, 3DBP implants have great potential to treat degenerative joint diseases and provide benefit to patients globally.
Collapse
Affiliation(s)
- Rachel L Pan
- College of Medicine, University of Central Florida, Orlando, FL, USA
| | - Kari Martyniak
- Biionix Cluster, College of Medicine, University of Central Florida, 6900 Lake Nona Blvd, Orlando, FL, 32827, USA
| | - Makan Karimzadeh
- Biionix Cluster, College of Medicine, University of Central Florida, 6900 Lake Nona Blvd, Orlando, FL, 32827, USA
| | - David G Gelikman
- College of Medicine, University of Central Florida, Orlando, FL, USA
| | - Jonathan DeVries
- College of Medicine, University of Central Florida, Orlando, FL, USA
| | - Kelly Sutter
- College of Medicine, University of Central Florida, Orlando, FL, USA
| | - Melanie Coathup
- Biionix Cluster, College of Medicine, University of Central Florida, 6900 Lake Nona Blvd, Orlando, FL, 32827, USA
| | - Mehdi Razavi
- Biionix Cluster, College of Medicine, University of Central Florida, 6900 Lake Nona Blvd, Orlando, FL, 32827, USA
| | - Rajendra Sawh-Martinez
- College of Medicine, University of Central Florida, Orlando, FL, USA.,Plastic and Reconstructive Surgery, AdventHealth, Orlando, FL, USA
| | - Thomas J Kean
- Biionix Cluster, College of Medicine, University of Central Florida, 6900 Lake Nona Blvd, Orlando, FL, 32827, USA.
| |
Collapse
|
11
|
Talimi R, Rabbani S, Mehryab F, Haeri A. Perivascular application of sirolimus multilayer nanofibrous mat for prevention of vascular stenosis: Preparation, In vitro characterization, and In vivo efficacy evaluation. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
12
|
Jang EH, Kim JH, Ryu JY, Lee J, Kim HH, Youn YN. Time-dependent pathobiological and physiological changes of implanted vein grafts in a canine model. J Cardiovasc Transl Res 2022; 15:1108-1118. [PMID: 35244875 DOI: 10.1007/s12265-022-10226-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 02/23/2022] [Indexed: 12/01/2022]
Abstract
Although autologous vein grafting is essential, the high vein failure rate and specific clinical interventions are not clear, so a potential treatment is critically needed; thus, complex analyses of the relationship between pathobiological and physiological processes in preclinical are essential. The interposition of the femoral vein was performed in a canine model. Maximized expansion and velocity were measured at 8 weeks post-implantation, and a relative decrease was observed at 12 weeks. However, NI formation and NI/Media ratio significantly increased time dependently, and differences between the mechanical properties were observed. Additionally, RhoA-mediated TNF-α induced by rapid structural changes and high shear stress was confirmed. After adaptation to the arterial environment, vascular remodeling occurred by SMC proliferation and differentiation, apoptosis and autophagy were induced through YAP activity without vasodilation and RhoA activity. Our results show that understanding pathobiological processes in which time-dependent physiological changes contribute to vein failure can lead to a potential strategy. The implanted vein graft within the arterial environment undergoes pathobiological processes through RhoA and YAP activity, leading to pathophysiological changes.
Collapse
Affiliation(s)
- Eui Hwa Jang
- Division of Cardiovascular Surgery, Department of Thoracic and Cardiovascular Surgery, Severance Cardiovascular Hospital, Yonsei University College of Medicine, 250 Seongsanro, Seodaemun-gu, Seoul, 03722, South Korea
| | - Jung-Hwan Kim
- Division of Cardiovascular Surgery, Department of Thoracic and Cardiovascular Surgery, Severance Cardiovascular Hospital, Yonsei University College of Medicine, 250 Seongsanro, Seodaemun-gu, Seoul, 03722, South Korea
| | - Ji-Yeon Ryu
- Division of Cardiovascular Surgery, Department of Thoracic and Cardiovascular Surgery, Severance Cardiovascular Hospital, Yonsei University College of Medicine, 250 Seongsanro, Seodaemun-gu, Seoul, 03722, South Korea
| | - Jiyong Lee
- Department of Mechanical Engineering, YONSEI University, Seoul, 03722, South Korea
| | - Hyo-Hyun Kim
- Division of Cardiovascular Surgery, Department of Thoracic and Cardiovascular Surgery, Severance Cardiovascular Hospital, Yonsei University College of Medicine, 250 Seongsanro, Seodaemun-gu, Seoul, 03722, South Korea
| | - Young-Nam Youn
- Division of Cardiovascular Surgery, Department of Thoracic and Cardiovascular Surgery, Severance Cardiovascular Hospital, Yonsei University College of Medicine, 250 Seongsanro, Seodaemun-gu, Seoul, 03722, South Korea.
| |
Collapse
|
13
|
A call to consider tight blood pressure control to maximize peripheral bypass vein graft patency. J Vasc Surg 2022; 75:389-390. [DOI: 10.1016/j.jvs.2021.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 10/03/2021] [Indexed: 11/19/2022]
|
14
|
Yangming-Fan, Jianjun-Ge. Pentoxifylline Prevents Restenosis by Inhibiting Cell Proliferation via p38MAPK Pathway in Rat Vein Graft Model. Cell Transplant 2022; 31:9636897221122999. [PMID: 36066039 PMCID: PMC9459444 DOI: 10.1177/09636897221122999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Coronary artery bypass grafting remains the gold standard in the therapy
of advanced-stage patients. But the vein grafts are prone to
restenosis or failure. Pentoxifylline (PTX) is a methylxanthine
derivative with a function of inhibiting cell proliferation. We thus
applied PTX locally to the vein grafts to study its effect on the
inhibition of graft restenosis using a rat vein graft model.
Morphometric results showed a significant decrease in the thickness of
vein grafts intimal and medial at day 28 after the bypass operation.
Results from Western blot and immunohistochemistry showed that PTX
also significantly reduced the proliferating cell nuclear antigen
(PCNA), alpha-smooth muscle actin (α-SMA) expression, and
phosphorylation of p38 in vein grafts. These results firstly
discovered the positive role of PTX in preventing the vein grafts
restenosis and the mechanism may be inhibition of vascular smooth
muscle cells (VSMCs) proliferation via the p38MAPK pathway.
Collapse
Affiliation(s)
- Yangming-Fan
- Department of Cardiac Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Jianjun-Ge
- Department of Cardiac Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| |
Collapse
|
15
|
Abstract
Tissue engineering is one of the most promising scientific breakthroughs of the late 20th century. Its objective is to produce in vitro tissues or organs to repair and replace damaged ones using various techniques, biomaterials, and cells. Tissue engineering emerged to substitute the use of native autologous tissues, whose quantities are sometimes insufficient to correct the most severe pathologies. Indeed, the patient’s health status, regulations, or fibrotic scars at the site of the initial biopsy limit their availability, especially to treat recurrence. This new technology relies on the use of biomaterials to create scaffolds on which the patient’s cells can be seeded. This review focuses on the reconstruction, by tissue engineering, of two types of tissue with tubular structures: vascular and urological grafts. The emphasis is on self-assembly methods which allow the production of tissue/organ substitute without the use of exogenous material, with the patient’s cells producing their own scaffold. These continuously improved techniques, which allow rapid graft integration without immune rejection in the treatment of severely burned patients, give hope that similar results will be observed in the vascular and urological fields.
