1
|
Zhou Z, Chen W, Cao Y, Abdi R, Tao W. Nanomedicine-based strategies for the treatment of vein graft disease. Nat Rev Cardiol 2025; 22:255-272. [PMID: 39501093 PMCID: PMC11925677 DOI: 10.1038/s41569-024-01094-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/27/2024] [Indexed: 01/03/2025]
Abstract
Autologous saphenous veins are the most frequently used conduits for coronary and peripheral artery bypass grafting. However, vein graft failure rates of 40-50% within 10 years of the implantation lead to poor long-term outcomes after bypass surgery. Currently, only a few therapeutic approaches for vein graft disease have been successfully translated into clinical practice. Building on the past two decades of advanced understanding of vein graft biology and the pathophysiological mechanisms underlying vein graft disease, nanomedicine-based strategies offer promising opportunities to address this important unmet clinical need. In this Review, we provide deep insight into the latest developments in the rational design and applications of nanoparticles that have the potential to target specific cells during various pathophysiological stages of vein graft disease, including early endothelial dysfunction, intermediate intimal hyperplasia and late-stage accelerated atherosclerosis. Additionally, we underscore the convergence of nanofabricated biomaterials, with a particular focus on hydrogels, external graft support devices and cell-based therapies, alongside bypass surgery to improve local delivery efficiency and therapeutic efficacy. Finally, we provide a specific discussion on the considerations, challenges and novel perspectives for the future clinical translation of nanomedicine for the treatment of vein graft disease.
Collapse
Affiliation(s)
- Zhuoming Zhou
- Center for Nanomedicine and Department of Anaesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Cardiac Surgery, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Wei Chen
- Center for Nanomedicine and Department of Anaesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Yihai Cao
- Department of Microbiology, Tumour and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Reza Abdi
- Transplantation Research Center and Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Wei Tao
- Center for Nanomedicine and Department of Anaesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
2
|
Sandner S, Antoniades C, Caliskan E, Czerny M, Dayan V, Fremes SE, Glineur D, Lawton JS, Thielmann M, Gaudino M. Intra-operative and post-operative management of conduits for coronary artery bypass grafting: a clinical consensus statement of the European Society of Cardiology Working Group on Cardiovascular Surgery and the European Association for Cardio-Thoracic Surgery Coronary Task Force. Eur Heart J 2025; 46:19-34. [PMID: 39412205 PMCID: PMC11695906 DOI: 10.1093/eurheartj/ehae654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2025] Open
Abstract
The structural and functional integrity of conduits used for coronary artery bypass grafting is critical for graft patency. Disruption of endothelial integrity and endothelial dysfunction are incurred during conduit harvesting subsequent to mechanical or thermal injury and during conduit storage prior to grafting, leading to acute thrombosis and early graft failure. Late graft failure, in particular that of vein grafts, is precipitated by progressive atherogenesis. Intra-operative management includes appropriate selection of conduit-specific harvesting techniques and storage solutions. Arterial grafts are prone to vasospasm subsequent to surgical manipulation, and application of intra-operative vasodilatory protocols is critical. Post-operative management includes continuation of oral vasodilator therapy and selection of antithrombotic and lipid-lowering agents to attenuate atherosclerotic disease progression in conduits. In this review, the scientific evidence underlying the key aspects of intra- and post-operative management of conduits for coronary artery bypass grafting is examined. Clinical consensus statements for best clinical practice are provided, and areas requiring further research are highlighted.
Collapse
Affiliation(s)
- Sigrid Sandner
- Department of Cardiac Surgery, Medical University of Vienna, Spitalgasse 23, 1090 Vienna, Austria
- Department of Cardiothoracic Surgery, Weill Cornell Medicine, 525 E 68th St, New York, NY 10065, USA
| | - Charalambos Antoniades
- Division of Cardiovascular Medicine, Radcliffe Department Medicine, University of Oxford, Oxford, UK
| | - Etem Caliskan
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité (DHZC), Berlin, Germany
| | - Martin Czerny
- Department of Cardiovascular Surgery, University Heart Center Freiburg—Bad Krozingen, Germany
- Faculty of Medicine, Albert Ludwigs University Freiburg, Freiburg, Germany
| | - Victor Dayan
- University Cardiovascular Center, National Institute of Cardiac Surgery, Montevideo, Uruguay
| | - Stephen E Fremes
- Schulich Heart Centre, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ontario, Canada
| | - David Glineur
- Division of Cardiac Surgery, Memorial University, St. John‘s, Newfoundland, Canada
- Department of Cardiac Surgery, University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - Jennifer S Lawton
- Department of Surgery, Division of Cardiac Surgery, Johns Hopkins University, Baltimore, MD, USA
| | - Matthias Thielmann
- Department of Thoracic and Cardiovascular Surgery, West-German Heart and Vascular Center, University of Duisburg-Essen, Essen, Germany
| | - Mario Gaudino
- Department of Cardiothoracic Surgery, Weill Cornell Medicine, 525 E 68th St, New York, NY 10065, USA
| |
Collapse
|
3
|
