1
|
Tierney JW, Francisco RP, Yu F, Ma J, Cheung-Flynn J, Keech MC, D'Arcy R, Shah VM, Kittel AR, Chang DJ, McCune JT, Bezold MG, Aligwekwe AN, Cook RS, Beckman JA, Brophy CM, Duvall CL. Intravascular delivery of an MK2 inhibitory peptide to prevent restenosis after angioplasty. Biomaterials 2025; 313:122767. [PMID: 39216327 DOI: 10.1016/j.biomaterials.2024.122767] [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: 03/07/2024] [Revised: 08/14/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024]
Abstract
Peripheral artery disease is commonly treated with balloon angioplasty, a procedure involving minimally invasive, transluminal insertion of a catheter to the site of stenosis, where a balloon is inflated to open the blockage, restoring blood flow. However, peripheral angioplasty has a high rate of restenosis, limiting long-term patency. Therefore, angioplasty is sometimes paired with delivery of cytotoxic drugs like paclitaxel to reduce neointimal tissue formation. We pursue intravascular drug delivery strategies that target the underlying cause of restenosis - intimal hyperplasia resulting from stress-induced vascular smooth muscle cell switching from the healthy contractile into a pathological synthetic phenotype. We have established MAPKAP kinase 2 (MK2) as a driver of this phenotype switch and seek to establish convective and contact transfer (coated balloon) methods for MK2 inhibitory peptide delivery to sites of angioplasty. Using a flow loop bioreactor, we showed MK2 inhibition in ex vivo arteries suppresses smooth muscle cell phenotype switching while preserving vessel contractility. A rat carotid artery balloon injury model demonstrated inhibition of intimal hyperplasia following MK2i coated balloon treatment in vivo. These studies establish both convective and drug coated balloon strategies as promising approaches for intravascular delivery of MK2 inhibitory formulations to improve efficacy of balloon angioplasty.
Collapse
Affiliation(s)
- J William Tierney
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA
| | - R Paolo Francisco
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA
| | - Fang Yu
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA
| | - Jinqi Ma
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA
| | - Joyce Cheung-Flynn
- Division of Vascular Surgery, Department of Surgery, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Megan C Keech
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA
| | - Richard D'Arcy
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA; Chemical Engineering, School of Engineering of Matter, Transport and Energy, Arizona State University, Tempe, AZ, USA
| | - Veeraj M Shah
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA
| | - Anna R Kittel
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA
| | - Devin J Chang
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA
| | - Joshua T McCune
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA
| | - Mariah G Bezold
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA
| | - Adrian N Aligwekwe
- Medical Scientist Training Program, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA; North Carolina State University, Raleigh, NC, 27695, USA
| | - Rebecca S Cook
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA; Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Joshua A Beckman
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Colleen M Brophy
- Division of Vascular Surgery, Department of Surgery, Vanderbilt University Medical Center, Nashville, TN, 37232, USA; Veterans Affairs Medical Center, VA Tennessee Valley Healthcare System, Nashville, TN, 37212, USA
| | - Craig L Duvall
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA.
| |
Collapse
|
2
|
Nazari-Shafti TZ, Thau H, Zacharova E, Beez CM, Exarchos V, Neuber S, Meyborg H, Puhl K, Wittig C, Szulcek R, Neumann K, Giampietro C, Krüger K, Cesarovic N, Falk V, Caliskan E, Rodriguez Cetina Biefer H, Emmert MY. Endothelial damage inhibitor preserves the integrity of venous endothelial cells from patients undergoing coronary bypass surgery. Eur J Cardiothorac Surg 2023; 64:ezad327. [PMID: 37740952 DOI: 10.1093/ejcts/ezad327] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 07/04/2023] [Accepted: 09/22/2023] [Indexed: 09/25/2023] Open
Abstract
OBJECTIVES Despite the success of coronary artery bypass graft (CABG) surgery using autologous saphenous vein grafts (SVGs), nearly 50% of patients experience vein graft disease within 10 years of surgery. One contributing factor to early vein graft disease is endothelial damage during short-term storage of SVGs in inappropriate solutions. Our aim was to evaluate the effects of a novel endothelial damage inhibitor (EDI) on SVGs from patients undergoing elective CABG surgery and on venous endothelial cells (VECs) derived from these SVGs. METHODS SVGs from 11 patients participating in an ongoing clinical registry (NCT02922088) were included in this study, and incubated with both full electrolyte solution (FES) or EDI for 1 h and then examined histologically. In 8 of 11 patients, VECs were isolated from untreated grafts, incubated with both FES and EDI for 2 h under hypothermic stress conditions and then analysed for activation of an inflammatory phenotype, cell damage and cytotoxicity, as well as endothelial integrity and barrier function. RESULTS The EDI was superior to FES in protecting the endothelium in SVGs (74 ± 8% versus 56 ± 8%, P < 0.001). Besides confirming that the EDI prevents apoptosis in SVG-derived VECs, we also showed that the EDI temporarily reduces adherens junctions in VECs while protecting focal adhesions compared to FES. CONCLUSIONS The EDI protects the connectivity and function of the SVG endothelium. Our data suggest that the EDI can preserve focal adhesions in VECs during short-term storage after graft harvesting. This might explain the superiority of the EDI in maintaining most of the endothelium in venous CABG surgery conduits.
Collapse
Affiliation(s)
- Timo Z Nazari-Shafti
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité (DHZC), Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Berlin, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
| | - Henriette Thau
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité (DHZC), Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Berlin, Germany
| | - Ema Zacharova
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité (DHZC), Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Berlin, Germany
- Department of Life Sciences, IMC University of Applied Sciences Krems, Krems an der Donau, Austria
| | - Christien M Beez
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité (DHZC), Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Berlin, Germany
| | - Vasileios Exarchos
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité (DHZC), Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Berlin, Germany
| | - Sebastian Neuber
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité (DHZC), Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Berlin, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
| | - Heike Meyborg
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité (DHZC), Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Berlin, Germany
| | - Kerstin Puhl
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité (DHZC), Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Berlin, Germany
| | - Corey Wittig
- German Centre for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
- Department of Cardiac Anesthesiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité (DHZC), Berlin, Germany
- Laboratory for in vitro modeling systems of pulmonary and thrombotic diseases, Institute of Physiology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Robert Szulcek
- German Centre for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
- Department of Cardiac Anesthesiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité (DHZC), Berlin, Germany
- Laboratory for in vitro modeling systems of pulmonary and thrombotic diseases, Institute of Physiology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Konrad Neumann
- Institute of Biometry and Clinical Epidemiology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Costanza Giampietro
- Experimental Continuum Mechanics, Empa Swiss Federal Laboratories for Materials Science and Technology, Dübendorf, Switzerland
| | - Katrin Krüger
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité (DHZC), Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Berlin, Germany
| | - Nikola Cesarovic
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité (DHZC), Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Volkmar Falk
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité (DHZC), Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
- Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Etem Caliskan
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité (DHZC), Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Hector Rodriguez Cetina Biefer
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité (DHZC), Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Department of Cardiac Surgery, City Hospital of Zurich, Site Triemli, Zurich, Switzerland
| | - Maximilian Y Emmert
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité (DHZC), Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Berlin, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
| |
Collapse
|
3
|
Layton GR, Ladak SS, Abbasciano R, McQueen LW, George SJ, Murphy GJ, Zakkar M. The Role of Preservation Solutions upon Saphenous Vein Endothelial Integrity and Function: Systematic Review and UK Practice Survey. Cells 2023; 12:815. [PMID: 36899951 PMCID: PMC10001248 DOI: 10.3390/cells12050815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 02/19/2023] [Accepted: 02/22/2023] [Indexed: 03/08/2023] Open
Abstract
The long saphenous vein is the most used conduit in cardiac surgery, but its long-term patency is limited by vein graft disease (VGD). Endothelial dysfunction is a key driver of VGD; its aetiology is multi-factorial. However emerging evidence identifies vein conduit harvest technique and preservation fluids as causal in their onset and propagation. This study aims to comprehensively review published data on the relationship between preservation solutions, endothelial cell integrity and function, and VGD in human saphenous veins harvested for CABG. The review was registered with PROSPERO (CRD42022358828). Electronic searches of Cochrane Central Register of Controlled Trials, MEDLINE, and EMBASE databases were undertaken from inception until August 2022. Papers were evaluated in line with registered inclusion and exclusion criteria. Searches identified 13 prospective, controlled studies for inclusion in the analysis. All studies used saline as a control solution. Intervention solutions included heparinised whole blood and saline, DuraGraft, TiProtec, EuroCollins, University of Wisconsin (UoW), buffered, cardioplegic and Pyruvate solutions. Most studies demonstrated that normal saline appears to have negative effects on venous endothelium and the most effective preservation solutions identified in this review were TiProtec and DuraGraft. The most used preservation solutions in the UK are heparinised saline or autologous whole blood. There is substantial heterogeneity both in practice and reporting of trials evaluating vein graft preservation solutions, and the quality of existing evidence is low. There is an unmet need for high quality trials evaluating the potential for these interventions to improve long-term patency in venous bypass grafts.
Collapse
Affiliation(s)
- Georgia R. Layton
- Department of Cardiovascular Sciences, University of Leicester, Leicester LE1 7RH, UK
| | - Shameem S. Ladak
- Department of Cardiovascular Sciences, University of Leicester, Leicester LE1 7RH, UK
| | | | - Liam W. McQueen
- Department of Cardiovascular Sciences, University of Leicester, Leicester LE1 7RH, UK
| | - Sarah J. George
- Department of Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol BS2 1UDD, UK
| | - Gavin J. Murphy
- Department of Cardiovascular Sciences, University of Leicester, Leicester LE1 7RH, UK
| | - Mustafa Zakkar
- Department of Cardiovascular Sciences, University of Leicester, Leicester LE1 7RH, UK
| |
Collapse
|
4
|
DEMİREL S. +4 °C’de Krebs-Henseleit Solüsyonunda Soğuk Depolamanın Sıçan Torasik Aortunun Vazoreaktivitesi Üzerindeki Etkileri. BALIKESIR HEALTH SCIENCES JOURNAL 2022. [DOI: 10.53424/balikesirsbd.1141819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2024]
Abstract
Aim: This study aimed to investigate whether endothelium-dependent and -independent relaxation responses can be preserved intact in the rat thoracic aorta after storage for 3, 6, and 18 hours in Krebs-Henseleit solution at +4 °C.
Methods: Isolated organ bath model and 10-12 weeks old male Wistar rats were used to perform the experiments. To investigate the effect of endothelium-dependent relaxation factors, the cyclooxygenase inhibitor INDO was added to the Krebs-Henseleit solution to inhibit endogenous prostanoid synthesis. Submaximal contraction response was obtained with a single dose of PE and then ACh was administered cumulatively (10-9-10-4 M) to induce endothelium-dependent relaxation responses. Besides, smooth muscle-dependent relaxation responses were obtained by applying SNP cumulatively (10-9-10-5 M) following precontraction induced by PE. The statistical significance level was considered as p0.05). Besides, cumulatively administered ACh did not cause a significant change in endothelium-dependent relaxation responses (p>0.05). Similarly, SNP did not modulate the endothelium-independent relaxation responses in aortic segments after storage for 3, 6, or 18 hours (p>0.05).