Collapse
|
16
|
Remes A, Basha D, Frey N, Wagner A, Müller O. Gene transfer to the vascular system: Novel translational perspectives for vascular diseases. Biochem Pharmacol 2020; 182:114265. [DOI: 10.1016/j.bcp.2020.114265] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 10/01/2020] [Accepted: 10/01/2020] [Indexed: 01/04/2023]
|
17
|
Loesch A, Dashwood MR. Saphenous Vein Vasa Vasorum as a Potential Target for Perivascular Fat-Derived Factors. Braz J Cardiovasc Surg 2020; 35:964-969. [PMID: 33306322 PMCID: PMC7731844 DOI: 10.21470/1678-9741-2020-0031] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Perivascular adipose tissue (PVAT) is a source of factors affecting vasomotor tone with the potential to play a role in the performance of saphenous vein (SV) bypass grafts. As these factors have been described as having constrictor or relaxant effects, they may be considered either beneficial or detrimental. The close proximity of PVAT to the adventitia provides an environment whereby adipose tissue-derived factors may affect the vasa vasorum, a microvascular network providing the vessel wall with oxygen and nutrients. Since medial ischaemia promotes aspects of graft occlusion the involvement of the PVAT/vasa vasorum axis in vein graft patency should be considered.
Collapse
Affiliation(s)
- Andrzej Loesch
- Centre for Rheumatology and Connective Tissue Diseases, University College London Medical School, Royal Free Campus, London, United Kingdom
| | - Michael Richard Dashwood
- Division of Surgery and Interventional Science, University College London Medical School, Royal Free Campus, London, United Kingdom
| |
Collapse
|
18
|
Ward AO, Angelini GD, Caputo M, Evans PC, Johnson JL, Suleiman MS, Tulloh RM, George SJ, Zakkar M. NF-κB inhibition prevents acute shear stress-induced inflammation in the saphenous vein graft endothelium. Sci Rep 2020; 10:15133. [PMID: 32934266 PMCID: PMC7492228 DOI: 10.1038/s41598-020-71781-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 08/14/2020] [Indexed: 12/17/2022] Open
Abstract
The long saphenous vein (LSV) is commonly used as a conduit in coronary artery bypass grafting. However, long term patency remains limited by the development of vascular inflammation, intimal hyperplasia and accelerated atherosclerosis. The impact of acute exposure of venous endothelial cells (ECs) to acute arterial wall shear stress (WSS) in the arterial circulation, and the subsequent activation of inflammatory pathways, remain poorly defined. Here, we tested the hypothesis that acute exposure of venous ECs to high shear stress is associated with inflammatory responses that are regulated by NF-κB both in-vitro and ex-vivo. Analysis of the LSV endothelium revealed that activation of NF-κB occurred within 30 min after exposure to arterial rates of shear stress. Activation of NF-κB was associated with increased levels of CCL2 production and enhanced binding of monocytes in LSVECs exposed to 6 h acute arterial WSS. Consistent with this, ex vivo exposure of LSVs to acute arterial WSS promoted monocyte interactions with the vessel lumen. Inhibition of the NF-κB pathway prevented acute arterial WSS-induced CCL2 production and reduced monocyte adhesion, both in vitro and in human LSV ex vivo, demonstrating that this pathway is necessary for the induction of the acute arterial WSS-induced pro-inflammatory response. We have identified NF-κB as a critical regulator of acute endothelial inflammation in saphenous vein in response to acute arterial WSS. Localised endothelial-specific inhibition of the NF-κB pathway may be beneficial to prevent vein graft inflammation and consequent failure.
Collapse
Affiliation(s)
- Alexander O Ward
- Bristol Medical School, University of Bristol, Research Floor Level 7, Queens' Building, Bristol Royal Infirmary, Upper Maudlin Street, Bristol, BS2 8HW, UK
| | - Gianni D Angelini
- Bristol Medical School, University of Bristol, Research Floor Level 7, Queens' Building, Bristol Royal Infirmary, Upper Maudlin Street, Bristol, BS2 8HW, UK
| | - Massimo Caputo
- Bristol Medical School, University of Bristol, Research Floor Level 7, Queens' Building, Bristol Royal Infirmary, Upper Maudlin Street, Bristol, BS2 8HW, UK
| | - Paul C Evans
- Department of Infection, Immunity and Cardiovascular Diseases, University of Sheffield, Sheffield, S10 2TN, UK
| | - Jason L Johnson
- Bristol Medical School, University of Bristol, Research Floor Level 7, Queens' Building, Bristol Royal Infirmary, Upper Maudlin Street, Bristol, BS2 8HW, UK
| | - M Saadeh Suleiman
- Bristol Medical School, University of Bristol, Research Floor Level 7, Queens' Building, Bristol Royal Infirmary, Upper Maudlin Street, Bristol, BS2 8HW, UK
| | - Robert M Tulloh
- Bristol Medical School, University of Bristol, Research Floor Level 7, Queens' Building, Bristol Royal Infirmary, Upper Maudlin Street, Bristol, BS2 8HW, UK
| | - Sarah J George
- Bristol Medical School, University of Bristol, Research Floor Level 7, Queens' Building, Bristol Royal Infirmary, Upper Maudlin Street, Bristol, BS2 8HW, UK
| | - Mustafa Zakkar
- Bristol Medical School, University of Bristol, Research Floor Level 7, Queens' Building, Bristol Royal Infirmary, Upper Maudlin Street, Bristol, BS2 8HW, UK.
| |
Collapse
|
19
|
Guida G, Ward AO, Bruno VD, George SJ, Caputo M, Angelini GD, Zakkar M. Saphenous vein graft disease, pathophysiology, prevention, and treatment. A review of the literature. J Card Surg 2020; 35:1314-1321. [PMID: 32353909 DOI: 10.1111/jocs.14542] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
BACKGROUND The saphenous vein remains the most frequently used conduit for coronary artery bypass grafting, despite reported unsatisfactory long-term patency rates. Understanding the pathophysiology of vein graft failure and attempting to improve its longevity has been a significant area of research for more than three decades. This article aims to review the current understanding of the pathophysiology and potential new intervention strategies. METHODS A search of three databases: MEDLINE, Web of Science, and Cochrane Library, was undertaken for the terms "pathophysiology," "prevention," and "treatment" plus the term "vein graft failure." RESULTS Saphenous graft failure is commonly the consequence of four different pathophysiological mechanisms, early acute thrombosis, vascular inflammation, intimal hyperplasia, and late accelerated atherosclerosis. Different methods have been proposed to inhibit or attenuate these pathological processes including modified surgical technique, topical pretreatment, external graft support, and postoperative pharmacological interventions. Once graft failure occurs, the available treatments are either surgical reintervention, angioplasty, or conservative medical management reserved for patients not eligible for either procedure. CONCLUSION Despite the extensive amount of research performed, the pathophysiology of saphenous vein graft is still not completely understood. Surgical and pharmacological interventions have improved early patency and different strategies for prevention seem to offer some hope in improving long-term patency.