Spadaccio C, Nenna A, Corrado D, Glenn C, Panza A, Vester R, Laskawski G, Rose D, Louis L. Use of Indocyanine Green (ICG) to Assess Myocardial Perfusion and Territorial Distribution of Vein Grafts Implanted on Coronary Arteries in an Ex-vivo Porcine Model. A Potential Adjunct to Assist Revascularization Strategies and Training in Coronary Artery Bypass Grafting. Rev Cardiovasc Med 2025; 26:25778. [PMID: 39867206 PMCID: PMC11759963 DOI: 10.31083/rcm25778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Revised: 11/04/2024] [Accepted: 11/14/2024] [Indexed: 01/28/2025] Open
Abstract
Background The fluorescent dye indocyanine green (ICG) has been used to identify anatomical structures intraoperatively in coronary artery bypass grafting (CABG). This study aimed to evaluate the feasibility of using ICG to assess graft patency and territorial distribution of myocardial reperfusion during CABG. Methods Porcine arrested hearts (n = 18) were used to evaluate territorial distribution of native coronary arteries and of a coronary bypass constructed with porcine saphenous vein graft (SVG) using ICG. Coronary ostia were dissected and selectively cannulated for ICG injection. Sequential fluorescence was assessed in the epicardial coronary arteries, myocardium and coronary veins using an infrared-sensitive charge-coupled device (CCD) camera system. In a separate set of experiments, SVG was used for anastomosis in end-to-side fashion to a terminal obtuse marginal (OM) branch. This approach was used to avoid bias in the assessment of territorial distribution. The anastomosis was injected with ICG; graft patency and territorial distribution was assessed using an infrared-sensitive CCD camera system from 30 cm above the field, as previously described. Native circulation and SVG grafts were assessed using real-time video recording and fluorescence intensity mapping that was averaged into a graded scoring system. The heart was divided into functional regions: anterior wall, lateral wall, inferior wall and right ventricle. All experiments were performed in triplicates. Results After ICG injection into the individual coronary ostia, perfusion of the native coronary artery was visible. Portions of the vessels embedded into the epicardial fat could be easily visualized on the surface of the heart and the dissection facilitated via fluorescence guidance. The territorial distribution reflected the expected regional perfusion. The SVG graft was anastomosed to an OM branch. ICG visualization allowed for assessment of graft patency excluding potential technical anastomosis problems or graft twisting or dissection. The myocardial perfusion observed in real-time confirmed regional distribution to the entire lateral wall and minimally to the inferior wall. These findings were confirmed in all the specimens used in the study. Conclusions Besides assisting the identification of intramyocardial vessels, ICG can provide information on the native coronary circulation status and the territorial distribution of the perfusion before and after grafting. It enables visualization of collaterals and the territory of distribution subtended by a graft offering real-time assessment and guidance on the grafting strategy.
Collapse
Affiliation(s)
- Cristiano Spadaccio
- Cardiac Surgery, University of Cincinnati Medical Center, Cincinnati, OH 45202, USA
| | - Antonio Nenna
- Cardiac Surgery, Università Campus Bio-Medico di Roma, 00128 Rome, Italy
| | - Diletta Corrado
- Cardiac Surgery, Università Campus Bio-Medico di Roma, 00128 Rome, Italy
| | - Carter Glenn
- Cardiac Surgery, University of Cincinnati Medical Center, Cincinnati, OH 45202, USA
| | - Antonio Panza
- Cardiac Surgery, University of Cincinnati Medical Center, Cincinnati, OH 45202, USA
| | - Russell Vester
- Cardiac Surgery, University of Cincinnati Medical Center, Cincinnati, OH 45202, USA
| | - Grzegorz Laskawski
- Cardiac Surgery, Blackpool Teaching Hospital - Lancashire Cardiac Center, FY3 8NP Blackpool, UK
| | - David Rose
- Cardiac Surgery, Blackpool Teaching Hospital - Lancashire Cardiac Center, FY3 8NP Blackpool, UK
| | - Louis Louis
- Cardiac Surgery, University of Cincinnati Medical Center, Cincinnati, OH 45202, USA
| |
Collapse
|
4
|
Sandner S, Antoniades C, Caliskan E, Czerny M, Dayan V, Fremes SE, Glineur D, Lawton JS, Thielmann M, Gaudino M. Intra-operative and post-operative management of conduits for coronary artery bypass grafting: a clinical consensus statement of the European Society of Cardiology Working Group on Cardiovascular Surgery and the European Association for Cardio-Thoracic Surgery Coronary Task Force. Eur J Cardiothorac Surg 2024; 66:ezae400. [PMID: 39656609 DOI: 10.1093/ejcts/ezae400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Indexed: 12/17/2024] Open
Abstract
The structural and functional integrity of conduits used for coronary artery bypass grafting is critical for graft patency. Disruption of endothelial integrity and endothelial dysfunction are incurred during conduit harvesting subsequent to mechanical or thermal injury and during conduit storage prior to grafting, leading to acute thrombosis and early graft failure. Late graft failure, in particular that of vein grafts, is precipitated by progressive atherogenesis. Intra-operative management includes appropriate selection of conduit-specific harvesting techniques and storage solutions. Arterial grafts are prone to vasospasm subsequent to surgical manipulation, and application of intra-operative vasodilatory protocols is critical. Post-operative management includes continuation of oral vasodilator therapy and selection of antithrombotic and lipid-lowering agents to attenuate atherosclerotic disease progression in conduits. In this review, the scientific evidence underlying the key aspects of intra- and post-operative management of conduits for coronary artery bypass grafting is examined. Clinical consensus statements for best clinical practice are provided, and areas requiring further research are highlighted.