Conclusion: In the present study, the first physiological findings have been obtained that the endothelium-dependent and -independent contraction-relaxation responses of rat thoracic aortas can be preserved intact after storage periods of 3, 6, or 18 hours in Krebs-Henseleit solution at +4°C.
Collapse
|
5
|
Pimentel MD, Lobo Filho JG, Lobo Filho HG, de Castro Miguel E, Pinheiro Paiva SK, Silva Matos JI, Mesquita Fernandes MA, Fechine Jamacaru FV. Effect of preservation solution and distension pressure on saphenous vein's endothelium. Interact Cardiovasc Thorac Surg 2022; 35:ivac124. [PMID: 35575424 PMCID: PMC9419693 DOI: 10.1093/icvts/ivac124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 03/29/2022] [Accepted: 04/23/2022] [Indexed: 11/23/2022] Open
Abstract
OBJECTIVES Approaches to improve saphenous vein (SV) patency in coronary artery bypass graft (CABG) surgery remain relevant. This study aimed to evaluate the effects of different preservation solutions and different pressures of intraluminal distention on the endothelium of SV segments in CABG. METHODS Forty-two SV segments obtained from 12 patients undergoing CABG were divided into 7 groups. Group 1 (control) was prepared without preservation or intraluminal distension, while the other 6 groups were preserved in autologous heparinized autologous arterial blood or normal saline (NS), with distention pressures 30, 100 and 300 mmHg. To assess the effects of using these solutions and pressures on the endothelium, the grafts were analysed by scanning electron microscopy, with the measurement of endothelial damage degree. RESULTS Segments in group 1 showed minimal endothelial damage. SV grafts preserved with NS had significantly greater endothelial damage both compared to the control group and compared to groups preserved with autologous arterial blood (P < 0.001). Segments distended with pressures up to 100 mmHg showed less damage when compared to those distended at 300 mmHg, with the ones subjected to higher pressures presenting a maximum degree of damage, with considerable loss and separation of endothelial cells, extensive foci of exposure of the basement membrane and numerous fractures of the intimate layer, without differences regarding the solution used. CONCLUSIONS Preparation of SV using NS and with intraluminal distension pressures above 100 mmHg is factors related to increased damage to the venous endothelium.
Collapse
Affiliation(s)
| | | | | | - Emílio de Castro Miguel
- Department of Metallurgical Engineering and Materials (DEMM) and Analytical Center, Federal University of Ceará, Fortaleza, Brazil
| | - Sergimar Kennedy Pinheiro Paiva
- Department of Metallurgical Engineering and Materials (DEMM) and Analytical Center, Federal University of Ceará, Fortaleza, Brazil
| | | | | | | |
Collapse
|
6
|
Tekin I, Demir M, Özdem S. Effect of different storage solutions on oxidative stress in human saphenous vein grafts. J Cardiothorac Surg 2022; 17:7. [PMID: 35034655 PMCID: PMC8762896 DOI: 10.1186/s13019-022-01752-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 01/06/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Ischemia-reperfusion injury of saphenous vein grafts (SVG) during coronary artery bypass grafting surgery negatively impacts endothelial integrity and functionality and is associated with vein graft failure. The aim of this study was to evaluate the level of oxidative stress in human SVG segments following ischemic storage in three intraoperative graft storage solutions: saline (S), autologous heparinized blood (HB) and DuraGraft (DG). METHODS 3 mm tissue rings derived from surplus SVG segments from 50 patients were stored at room temperature for 30 min in DG, S or HB. Total oxidative status (TOS) and total antioxidant status (TAS) levels were determined from which the oxidative stress index (OSI: TOS/TAS ratio) was calculated. A p-value < 0.017 was considered significant implementing a Bonferroni correction. RESULTS TOS values were significantly lower for DG stored samples in comparison to both S and HB; there was no difference between S and HB (DG: 32.6 ± 1.8, S: 39.6 ± 2.8 and HB: 40.6 ± 2.4 µmol H2O2 eqv.; DG vs. S and DG vs. HB p < 0.0001, S vs. HB p = 0.047). TAS was higher for both DG and HB in comparison to S (DG: 8.9 ± 0.9, S: 6.9 ± 1.0 and HB: 8.6 ± 0.9 mmol Trolox eqv.; DG vs S p < 0.0001, DG vs. HB p = 0.263, S vs. HB p < 0.0001). OSI differed between all groups with the lowest value for DG (DG: 3.7 ± 0.2, S: 5.8 ± 0.4 and HB: 4.7 ± 0.2 µmol H2O2 eqv./mmol Trolox eqv.; all p < 0.0001). CONCLUSIONS Saphenous veins grafts stored in DuraGraft had a lower oxidative level, higher antioxidant level and a lower oxidative stress index in comparison to saphenous vein grafts stored in saline or heparinized blood. ClinicalTrials.gov Identifier NCT02922088.
Collapse
Affiliation(s)
- Ilker Tekin
- Department of Cardiovascular Surgery, Manavgat Government Hospital, Manavgat, Turkey.
- Department of Cardiovascular Surgery, Faculty of Medicine, Bahçeşehir University, İstanbul, Turkey.
| | - Meltem Demir
- Department of Biochemistry, Medicalpark Hospital Complex, Antalya, Turkey
- Faculty of Health Science, Bilim University, Antalya, Turkey
| | - Sebahat Özdem
- Department of Medical Biochemistry, Faculty of Medicine, Akdeniz University, Antalya, Turkey
| |
Collapse
|
7
|
Aschacher T, Baranyi U, Aschacher O, Eichmair E, Messner B, Zimpfer D, Moayedifar R, Laufer G, Emmert MY, Sandner SE. A Novel Endothelial Damage Inhibitor Reduces Oxidative Stress and Improves Cellular Integrity in Radial Artery Grafts for Coronary Artery Bypass. Front Cardiovasc Med 2021; 8:736503. [PMID: 34692789 PMCID: PMC8527012 DOI: 10.3389/fcvm.2021.736503] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 09/07/2021] [Indexed: 11/13/2022] Open
Abstract
The radial artery (RA) is a frequently used conduit in coronary artery bypass grafting (CABG). Endothelial injury incurred during graft harvesting promotes oxidative damage, which leads to graft disease and graft failure. We evaluated the protective effect of DuraGraft®, an endothelial damage inhibitor (EDI), on RA grafts. We further compared the protective effect of the EDI between RA grafts and saphenous vein grafts (SVG). Samples of RA (n = 10) and SVG (n = 13) from 23 patients undergoing CABG were flushed and preserved with either EDI or heparinized Ringer's lactate solution (RL). The effect of EDI vs. RL on endothelial damage was evaluated ex vivo and in vitro using histological analysis, immunofluorescence staining, Western blot, and scanning electron microscopy. EDI-treated RA grafts showed a significant reduction of endothelial and sub-endothelial damage. Lower level of reactive oxygen species (ROS) after EDI treatment was correlated with a reduction of hypoxic damage (eNOS and Caveolin-1) and significant increase of oxidation-reduction potential. Additionally, an increased expression of TGFβ, PDGFα/β, and HO-1 which are indicative for vascular protective function were observed after EDI exposure. EDI treatment preserves functionality and integrity of endothelial and intimal cells. Therefore, EDI may have the potential to reduce the occurrence of graft disease and failure in RA grafts in patients undergoing CABG.
Collapse
Affiliation(s)
- Thomas Aschacher
- Department of Cardio-Vascular Surgery, Clinic Floridsdorf and Karl Landsteiner Institute for Cardio-Vascular Research, Vienna, Austria.,Department of Cardiac Surgery, Medical University Vienna, Vienna, Austria
| | - Ulrike Baranyi
- Department of Cardiac Surgery, Medical University Vienna, Vienna, Austria
| | - Olivia Aschacher
- Department of Plastic, Reconstructive and Plastic Surgery, Medical University Vienna, Vienna, Austria
| | - Eva Eichmair
- Department of Cardiac Surgery, Medical University Vienna, Vienna, Austria
| | - Barbara Messner
- Department of Cardiac Surgery, Medical University Vienna, Vienna, Austria
| | - Daniel Zimpfer
- Department of Cardiac Surgery, Medical University Vienna, Vienna, Austria
| | - Roxana Moayedifar
- Department of Cardiac Surgery, Medical University Vienna, Vienna, Austria
| | - Guenther Laufer
- Department of Cardiac Surgery, Medical University Vienna, Vienna, Austria
| | - Maximilian Y Emmert
- Cardiovascular Surgery, Charite-Universitätsmedizin Berlin, Berlin, Germany.,Department of Cardiothoracic and Vascular Surgery, German Heart Institute Berlin, Berlin, Germany
| | - Sigrid E Sandner
- Department of Cardiac Surgery, Medical University Vienna, Vienna, Austria
| |
Collapse
|
8
|
Storage solutions to improve grafts preservation and longevity in coronary artery bypass grafting surgery: hype or hope? Curr Opin Cardiol 2021; 36:616-622. [PMID: 34397467 DOI: 10.1097/hco.0000000000000875] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW Saphenous vein grafts (SVGs) remain the most-commonly used conduits for coronary artery bypass grafting (CABG).Significant rates of vein graft failure (VGF) remain a limitation of their use as this diminishes the long-term benefits of CABG. The choice of intraoperative SVGs preservation solution is believed to have an impact on graft patency; however, the superiority of one solution over the others remains in question. RECENT FINDINGS In the present review, we describe the pathophysiological mechanisms underlying the different phases of VGF. We also reviewed the most recent literature comparing and evaluating the efficacy of various storage solutions. These include heparinized saline, autologous heparinized blood, buffered solutions, and crystalloid cardioplegia. SUMMARY It is clear that the composition of the SVGs storage solution has an impact on vessel wall structure and function. There is a lack of translational and clinical research on the topic; thus, conclusions cannot be drawn regarding the superiority of one solution over the others in terms of VGF. Future research needs to be conducted to address this gap in the literature in order to make meaningful evidence-based recommendations on intraoperative graft storage.
Collapse
|
9
|
Spadaccio C, Antoniades C, Nenna A, Chung C, Will R, Chello M, Gaudino MFL. Preventing treatment failures in coronary artery disease: what can we learn from the biology of in-stent restenosis, vein graft failure, and internal thoracic arteries? Cardiovasc Res 2020; 116:505-519. [PMID: 31397850 DOI: 10.1093/cvr/cvz214] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 06/01/2019] [Accepted: 08/08/2019] [Indexed: 12/18/2022] Open
Abstract
Coronary artery disease (CAD) remains one of the most important causes of morbidity and mortality worldwide, and the availability of percutaneous or surgical revascularization procedures significantly improves survival. However, both strategies are daunted by complications which limit long-term effectiveness. In-stent restenosis (ISR) is a major drawback for intracoronary stenting, while graft failure is the limiting factor for coronary artery bypass graft surgery (CABG), especially using veins. Conversely, internal thoracic artery (ITA) is known to maintain long-term patency in CABG. Understanding the biology and pathophysiology of ISR and vein graft failure (VGF) and mechanisms behind ITA resistance to failure is crucial to combat these complications in CAD treatment. This review intends to provide an overview of the biological mechanisms underlying stent and VGF and of the potential therapeutic strategy to prevent these complications. Interestingly, despite being different modalities of revascularization, mechanisms of failure of stent and saphenous vein grafts are very similar from the biological standpoint.