Collapse
Affiliation(s)
- Gustavo Guida
- Faculty of Health Sciences, Bristol Heart Institute, Bristol Royal Infirmary, University of Bristol, Bristol, UK
| | - Alex O Ward
- Faculty of Health Sciences, Bristol Heart Institute, Bristol Royal Infirmary, University of Bristol, Bristol, UK
| | - Vito D Bruno
- Faculty of Health Sciences, Bristol Heart Institute, Bristol Royal Infirmary, University of Bristol, Bristol, UK
| | - Sarah J George
- Faculty of Health Sciences, Bristol Heart Institute, Bristol Royal Infirmary, University of Bristol, Bristol, UK
| | - Massimo Caputo
- Faculty of Health Sciences, Bristol Heart Institute, Bristol Royal Infirmary, University of Bristol, Bristol, UK
| | - Gianni D Angelini
- Faculty of Health Sciences, Bristol Heart Institute, Bristol Royal Infirmary, University of Bristol, Bristol, UK
| | - Mustafa Zakkar
- Faculty of Health Sciences, Bristol Heart Institute, Bristol Royal Infirmary, University of Bristol, Bristol, UK.,Department of Cardiovascular Sciences, Clinical Sciences Wing, University of Leicester, Glenfield Hospital, Leicester, England
| |
Collapse
|
20
|
Adventitial Collagen Crosslink Reduces Intimal Hyperplasia in a Rabbit Arteriovenous Graft Model. J Surg Res 2019; 246:550-559. [PMID: 31668608 DOI: 10.1016/j.jss.2019.09.047] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 08/19/2019] [Accepted: 09/19/2019] [Indexed: 11/22/2022]
Abstract
BACKGROUND Intimal hyperplasia (IH) is the initial lesion of vein graft failure after coronary artery bypass grafting. The weak venous wall is likely one of the primary reasons for IH after exposure to the arterial environment. We investigate whether adventitial collagen cross-link by glutaraldehyde (GA) reinforces the venous wall and then reduces IH. MATERIALS AND METHODS Adventitial collagen cross-link by 0.3% GA was performed on the rabbit jugular veins. The degree of cross-link was accessed by tensile test. The jugular vein with or without cross-link was implanted into the carotid artery of rabbit. Vein dilatation at the immediate anastomosis and pathological remodeling of vein graft after 4 wk was assessed. RESULTS Tensile test indicated that the mechanical property of 3-min cross-linked veins more closely resembled that of the carotid artery. In rabbit arteriovenous graft models, 3-min adventitial collagen cross-link limited overdistension (diameter: 3.24 mm versus 4.65 mm, P < 0.01) at the immediate anastomosis and reduced IH (intima thickness: 78.83 μm versus 140.19 μm, P < 0.01) of vein grafts 4 wk after implantation in the cross-link group as compared with the graft group (without cross-link). Compared with the cross-link group, the expression of proliferating cell nuclear antigen and vascular cell adhesion molecule-1 increased significantly at both the mRNA and protein levels within the graft group (P < 0.01), but the expression of smooth muscle-22α decreased significantly (P < 0.01). CONCLUSIONS Adventitial collagen cross-link by GA increased the vessel stiffness and remarkably reduced IH in a rabbit arteriovenous graft model.
Collapse
|
21
|
Dedicator of cytokinesis 2 silencing therapy inhibits neointima formation and improves blood flow in rat vein grafts. J Mol Cell Cardiol 2019; 128:134-144. [DOI: 10.1016/j.yjmcc.2019.01.030] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2018] [Revised: 01/02/2019] [Accepted: 01/31/2019] [Indexed: 01/01/2023]
|
22
|
Cao BJ, Zhu L, Wang XW, Zou RJ, Lu ZQ. MicroRNA-365 promotes the contractile phenotype of venous smooth muscle cells and inhibits neointimal formation in rat vein grafts. IUBMB Life 2019; 71:908-916. [PMID: 30746857 DOI: 10.1002/iub.2022] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 01/22/2019] [Accepted: 01/25/2019] [Indexed: 12/26/2022]
Abstract
The high rate of autologous vein graft failure caused by neointimal hyperplasia remains an unresolved issue in the field of cardiovascular surgery; therefore, it is important to explore new methods for protecting against neointimal hyperplasia. MicroRNA-365 has been reported to inhibit the proliferation of vascular smooth muscle cells (SMCs). This study aimed to test whether adenovirus-mediated miR-365 was able to attenuate neointimal formation in rat vein grafts. We found that miR-365 expression was substantially reduced in vein grafts following engraftment. In vitro, overexpression of miR-365 promoted smooth muscle-specific gene expression and inhibited venous SMC proliferation and migration. Consistent with this, overexpression of miR-365 in a rat vein graft model significantly reduced grafting-induced neointimal formation and effectively improved the hemodynamics of the vein grafts. Mechanistically, we identified that cyclin D1 as a potential downstream target of miR-365 in vein grafts. Specially, to increase the efficiency of miR-365 gene transfection, a 30% poloxamer F-127 gel containing 0.25% trypsin was mixed with adenovirus and spread around the vein grafts to increase the adenovirus contact time and penetration. We showed that adenovirus-mediated miR-365 attenuated venous SMC proliferation and migration in vitro and effectively inhibited neointimal formation in rat vein grafts. Restoring expression of miR-365 is a potential therapeutic approach for the treatment of vein graft failure. © 2019 IUBMB Life, 2019.
Collapse
Affiliation(s)
- Bo-Jun Cao
- Department of Cardiothoracic Surgery, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai, 200233, China
| | - Lei Zhu
- Department of Oncological Surgery, Anqing Hospital of Anhui Medical University, Anhui, 246000, China
| | - Xiao-Wen Wang
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Rong-Jiang Zou
- Department of Cardiovascular Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200001, China
| | - Zhi-Qian Lu
- Department of Cardiothoracic Surgery, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai, 200233, China
| |
Collapse
|
23
|
Kovacic JC, Dimmeler S, Harvey RP, Finkel T, Aikawa E, Krenning G, Baker AH. Endothelial to Mesenchymal Transition in Cardiovascular Disease: JACC State-of-the-Art Review. J Am Coll Cardiol 2019; 73:190-209. [PMID: 30654892 PMCID: PMC6865825 DOI: 10.1016/j.jacc.2018.09.089] [Citation(s) in RCA: 390] [Impact Index Per Article: 65.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 08/20/2018] [Accepted: 09/06/2018] [Indexed: 12/15/2022]
Abstract
Endothelial to mesenchymal transition (EndMT) is a process whereby an endothelial cell undergoes a series of molecular events that lead to a change in phenotype toward a mesenchymal cell (e.g., myofibroblast, smooth muscle cell). EndMT plays a fundamental role during development, and mounting evidence indicates that EndMT is involved in adult cardiovascular diseases (CVDs), including atherosclerosis, pulmonary hypertension, valvular disease, and fibroelastosis. Therefore, the targeting of EndMT may hold therapeutic promise for treating CVD. However, the field faces a number of challenges, including the lack of a precise functional and molecular definition, a lack of understanding of the causative pathological role of EndMT in CVDs (versus being a "bystander-phenomenon"), and a lack of robust human data corroborating the extent and causality of EndMT in adult CVDs. Here, we review this emerging but exciting field, and propose a framework for its systematic advancement at the molecular and translational levels.