Collapse
Affiliation(s)
- Sigrid Sandner
- Department of Cardiac Surgery, Medical University of Vienna, Spitalgasse 23, 1090 Vienna, Austria
- Department of Cardiothoracic Surgery, Weill Cornell Medicine, 525 E 68th St, New York, NY 10065, USA
| | - Charalambos Antoniades
- Division of Cardiovascular Medicine, Radcliffe Department Medicine, University of Oxford, Oxford, UK
| | - Etem Caliskan
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité (DHZC), Berlin, Germany
| | - Martin Czerny
- Department of Cardiovascular Surgery, University Heart Center Freiburg-Bad Krozingen, Germany
- Faculty of Medicine, Albert Ludwigs University Freiburg, Freiburg, Germany
| | - Victor Dayan
- University Cardiovascular Center, National Institute of Cardiac Surgery, Montevideo, Uruguay
| | - Stephen E Fremes
- Schulich Heart Centre, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ontario, Canada
| | - David Glineur
- Division of Cardiac Surgery, Memorial University, St John's, Newfoundland, Canada
- Department of Cardiac Surgery, University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - Jennifer S Lawton
- Department of Surgery, Division of Cardiac Surgery, Johns Hopkins University, Baltimore, MD, USA
| | - Matthias Thielmann
- Department of Thoracic and Cardiovascular Surgery, West-German Heart and Vascular Center, University of Duisburg-Essen, Essen, Germany
| | - Mario Gaudino
- Department of Cardiothoracic Surgery, Weill Cornell Medicine, 525 E 68th St, New York, NY 10065, USA
| |
Collapse
|
5
|
Vahedi SM, Azaiez J. Optimization of polymer coating thickness and strut width in drug Eluting stents using Magnetic field. Eur J Pharm Biopharm 2024; 203:114455. [PMID: 39147171 DOI: 10.1016/j.ejpb.2024.114455] [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: 06/06/2024] [Revised: 07/28/2024] [Accepted: 08/12/2024] [Indexed: 08/17/2024]
Abstract
The transport of drug/magnetic particle (MP) conjugates in the presence of a Magnetic Field (MF) in Drug Eluting Stents (DESs) is modeled numerically using the Finite Volume Method (FVM). The effects of physiological conditions corresponding to different degrees of calcification, drug particles sizes and hematocrit level, were analyzed by investigating the roles of the tissue permeability, its anisotropy and the plasma viscosity. It was found that both in the absence and presence of the MF, as the tissue permeability decreases or the plasma viscosity increases, the free-phase drug and Extracellular Matrix (ECM)-bound phase contents increase. Stronger tissue anisotropy leads to a decrease of the free-phase drug content and an increase of the ECM-bound phase content. Within the explored ranges, the Specific Receptor (SR)-bound phase of the drug was found to be insensitive to the tissue permeability and plasma viscosity, and to be larger in anisotropic tissues. The activation of the MF leads systematically to larger free-phase drug contents, with the increases most prominent at smaller tissue permeability, anisotropy and plasma viscosity. On the other hand, the effects on the ECM-bound phase content are found to be stronger at larger permeability, smaller plasma viscosity and lower tissue anisotropy. For an isotropic tissue, the MF induces a decrease of the ECM-bound phase content at early times, followed by an increase at later times. For the considered ranges of permeability and viscosity, the MF does not seem to have any noticeable effects on the SR-bound phase. However, this phase of the drug tends to increase with the activation of the MF in isotropic tissues and is unchanged in anisotropic ones. These reported effects of the MF hold promise for alleviating two factors contributing to In-Stent Restenosis, namely the polymer coating width and thickness. The study reveals that a narrower or thinner polymer layer, in combination with the MF, can mimic the drug release dynamics of a wider or thicker polymer layer in the absence of the MF. The corresponding width and thickness of the magnetized stents, that we referred to as the equivalent polymer width (EPW) and equivalent polymer thickness (EPT) were determined and their dependence on the tissue permeability, isotropy and the plasma viscosity, was investigated. The study shows that it is possible to achieve the same drug delivery with polymer coating of half the width or half the thickness of the non-magnetized stent when an electric intensity of 3A is used.
Collapse
Affiliation(s)
- Seyed Masoud Vahedi
- Department of Chemical and Petroleum Engineering, University of Calgary, Calgary, Alberta, Canada.
| | - Jalel Azaiez
- Department of Chemical and Petroleum Engineering, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
6
|
Madiyeva MI, Aripov MA, Goncharov AY, Zholdasbekova RY. Outcomes of myocardial revascularization in patients with obesity and multivessel coronary artery disease. Egypt Heart J 2024; 76:114. [PMID: 39198363 PMCID: PMC11358579 DOI: 10.1186/s43044-024-00548-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Accepted: 08/22/2024] [Indexed: 09/01/2024] Open
Abstract
BACKGROUND There is uncertainty regarding the best revascularization approach-whether coronary artery bypass grafting (CABG) or percutaneous coronary intervention (PCI)-for obese patients suffering from multi-vessel coronary artery disease. RESULTS 406 patients with low and intermediate SYNTAX scores (SS) underwent PCI with drug-eluting stents (n = 200, 100 with SS ≤ 22, and 100 with SS 23-32) and CABG (n = 206, 100 with SS ≤ 22, and 106 with SS 23-32). Patients were also categorized by body mass index (BMI): normal weight (12%, 48 patients), overweight (41.6%, 169 patients), and obese (46.6%, 189 patients). The follow-up period averaged 9 ± 1.9 years. The endpoints of the study were as follows: major adverse cardiac and cerebrovascular events, a repeat revascularization, diminished left ventricular ejection fraction (LVEF), and high SS (≥ 33) observed over time. When comparing PCI and CABG in overweight individuals, the risk of myocardial infarction (MI) following PCI was greater than after CABG (Hazard Ratio [HR] 2.7, 95% Confidence Interval [CI] 1.1-6.7, p = 0.03). In patients with overweight and Class I obesity, CABG was associated with the risk of coronary atherosclerosis progression (SS ≥ 33) (HR 4.4, 95% CI 1.5-13, p = 0.009 and HR 4.9, 95% CI 1.9-12, p = 0.001, respectively); whereas PCI was connected with the likelihood of repeat revascularization (HR 2.7, 95% CI 1.6-4.55, p < 0.0001 and HR 2, 95% CI 1.3-3.1, p = 0.002, respectively). At the same time, for stented patients, Class III obesity was associated with the risk of repeat revascularization (HR 2.5, 95% CI 1.02-6, p = 0.044). CONCLUSION There were no significant weight-related impacts on long-term outcomes among patients who underwent surgery. Whereas in stented patients, Class III obesity was associated with the risk of repeat revascularization. When comparing PCI and CABG, for overweight and Class I obesity patients, CABG was associated with a likelihood of coronary atherosclerosis progression (SS ≥ 33), while PCI was linked to the risk of repeat revascularization. For overweight patients, CABG outperformed PCI in terms of the risk of MI. For other adverse events in patients of different weight categories, PCI and CABG did not reveal any significant benefits.