Collapse
Affiliation(s)
- Cristiano Spadaccio
- Department of Cardiac Surgery, Golden Jubilee National Hospital, Agamemnon St, Clydebank, G81 4DY Glasgow, UK
| | | | - Antonio Nenna
- Department of Cardiovascular Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Calvin Chung
- Department of Cardiac Surgery, Golden Jubilee National Hospital, Agamemnon St, Clydebank, G81 4DY Glasgow, UK
| | - Ricardo Will
- Department of Cardiac Surgery, Golden Jubilee National Hospital, Agamemnon St, Clydebank, G81 4DY Glasgow, UK
| | - Massimo Chello
- Department of Cardiovascular Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Mario F L Gaudino
- Department of Cardiothoracic Surgery, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
10
|
Yim TW, Perling D, Polcz M, Komalavilas P, Brophy C, Cheung-Flynn J. A cell permeant phosphopeptide mimetic of Niban inhibits p38 MAPK and restores endothelial function after injury. FASEB J 2020; 34:9180-9191. [PMID: 32396246 PMCID: PMC7383822 DOI: 10.1096/fj.201902745r] [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] [Received: 11/01/2019] [Revised: 04/17/2020] [Accepted: 04/21/2020] [Indexed: 12/11/2022]
Abstract
Vascular injury leads to membrane disruption, ATP release, and endothelial dysfunction. Increases in the phosphorylation of p38 mitogen‐activated protein kinase (p38 MAPK) and decreases in the phosphorylation of Niban, a protein implicated in ER stress and apoptosis, are associated with vascular injury. A cell permeant phosphopeptide mimetic of Niban (NiPp) was generated. The effects of NiPp in restoring endothelial function were determined ex vivo using intact rat aortic tissue (RA) after pharmacological activation of p38 MAPK and also in multiple clinically relevant injury models. Anisomycin (Aniso) increased p38 MAPK phosphorylation and reduced endothelial‐dependent relaxation in RA. Treatment with NiPp prevented Ansio‐induced reduction in endothelial function and increases in p38 MAPK phosphorylation. NiPp treatment also restored endothelial function after stretch injury (subfailure stretch), treatment with acidic Normal Saline (NS), and P2X7R activation with 2′(3′)‐O‐(4‐Benzoylbenzoyl)adenosine 5′‐triphosphate (BzATP). Aged, diseased, human saphenous vein (HSV) remnants obtained from patients undergoing coronary bypass surgical procedures have impaired endothelial function. Treatment of these HSV segments with NiPp improved endothelial‐dependent relaxation. Kinome screening experiments indicated that NiPp inhibits p38 MAPK. These data demonstrate that p38 MAPK and Niban signaling have a role in endothelial function, particularly in response to injury. Niban may represent an endogenous regulator of p38 MAPK activation. The NiPp peptide may serve as an experimental tool to further elucidate p38 MAPK regulation and as a potential therapeutic for endothelial dysfunction.
Collapse
Affiliation(s)
- Tsz Wing Yim
- Department of Surgery, Vanderbilt University, Nashville, TN, USA
| | - Daniel Perling
- Department of Surgery, Vanderbilt University, Nashville, TN, USA
| | - Monica Polcz
- Department of Surgery, Vanderbilt University, Nashville, TN, USA
| | - Padmini Komalavilas
- Department of Surgery, Vanderbilt University, Nashville, TN, USA.,VA Tennessee Valley Healthcare System, Nashville, TN, USA
| | - Colleen Brophy
- Department of Surgery, Vanderbilt University, Nashville, TN, USA.,VA Tennessee Valley Healthcare System, Nashville, TN, USA
| | | |
Collapse
|
11
|
Fouquet O, Blossier JD, Dang Van S, Robert P, Barbelivien A, Pinaud F, Binuani P, Eid M, Henrion D, Baufreton C, Loufrani L. Do storage solutions protect endothelial function of arterialized vein graft in an experimental rat model? J Cardiothorac Surg 2020; 15:34. [PMID: 32041642 PMCID: PMC7011455 DOI: 10.1186/s13019-020-1077-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 01/30/2020] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND This study aims to compare the effects of storage solutions commonly used in coronary artery bypass grafting on the vascular reactivity in vein graft interposed in arterial position in syngeneic rats. METHODS Twenty-seven male Lewis rats were sacrified to sample a vein graft implanted 6 weeks ago into abdominal aorta position. The vein grafts were inferior venae cavae initially pretreated with heparinized saline solution (HS) or autologous heparinized blood (AHB) or our referent solution, GALA. The endothelial functionality, the in situ Reactive Oxygen Species (ROS) levels and the histological characteristics were conducted from segments of arterialized vein graft. RESULTS At 6 weeks, graft thrombosis occurred respectively in 22% of AHB group, 62.5% in the HS group and 82.5% in the GALA group. In each group, significative intimal hyperplasia was observed. After 6 weeks, an endothelium-remodeling layer associated with an increase of wall thickness was observed in each group. Endothelium-dependent tone was reduced in the vein graft regardless of the group. No difference was observed concerning the ROS in vein graft between the different groups. In distal aortic sections, ROS levels were increased in HS and GALA groups. CONCLUSIONS Storage solutions used in this experimental model of vein graft implanted in arterial position cause graft injury and a complete disappearance of vascular reactivity. GALA solution did not reduce intimal risk hyperplasia when the vein graft was exposed to arterial flow in a rat model.
Collapse
Affiliation(s)
- Olivier Fouquet
- Department of Cardiac Surgery, University Hospital of Angers, France, 4 rue Larrey, 49933 CHU, Angers Cedex 9, France.
- Institute MITOVASC CNRS UMR 6015, INSERM 1083, Angers, France.
| | - Jean-David Blossier
- Institute MITOVASC CNRS UMR 6015, INSERM 1083, Angers, France
- Department of Cardiac Surgery, CHU Dupuytren, Limoges, France
| | - Simon Dang Van
- Department of Cardiac Surgery, University Hospital of Angers, France, 4 rue Larrey, 49933 CHU, Angers Cedex 9, France
- Institute MITOVASC CNRS UMR 6015, INSERM 1083, Angers, France
| | - Pauline Robert
- Institute MITOVASC CNRS UMR 6015, INSERM 1083, Angers, France
| | | | - Frédéric Pinaud
- Department of Cardiac Surgery, University Hospital of Angers, France, 4 rue Larrey, 49933 CHU, Angers Cedex 9, France
- Institute MITOVASC CNRS UMR 6015, INSERM 1083, Angers, France
| | - Patrice Binuani
- Department of Cardiac Surgery, University Hospital of Angers, France, 4 rue Larrey, 49933 CHU, Angers Cedex 9, France
| | - Maroua Eid
- Department of Cardiac Surgery, University Hospital of Angers, France, 4 rue Larrey, 49933 CHU, Angers Cedex 9, France
| | - Daniel Henrion
- Institute MITOVASC CNRS UMR 6015, INSERM 1083, Angers, France
- University Hospital of Angers, Angers, France
| | - Christophe Baufreton
- Department of Cardiac Surgery, University Hospital of Angers, France, 4 rue Larrey, 49933 CHU, Angers Cedex 9, France
- Institute MITOVASC CNRS UMR 6015, INSERM 1083, Angers, France
| | - Laurent Loufrani
- Institute MITOVASC CNRS UMR 6015, INSERM 1083, Angers, France
- University Hospital of Angers, Angers, France
| |
Collapse
|
12
|
Structure and Function of Porcine Arteries Are Preserved for up to 6 Days Using the HypoRP Cold-storage Solution. Transplantation 2020; 104:e125-e134. [PMID: 32000259 DOI: 10.1097/tp.0000000000003141] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Maintaining functional vessels during preservation of vascularized composite allografts (VCAs) remains a major challenge. The University of Wisconsin (UW) solution has demonstrated significant short-term benefits (4-6 h). Here we determined whether the new hypothermic resuscitation and preservation solution HypoRP improves both structure, survival, and function of pig arteries during storage for up to 6 days. METHODS Using porcine swine mesenteric arteries, the effects of up to 6-day incubation in a saline (PBS), UW, or HypoRP solution on the structure, cell viability, metabolism, and function were determined. RESULTS After incubation at 4°C, for up to 6 days, the structures of the arteries were significantly disrupted, especially the tunica media, following incubation in PBS, in contrast with incubation in the HypoRP solution and to a lesser extent, in UW solution. Those disruptions were associated with increased active caspase 3 indicative of apoptosis. Additionally, while incubation in PBS led to a significant decrease in the metabolic activity, UW and HypoRP solutions allowed a stable to increased metabolic activity following 6 days of cold storage. Functional responsiveness to phenylephrine (PE) and sodium nitroprusside (SNP) decreased over time for artery rings stored in PBS and UW solution but not for those stored in HypoRP solution. Moreover, artery rings cold-stored in HypoRP solution were more sensitive to ATP. CONCLUSIONS The HypoRP solution improved long-term cold storage of porcine arteries by limiting structural alterations, including the collagen matrix, reducing apoptosis, and maintaining artery contraction-relaxation functions for up to 6 days.
Collapse
|
13
|
Sequential multidetector computed tomography assessments after venous graft treatment solution in coronary artery bypass grafting. J Thorac Cardiovasc Surg 2019; 161:96-106.e2. [PMID: 31866081 DOI: 10.1016/j.jtcvs.2019.10.115] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 10/21/2019] [Accepted: 10/21/2019] [Indexed: 11/20/2022]
Abstract
OBJECTIVES To assess the effect of DuraGraft (Somahlution Inc, Jupiter, Fla), an intraoperative graft treatment, on saphenous vein grafts in patients undergoing isolated coronary artery bypass grafting. METHODS Within patients, 2 saphenous vein grafts were randomized to DuraGraft or heparinized saline. Multidetector computed tomography angiography at 1, 3, and 12 months assessed change in wall thickness (primary end point at 3 months), lumen diameter, and maximum narrowing for the whole graft and the proximal 5-cm segment. Safety end points included graft occlusion, death, myocardial infarction, and repeat revascularization. RESULTS At 3 months, no significant changes were observed between DuraGraft- and saline-treated grafts (125 each) for wall thickness, lumen diameter, and maximum narrowing. At 12 months, DuraGraft-treated grafts demonstrated smaller mean wall thickness, overall (0.12 ± 0.06 vs 0.20 ± 0.31 mm; P = .02) and in the proximal segment (0.11 ± 0.03 vs 0.21 ± 0.33 mm; P = .01). Changes in wall thickness were greater in the proximal segment of saline-treated grafts (0.09 ± 0.29 vs 0.00 ± 0.03 mm; P = .04). Increase in maximum graft narrowing was larger in the proximal segment in the saline-treated grafts (4.7% ± 12.7% vs 0.2% ± 3.8%; P = .01). Nine DuraGraft and 11 saline grafts had occluded or thrombosed. One myocardial infarction was associated with a saline graft occlusion. No deaths or revascularizations were observed. CONCLUSIONS DuraGraft demonstrated a favorable effect on wall thickness at 12 months, particularly in the proximal segment. Longer-term follow-up in larger studies is needed to evaluate the effect on clinical outcomes.