Collapse
Affiliation(s)
- Jason C Kovacic
- The Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, New York.
| | - Stefanie Dimmeler
- Institute for Cardiovascular Regeneration, Goethe University, and German Center of Cardiovascular Research, Frankfurt, Germany
| | - Richard P Harvey
- Developmental and Stem Cell Biology Division, Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia; St. Vincent's Clinical School and School of Biotechnology and Biomolecular Science, University of New South Wales, Kensington, New South Wales, Australia
| | - Toren Finkel
- Aging Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Elena Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, and Center for Excellence in Vascular Biology, Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Guido Krenning
- Laboratory for Cardiovascular Regenerative Medicine, Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Andrew H Baker
- UoE/BHF Center for Cardiovascular Science, Queen's Medical Research Institute, Edinburgh, United Kingdom.
| |
Collapse
|
24
|
Wan S, Cheng ZY. Taking care of the soldiers. J Thorac Dis 2019; 10:S4002-S4005. [PMID: 30631539 DOI: 10.21037/jtd.2018.09.25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Song Wan
- Division of Cardiothoracic Surgery, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
| | - Zhao-Yun Cheng
- Department of Cardiovascular Surgery, Fuwai Central China Cardiovascular Hospital, Zhengzhou 450046, China
| |
Collapse
|
25
|
Cao BJ, Wang XW, Zhu L, Zou RJ, Lu ZQ. MicroRNA-146a sponge therapy suppresses neointimal formation in rat vein grafts. IUBMB Life 2018; 71:125-133. [PMID: 30291803 DOI: 10.1002/iub.1946] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2018] [Revised: 08/14/2018] [Accepted: 08/31/2018] [Indexed: 12/13/2022]
Affiliation(s)
- Bo-Jun Cao
- Department of Cardiothoracic Surgery, Shanghai Sixth People's Hospital; Shanghai Jiao Tong University; Shanghai 200233 China
| | - Xiao-Wen Wang
- Department of Cardiothoracic Surgery; The First Affiliated Hospital of Chongqing Medical University; Chongqing 400016 China
| | - Lei Zhu
- Department of Oncological Surgery; Anqing Hospital of Anhui Medical University; Anhui 246000 China
| | - Rong-Jiang Zou
- Department of Cardiovascular Surgery, Ren Ji Hospital, School of Medicine; Shanghai Jiao Tong University; Shanghai 200001 China
| | - Zhi-Qian Lu
- Department of Cardiothoracic Surgery, Shanghai Sixth People's Hospital; Shanghai Jiao Tong University; Shanghai 200233 China
| |
Collapse
|
26
|
Activation and inflammation of the venous endothelium in vein graft disease. Atherosclerosis 2017; 265:266-274. [PMID: 28865843 DOI: 10.1016/j.atherosclerosis.2017.08.023] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Revised: 07/19/2017] [Accepted: 08/23/2017] [Indexed: 01/13/2023]
Abstract
The long saphenous vein is the most commonly used conduit in coronary artery bypass graft (CABG) surgery when bypassing multiple diseased arteries; however, its use is complicated by the development of vascular inflammation, intimal hyperplasia and accelerated atherosclerosis leading to compromised graft efficacy. Despite refinement of surgical techniques to improve graft patency, late vein graft failure remains a significant problem. Moreover, there is a lack of pharmacological interventions proven to be effective in the treatment of late vein graft failure. A greater understanding of the molecular nature of the disease and the interactions between endothelial and smooth muscle cells as a result of alterations in local haemodynamics may assist with designing future beneficial pharmacological interventions. Venous endothelial cells (ECs) are physiologically adapted to chronic low shear stress; however, once the graft is implanted into the arterial circulation, they become suddenly exposed to acute high levels of shear stress. A small number of in vitro and ex vivo studies have demonstrated that acute high shear stress is associated with the activation of a pro-inflammatory profile in saphenous vein ECs, which may be mediated by mitogen-activated protein kinase (MAPK) and nuclear factor-κB (NF-κB) signalling pathways. The impact of acute changes in shear stress on venous ECs and the role of ECs in the development of intimal hyperplasia remains incomplete and is the subject of this review.
Collapse
|
27
|
Tan J, Yang L, Liu C, Yan Z. MicroRNA-26a targets MAPK6 to inhibit smooth muscle cell proliferation and vein graft neointimal hyperplasia. Sci Rep 2017; 7:46602. [PMID: 28429763 PMCID: PMC5399463 DOI: 10.1038/srep46602] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Accepted: 03/21/2017] [Indexed: 12/21/2022] Open
Abstract
Neointima formation is the major reason for vein graft failure. However, the underlying mechanism is unclear. The aim of this study was to determine the role of miR-26a in the development of neointimal hyperplasia of autogenous vein grafts. Using autologous jugular vein grafts in the rat carotid artery as a model, we found that miR-26a was significantly downregulated in grafted veins as well as proliferating vascular smooth muscle cells (VSMCs) stimulated with platelet-derived growth factor-BB (PDGF-BB). Overexpression of miR-26a reduced the proliferation and migration of VSMCs. Further analysis revealed that the effects of miR-26a in VSMCs were mediated by targeting MAPK6 at the mRNA and protein levels. Luciferase assays showed that miR-26a repressed wild type (WT) MAPK6-3′-UTR-luciferase activity but not mutant MAPK6-3′-UTR-luciferease reporter. MAPK6 deficiency reduced proliferation and migration; in contrast, overexpression of MAPK6 enhanced the proliferation and migration of VSMCs. This study confirmed that neointimal hyperplasia in vein grafts was reduced in vivo by up-regulated miR-26a expression. In conclusion, our results showed that miR-26a is an important regulator of VSMC functions and neointimal hyperplasia, suggesting that miR-26a may be a potential therapeutic target for autologous vein graft diseases.