Collapse
Affiliation(s)
- Maira I Madiyeva
- Department of Emergency Cardiology, Pavlodar Regional Cardiology Center, 10/3 Tkacheva St, Pavlodar, 140000, Kazakhstan.
- Department of Therapy, Semey Medical University, Semey, Kazakhstan.
| | - Marat A Aripov
- Department of Interventional Cardiology, National Research Cardiac Surgery Center, Astana, Kazakhstan
| | - Alexey Y Goncharov
- Department of Interventional Cardiology, National Research Cardiac Surgery Center, Astana, Kazakhstan
| | - Rakhima Y Zholdasbekova
- Department of Emergency Cardiology, Pavlodar Regional Cardiology Center, 10/3 Tkacheva St, Pavlodar, 140000, Kazakhstan
| |
Collapse
|
7
|
Jackson ML, Bond AR, Ascione R, Johnson JL, George SJ. FGL2/FcγRIIB Signalling Mediates Arterial Shear Stress-Mediated Endothelial Cell Apoptosis: Implications for Coronary Artery Bypass Vein Graft Pathogenesis. Int J Mol Sci 2024; 25:7638. [PMID: 39062880 PMCID: PMC11277082 DOI: 10.3390/ijms25147638] [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: 04/09/2024] [Revised: 07/05/2024] [Accepted: 07/10/2024] [Indexed: 07/28/2024] Open
Abstract
The sudden exposure of venous endothelial cells (vECs) to arterial fluid shear stress (FSS) is thought to be a major contributor to coronary artery bypass vein graft failure (VGF). However, the effects of arterial FSS on the vEC secretome are poorly characterised. We propose that analysis of the vEC secretome may reveal potential therapeutic approaches to suppress VGF. Human umbilical vein endothelial cells (HUVECs) pre-conditioned to venous FSS (18 h; 1.5 dynes/cm2) were exposed to venous or arterial FSS (15 dynes/cm2) for 24 h. Tandem Mass Tagging proteomic analysis of the vEC secretome identified significantly increased fibroleukin (FGL2) in conditioned media from HUVECs exposed to arterial FSS. This increase was validated by Western blotting. Application of the NFκB inhibitor BAY 11-7085 (1 µM) following pre-conditioning reduced FGL2 release from vECs exposed to arterial FSS. Exposure of vECs to arterial FSS increased apoptosis, measured by active cleaved caspase-3 (CC3) immunocytochemistry, which was likewise elevated in HUVECs treated with recombinant FGL2 (20 ng/mL) for 24 h under static conditions. To determine the mechanism of FGL2-induced apoptosis, HUVECs were pre-treated with a blocking antibody to FcγRIIB, a receptor FGL2 is proposed to interact with, which reduced CC3 levels. In conclusion, our findings indicate that the exposure of vECs to arterial FSS results in increased release of FGL2 via NFκB signalling, which promotes endothelial apoptosis via FcγRIIB signalling. Therefore, the inhibition of FGL2/FcγRIIB signalling may provide a novel approach to reduce arterial FSS-induced vEC apoptosis in vein grafts and suppress VGF.
Collapse
Affiliation(s)
| | | | | | | | - Sarah J. George
- Translational Health Sciences, Bristol Medical School, Faculty of Health and Life Sciences, University of Bristol, Bristol BS2 8HW, UK; (M.L.J.); (A.R.B.); (R.A.); (J.L.J.)
| |
Collapse
|
8
|
Zong Q, He C, Long B, Huang Q, Chen Y, Li Y, Dang Y, Cai C. Targeted Delivery of Nanoparticles to Blood Vessels for the Treatment of Atherosclerosis. Biomedicines 2024; 12:1504. [PMID: 39062077 PMCID: PMC11275173 DOI: 10.3390/biomedicines12071504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 06/21/2024] [Accepted: 07/04/2024] [Indexed: 07/28/2024] Open
Abstract
Atherosclerosis is a common form of cardiovascular disease, which is one of the most prevalent causes of death worldwide, particularly among older individuals. Surgery is the mainstay of treatment for severe stenotic lesions, though the rate of restenosis remains relatively high. Current medication therapy for atherosclerosis has limited efficacy in reversing the formation of atherosclerotic plaques. The search for new drug treatment options is imminent. Some potent medications have shown surprising therapeutic benefits in inhibiting inflammation and endothelial proliferation in plaques. Unfortunately, their use is restricted due to notable dose-dependent systemic side effects or degradation. Nevertheless, with advances in nanotechnology, an increasing number of nano-related medical applications are emerging, such as nano-drug delivery, nano-imaging, nanorobots, and so forth, which allow for restrictions on the use of novel atherosclerotic drugs to be lifted. This paper reviews new perspectives on the targeted delivery of nanoparticles to blood vessels for the treatment of atherosclerosis in both systemic and local drug delivery. In systemic drug delivery, nanoparticles inhibit drug degradation and reduce systemic toxicity through passive and active pathways. To further enhance the precise release of drugs, the localized delivery of nanoparticles can also be accomplished through blood vessel wall injection or using endovascular interventional devices coated with nanoparticles. Overall, nanotechnology holds boundless potential for the diagnosis and treatment of atherosclerotic diseases in the future.