Collapse
|
14
|
Caliskan E, Sandner S, Misfeld M, Aramendi J, Salzberg SP, Choi YH, Satishchandran V, Iyer G, Perrault LP, Böning A, Emmert MY. A novel endothelial damage inhibitor for the treatment of vascular conduits in coronary artery bypass grafting: protocol and rationale for the European, multicentre, prospective, observational DuraGraft registry. J Cardiothorac Surg 2019; 14:174. [PMID: 31615560 PMCID: PMC6794868 DOI: 10.1186/s13019-019-1010-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 09/30/2019] [Indexed: 11/15/2022] Open
Abstract
Background Vein graft disease (VGD) impairs graft patency rates and long-term outcomes after coronary artery bypass grafting (CABG). DuraGraft is a novel endothelial-damage inhibitor developed to efficiently protect the structural and functional integrity of the vascular endothelium. The DuraGraft registry will evaluate the long-term clinical outcomes of DuraGraft in patients undergoing CABG procedures. Methods This ongoing multicentre, prospective observational registry will enrol 3000 patients undergoing an isolated CABG procedure or a combined procedure (ie, CABG plus valve surgery or other surgery) with at least one saphenous vein grafts or one free arterial graft (ie, radial artery or mammary artery). If a patient is enrolled, all free grafts (SVG and arterial will be treated with DuraGraft. Data on baseline, clinical, and angiographic characteristics as well as procedural and clinical events will be collected. The primary outcome measure is the occurrence of a major adverse cardiac event (MACE; defined as death, non-fatal myocardial-infarction, or need for repeat-revascularisation). Secondary outcome measures are the occurrence of major adverse cardiac and cerebrovascular events (MACCE; defined as death, non-fatal myocardial-infarction, repeat-revascularisation, or stroke), patient-reported quality of life, and health-economic data. Patient assessments will be performed during hospitalisation, at 1-month, 1-year, and annually thereafter to 5 years post-CABG. Events will be adjudicated by an independent clinical events committee. This European, multi-institutional registry will provide detailed insights into clinical outcome associated with DuraGraft. Discussion This European, multi-institutional registry will provide detailed insights into clinical outcome associated with the use of DuraGraft. Beyond that, and given the comprehensive data sets comprising of patient, procedural, and graft parameters that are being collected, the registry will enable for multiple subgroup analyses targeting focus groups or specific clinical questions. These may include analysis of subpopulations such as patients with diabetes or multimorbid high-risk patients (patient level), evaluation of relevance of harvesting technique including endoscopic versus open conduit harvesting (procedural level), or particular graft-specific aspects (conduit level). Trial registration ClinicalTrials.gov NCT02922088. Registered October 3, 2016. Ethics and dissemination The regional ethics committees have approved the registry. Results will be submitted for publication.
Collapse
Affiliation(s)
- Etem Caliskan
- Department of Cardiovascular Surgery, Charité Universitätsmedizin Berlin, Berlin, Germany.,Department of Cardiothoracic and Vascular Surgery, German Heart Center Berlin, Berlin, Germany
| | - Sigrid Sandner
- Department of Cardiac Surgery, Vienna General Hospital, Medical University of Vienna, Vienna, Austria
| | - Martin Misfeld
- University Clinic of Cardiac Surgery, Heart Center Leipzig, Leipzig, Germany
| | - Jose Aramendi
- Division of Cardiac Surgery, Hospital de Cruces, Barakaldo, Spain
| | | | - Yeong-Hoon Choi
- Department of Cardiothoracic Surgery, Heart Center of the University Hospital of Cologne, Cologne, Germany
| | | | | | - Louis P Perrault
- Department of Surgery, Montreal Heart Institute, Université de Montréal, Montreal, Québec, Canada
| | - Andreas Böning
- Department of Cardiovascular Surgery, Justus-Liebig University Gießen, Gießen, Germany
| | - Maximilian Y Emmert
- Department of Cardiovascular Surgery, Charité Universitätsmedizin Berlin, Berlin, Germany. .,Department of Cardiothoracic and Vascular Surgery, German Heart Center Berlin, Berlin, Germany.
| |
Collapse
|
15
|
Normal Saline solutions cause endothelial dysfunction through loss of membrane integrity, ATP release, and inflammatory responses mediated by P2X7R/p38 MAPK/MK2 signaling pathways. PLoS One 2019; 14:e0220893. [PMID: 31412063 PMCID: PMC6693757 DOI: 10.1371/journal.pone.0220893] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Accepted: 07/25/2019] [Indexed: 12/12/2022] Open
Abstract
Resuscitation with 0.9% Normal Saline (NS), a non-buffered acidic solution, leads to increased morbidity and mortality in the critically ill. The goal of this study was to determine the molecular mechanisms of endothelial injury after exposure to NS. The hypothesis of this investigation is that exposure of endothelium to NS would lead to loss of cell membrane integrity, resulting in release of ATP, activation of the purinergic receptor (P2X7R), and subsequent activation of stress activated signaling pathways and inflammation. Human saphenous vein endothelial cells (HSVEC) incubated in NS, but not buffered electrolyte solution (Plasma-Lyte, PL), exhibited abnormal morphology and increased release of lactate dehydrogenase (LDH), adenosine triphosphate (ATP), and decreased transendothelial resistance (TEER), suggesting loss of membrane integrity. Incubation of intact rat aorta (RA) or human saphenous vein in NS but not PL led to impaired endothelial-dependent relaxation which was ameliorated by apyrase (hydrolyzes ATP) or SB203580 (p38 MAPK inhibitor). Exposure of HSVEC to NS but not PL led to activation of p38 MAPK and its downstream substrate, MAPKAP kinase 2 (MK2). Treatment of HSVEC with exogenous ATP led to interleukin 1β (IL-1β) release and increased vascular cell adhesion molecule (VCAM) expression. Treatment of RA with IL-1β led to impaired endothelial relaxation. IL-1β treatment of HSVEC led to increases in p38 MAPK and MK2 phosphorylation, and increased levels of arginase II. Incubation of porcine saphenous vein (PSV) in PL with pH adjusted to 6.0 or less also led to impaired endothelial function, suggesting that the acidic nature of NS is what contributes to endothelial dysfunction. Volume overload resuscitation in a porcine model after hemorrhage with NS, but not PL, led to acidosis and impaired endothelial function. These data suggest that endothelial dysfunction caused by exposure to acidic, non-buffered NS is associated with loss of membrane integrity, release of ATP, and is modulated by P2X7R-mediated inflammatory responses.
Collapse
|
16
|
Vecherskiy YY, Manvelyan DV, Zatolokin VV, Shipulin VM. VENOUS CONDUITS IN CORONARY SURGERY: OLD PROBLEMS — NEW SOLUTIONS. ACTA ACUST UNITED AC 2019. [DOI: 10.29001/2073-8552-2018-34-1-24-32] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
The introduction of autovenous coronary artery bypass grafting (CABG) marked the era of surgical revascularization in patients with coronary artery disease. It provided effective treatment for angina and significantly improved the long-term prognosis. Venous transplants today remain the most popular conduits in coronary surgery due to their availability, ease of harvesting, and the absence of length restrictions. Despite the advantages of autovenous CABG, the main disadvantage is the high incidence of venous graft failure, which represents an important and unresolved problem in cardiac and cardiovascular surgery. On the other hand, the traditional allocation of a large saphenous vein implies the dissection of soft tissues throughout the length of the isolated conduit. Traumatic dissection causes a long-lasting persistent pain syndrome after surgery, frequent abnormalities in skin sensitivity, and a high incidence of wound complications in the lower extremities. These complications lengthen the period of rehabilitation of patients and worsen the quality of life. There is an approach of isolating the vein in a block with surrounding tissues to optimize the long-term functioning of the venous shunt, however, this technique is even more traumatic than the traditional method, and therefore its use is limited in practice. On the other hand, the introduction of minimally invasive methods of isolation allowed to reduce the incidence of wound complications and to improve the cosmetic result, but there is no convincing data regarding the effect on the consistency of shunts in the long-term postoperative period. The problems associated with the use of venous conduits in CABG are multifaceted, and their solutions are necessary to improve the effectiveness of surgical revascularization.
Collapse
Affiliation(s)
- Y. Y. Vecherskiy
- Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences
| | - D. V. Manvelyan
- Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences
| | - V. V. Zatolokin
- Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences
| | - V. M. Shipulin
- Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences
| |
Collapse
|
17
|
Aavik A, Kibur RT, Lieberg J, Lepner U, Aunapuu M, Arend A. Cold-Stored Venous Allografts In Different Preserving Solutions: A Study On Changes In Vein Wall Morphology. Scand J Surg 2018; 108:67-75. [PMID: 30319041 DOI: 10.1177/1457496918783728] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND: The saphenous vein is the most frequently used bypass conduit for vascular reconstructions, which may need to be stored for a prolonged time. The aim of this study was to compare the effect of different preservation solutions on the morphology of saphenous veins during the long-term cold storage. DESIGN: An in vitro study. MATERIAL AND METHODS: Saphenous vein samples, collected from 29 patients undergoing varicose vein surgery, were stored at +4°C in (1) 10% formalin, (2) isotonic saline with heparin and antibiotic, (3) phosphate-buffered saline, (4) 2.5% glutaraldehyde + phosphate-buffered saline, and (5) Custodiol (histidine-tryptophan-ketoglutarate). Changes in the vein wall were histologically investigated up to day 35. Possible retention of the capacity of endothelial function was evaluated by immunohistochemical detection of endothelial nitric oxide synthase. RESULTS: Formalin as the control medium well preserved the vein wall morphology, but endothelial nitric oxide synthase immunostaining was very weak. Phosphate-buffered saline and isotonic saline with heparin and antibiotic poorly preserved vein wall morphology. Phosphate-buffered saline endothelial nitric oxide synthase staining decreased dramatically throughout the study period. Compared to phosphate-buffered saline, stronger isotonic saline with heparin and antibiotic endothelial nitric oxide synthase staining was noted at day 35 (p < 0.001). Custodiol and glutaraldehyde better preserved vein morphology compared to ISHA and PBS at day 5 and later (p < 0.001), but compared to stronger isotonic saline with heparin and antibiotic their endothelial nitric oxide synthase staining was weaker. CONCLUSION: In terms of preserving the morphology of saphenous veins, phosphate-buffered saline and isotonic saline with heparin and antibiotic were the poorest, while Custodiol and glutaraldehyde were the best. Demonstrating good retention of endothelial nitric oxide synthase staining throughout the study period, isotonic saline with heparin and antibiotic seems to have the best potential to retain vein wall functionality, despite relatively poor morphological preservation.