Collapse
Affiliation(s)
- Juanjuan Tan
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| | - Liguo Yang
- Department of Cardiology, Shanghai Jiao Tong University afliated Sixth People's Hospital South Campus, Shanghai, 201400, P. R. China
| | - Cuicui Liu
- Central laboratory, Shanghai Jiao Tong University afliated Sixth People's Hospital South Campus, Shanghai, 201400, P. R. China
| | - Zhiqiang Yan
- Central laboratory, Shanghai Jiao Tong University afliated Sixth People's Hospital South Campus, Shanghai, 201400, P. R. China
| |
Collapse
|
28
|
Wang Q, Wan L, Liu L, Liu J. Role of the mTOR Signalling Pathway in Experimental Rabbit Vein Grafts. Heart Lung Circ 2016; 25:1124-1132. [DOI: 10.1016/j.hlc.2016.03.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Revised: 01/29/2016] [Accepted: 03/21/2016] [Indexed: 11/29/2022]
|
29
|
Bodewes TCF, Johnson JM, Auster M, Huynh C, Muralidharan S, Contreras M, LoGerfo FW, Pradhan-Nabzdyk L. Intraluminal delivery of thrombospondin-2 small interfering RNA inhibits the vascular response to injury in a rat carotid balloon angioplasty model. FASEB J 2016; 31:109-119. [PMID: 27671229 DOI: 10.1096/fj.201600501r] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 09/16/2016] [Indexed: 01/06/2023]
Abstract
In an effort to inhibit the response to vascular injury that leads to intimal hyperplasia, this study investigated the in vivo efficacy of intraluminal delivery of thrombospondin-2 (TSP-2) small interfering RNA (siRNA). Common carotid artery (CCA) balloon angioplasty injury was performed in rats. Immediately after denudation, CCA was transfected intraluminally (15 min) with one of the following: polyethylenimine (PEI)+TSP-2 siRNA, saline, PEI only, or PEI+control siRNA. CCA was analyzed at 24 h or 21 d by using quantitative real-time PCR and immunohistochemistry. TSP-2 gene and protein expression were significantly up-regulated after endothelial denudation at 24 h and 21 d compared with contralateral untreated, nondenuded CCA. Treatment with PEI+TSP-2 siRNA significantly suppressed TSP-2 gene expression (3.1-fold) at 24 h and TSP-2 protein expression, cell proliferation, and collagen deposition up to 21 d. These changes could be attributed to changes in TGF-β and matrix metalloproteinase-9, the downstream effectors of TSP-2. TSP-2 knockdown induced anti-inflammatory M2 macrophage polarization at 21 d; however, it did not significantly affect intima/media ratios. In summary, these data demonstrate effective siRNA transfection of the injured arterial wall and provide a clinically effective and translationally applicable therapeutic strategy that involves nonviral siRNA delivery to ameliorate the response to vascular injury.-Bodewes, T. C. F., Johnson, J. M., Auster, M., Huynh, C., Muralidharan, S., Contreras, M., LoGerfo, F. W., Pradhan-Nabzdyk, L. Intraluminal delivery of thrombospondin-2 small interfering RNA inhibits the vascular response to injury in a rat carotid balloon angioplasty model.
Collapse
Affiliation(s)
- Thomas C F Bodewes
- Division of Vascular and Endovascular Surgery, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA.,Department of Vascular Surgery, University Medical Center, Utrecht, The Netherlands; and
| | - Joel M Johnson
- Division of Vascular and Endovascular Surgery, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Michael Auster
- Division of Vascular and Endovascular Surgery, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Cindy Huynh
- Division of Vascular and Endovascular Surgery, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA.,Department of Surgery, State University of New York (SUNY) Upstate Medical University, Syracuse, New York, USA
| | - Sriya Muralidharan
- Division of Vascular and Endovascular Surgery, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Mauricio Contreras
- Division of Vascular and Endovascular Surgery, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Frank W LoGerfo
- Division of Vascular and Endovascular Surgery, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Leena Pradhan-Nabzdyk
- Division of Vascular and Endovascular Surgery, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA;
| |
Collapse
|
30
|
de Vries MR, Simons KH, Jukema JW, Braun J, Quax PHA. Vein graft failure: from pathophysiology to clinical outcomes. Nat Rev Cardiol 2016; 13:451-70. [PMID: 27194091 DOI: 10.1038/nrcardio.2016.76] [Citation(s) in RCA: 203] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Occlusive arterial disease is a leading cause of morbidity and mortality worldwide. Aside from balloon angioplasty, bypass graft surgery is the most commonly performed revascularization technique for occlusive arterial disease. Coronary artery bypass graft surgery is performed in patients with left main coronary artery disease and three-vessel coronary disease, whereas peripheral artery bypass graft surgery is used to treat patients with late-stage peripheral artery occlusive disease. The great saphenous veins are commonly used conduits for surgical revascularization; however, they are associated with a high failure rate. Therefore, preservation of vein graft patency is essential for long-term surgical success. With the exception of 'no-touch' techniques and lipid-lowering and antiplatelet (aspirin) therapy, no intervention has hitherto unequivocally proven to be clinically effective in preventing vein graft failure. In this Review, we describe both preclinical and clinical studies evaluating the pathophysiology underlying vein graft failure, and the latest therapeutic options to improve patency for both coronary and peripheral grafts.
Collapse
Affiliation(s)
- Margreet R de Vries
- Department of Surgery, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, Netherlands
| | - Karin H Simons
- Department of Surgery, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, Netherlands
| | - J Wouter Jukema
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, Netherlands.,Department of Cardiology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, Netherlands
| | - Jerry Braun
- Department of Cardiothoracic Surgery, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, Netherlands
| | - Paul H A Quax
- Department of Surgery, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, Netherlands
| |
Collapse
|
31
|
Wilson R, Aung H, Ju X, Paterson C, Sayer R, McIntyre A, Going J, Irvine R, Berry C. Optical coherence tomography: an experimental validation for vascular imaging of saphenous vein bypass grafts. Biomed Phys Eng Express 2016. [DOI: 10.1088/2057-1976/2/2/025002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|
32
|
Wang M, Collins MJ, Foster TR, Bai H, Hashimoto T, Santana JM, Shu C, Dardik A. Eph-B4 mediates vein graft adaptation by regulation of endothelial nitric oxide synthase. J Vasc Surg 2016; 65:179-189. [PMID: 26817610 DOI: 10.1016/j.jvs.2015.11.041] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Accepted: 11/10/2015] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Vein graft adaptation is characterized by loss of expression of the tyrosine kinase receptor Eph-B4, the embryonic determinant of venous identity, without increased expression of its ligand ephrin-B2, the embryonic determinant of arterial identity. Endothelial nitric oxide synthase (eNOS) is an important mediator of vessel remodeling. We hypothesized that the mechanism of action of Eph-B4 during vein graft adaptation might be through regulation of downstream eNOS activity. METHODS Mouse lung endothelial cells were stimulated with ephrin-B2/Fc, without and with preclustering, without and with the eNOS inhibitor Nω-nitro-l-arginine methyl ester hydrochloride or the Eph-B4 inhibitor NVP-BHG712, and assessed by Western blot and immunofluorescence for eNOS and Eph-B4 phosphorylation. Nitric oxide (NO) production was assessed using an NO-specific chemiluminescence analyzer. Cell migration was assessed using a Transwell assay. Human and mouse vein graft specimens were examined for eNOS activity by Western blot, and vessel remodeling was assessed in vein grafts in wild-type or eNOS knockout mice. RESULTS Ephrin-B2/Fc stimulated both Eph-B4 and eNOS phosphorylation in a bimodal temporal distribution (n = 4; P < .05), with preclustered ephrin-B2/Fc causing prolonged peak Eph-B4 and eNOS phosphorylation as well as altered subcellular localization (n = 4; P < .05). Ephrin-B2/Fc increased NO release (n = 3; P < .01) as well as increased endothelial cell migration (n = 6; P < .05) in an eNOS-dependent fashion. Both human and mouse vein grafts showed increased eNOS phosphorylation compared with normal veins (n = 3; P < .05). Vein grafts from eNOS knockout mice showed less dilation and less wall thickening compared with wild-type vein grafts (n = 7; P < .05). CONCLUSIONS eNOS is a mediator of vein graft adaptation to the arterial environment. Eph-B4 stimulates eNOS phosphorylation in vitro and may mediate vein graft adaptation by regulation of eNOS activity in vivo.