Collapse
Affiliation(s)
- Qiushuo Zong
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (Q.Z.); (Y.C.); (Y.L.)
| | - Chengyi He
- Department of Vascular Surgery, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China;
| | - Binbin Long
- Department of General Surgery, Taihe Hospital Affiliated to Hubei University of Medicine, Shiyan 442099, China;
| | - Qingyun Huang
- Department of Cardiothoracic Surgery, The First Hospital of Putian Affiliated to Fujian Medical University, Putian 351106, China;
| | - Yunfei Chen
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (Q.Z.); (Y.C.); (Y.L.)
| | - Yiqing Li
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (Q.Z.); (Y.C.); (Y.L.)
| | - Yiping Dang
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (Q.Z.); (Y.C.); (Y.L.)
| | - Chuanqi Cai
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (Q.Z.); (Y.C.); (Y.L.)
| |
Collapse
|
9
|
Xu Z, Mou C, Ji R, Chen H, Ding Y, Jiang X, Meng F, He F, Luo B, Yu J. Alterations in metabolome and lipidome in patients with in-stent restenosis. CNS Neurosci Ther 2024; 30:e14832. [PMID: 39009504 PMCID: PMC11249805 DOI: 10.1111/cns.14832] [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: 02/24/2024] [Revised: 05/23/2024] [Accepted: 06/20/2024] [Indexed: 07/17/2024] Open
Abstract
CONTEXT In-stent restenosis (ISR) can lead to blood flow obstruction, insufficient blood supply to the brain, and may even result in serious complications such as stroke. Endothelial cell hyperproliferation and thrombosis are the primary etiologies, frequently resulting in alterations in intravascular metabolism. However, the metabolic changes related to this process are still undermined. OBJECTIVE We tried to characterize the serum metabolome of patients with ISR and those with non-restenosis (NR) using metabolomics and lipidomics, exploring the key metabolic pathways of this pathological phenomenon. RESULTS We observed that the cysteine and methionine pathways, which are associated with cell growth and oxidative homeostasis, showed the greatest increase in the ISR group compared to the NR group. Within this pathway, the levels of N-formyl-l-methionine and L-methionine significantly increased in the ISR group, along with elevated levels of downstream metabolites such as 2-ketobutyric acid, pyruvate, and taurocholate. Additionally, an increase in phosphatidylcholine (PC) and phosphatidylserine (PS), as well as a decrease in triacylglycerol in the ISR group, indicated active lipid metabolism in these patients, which could be a significant factor contributing to the recurrence of blood clots after stent placement. Importantly, phenol sulfate and PS(38:4) were identified as potential biomarkers for distinguishing ISR, with an area under the curve of more than 0.85. CONCLUSIONS Our study revealed significant metabolic alterations in patients with ISR, particularly in the cysteine and methionine pathways, with phenol sulfate and PS(38:4) showing promise for ISR identification.
Collapse
Affiliation(s)
- Ziqi Xu
- Department of Neurology, First Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouChina
| | - Chenye Mou
- Department of Neurology, First Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouChina
| | - Renjie Ji
- Department of Neurology, First Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouChina
| | - Hanfen Chen
- Department of Neurology, First Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouChina
| | - Yuge Ding
- Department of Neurology, First Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouChina
| | - Xiaoyi Jiang
- Department of Neurology, First Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouChina
| | - Fanxia Meng
- Department of Neurology, First Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouChina
| | - Fangping He
- Department of Neurology, First Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouChina
| | - Benyan Luo
- Department of Neurology, First Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouChina
| | - Jie Yu
- Department of Neurology, First Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouChina
| |
Collapse
|
10
|
Saleem MS, Samad S, Ahmed SS, Mehmood N. Optimizing PCSK9 Inhibitor Integration for Cardiovascular Disease Management in Pakistan. CLINICAL MEDICINE INSIGHTS-CARDIOLOGY 2024; 18:11795468241262648. [PMID: 38911696 PMCID: PMC11191606 DOI: 10.1177/11795468241262648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Accepted: 05/29/2024] [Indexed: 06/25/2024]
Abstract
Cardiovascular illnesses (CVDs), particularly Coronary Artery Disease (CAD) and Ischemic Heart Disease (IHD), are major global health burdens, with a growing incidence in Pakistan. The development of PCSK9 inhibitors offers encouraging advantages in lowering LDL cholesterol and lowering cardiovascular risk, even though conservative treatments are still essential. However, access to them is severely hampered by their high cost, especially in environments with little resources. The financial limitations and scarcity of healthcare resources while examining the difficulties in obtaining PCSK9 inhibitors in Pakistan is essential. In order to develop solutions for affordability and fair access, it emphasizes the urgent need for multi-stakeholder collaboration, including governmental action, healthcare sector involvement, and pharmaceutical company engagement. It also emphasizes the need for data-specific research and the use of PCSK9 inhibitors in conventional treatment protocols.