Collapse
Affiliation(s)
- A Aavik
- 1 Department of Vascular Surgery, Surgery Clinic, Tartu University Hospital, Tartu, Estonia
| | - R-T Kibur
- 2 Department of Anatomy, University of Tartu, Tartu, Estonia
| | - J Lieberg
- 1 Department of Vascular Surgery, Surgery Clinic, Tartu University Hospital, Tartu, Estonia
| | - U Lepner
- 1 Department of Vascular Surgery, Surgery Clinic, Tartu University Hospital, Tartu, Estonia
| | - M Aunapuu
- 2 Department of Anatomy, University of Tartu, Tartu, Estonia.,3 Department of Basic Veterinary Sciences and Population Medicine, Estonian University of Life Sciences, Tartu, Estonia
| | - A Arend
- 2 Department of Anatomy, University of Tartu, Tartu, Estonia
| |
Collapse
|
18
|
Haime M, McLean RR, Kurgansky KE, Emmert MY, Kosik N, Nelson C, Gaziano MJ, Cho K, Gagnon DR. Relationship between intra-operative vein graft treatment with DuraGraft® or saline and clinical outcomes after coronary artery bypass grafting. Expert Rev Cardiovasc Ther 2018; 16:963-970. [PMID: 30285502 DOI: 10.1080/14779072.2018.1532289] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Affiliation(s)
- Miguel Haime
- VA Boston Healthcare System, Harvard Medical School, West Roxbury, MA, USA
| | - Robert R. McLean
- Hebrew SeniorLife, Institute for Aging Research, Roslindale, MA, USA
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
- Massachusetts Veterans Epidemiology Research and Information Center (MAVERIC), VA Boston Healthcare System, Boston, MA, USA
| | - Katherine E. Kurgansky
- Massachusetts Veterans Epidemiology Research and Information Center (MAVERIC), VA Boston Healthcare System, Boston, MA, USA
| | - Maximilian Y. Emmert
- Clinic for Cardiovascular Surgery, University Hospital of Zurich, Zurich, Switzerland
| | - Nicole Kosik
- Massachusetts Veterans Epidemiology Research and Information Center (MAVERIC), VA Boston Healthcare System, Boston, MA, USA
| | - Constance Nelson
- Massachusetts Veterans Epidemiology Research and Information Center (MAVERIC), VA Boston Healthcare System, Boston, MA, USA
| | - Michael J. Gaziano
- VA Boston Healthcare System, Harvard Medical School, West Roxbury, MA, USA
- Massachusetts Veterans Epidemiology Research and Information Center (MAVERIC), VA Boston Healthcare System, Boston, MA, USA
- Division of Aging, Brigham and Women’s Hospital, Boston, MA, USA
| | - Kelly Cho
- VA Boston Healthcare System, Harvard Medical School, West Roxbury, MA, USA
- Massachusetts Veterans Epidemiology Research and Information Center (MAVERIC), VA Boston Healthcare System, Boston, MA, USA
- Division of Aging, Brigham and Women’s Hospital, Boston, MA, USA
| | - David R. Gagnon
- Division of Aging, Brigham and Women’s Hospital, Boston, MA, USA
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| |
Collapse
|
19
|
Sobel M, Kikuchi S, Chen L, Tang GL, Wight TN, Kenagy RD. Clinical factors that influence the cellular responses of saphenous veins used for arterial bypass. J Vasc Surg 2018; 68:165S-176S.e6. [PMID: 29914830 DOI: 10.1016/j.jvs.2018.03.436] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 03/22/2018] [Indexed: 02/01/2023]
Abstract
OBJECTIVE When an autogenous vein is harvested and used for arterial bypass, it suffers physical and biologic injuries that may set in motion the cellular processes that lead to wall thickening, fibrosis, stenosis, and ultimately graft failure. Whereas the injurious effects of surgical preparation of the vein conduit have been extensively studied, little is known about the influence of the clinical environment of the donor leg from which the vein is obtained. METHODS We studied the cellular responses of fresh saphenous vein samples obtained before implantation in 46 patients undergoing elective lower extremity bypass surgery. Using an ex vivo model of response to injury, we quantified the outgrowth of cells from explants of the adventitial and medial layers of the vein. We correlated this cellular outgrowth with the clinical characteristics of the patients, including the Wound, Ischemia, and foot Infection classification of the donor leg for ischemia, wounds, and infection as well as smoking and diabetes. RESULTS Cellular outgrowth was significantly faster and more robust from the adventitial layer than from the medial layer. The factors of leg ischemia (P < .001), smoking (P = .042), and leg infection (P = .045) were associated with impaired overall outgrowth from the adventitial tissue on multivariable analysis. Only ischemia (P = .046) was associated with impaired outgrowth of smooth muscle cells (SMCs) from the medial tissue. Co-culture of adventitial cells and SMCs propagated from vein explants revealed that adventitial cells significantly inhibited the growth of SMCs, whereas SMCs promoted the growth of adventitial cells. The AA genotype of the -838C>A p27 polymorphism (previously associated with superior graft patency) enhanced these effects, whereas the factor of smoking attenuated adventitial cell inhibition of SMC growth. Comparing gene expression, the cells cultured from the media overexpress Kyoto Encyclopedia of Genes and Genomes pathways associated with inflammation and infection, whereas those from the adventitia overexpress gene families associated with development and stem/progenitor cell maintenance. CONCLUSIONS The adverse clinical environment of the leg may influence the biologic behavior of the cells in the vein wall, especially the adventitial cells. Chronic ischemia was the most significant factor that retards adventitial cell outgrowth. The cells arising from the vein adventitia may be key players in determining a healthy adaptive or a pathologic response to the injuries associated with vein grafting.
Collapse
Affiliation(s)
- Michael Sobel
- Division of Vascular Surgery, VA Puget Sound Health Care System, Seattle, Wash; Division of Vascular Surgery, University of Washington, Seattle, Wash
| | - Shinsuke Kikuchi
- Department of Vascular Surgery, Asahikawa Medical University, Asahikawa, Japan
| | - Lihua Chen
- Division of Vascular Surgery, University of Washington, Seattle, Wash
| | - Gale L Tang
- Division of Vascular Surgery, VA Puget Sound Health Care System, Seattle, Wash; Division of Vascular Surgery, University of Washington, Seattle, Wash
| | - Tom N Wight
- Matrix Biology Program, Benaroya Research Institute, Seattle, Wash
| | - Richard D Kenagy
- Division of Vascular Surgery, University of Washington, Seattle, Wash.
| |
Collapse
|
20
|
Ben Ali W, Voisine P, Olsen PS, Jeanmart H, Noiseux N, Goeken T, Satishchandran V, Cademartiri F, Cutter G, Veerasingam D, Brown C, Emmert MY, Perrault LP. DuraGraft vascular conduit preservation solution in patients undergoing coronary artery bypass grafting: rationale and design of a within-patient randomised multicentre trial. Open Heart 2018; 5:e000780. [PMID: 29682294 PMCID: PMC5905829 DOI: 10.1136/openhrt-2018-000780] [Citation(s) in RCA: 12] [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: 01/13/2018] [Revised: 02/21/2018] [Accepted: 03/06/2018] [Indexed: 11/25/2022] Open
Abstract
Introduction Saphenous vein grafts (SVGs) remain the most often used conduits in coronary artery bypass grafting (CABG). However, they are prone to vein graft disease (VGD) during follow-up, which may compromise clinical outcomes. Injury to the SVG endothelium during harvesting and storage promotes neointimal hyperplasia that can advance to atherosclerosis characterised by SVG failure. This trial investigates the potential benefit of DuraGraft, a novel, one-time intraoperative graft treatment developed to efficiently protect the structural and functional integrity of the vascular endothelium, on the development and progression of VGD in CABG patients. Methods and analysis This ongoing prospective randomised, double-blinded multicentre trial (NCT02272582/NCT02774824) includes patients undergoing isolated CABG requiring at least two SVGs. It compares the impact of DuraGraft, a novel treatment against VGD versus the standard-of-care (SOC; heparinised saline) using a within-patient randomisation (with one SVG treated with DuraGraft and the other treated with SOC). Besides clinical assessments, patients undergo longitudinal 64-slice or better multidetector CT (MDCT) angiography of paired grafts (within each patient) at 4–6 weeks, 3 months and 12 months. Primary endpoints will be the magnitude of change in mean wall thickness and lumen diameter (stenosis) of paired grafts, at 3 and 12 months, respectively. Besides the evaluation of overall safety, longitudinal assessment of each graft (secondary endpoint) is performed in order to obtain insight into graft behaviour after CABG. Enrolment of 119 patients was successfully completed, and analysis of MDCT angiography follow-up is ongoing with the completed analysis becoming available by end of first quarter of 2018. Ethics and dissemination The regional ethics committees have approved the trial. Results will be submitted for publication. Clinical trial identifier NCT02272582 and NCT02774824.
Collapse
Affiliation(s)
- Walid Ben Ali
- Department of Surgery, Montreal Heart Institute, Montréal, Canada
| | - Pierre Voisine
- Institut Universitaire de Cardiologie et de Pneumologie, Quebec City, Canada
| | - Peter Skov Olsen
- Dept. of Cardiac Surgery, Rigshopitalet University of Copenhagen, Copenhagen, Denmark
| | | | - Nicolas Noiseux
- Department of Surgery, Montreal Heart Institute, Montréal, Canada
| | | | | | | | | | | | - Craig Brown
- New Brunswick Heart Centre, Saint John, Canada
| | | | - Louis P Perrault
- Department of Surgery, Montreal Heart Institute, Montréal, Canada
| |
Collapse
|
21
|
Ben Ali W, Bouhout I, Perrault LP. The effect of storage solutions, gene therapy, and antiproliferative agents on endothelial function and saphenous vein graft patency. J Card Surg 2018; 33:235-242. [DOI: 10.1111/jocs.13608] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Walid Ben Ali
- Department of Cardiac Surgery; Montreal Heart Institute; Quebec Canada
| | - Ismail Bouhout
- Department of Surgery; Université de Montréal; Quebec Canada
| | - Louis P. Perrault
- Department of Cardiovascular Surgery; Montreal Heart Institute; Quebec Canada
| |
Collapse
|
22
|
Abstract
Crystalloid infusion is widely employed in patient care for volume replacement and resuscitation. In the United States the crystalloid of choice is often normal saline. Surgeons and anesthesiologists have long preferred buffered solutions such as Ringer's Lactate and Plasma-Lyte A. Normal saline is the solution most widely employed in medical and pediatric care, as well as in hematology and transfusion medicine. However, there is growing concern that normal saline is more toxic than balanced, buffered crystalloids such as Plasma-Lyte and Lactated Ringer's. Normal saline is the only solution recommended for red cell washing, administration and salvage in the USA, but Plasma-Lyte A is also FDA approved for these purposes. Lactated Ringer's has been traditionally avoided in these applications due to concerns over clotting, but existing research suggests this is not likely a problem. In animal models and clinical studies in various settings, normal saline can cause metabolic acidosis, vascular and renal function changes, as well as abdominal pain in comparison with balanced crystalloids. The one extant randomized trial suggests that in very small volumes (2 l or less) normal saline is not more toxic than other crystalloids. Recent evidence suggests that normal saline causes substantially more in vitro hemolysis than Plasma-Lyte A and similar solutions during short term storage (24 hours) after washing or intraoperative salvage. There are now abundant data to raise concerns as to whether normal saline is the safest replacement solution in infusion therapy, red cell washing and salvage, apheresis and similar uses. In the USA, Plasma-Lyte A is also FDA approved for use with blood components and is likely a safer solution for these purposes. Its only disadvantage is a higher cost. Additional studies of the safety of normal saline for virtually all current clinical uses are needed. It seems likely that normal saline will eventually be abandoned in favor of safer, more physiologic crystalloid solutions in the coming years.