Collapse
Affiliation(s)
- Mo Wang
- Department of Vascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, China; Vascular Biology and Therapeutics Program and Department of Surgery, Yale University School of Medicine, New Haven, Conn
| | - Michael J Collins
- Vascular Biology and Therapeutics Program and Department of Surgery, Yale University School of Medicine, New Haven, Conn
| | - Trenton R Foster
- Vascular Biology and Therapeutics Program and Department of Surgery, Yale University School of Medicine, New Haven, Conn
| | - Hualong Bai
- Vascular Biology and Therapeutics Program and Department of Surgery, Yale University School of Medicine, New Haven, Conn
| | - Takuya Hashimoto
- Vascular Biology and Therapeutics Program and Department of Surgery, Yale University School of Medicine, New Haven, Conn
| | - Jeans M Santana
- Vascular Biology and Therapeutics Program and Department of Surgery, Yale University School of Medicine, New Haven, Conn
| | - Chang Shu
- Department of Vascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Alan Dardik
- Vascular Biology and Therapeutics Program and Department of Surgery, Yale University School of Medicine, New Haven, Conn; Department of Surgery, VA Connecticut Healthcare System, West Haven, Conn.
| |
Collapse
|
33
|
Dakin RS, Parker AL, Delles C, Nicklin SA, Baker AH. Efficient transduction of primary vascular cells by the rare adenovirus serotype 49 vector. Hum Gene Ther 2015; 26:312-9. [PMID: 25760682 PMCID: PMC4442572 DOI: 10.1089/hum.2015.019] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Accepted: 03/06/2015] [Indexed: 01/16/2023] Open
Abstract
Neointima formation and vascular remodeling through vascular smooth muscle cell migration and proliferation can limit the long-term success of coronary interventions, for example, in coronary artery bypass grafting (CABG). Ex vivo gene therapy has the potential to reduce unnecessary cell proliferation and limit neointima formation in vascular pathologies. To date, the species C adenovirus serotype 5 has been commonly used for preclinical gene therapy; however, its suitability is potentially limited by relatively poor tropism for vascular cells and high levels of preexisting immunity in the population. To avoid these limitations, novel species of adenovirus are being tested; here we investigate the potential of adenovirus 49 (Ad49) for use in gene therapy. Transduction of primary human vascular cells by a range of adenovirus serotypes was assessed; Ad49 demonstrated highest transduction of both vascular smooth muscle and endothelial cells. Gene transfer with Ad49 in vascular smooth muscle and endothelial cells was possible following short exposure times (<1 hr) and with low MOI, which is clinically relevant. Ex vivo delivery to surplus CABG tissue showed efficient gene transfer with Ad49, consistent with the in vitro findings. Luminal infusion of Ad49GFP into intact CABG samples ex vivo resulted in efficient vessel transduction. In addition, no seroprevalence rates to Ad49 were observed in a Scottish cohort of patients from cardiovascular clinics, thus circumventing issues with preexisting immunity. Our results show that Ad49 has tropism for vascular cells in vitro and ex vivo and demonstrate that Ad49 may be an improved vector for local vascular gene therapy compared with current alternatives.
Collapse
Affiliation(s)
- Rachel S. Dakin
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow G12 8TA, United Kingdom
| | - Alan L. Parker
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow G12 8TA, United Kingdom
| | - Christian Delles
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow G12 8TA, United Kingdom
| | - Stuart A. Nicklin
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow G12 8TA, United Kingdom
| | - Andrew H. Baker
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow G12 8TA, United Kingdom
| |
Collapse
|
34
|
Abstract
Recent advances in our understanding of the pathophysiology of myocardial dysfunction in the setting of congestive heart failure have created a new opportunity in developing nonpharmacological approaches to treatment. Gene therapy has emerged as a powerful tool in targeting the molecular mechanisms of disease by preventing the ventricular remodeling and improving bioenergetics in heart failure. Refinements in vector technology, including the creation of recombinant adeno-associated viruses, have allowed for safe and efficient gene transfer. These advancements have been coupled with evolving delivery methods that include vascular, pericardial, and direct myocardial approaches. One of the most promising targets, SERCA2a, is currently being used in clinical trials. The recent success of the Calcium Upregulation by Percutaneous Administration of Gene Therapy in Cardiac Disease phase 2 trials using adeno-associated virus 1-SERCA2a in improving outcomes highlights the importance of gene therapy as a future tool in treating congestive heart failure.
Collapse
|
35
|
Icli B, Dorbala P, Feinberg MW. An emerging role for the miR-26 family in cardiovascular disease. Trends Cardiovasc Med 2014; 24:241-8. [PMID: 25066487 DOI: 10.1016/j.tcm.2014.06.003] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2014] [Revised: 06/06/2014] [Accepted: 06/06/2014] [Indexed: 12/26/2022]
Abstract
In response to acute myocardial infarction (MI), a complex series of cellular and molecular signaling events orchestrate the myocardial remodeling that ensues weeks to months after injury. Clinical, epidemiological, and pathological studies demonstrate that inadequate or impaired angiogenesis after myocardial injury is often associated with decreased left ventricular (LV) function and clinical outcomes. The microRNA family, miR-26, plays diverse roles in regulating key aspects of cellular growth, development, and activation. Recent evidence supports a central role for the miR-26 family in cardiovascular disease by controlling critical signaling pathways, such as BMP/SMAD1 signaling, and targets relevant to endothelial cell growth, angiogenesis, and LV function post-MI. Emerging studies of the miR-26 family in other cell types including vascular smooth muscle cells, cardiac fibroblasts, and cardiomyocytes suggest that miR-26 may bear important implications for a range of cardiovascular repair mechanisms. This review examines the current knowledge of the miR-26 family's role in key cell types that critically control cardiovascular disease under pathological and physiological stimuli.
Collapse
Affiliation(s)
- Basak Icli
- Department of Medicine, Cardiovascular Division, Brigham and Women׳s Hospital, Harvard Medical School, Boston, MA
| | - Pranav Dorbala
- Department of Medicine, Cardiovascular Division, Brigham and Women׳s Hospital, Harvard Medical School, Boston, MA
| | - Mark W Feinberg
- Department of Medicine, Cardiovascular Division, Brigham and Women׳s Hospital, Harvard Medical School, Boston, MA.
| |
Collapse
|
36
|
Lu DY, Chen EY, Wong DJ, Yamamoto K, Protack CD, Williams WT, Assi R, Hall MR, Sadaghianloo N, Dardik A. Vein graft adaptation and fistula maturation in the arterial environment. J Surg Res 2014; 188:162-73. [PMID: 24582063 DOI: 10.1016/j.jss.2014.01.042] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2013] [Revised: 12/25/2013] [Accepted: 01/24/2014] [Indexed: 12/21/2022]
Abstract
Veins are exposed to the arterial environment during two common surgical procedures, creation of vein grafts and arteriovenous fistulae (AVF). In both cases, veins adapt to the arterial environment that is characterized by different hemodynamic conditions and increased oxygen tension compared with the venous environment. Successful venous adaptation to the arterial environment is critical for long-term success of the vein graft or AVF and, in both cases, is generally characterized by venous dilation and wall thickening. However, AVF are exposed to a high flow, high shear stress, low-pressure arterial environment and adapt mainly via outward dilation with less intimal thickening. Vein grafts are exposed to a moderate flow, moderate shear stress, high-pressure arterial environment and adapt mainly via increased wall thickening with less outward dilation. We review the data that describe these differences, as well as the underlying molecular mechanisms that mediate these processes. Despite extensive research, there are few differences in the molecular pathways that regulate cell proliferation and migration or matrix synthesis, secretion, or degradation currently identified between vein graft adaptation and AVF maturation that account for the different types of venous adaptation to arterial environments.