Collapse
Affiliation(s)
| | | | | | - Nadia Mehmood
- Karachi Medical and Dental College, Karachi, Pakistan
| |
Collapse
|
11
|
Yuki H, Sundt TM, Niida T, Suzuki K, Kinoshita D, Fujimoto D, Dey D, Lee H, McNulty I, Naganuma T, Nakamura S, Usui E, Kakuta T, Jang I. Level of Perivascular Inflammation Is Significantly Lower Around the Left Internal Mammary Artery Than Around Native Coronary Arteries. J Am Heart Assoc 2024; 13:e033224. [PMID: 38879462 PMCID: PMC11255748 DOI: 10.1161/jaha.123.033224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Accepted: 05/16/2024] [Indexed: 06/19/2024]
Abstract
BACKGROUND The left internal mammary artery (LIMA) is protected from developing atherosclerosis. Perivascular inflammation, which is closely associated with atherosclerosis, can be measured by perivascular adipose tissue attenuation on computed tomography angiography. Whether the absence of atherosclerosis in LIMA is related to the lower level of perivascular inflammation is unknown. This study was performed to compare the level of perivascular inflammation between LIMA in situ and native coronary arteries in patients with coronary artery disease. METHODS AND RESULTS A total of 573 patients who underwent both computed tomography angiography and optical coherence tomography imaging were included. The level of perivascular adipose tissue attenuation between LIMA in situ and coronary arteries was compared. Perivascular adipose tissue attenuation around LIMA in situ was significantly lower around the 3 coronary arteries (-82.9 [-87.3 to -78.0] versus -70.8 [-75.9 to -65.9]; P<0.001), irrespective of the level of pericoronary inflammation or the number of vulnerable features on optical coherence tomography. When patients were divided into high and low pericoronary inflammation groups, those in the high inflammation group had more target vessel failure (hazard ratio, 2.97 [95% CI, 1.16-7.59]; P=0.017). CONCLUSIONS The current study demonstrated that perivascular adipose tissue attenuation was significantly lower around LIMA in situ than around native coronary arteries. The lower level of perivascular inflammation may be related to the low prevalence of atherosclerosis in LIMA. REGISTRATION URL: https://www.clinicaltrials.gov; Unique Identifier: NCT04523194.
Collapse
Affiliation(s)
- Haruhito Yuki
- Cardiology DivisionMassachusetts General Hospital, Harvard Medical SchoolBostonMA
| | - Thoralf M. Sundt
- Cardiac Surgery Division, Department of SurgeryMassachusetts General Hospital, Harvard Medical SchoolBostonMA
| | - Takayuki Niida
- Cardiology DivisionMassachusetts General Hospital, Harvard Medical SchoolBostonMA
| | - Keishi Suzuki
- Cardiology DivisionMassachusetts General Hospital, Harvard Medical SchoolBostonMA
| | - Daisuke Kinoshita
- Cardiology DivisionMassachusetts General Hospital, Harvard Medical SchoolBostonMA
| | - Daichi Fujimoto
- Cardiology DivisionMassachusetts General Hospital, Harvard Medical SchoolBostonMA
| | - Damini Dey
- Biomedical Imaging Research Institute, Cedars‐Sinai Medical CenterLos AngelesCA
| | - Hang Lee
- Biostatistics CenterMassachusetts General Hospital, Harvard Medical SchoolBostonMA
| | - Iris McNulty
- Cardiology DivisionMassachusetts General Hospital, Harvard Medical SchoolBostonMA
| | - Toru Naganuma
- Interventional Cardiology UnitNew Tokyo HospitalChibaJapan
| | - Sunao Nakamura
- Interventional Cardiology UnitNew Tokyo HospitalChibaJapan
| | - Eisuke Usui
- Department of CardiologyTsuchiura Kyodo General HospitalTsuchiuraJapan
| | - Tsunekazu Kakuta
- Department of CardiologyTsuchiura Kyodo General HospitalTsuchiuraJapan
| | - Ik‐Kyung Jang
- Cardiology DivisionMassachusetts General Hospital, Harvard Medical SchoolBostonMA
| |
Collapse
|
12
|
Wang X, Li K, Yuan Y, Zhang N, Zou Z, Wang Y, Yan S, Li X, Zhao P, Li Q. Nonlinear Elasticity of Blood Vessels and Vascular Grafts. ACS Biomater Sci Eng 2024; 10:3631-3654. [PMID: 38815169 DOI: 10.1021/acsbiomaterials.4c00326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/01/2024]
Abstract
The transplantation of vascular grafts has emerged as a prevailing approach to address vascular disorders. However, the development of small-diameter vascular grafts is still in progress, as they serve in a more complicated mechanical environment than their counterparts with larger diameters. The biocompatibility and functional characteristics of small-diameter vascular grafts have been well developed; however, mismatch in mechanical properties between the vascular grafts and native arteries has not been accomplished, which might facilitate the long-term patency of small-diameter vascular grafts. From a point of view in mechanics, mimicking the nonlinear elastic mechanical behavior exhibited by natural blood vessels might be the state-of-the-art in designing vascular grafts. This review centers on elucidating the nonlinear elastic behavior of natural blood vessels and vascular grafts. The biological functionality and limitations associated with as-reported vascular grafts are meticulously reviewed and the future trajectory for fabricating biomimetic small-diameter grafts is discussed. This review might provide a different insight from the traditional design and fabrication of artificial vascular grafts.