Collapse
|
23
|
Vestergaard LP, Benhassen L, Modrau IS, de Paoli F, Boedtkjer E. Increased Contractile Function of Human Saphenous Vein Grafts Harvested by "No-Touch" Technique. Front Physiol 2018; 8:1135. [PMID: 29379447 PMCID: PMC5770882 DOI: 10.3389/fphys.2017.01135] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 12/22/2017] [Indexed: 01/21/2023] Open
Abstract
Saphenous vein grafts are the most common conduits used for coronary artery bypass grafting (CABG); however, no more than 60% of vein grafts remain open after 10 years and graft failure is associated with poor clinical outcome. The “no-touch” harvesting technique—where a sheet of perivascular tissue is retained around the vein—improves graft patency to over 80% after 16 years of follow-up, but the mechanism for the improved patency rate is unclear. In this study, we investigated acute functional differences between vein grafts harvested conventionally and by “no-touch” technique and explored the importance of perivascular tissue for reducing surgical trauma, minimizing excessive distension, and releasing vasoactive paracrine factors. Segments of human saphenous veins were obtained from CABG surgery and their functional properties investigated by isometric and isobaric myography. We found a broad diameter-tension relationship for human saphenous veins, with peak capacity for active tension development at diameters corresponding to transmural pressures around 60 mmHg. Across the investigated transmural pressure range between 10 and 120 mmHg, maximal tension development was higher for “no-touch” compared to conventionally harvested saphenous veins. Contractile responses to serotonin, noradrenaline, and depolarization induced with elevated extracellular [K+] were significantly larger for saphenous veins harvested by “no-touch” compared to conventional technique. Conventional vein grafts are routinely pressurized manually in order to test for leaks; however, avoiding this distension procedure did not change the acute contractile function of the conventionally excised saphenous veins. In contrast, even though surgical removal of perivascular tissue during conventional harvesting was associated with a substantial decrease in force development, removal of perivascular tissue by careful dissection under a stereomicroscope only marginally affected contractile responses of veins harvested by “no-touch” technique. In conclusion, we show that saphenous veins harvested by “no-touch” technique have greater contractile capacity than veins harvested by conventional technique. The different capacity for smooth muscle contraction is not due to vasoactive substances released by the perivascular tissue. Instead, we propose that the larger tension development of saphenous veins harvested by “no-touch” technique reflects reduced surgical damage, which may have long-term consequences that contribute to the superior graft patency.
Collapse
Affiliation(s)
| | - Leila Benhassen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark.,Department of Cardiothoracic Surgery, Aarhus University Hospital, Aarhus, Denmark
| | - Ivy S Modrau
- Department of Cardiothoracic Surgery, Aarhus University Hospital, Aarhus, Denmark
| | - Frank de Paoli
- Department of Biomedicine, Aarhus University, Aarhus, Denmark.,Department of Cardiothoracic Surgery, Aarhus University Hospital, Aarhus, Denmark
| | - Ebbe Boedtkjer
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
24
|
Luo W, Feldman D, McCallister R, Brophy C, Cheung-Flynn J. P2X7R antagonism after subfailure overstretch injury of blood vessels reverses vasomotor dysfunction and prevents apoptosis. Purinergic Signal 2017; 13:579-590. [PMID: 28905300 PMCID: PMC5714848 DOI: 10.1007/s11302-017-9585-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Accepted: 08/25/2017] [Indexed: 12/13/2022] Open
Abstract
Human saphenous vein (HSV) is harvested and prepared prior to implantation as an arterial bypass graft. Injury and the response to injury from surgical harvest and preparation trigger cascades of molecular events and contribute to graft remodeling and intimal hyperplasia. Apoptosis is an early response after implantation that contributes the development of neointimal lesions. Here, we showed that surgical harvest and preparation of HSV leads to vasomotor dysfunction, increased apoptosis and downregulation of the phosphorylation of the anti-apoptotic protein, Niban. A model of subfailure overstretch injury in rat aorta (RA) was used to demonstrate impaired vasomotor function, increased extracellular ATP (eATP) release, and increased apoptosis following pathological vascular injury. The subfailure overstretch injury was associated with activation of p38 MAPK stress pathway and decreases in the phosphorylation of the anti-apoptotic protein Niban. Treatment of RA after overstretch injury with antagonists to purinergic P2X7 receptor (P2X7R) antagonists or P2X7R/pannexin (PanX1) complex, but not PanX1 alone, restored vasomotor function. Inhibitors to P2X7R and PanX1 reduced stretch-induced eATP release. P2X7R/PanX1 antagonism led to decrease in p38 MAPK phosphorylation, restoration of Niban phosphorylation and increases in the phosphorylation of the anti-apoptotic protein Akt in RA and reduced TNFα-stimulated caspase 3/7 activity in cultured rat vascular smooth muscle cells. In conclusion, inhibition of P2X7R after overstretch injury restored vasomotor function and inhibited apoptosis. Treatment with P2X7R/PanX1 complex inhibitors after harvest and preparation injury of blood vessels used for bypass conduits may prevent the subsequent response to injury that lead to apoptosis and represents a novel therapeutic approach to prevent graft failure.
Collapse
Affiliation(s)
- Weifeng Luo
- Department of Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Daniel Feldman
- Department of Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Reid McCallister
- Department of Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Colleen Brophy
- Department of Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
- VA Tennessee Valley Healthcare System, Nashville, TN, USA
| | - Joyce Cheung-Flynn
- Department of Surgery, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
25
|
Komalavilas P, Luo W, Guth CM, Jolayemi O, Bartelson RI, Cheung-Flynn J, Brophy CM. Vascular surgical stretch injury leads to activation of P2X7 receptors and impaired endothelial function. PLoS One 2017; 12:e0188069. [PMID: 29136654 PMCID: PMC5685620 DOI: 10.1371/journal.pone.0188069] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 10/31/2017] [Indexed: 12/17/2022] Open
Abstract
A viable vascular endothelial layer prevents vasomotor dysfunction, thrombosis, inflammation, and intimal hyperplasia. Injury to the endothelium occurs during harvest and “back table” preparation of human saphenous vein prior to implantation as an arterial bypass conduit. A subfailure overstretch model of rat aorta was used to show that subfailure stretch injury of vascular tissue leads to impaired endothelial-dependent relaxation. Stretch-induced impaired relaxation was mitigated by treatment with purinergic P2X7 receptor (P2X7R) inhibitors, brilliant blue FCF (FCF) and A740003, or apyrase, an enzyme that catalyzes the hydrolysis of ATP. Alternatively, treatment of rat aorta with exogenous ATP or 2’(3’)-O-(4-Benzoyl benzoyl)-ATP (BzATP) also impaired endothelial-dependent relaxation. Treatment of human saphenous vein endothelial cells (HSVEC) with exogenous ATP led to reduced nitric oxide production which was associated with increased phosphorylation of the stress activated protein kinase, p38 MAPK. ATP- stimulated p38 MAPK phosphorylation of HSVEC was inhibited by FCF and SB203580. Moreover, ATP inhibition of nitric oxide production in HSVEC was prevented by FCF, SB203580, L-arginine supplementation and arginase inhibition. Finally, L-arginine supplementation and arginase inhibition restored endothelial dependent relaxation after stretch injury of rat aorta. These results suggest that vascular stretch injury leads to ATP release, activation of P2X7R and p38 MAPK resulting in endothelial dysfunction due to arginase activation. Endothelial function can be restored in both ATP treated HSVEC and intact stretch injured rat aorta by P2X7 receptor inhibition with FCF or L-arginine supplementation, implicating straightforward therapeutic options for treatment of surgical vascular injury.