Collapse
Affiliation(s)
- Daniel Y Lu
- Yale University Vascular Biology and Therapeutics Program, New Haven, Connecticut; Department of Surgery, Yale University School of Medicine, New Haven, Connecticut
| | - Elizabeth Y Chen
- Yale University Vascular Biology and Therapeutics Program, New Haven, Connecticut; Department of Surgery, Yale University School of Medicine, New Haven, Connecticut
| | - Daniel J Wong
- Yale University Vascular Biology and Therapeutics Program, New Haven, Connecticut; Department of Surgery, Yale University School of Medicine, New Haven, Connecticut
| | - Kota Yamamoto
- Yale University Vascular Biology and Therapeutics Program, New Haven, Connecticut; Department of Surgery, Yale University School of Medicine, New Haven, Connecticut; VA Connecticut Healthcare System, West Haven, Connecticut
| | - Clinton D Protack
- Yale University Vascular Biology and Therapeutics Program, New Haven, Connecticut; Department of Surgery, Yale University School of Medicine, New Haven, Connecticut
| | - Willis T Williams
- Yale University Vascular Biology and Therapeutics Program, New Haven, Connecticut; Department of Surgery, Yale University School of Medicine, New Haven, Connecticut
| | - Roland Assi
- Yale University Vascular Biology and Therapeutics Program, New Haven, Connecticut; Department of Surgery, Yale University School of Medicine, New Haven, Connecticut
| | - Michael R Hall
- Yale University Vascular Biology and Therapeutics Program, New Haven, Connecticut; Department of Surgery, Yale University School of Medicine, New Haven, Connecticut
| | - Nirvana Sadaghianloo
- Yale University Vascular Biology and Therapeutics Program, New Haven, Connecticut; Department of Surgery, Yale University School of Medicine, New Haven, Connecticut; Department of Vascular Surgery, University Hospital of Nice, Nice, France
| | - Alan Dardik
- Yale University Vascular Biology and Therapeutics Program, New Haven, Connecticut; Department of Surgery, Yale University School of Medicine, New Haven, Connecticut; VA Connecticut Healthcare System, West Haven, Connecticut.
| |
Collapse
|
37
|
Hu J, Zhao JJ. Bone morphogenic protein-4: a potential novel target for preventing vein graft failure in coronary revascularization. Med Hypotheses 2013; 81:1025-8. [PMID: 24119764 DOI: 10.1016/j.mehy.2013.09.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2012] [Revised: 08/15/2013] [Accepted: 09/18/2013] [Indexed: 02/05/2023]
Abstract
Coronary artery bypass surgery is an effective and durable therapy in both acute coronary syndrome and chronic coronary stenotic disease refractory to pharmacological treatment. Despite rapid development in operation-specific technologies and secondary prevention measures, the benefits of surgical revascularization are largely limited by inadequate patency of one of the most commonly used conduits, namely the autologous saphenous vein. However, apart from antiplatelet and lipid-lowering drugs, no other pharmacologic agent has hitherto proven clinically effective in preventing short- and long-term vein graft failure. Aiming at a large number of known biomolecules, multiple promising strategies failed to translate their beneficial effects observed in animal models into the clinical settings. Bone morphogenic protein-4 (BMP4), originally identified as a mediator in bone formation, has been recently demonstrated to participate in the process of arterial post-injury remodeling. Existing evidence has demonstrated that BMP4 is closely involved in the pathogenesis of thrombus formation, neointimal hyperplasia and superimposed atherosclerosis, all of which significantly contribute to arterial stenotic lesions. Although the post-injury responses inherent to arterial and venous vessel are unique, they share common elements and present with similar physiologic characteristics and clinical sequelae. Therefore, with regard to the multifaceted effects of BMP4 in regulating arterial wall remodeling, we hypothesize that BMP4 may play an important role in mediating the pathological responses of the venous wall to the arterial circulation. If our hypothesis is demonstrated correct, BMP4 inhibition could presumably serve as a novel strategy for preventing vein graft failure in coronary revascularization.
Collapse
Affiliation(s)
- Jia Hu
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu, People's Republic of China.
| | | |
Collapse
|
38
|
Meng QH, Irvine S, Tagalakis AD, McAnulty RJ, McEwan JR, Hart SL. Inhibition of neointimal hyperplasia in a rabbit vein graft model following non-viral transfection with human iNOS cDNA. Gene Ther 2013; 20:979-86. [PMID: 23636244 PMCID: PMC3795475 DOI: 10.1038/gt.2013.20] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2012] [Revised: 03/04/2013] [Accepted: 03/22/2013] [Indexed: 11/13/2022]
Abstract
Vein graft failure caused by neointimal hyperplasia (IH) after coronary artery bypass grafting with saphenous veins is a major clinical problem. The lack of safe and efficient vectors for vascular gene transfer has significantly hindered progress in this field. We have developed a Receptor-Targeted Nanocomplex (RTN) vector system for this purpose and assessed its therapeutic efficacy in a rabbit vein graft model of bypass grafting. Adventitial delivery of β-Galactosidase showed widespread transfection throughout the vein wall on day 7, estimated at about 10% of cells in the adventitia and media. Vein grafts were then transfected with a plasmid encoding inducible nitric oxide synthase (iNOS) and engrafted into the carotid artery. Fluorescent immunohistochemistry analysis of samples from rabbits killed at 7 days after surgery showed that mostly endothelial cells and macrophages were transfected. Morphometric analysis of vein graft samples from the 28-day groups showed approximately a 50% reduction of neointimal thickness and 64% reduction of neointimal area in the iNOS-treated group compared with the surgery control groups. This study demonstrates efficacy of iNOS gene delivery by the RTN formulation in reducing IH in the rabbit model of vein graft disease.
Collapse
Affiliation(s)
- Q-H Meng
- Molecular Immunology Unit, UCL Institute of Child Health, University College London, London, UK
| | - S Irvine
- Molecular Immunology Unit, UCL Institute of Child Health, University College London, London, UK
| | - A D Tagalakis
- Molecular Immunology Unit, UCL Institute of Child Health, University College London, London, UK
| | - R J McAnulty
- Centre for Inflammation and Tissue Repair, UCL Respiratory, University College London, London, UK
| | - J R McEwan
- Centre for Cardiovascular Medicine and Biology, University College London, London, UK
| | - S L Hart
- Molecular Immunology Unit, UCL Institute of Child Health, University College London, London, UK
| |
Collapse
|
39
|
Abstract
Saphenous vein remains a widely used conduit in coronary surgery. However, the long-term success of surgical myocardial revascularization is largely limited by the development of neointimal hyperplasia and superimposed atherosclerosis in vein grafts. Although strategies for preventing vein graft failure have been constantly explored, few therapeutic interventions to date have shown sustained benefits in the clinical setting. The application of external support has emerged as a promising strategy for modulating the overall biomechanical responses in venous wall. Nonetheless, clinical translation of this intervention has been formerly challenged, primarily due to several technique limitations. The purpose of the current review is to summarize the possible mechanisms involved in the external support strategy for preventing vein graft failure. Furthermore, several previously tested biomaterials and delivery techniques are also highlighted.