Collapse
Affiliation(s)
- Xiaofeng Wang
- School of Mechanics and Safety Engineering, National Center for International Research of Micro-nano Molding Technology, Zhengzhou University, Zhengzhou 450001, China
- The State Key Laboratory of Fluid Power and Mechatronic Systems, Zhejiang University, Hangzhou 310027, China
| | - Kecheng Li
- School of Mechanics and Safety Engineering, National Center for International Research of Micro-nano Molding Technology, Zhengzhou University, Zhengzhou 450001, China
| | - Yuan Yuan
- School of Mechanics and Safety Engineering, National Center for International Research of Micro-nano Molding Technology, Zhengzhou University, Zhengzhou 450001, China
| | - Ning Zhang
- School of Mechanics and Safety Engineering, National Center for International Research of Micro-nano Molding Technology, Zhengzhou University, Zhengzhou 450001, China
| | - Zifan Zou
- School of Mechanics and Safety Engineering, National Center for International Research of Micro-nano Molding Technology, Zhengzhou University, Zhengzhou 450001, China
| | - Yun Wang
- School of Mechanics and Safety Engineering, National Center for International Research of Micro-nano Molding Technology, Zhengzhou University, Zhengzhou 450001, China
| | - Shujie Yan
- School of Mechanics and Safety Engineering, National Center for International Research of Micro-nano Molding Technology, Zhengzhou University, Zhengzhou 450001, China
| | - Xiaomeng Li
- School of Mechanics and Safety Engineering, National Center for International Research of Micro-nano Molding Technology, Zhengzhou University, Zhengzhou 450001, China
| | - Peng Zhao
- The State Key Laboratory of Fluid Power and Mechatronic Systems, Zhejiang University, Hangzhou 310027, China
| | - Qian Li
- School of Mechanics and Safety Engineering, National Center for International Research of Micro-nano Molding Technology, Zhengzhou University, Zhengzhou 450001, China
| |
Collapse
|
13
|
Szafron JM, Heng EE, Boyd J, Humphrey JD, Marsden AL. Hemodynamics and Wall Mechanics of Vascular Graft Failure. Arterioscler Thromb Vasc Biol 2024; 44:1065-1085. [PMID: 38572650 PMCID: PMC11043008 DOI: 10.1161/atvbaha.123.318239] [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: 09/04/2023] [Accepted: 03/12/2024] [Indexed: 04/05/2024]
Abstract
Blood vessels are subjected to complex biomechanical loads, primarily from pressure-driven blood flow. Abnormal loading associated with vascular grafts, arising from altered hemodynamics or wall mechanics, can cause acute and progressive vascular failure and end-organ dysfunction. Perturbations to mechanobiological stimuli experienced by vascular cells contribute to remodeling of the vascular wall via activation of mechanosensitive signaling pathways and subsequent changes in gene expression and associated turnover of cells and extracellular matrix. In this review, we outline experimental and computational tools used to quantify metrics of biomechanical loading in vascular grafts and highlight those that show potential in predicting graft failure for diverse disease contexts. We include metrics derived from both fluid and solid mechanics that drive feedback loops between mechanobiological processes and changes in the biomechanical state that govern the natural history of vascular grafts. As illustrative examples, we consider application-specific coronary artery bypass grafts, peripheral vascular grafts, and tissue-engineered vascular grafts for congenital heart surgery as each of these involves unique circulatory environments, loading magnitudes, and graft materials.
Collapse
Affiliation(s)
- Jason M Szafron
- Departments of Pediatrics (J.M.S., A.L.M.), Stanford University, CA
| | - Elbert E Heng
- Cardiothoracic Surgery (E.E.H., J.B.), Stanford University, CA
| | - Jack Boyd
- Cardiothoracic Surgery (E.E.H., J.B.), Stanford University, CA
| | - Jay D Humphrey
- Department of Biomedical Engineering, Yale University, New Haven, CT (J.D.H.)
| | | |
Collapse
|
14
|
Sun X, Wu J, Zhang X, Xie C, Wei H, Li P, Yang Y, Yuan H, Cai J, Xiao Q, Cheng J, Xu Q. Atlas of Cell Repertoire Within Neointimal Lesions Is Metabolically Altered in Hypertensive Rats. Hypertension 2024; 81:787-800. [PMID: 38240164 DOI: 10.1161/hypertensionaha.123.22057] [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: 09/14/2023] [Accepted: 01/09/2024] [Indexed: 02/22/2024]
Abstract
BACKGROUND High blood pressure has been suggested to accelerate vascular injury-induced neointimal formation and progression. However, little is known about the intricate relationships between vascular injury and hypertension in the context of arterial remodeling. METHODS Single-cell RNA-sequencing analysis was used to depict the cell atlas of carotid arteries of Wistar Kyoto rats and spontaneously hypertensive rats with or without balloon injury. RESULTS We found that hypertension significantly aggravated balloon injury-induced arterial stenosis. A total of 36 202 cells from carotid arteries with or without balloon injury were included in single-cell RNA-sequencing analysis. Cell composition analysis showed that vascular injury and hypertension independently induced distinct aortic cell phenotypic alterations including immune cells, endothelial cells (ECs), and smooth muscle cells. Specifically, our data showed that injury and hypertension-induced specific EC phenotypic alterations, and revealed a transition from functional ECs to hypermetabolic, and eventually dysfunctional ECs in hypertensive rats upon balloon injury. Importantly, our data also showed that vascular injury and hypertension-induced different smooth muscle cell phenotypic alterations, characterized by deferential expression of synthetic signatures. Interestingly, pathway analysis showed that dysregulated metabolic pathways were a common feature in monocytes/macrophages, ECs, and smooth muscle cells in response to injury and hypertension. Functionally, we demonstrate that inhibition of mitochondrial respiration significantly ameliorated injury-induced neointimal formation in spontaneously hypertensive rats. CONCLUSIONS This study provides the cell landscape changes of the main aortic cell phenotypic alterations in response to injury and hypertension. Our findings suggest that targeting cellular mitochondrial respiration could be a novel therapeutic for patients with hypertension undergoing vascular angioplasty.
Collapse
Affiliation(s)
- Xiaolei Sun
- Department of General Surgery (Vascular Surgery), Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, The Affiliated Hospital of Southwest Medical University, Luzhou, China (X.S., H.W.)