Collapse
Affiliation(s)
- Padmini Komalavilas
- Vanderbilt University Medical Center, Department of Surgery, Nashville, TN, United States of America
- VA Tennessee Valley Healthcare System, Nashville, TN, United States of America
- * E-mail:
| | - Weifeng Luo
- Vanderbilt University Medical Center, Department of Surgery, Nashville, TN, United States of America
| | - Christy M. Guth
- Vanderbilt University Medical Center, Department of Surgery, Nashville, TN, United States of America
| | - Olukemi Jolayemi
- Vanderbilt University Medical Center, Department of Surgery, Nashville, TN, United States of America
| | - Rachel I. Bartelson
- Vanderbilt University Medical Center, Department of Surgery, Nashville, TN, United States of America
| | - Joyce Cheung-Flynn
- Vanderbilt University Medical Center, Department of Surgery, Nashville, TN, United States of America
| | - Colleen M. Brophy
- Vanderbilt University Medical Center, Department of Surgery, Nashville, TN, United States of America
- VA Tennessee Valley Healthcare System, Nashville, TN, United States of America
| |
Collapse
|
26
|
Gaudino M, Antoniades C, Benedetto U, Deb S, Di Franco A, Di Giammarco G, Fremes S, Glineur D, Grau J, He GW, Marinelli D, Ohmes LB, Patrono C, Puskas J, Tranbaugh R, Girardi LN, Taggart DP, Ruel M, Bakaeen FG. Mechanisms, Consequences, and Prevention of Coronary Graft Failure. Circulation 2017; 136:1749-1764. [DOI: 10.1161/circulationaha.117.027597] [Citation(s) in RCA: 138] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Mario Gaudino
- From Department of Cardiothoracic Surgery, @Weill Cornell Medicine, New York (M.G., A.D.F., L.B.O., R.T., L.N.G.); Department of Medicine and Department of Cardiovascular Surgery, John Radcliffe Hospital, University of Oxford, UK (C.A., D.P.T.); Bristol Heart Institute, University of Bristol, School of Clinical Sciences, UK (U.B.); Schulich Heart Centre, Sunnybrook Health Science, University of Toronto, Canada (S.D., S.F.); University “G. D’Annunzio,” Chieti, Italy (G.D.G., D.M.); Division of
| | - Charalambos Antoniades
- From Department of Cardiothoracic Surgery, @Weill Cornell Medicine, New York (M.G., A.D.F., L.B.O., R.T., L.N.G.); Department of Medicine and Department of Cardiovascular Surgery, John Radcliffe Hospital, University of Oxford, UK (C.A., D.P.T.); Bristol Heart Institute, University of Bristol, School of Clinical Sciences, UK (U.B.); Schulich Heart Centre, Sunnybrook Health Science, University of Toronto, Canada (S.D., S.F.); University “G. D’Annunzio,” Chieti, Italy (G.D.G., D.M.); Division of
| | - Umberto Benedetto
- From Department of Cardiothoracic Surgery, @Weill Cornell Medicine, New York (M.G., A.D.F., L.B.O., R.T., L.N.G.); Department of Medicine and Department of Cardiovascular Surgery, John Radcliffe Hospital, University of Oxford, UK (C.A., D.P.T.); Bristol Heart Institute, University of Bristol, School of Clinical Sciences, UK (U.B.); Schulich Heart Centre, Sunnybrook Health Science, University of Toronto, Canada (S.D., S.F.); University “G. D’Annunzio,” Chieti, Italy (G.D.G., D.M.); Division of
| | - Saswata Deb
- From Department of Cardiothoracic Surgery, @Weill Cornell Medicine, New York (M.G., A.D.F., L.B.O., R.T., L.N.G.); Department of Medicine and Department of Cardiovascular Surgery, John Radcliffe Hospital, University of Oxford, UK (C.A., D.P.T.); Bristol Heart Institute, University of Bristol, School of Clinical Sciences, UK (U.B.); Schulich Heart Centre, Sunnybrook Health Science, University of Toronto, Canada (S.D., S.F.); University “G. D’Annunzio,” Chieti, Italy (G.D.G., D.M.); Division of
| | - Antonino Di Franco
- From Department of Cardiothoracic Surgery, @Weill Cornell Medicine, New York (M.G., A.D.F., L.B.O., R.T., L.N.G.); Department of Medicine and Department of Cardiovascular Surgery, John Radcliffe Hospital, University of Oxford, UK (C.A., D.P.T.); Bristol Heart Institute, University of Bristol, School of Clinical Sciences, UK (U.B.); Schulich Heart Centre, Sunnybrook Health Science, University of Toronto, Canada (S.D., S.F.); University “G. D’Annunzio,” Chieti, Italy (G.D.G., D.M.); Division of
| | - Gabriele Di Giammarco
- From Department of Cardiothoracic Surgery, @Weill Cornell Medicine, New York (M.G., A.D.F., L.B.O., R.T., L.N.G.); Department of Medicine and Department of Cardiovascular Surgery, John Radcliffe Hospital, University of Oxford, UK (C.A., D.P.T.); Bristol Heart Institute, University of Bristol, School of Clinical Sciences, UK (U.B.); Schulich Heart Centre, Sunnybrook Health Science, University of Toronto, Canada (S.D., S.F.); University “G. D’Annunzio,” Chieti, Italy (G.D.G., D.M.); Division of
| | - Stephen Fremes
- From Department of Cardiothoracic Surgery, @Weill Cornell Medicine, New York (M.G., A.D.F., L.B.O., R.T., L.N.G.); Department of Medicine and Department of Cardiovascular Surgery, John Radcliffe Hospital, University of Oxford, UK (C.A., D.P.T.); Bristol Heart Institute, University of Bristol, School of Clinical Sciences, UK (U.B.); Schulich Heart Centre, Sunnybrook Health Science, University of Toronto, Canada (S.D., S.F.); University “G. D’Annunzio,” Chieti, Italy (G.D.G., D.M.); Division of
| | - David Glineur
- From Department of Cardiothoracic Surgery, @Weill Cornell Medicine, New York (M.G., A.D.F., L.B.O., R.T., L.N.G.); Department of Medicine and Department of Cardiovascular Surgery, John Radcliffe Hospital, University of Oxford, UK (C.A., D.P.T.); Bristol Heart Institute, University of Bristol, School of Clinical Sciences, UK (U.B.); Schulich Heart Centre, Sunnybrook Health Science, University of Toronto, Canada (S.D., S.F.); University “G. D’Annunzio,” Chieti, Italy (G.D.G., D.M.); Division of
| | - Juan Grau
- From Department of Cardiothoracic Surgery, @Weill Cornell Medicine, New York (M.G., A.D.F., L.B.O., R.T., L.N.G.); Department of Medicine and Department of Cardiovascular Surgery, John Radcliffe Hospital, University of Oxford, UK (C.A., D.P.T.); Bristol Heart Institute, University of Bristol, School of Clinical Sciences, UK (U.B.); Schulich Heart Centre, Sunnybrook Health Science, University of Toronto, Canada (S.D., S.F.); University “G. D’Annunzio,” Chieti, Italy (G.D.G., D.M.); Division of
| | - Guo-Wei He
- From Department of Cardiothoracic Surgery, @Weill Cornell Medicine, New York (M.G., A.D.F., L.B.O., R.T., L.N.G.); Department of Medicine and Department of Cardiovascular Surgery, John Radcliffe Hospital, University of Oxford, UK (C.A., D.P.T.); Bristol Heart Institute, University of Bristol, School of Clinical Sciences, UK (U.B.); Schulich Heart Centre, Sunnybrook Health Science, University of Toronto, Canada (S.D., S.F.); University “G. D’Annunzio,” Chieti, Italy (G.D.G., D.M.); Division of
| | - Daniele Marinelli
- From Department of Cardiothoracic Surgery, @Weill Cornell Medicine, New York (M.G., A.D.F., L.B.O., R.T., L.N.G.); Department of Medicine and Department of Cardiovascular Surgery, John Radcliffe Hospital, University of Oxford, UK (C.A., D.P.T.); Bristol Heart Institute, University of Bristol, School of Clinical Sciences, UK (U.B.); Schulich Heart Centre, Sunnybrook Health Science, University of Toronto, Canada (S.D., S.F.); University “G. D’Annunzio,” Chieti, Italy (G.D.G., D.M.); Division of
| | - Lucas B. Ohmes
- From Department of Cardiothoracic Surgery, @Weill Cornell Medicine, New York (M.G., A.D.F., L.B.O., R.T., L.N.G.); Department of Medicine and Department of Cardiovascular Surgery, John Radcliffe Hospital, University of Oxford, UK (C.A., D.P.T.); Bristol Heart Institute, University of Bristol, School of Clinical Sciences, UK (U.B.); Schulich Heart Centre, Sunnybrook Health Science, University of Toronto, Canada (S.D., S.F.); University “G. D’Annunzio,” Chieti, Italy (G.D.G., D.M.); Division of
| | - Carlo Patrono
- From Department of Cardiothoracic Surgery, @Weill Cornell Medicine, New York (M.G., A.D.F., L.B.O., R.T., L.N.G.); Department of Medicine and Department of Cardiovascular Surgery, John Radcliffe Hospital, University of Oxford, UK (C.A., D.P.T.); Bristol Heart Institute, University of Bristol, School of Clinical Sciences, UK (U.B.); Schulich Heart Centre, Sunnybrook Health Science, University of Toronto, Canada (S.D., S.F.); University “G. D’Annunzio,” Chieti, Italy (G.D.G., D.M.); Division of
| | - John Puskas
- From Department of Cardiothoracic Surgery, @Weill Cornell Medicine, New York (M.G., A.D.F., L.B.O., R.T., L.N.G.); Department of Medicine and Department of Cardiovascular Surgery, John Radcliffe Hospital, University of Oxford, UK (C.A., D.P.T.); Bristol Heart Institute, University of Bristol, School of Clinical Sciences, UK (U.B.); Schulich Heart Centre, Sunnybrook Health Science, University of Toronto, Canada (S.D., S.F.); University “G. D’Annunzio,” Chieti, Italy (G.D.G., D.M.); Division of
| | - Robert Tranbaugh
- From Department of Cardiothoracic Surgery, @Weill Cornell Medicine, New York (M.G., A.D.F., L.B.O., R.T., L.N.G.); Department of Medicine and Department of Cardiovascular Surgery, John Radcliffe Hospital, University of Oxford, UK (C.A., D.P.T.); Bristol Heart Institute, University of Bristol, School of Clinical Sciences, UK (U.B.); Schulich Heart Centre, Sunnybrook Health Science, University of Toronto, Canada (S.D., S.F.); University “G. D’Annunzio,” Chieti, Italy (G.D.G., D.M.); Division of
| | - Leonard N. Girardi
- From Department of Cardiothoracic Surgery, @Weill Cornell Medicine, New York (M.G., A.D.F., L.B.O., R.T., L.N.G.); Department of Medicine and Department of Cardiovascular Surgery, John Radcliffe Hospital, University of Oxford, UK (C.A., D.P.T.); Bristol Heart Institute, University of Bristol, School of Clinical Sciences, UK (U.B.); Schulich Heart Centre, Sunnybrook Health Science, University of Toronto, Canada (S.D., S.F.); University “G. D’Annunzio,” Chieti, Italy (G.D.G., D.M.); Division of
| | - David P. Taggart
- From Department of Cardiothoracic Surgery, @Weill Cornell Medicine, New York (M.G., A.D.F., L.B.O., R.T., L.N.G.); Department of Medicine and Department of Cardiovascular Surgery, John Radcliffe Hospital, University of Oxford, UK (C.A., D.P.T.); Bristol Heart Institute, University of Bristol, School of Clinical Sciences, UK (U.B.); Schulich Heart Centre, Sunnybrook Health Science, University of Toronto, Canada (S.D., S.F.); University “G. D’Annunzio,” Chieti, Italy (G.D.G., D.M.); Division of
| | - Marc Ruel
- Division of Cardiac Surgery, School of Epidemiology, Public Health, and Preventive Medicine, University of Ottawa, Canada
| | - Faisal G. Bakaeen
- Department of Cardiovascular Surgery, Texas Heart Institute, Houston, and Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX
| |
Collapse
|
27
|
Limiting Injury During Saphenous Vein Graft Preparation For Coronary Arterial Bypass Prevents Metabolic Decompensation. Sci Rep 2017; 7:14179. [PMID: 29079734 PMCID: PMC5660200 DOI: 10.1038/s41598-017-13819-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Accepted: 10/02/2017] [Indexed: 02/01/2023] Open
Abstract
Standard harvest and preparation of human saphenous vein (HSV) for autologous coronary and peripheral arterial bypass procedures is associated with injury and increased oxidative stress that negatively affect graft performance. In this study we investigated the global metabolomic profiles of HSV before (unprepared; UP) and after standard vein graft preparation (AP). AP-HSV showed impaired vasomotor function that was associated with increased oxidative stress, phospholipid hydrolysis and energy depletion that are characteristic of mechanical and chemical injury. A porcine model (PSV) was utilized to validate these metabolomic changes in HSV and to determine the efficacy of an improved preparation technique (OP) using pressure-regulated distension, a non-toxic vein marker, and graft storage in buffered PlasmaLyte solution in limiting metabolic decompensation due to graft preparation. Deficits in vasomotor function and metabolic signature observed in AP-PSV could be largely mitigated with the OP procedure. These findings suggest that simple strategies aimed at reducing injury during graft harvest and preparation represents a straightforward and viable strategy to preserve conduit function and possibly improve graft patency.