Collapse
Affiliation(s)
- Jia Hu
- Division of Cardiothoracic Surgery, Department of Surgery, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, People's Republic of China
| | | |
Collapse
|
40
|
Southerland KW, Frazier SB, Bowles DE, Milano CA, Kontos CD. Gene therapy for the prevention of vein graft disease. Transl Res 2013; 161:321-38. [PMID: 23274305 PMCID: PMC3602161 DOI: 10.1016/j.trsl.2012.12.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2012] [Revised: 12/04/2012] [Accepted: 12/04/2012] [Indexed: 11/20/2022]
Abstract
Ischemic cardiovascular disease remains the leading cause of death worldwide. Despite advances in the medical management of atherosclerosis over the past several decades, many patients require arterial revascularization to reduce mortality and alleviate ischemic symptoms. Technological advancements have led to dramatic increases in the use of percutaneous and endovascular approaches, yet surgical revascularization (bypass surgery) with autologous vein grafts remains a mainstay of therapy for both coronary and peripheral artery disease. Although bypass surgery is highly efficacious in the short term, long-term outcomes are limited by relatively high failure rates as a result of intimal hyperplasia, which is a common feature of vein graft disease. The supply of native veins is limited, and many individuals require multiple grafts and repeat procedures. The need to prevent vein graft failure has led to great interest in gene therapy approaches to this problem. Bypass grafting presents an ideal opportunity for gene therapy, as surgically harvested vein grafts can be treated with gene delivery vectors ex vivo, thereby maximizing gene delivery while minimizing the potential for systemic toxicity and targeting the pathogenesis of vein graft disease at its onset. Here we will review the pathogenesis of vein graft disease and discuss vector delivery strategies and potential molecular targets for its prevention. We will summarize the preclinical and clinical literature on gene therapy in vein grafting and discuss additional considerations for future therapies to prevent vein graft disease.
Collapse
Affiliation(s)
- Kevin W Southerland
- Department of Surgery, Division of Surgical Sciences, Duke University Medical Center, Durham, North Carolina, USA
| | | | | | | | | |
Collapse
|
41
|
McDonald RA, White KM, Wu J, Cooley BC, Robertson KE, Halliday CA, McClure JD, Francis S, Lu R, Kennedy S, George SJ, Wan S, van Rooij E, Baker AH. miRNA-21 is dysregulated in response to vein grafting in multiple models and genetic ablation in mice attenuates neointima formation. Eur Heart J 2013; 34:1636-43. [PMID: 23530023 PMCID: PMC3675389 DOI: 10.1093/eurheartj/eht105] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Aims The long-term failure of autologous saphenous vein bypass grafts due to neointimal thickening is a major clinical burden. Identifying novel strategies to prevent neointimal thickening is important. Thus, this study aimed to identify microRNAs (miRNAs) that are dysregulated during neointimal formation and determine their pathophysiological relevance following miRNA manipulation. Methods and results We undertook a microarray approach to identify dysregulated miRNAs following engraftment in an interpositional porcine graft model. These profiling experiments identified a number of miRNAs which were dysregulated following engraftment. miR-21 levels were substantially elevated following engraftment and these results were confirmed by quantitative real-time PCR in mouse, pig, and human models of vein graft neointimal formation. Genetic ablation of miR-21 in mice or grafted veins dramatically reduced neointimal formation in a mouse model of vein grafting. Furthermore, pharmacological knockdown of miR-21 in human veins resulted in target gene de-repression and a significant reduction in neointimal formation. Conclusion This is the first report demonstrating that miR-21 plays a pathological role in vein graft failure. Furthermore, we also provided evidence that knockdown of miR-21 has therapeutic potential for the prevention of pathological vein graft remodelling.
Collapse
Affiliation(s)
- Robert A McDonald
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow G12 8TA, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Gene therapy for cardiovascular disease: perspectives and potential. Vascul Pharmacol 2012; 58:174-81. [PMID: 23142171 DOI: 10.1016/j.vph.2012.10.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2012] [Revised: 10/24/2012] [Accepted: 10/29/2012] [Indexed: 01/01/2023]
Abstract
Cardiovascular disease is the most frequent cause of mortality in the western world, accounting for over 800,000 premature deaths per year in the EU alone. Cardiovascular disease is the second most common application for gene therapy clinical trials, which most frequently employ adenovirus serotype 5 (Ad5)-based vectors as delivery vehicles. Although interactions of Ad5 vectors with circulating proteins and cells can limit their efficacy after systemic administration, local gene delivery strategies show great potential in the cardiovascular setting, notably in the context of vascular delivery. Here we review the pathogenesis of bypass graft failure and in-stent restenosis, identifying potential therapeutic targets and discussing recent advances in the field of adenovirus biology and retargeting that, in concert, will potentially translate in coming years to more effective gene therapies for cardiovascular applications.
Collapse
|
43
|
Robertson KE, McDonald RA, Oldroyd KG, Nicklin SA, Baker AH. Prevention of coronary in-stent restenosis and vein graft failure: does vascular gene therapy have a role? Pharmacol Ther 2012; 136:23-34. [PMID: 22796519 DOI: 10.1016/j.pharmthera.2012.07.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2012] [Accepted: 06/28/2012] [Indexed: 12/19/2022]
Abstract
Coronary artery bypass grafting (CABG) and percutaneous coronary intervention (PCI), including stent insertion, are established therapies in both acute coronary syndromes (ACS) and symptomatic chronic coronary artery disease refractory to pharmacological therapy. These continually advancing treatments remain limited by failure of conduit grafts in CABG and by restenosis or thrombosis of stented vessel segments in PCI caused by neointimal hyperplasia, impaired endothelialisation and accelerated atherosclerosis. While pharmacological and technological advancements have improved patient outcomes following both procedures, when grafts or stents fail these result in significant health burdens. In this review we discuss the pathophysiology of vein graft disease and in-stent restenosis, gene therapy vector development and design, and translation from pre-clinical animal models through human clinical trials. We identify the key issues that are currently preventing vascular gene therapy from interfacing with clinical use and introduce the areas of research attempting to overcome these.
Collapse
Affiliation(s)
- Keith E Robertson
- British Heart Foundation Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | | | | | | | | |
Collapse
|
44
|
Andrews RK. FoxO function PAR excellence... Thromb Haemost 2012; 108:11. [PMID: 22628011 DOI: 10.1160/th12-05-0305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2012] [Accepted: 05/11/2012] [Indexed: 11/05/2022]
Affiliation(s)
- Robert K Andrews
- Robert K. Andrews, Australian Centre for Blood Diseases, Monash University, Commercial Road, Melbourne, Victoria 3004, Australia.
| |
Collapse
|