Collapse
|
28
|
Wheeler AR, Kendrick DE, Allemang MT, Gosling AF, Kim AH, Hausladen A, Kashyap VS. Endothelial Function Is Preserved in Veins Harvested by Either Endoscopic or Surgical Techniques. Ann Vasc Surg 2017; 44:317-324. [PMID: 28495542 DOI: 10.1016/j.avsg.2017.05.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Accepted: 05/01/2017] [Indexed: 10/19/2022]
Abstract
BACKGROUND Endoscopic vein harvest for lower extremity arterial bypass grafting has been questioned due to concern for endothelial damage during procurement. We sought to compare nitric oxide (NO)-mediated endothelial-dependent relaxation (EDR) in vein segments harvested using open surgical techniques (OH) versus endoscopic vein harvest (EH) techniques. METHODS Saphenous vein segments were harvested for lower extremity bypass, and a single, minimally handled section of saphenous vein, free of branches, was taken from the end of the graft. Four 4-mm venous ring segments were then cut and mounted on force transducers. Segments were mounted in 37° oxygenated Krebs-Henseleit solution and maximally contracted using KCl. Individual ring segments that did not react to KCl were excluded from the study. Norepinephrine (NE) was used to achieve submaximal contraction. EDR was determined using increasing concentrations of bradykinin (BDK). Endothelial-independent relaxation (EIR) was confirmed using sodium nitroprusside. Two-way analysis of variance (ANOVA) was used to analyze differences between harvest techniques across BDK concentration and a Student's t-test was used to analyze single comparisons. RESULTS Vein segments harvested from patients (n = 13) led to 28 viable rings that exhibited a positive reaction to KCl (11 rings; 5 patients EH vs. 17 rings; 8 patients OH). Both vein groups achieved moderate relaxation to maximal BDK concentration, [10-6 M]; (49.5% EH vs. 40.55% OH, P = 0.270). Analysis by 2-way ANOVA for mean % relaxation for BDK concentration [10-11-10-6 M] showed improved EDR in EH samples compared with OH (P = 0.029). Mean nitrite/nitrate (NO(x)) tissue bath concentration measurements post-BDK were 139.8 nM (EH) vs. 97.2 nM (OH; P = 0.264). Histology and positive factor VIII immunohistochemistry staining provided evidence for the presence of intact endothelium in our sample segments. EIR was preserved and was similar in the two groups. CONCLUSIONS Endothelial function is preserved when utilizing endoscopic harvesting techniques. The advantages of minimally invasive vein procurement for lower extremity bypass can be obtained without concern for damaging venous endothelium.
Collapse
Affiliation(s)
- Adam R Wheeler
- Department of Vascular Surgery, University Hospitals, Cleveland, OH
| | | | - Matt T Allemang
- Department of Vascular Surgery, University Hospitals, Cleveland, OH
| | - Andre F Gosling
- Department of Vascular Surgery, University Hospitals, Cleveland, OH
| | - Ann H Kim
- Department of Vascular Surgery, University Hospitals, Cleveland, OH
| | - Alfred Hausladen
- Institute for Transformative Molecular Medicine, Case Western Reserve University School of Medicine, University Hospitals Case Medical Center, Cleveland, OH
| | - Vikram S Kashyap
- Department of Vascular Surgery, University Hospitals, Cleveland, OH.
| |
Collapse
|
29
|
Guth CM, Luo W, Jolayemi O, Chadalavada KS, Komalavilas P, Cheung-Flynn J, Brophy CM. Adenosine triphosphate as a molecular mediator of the vascular response to injury. J Surg Res 2017; 216:80-86. [PMID: 28807217 DOI: 10.1016/j.jss.2017.03.025] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 03/03/2017] [Accepted: 03/23/2017] [Indexed: 11/29/2022]
Abstract
BACKGROUND Human saphenous veins used for arterial bypass undergo stretch injury at the time of harvest and preimplant preparation. Vascular injury promotes intimal hyperplasia, the leading cause of graft failure, but the molecular events leading to this response are largely unknown. This study investigated adenosine triphosphate (ATP) as a potential molecular mediator in the vascular response to stretch injury, and the downstream effects of the purinergic receptor, P2X7R, and p38 MAPK activation. MATERIALS AND METHODS A subfailure stretch rat aorta model was used to determine the effect of stretch injury on release of ATP and vasomotor responses. Stretch-injured tissues were treated with apyrase, the P2X7R antagonist, A438079, or the p38 MAPK inhibitor, SB203580, and subsequent contractile forces were measured using a muscle bath. An exogenous ATP (eATP) injury model was developed and the experiment repeated. Change in p38 MAPK phosphorylation after stretch and eATP tissue injury was determined using Western blotting. Noninjured tissue was incubated in the p38 MAPK activator, anisomycin, and subsequent contractile function and p38 MAPK phosphorylation were analyzed. RESULTS Stretch injury was associated with release of ATP. Contractile function was decreased in tissue subjected to subfailure stretch, eATP, and anisomycin. Contractile function was restored by apyrase, P2X7R antagonism, and p38-MAPK inhibition. Stretch, eATP, and anisomycin-injured tissue demonstrated increased phosphorylation of p38 MAPK. CONCLUSIONS Taken together, these data suggest that the vascular response to stretch injury is associated with release of ATP and activation of the P2X7R/P38 MAPK pathway, resulting in contractile dysfunction. Modulation of this pathway in vein grafts after harvest and before implantation may reduce the vascular response to injury.
Collapse
Affiliation(s)
- Christy M Guth
- Department of Surgery, Vanderbilt University, Nashville, Tennessee.
| | - Weifung Luo
- Department of Surgery, Vanderbilt University, Nashville, Tennessee
| | - Olukemi Jolayemi
- Department of Surgery, Vanderbilt University, Nashville, Tennessee
| | | | | | | | - Colleen M Brophy
- Department of Surgery, Vanderbilt University, Nashville, Tennessee; VA Tennessee Valley Healthcare System, Nashville, Tennessee
| |
Collapse
|
30
|
Luo W, Guth CM, Jolayemi O, Duvall CL, Brophy CM, Cheung-Flynn J. Subfailure Overstretch Injury Leads to Reversible Functional Impairment and Purinergic P2X7 Receptor Activation in Intact Vascular Tissue. Front Bioeng Biotechnol 2016; 4:75. [PMID: 27747211 PMCID: PMC5040722 DOI: 10.3389/fbioe.2016.00075] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 09/13/2016] [Indexed: 11/30/2022] Open
Abstract
Vascular stretch injury is associated with blunt trauma, vascular surgical procedures, and harvest of human saphenous vein for use in vascular bypass grafting. A model of subfailure overstretch in rat abdominal aorta was developed to characterize surgical vascular stretch injury. Longitudinal stretch of rat aorta was characterized ex vivo. Stretch to the haptic endpoint, where the tissues would no longer lengthen, occurred at twice the resting length. The stress produced at this length was greater than physiologic mechanical forces but well below the level of mechanical disruption. Functional responses were determined in a muscle bath, and this subfailure overstretch injury led to impaired smooth muscle function that was partially reversed by treatment with purinergic receptor (P2X7R) antagonists. These data suggest that vasomotor dysfunction caused by subfailure overstretch injury may be due to the activation of P2X7R. These studies have implications for our understanding of mechanical stretch injury of blood vessels and offer novel therapeutic opportunities.
Collapse
Affiliation(s)
- Weifeng Luo
- Department of Surgery, Vanderbilt University, Nashville, TN, USA
| | - Christy M. Guth
- Department of Surgery, Vanderbilt University, Nashville, TN, USA
| | - Olukemi Jolayemi
- Department of Surgery, Vanderbilt University, Nashville, TN, USA
| | - Craig L. Duvall
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Colleen Marie Brophy
- Department of Surgery, Vanderbilt University, Nashville, TN, USA
- VA Tennessee Valley Healthcare System, Nashville, TN, USA
| | | |
Collapse
|
31
|
Wise ES, Brophy CM. The Case for Endothelial Preservation via Pressure-Regulated Distension in the Preparation of Autologous Saphenous Vein Conduits in Cardiac and Peripheral Bypass Operations. Front Surg 2016; 3:54. [PMID: 27713879 PMCID: PMC5031700 DOI: 10.3389/fsurg.2016.00054] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Accepted: 09/12/2016] [Indexed: 12/04/2022] Open
Affiliation(s)
- Eric S Wise
- Department of Surgery, University of Maryland Medical Center , Baltimore, MD , USA
| | - Colleen M Brophy
- VA Tennessee Valley Healthcare System, Vanderbilt University, Nashville, TN, USA; Department of Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
32
|
Wise ES, Hocking KM, Luo W, Feldman DL, Song J, Komalavilas P, Cheung-Flynn J, Brophy CM. Traditional graft preparation decreases physiologic responses, diminishes viscoelasticity, and reduces cellular viability of the conduit: A porcine saphenous vein model. Vasc Med 2016; 21:413-421. [PMID: 27216870 DOI: 10.1177/1358863x16649040] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Traditional methods of intraoperative human saphenous vein preparation for use as bypass grafts can be deleterious to the conduit. The purpose of this study was to characterize acute graft preparation injury, and to mitigate this harm via an improved preparation technique. Porcine saphenous veins were surgically harvested (unprepared controls, UnP) and prepared using traditional (TraP) and improved preparations (ImP). The TraP used unregulated radial distension, marking with a surgical skin marker and preservation in heparinized normal saline. ImP used pressure-regulated distension, brilliant blue FCF-based pen marking and preservation in heparinized Plasma-Lyte A. Rings from each preparation were suspended in a muscle bath for characterization of physiologic responses to vasoactive agents and viscoelasticity. Cellular viability was assessed using the methyl thiazolyl tetrazolium (MTT) assay and the terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL) assay for apoptosis. Contractile responses to potassium chloride (110 mM) and phenylephrine (10 µM), and endothelial-dependent and independent vasodilatory responses to carbachol (0.5 µM) and sodium nitroprusside (1 µM), respectively, were decreased in TraP tissues compared to both UnP and ImP tissues (p ⩽ 0.05). TraP tissues demonstrated diminished viscoelasticity relative to UnP and ImP tissues (p ⩽ 0.05), and reduced cellular viability relative to UnP control (p ⩽ 0.01) by the MTT assay. On the TUNEL assay, TraP tissues demonstrated a greater degree of apoptosis relative to UnP and ImP tissues (p ⩽ 0.01). In conclusion, an improved preparation technique prevents vascular graft smooth muscle and endothelial injury observed in tissues prepared using a traditional approach.
Collapse
Affiliation(s)
- Eric S Wise
- Department of Surgery, Vanderbilt University, Nashville, TN, USA
| | - Kyle M Hocking
- Department of Surgery, Vanderbilt University, Nashville, TN, USA.,Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Weifeng Luo
- Department of Surgery, Vanderbilt University, Nashville, TN, USA
| | - Daniel L Feldman
- Department of Surgery, Vanderbilt University, Nashville, TN, USA
| | - Jun Song
- Department of Surgery, Vanderbilt University, Nashville, TN, USA
| | - Padmini Komalavilas
- Department of Surgery, Vanderbilt University, Nashville, TN, USA.,VA Tennessee Valley Healthcare System, Nashville, TN, USA
| | | | - Colleen M Brophy
- Department of Surgery, Vanderbilt University, Nashville, TN, USA.,VA Tennessee Valley Healthcare System, Nashville, TN, USA
| |
Collapse
|
33
|
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
|
34
|
Winkler B, Reineke D, Heinisch PP, Schönhoff F, Huber C, Kadner A, Englberger L, Carrel T. Graft preservation solutions in cardiovascular surgery. Interact Cardiovasc Thorac Surg 2016; 23:300-9. [DOI: 10.1093/icvts/ivw056] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Accepted: 02/10/2016] [Indexed: 01/28/2023] Open
|