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Public Center of Experimental Technology, Southwest Medical University, Luzhou, China (X.S., X.Z., C.X., P.L., Y.Y., J. Cheng, Q. Xu)
| | - Junru Wu
- Department of Cardiology and Center of Pharmacology, Postdoctoral Station of Clinical Medicine, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China (J.W., H.Y., J. Cai)
| | - Xiaolin Zhang
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Public Center of Experimental Technology, Southwest Medical University, Luzhou, China (X.S., X.Z., C.X., P.L., Y.Y., J. Cheng, Q. Xu)
| | - Cheng Xie
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Public Center of Experimental Technology, Southwest Medical University, Luzhou, China (X.S., X.Z., C.X., P.L., Y.Y., J. Cheng, Q. Xu)
| | - Haijun Wei
- Department of General Surgery (Vascular Surgery), Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, The Affiliated Hospital of Southwest Medical University, Luzhou, China (X.S., H.W.)
| | - Pengyun Li
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Public Center of Experimental Technology, Southwest Medical University, Luzhou, China (X.S., X.Z., C.X., P.L., Y.Y., J. Cheng, Q. Xu)
| | - Yan Yang
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Public Center of Experimental Technology, Southwest Medical University, Luzhou, China (X.S., X.Z., C.X., P.L., Y.Y., J. Cheng, Q. Xu)
| | - Hong Yuan
- Department of Cardiology and Center of Pharmacology, Postdoctoral Station of Clinical Medicine, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China (J.W., H.Y., J. Cai)
| | - Jingjing Cai
- Department of Cardiology and Center of Pharmacology, Postdoctoral Station of Clinical Medicine, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China (J.W., H.Y., J. Cai)
| | - Qingzhong Xiao
- Centre for Clinical Pharmacology and Precision Medicine, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, United Kingdom (Q. Xiao, Q. Xu)
| | - Jun Cheng
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Public Center of Experimental Technology, Southwest Medical University, Luzhou, China (X.S., X.Z., C.X., P.L., Y.Y., J. Cheng, Q. Xu)
| | - Qingbo Xu
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Public Center of Experimental Technology, Southwest Medical University, Luzhou, China (X.S., X.Z., C.X., P.L., Y.Y., J. Cheng, Q. Xu)
- Centre for Clinical Pharmacology and Precision Medicine, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, United Kingdom (Q. Xiao, Q. Xu)
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China (Q. Xu)
| |
Collapse
|
15
|
Peng K, Wang M, Wang J, Wang Q, Li D, Sun X, Yang Y, Yang D. Nuclear receptor subfamily 1 group D member 1 suppresses the proliferation, migration of adventitial fibroblasts, and vascular intimal hyperplasia via mammalian target of rapamycin complex 1/β-catenin pathway. Clin Exp Hypertens 2023; 45:2178659. [PMID: 36794491 DOI: 10.1080/10641963.2023.2178659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2023]
Abstract
BACKGROUND In-stent restenosis hardly limits the therapeutic effect of the percutaneous vascular intervention. Although the restenosis is significantly ameliorated after the application of new drug-eluting stents, the incidence of restenosis remains at a high level. OBJECTIVE Vascular adventitial fibroblasts (AFs) play an important role in intimal hyperplasia and subsequent restenosis. The current study was aimed to investigate the role of nuclear receptor subfamily 1, group D, member 1 (NR1D1) in the vascular intimal hyperplasia. METHODS AND RESULTS We observed increased expression of NR1D1 after the transduction of adenovirus carrying Nr1d1 gene (Ad-Nr1d1) in AFs. Ad-Nr1d1 transduction significantly reduced the numbers of total AFs, Ki-67-positive AFs, and the migration rate of AFs. NR1D1 overexpression decreased the expression level of β-catenin and attenuated the phosphorylation of the effectors of mammalian target of rapamycin complex 1 (mTORC1), including mammalian target of rapamycin (mTOR) and 4E binding protein 1 (4EBP1). Restoration of β-catenin by SKL2001 abolished the inhibitory effects of NR1D1 overexpression on the proliferation and migration of AFs. Surprisingly, the restoration of mTORC1 activity by insulin could also reverse the decreased expression of β-catenin, attenuated proliferation, and migration in AFs induced by NR1D1 overexpression. In vivo, we found that SR9009 (an agonist of NR1D1) ameliorated the intimal hyperplasia at days 28 after injury of carotid artery. We further observed that SR9009 attenuated the increased Ki-67-positive AFs, an essential part of vascular restenosis at days 7 after injury to the carotid artery. CONCLUSION These data suggest that NR1D1 inhibits intimal hyperplasia by suppressing the proliferation and migration of AFs in a mTORC1/β-catenin-dependent manner.
Collapse
Affiliation(s)
- Ke Peng
- School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, China.,Department of Cardiovascular Medicine, General Hospital of Western Theater Command, Chengdu, China
| | - Mingliang Wang
- Department of Cardiovascular Medicine, General Hospital of Western Theater Command, Chengdu, China
| | - Jun Wang
- Central Sterile Supply Department, General Hospital of Western Theater Command, Chengdu, China
| | - Qiang Wang
- Department of Cardiovascular Medicine, General Hospital of Western Theater Command, Chengdu, China
| | - De Li
- Department of Cardiovascular Medicine, General Hospital of Western Theater Command, Chengdu, China
| | - Xiongshan Sun
- Department of Cardiovascular Medicine, General Hospital of Western Theater Command, Chengdu, China
| | - Yongjian Yang
- School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, China.,Department of Cardiovascular Medicine, General Hospital of Western Theater Command, Chengdu, China
| | - Dachun Yang
- Department of Cardiovascular Medicine, General Hospital of Western Theater Command, Chengdu, China
| |
Collapse
|
16
|
Chen TQ, Guo X, Huo B, Zhong XX, Wang QH, Chen Y, Zhu XH, Feng GK, Jiang DS, Fang ZM, Wei X. BRD4770 inhibits vascular smooth muscle cell proliferation via SUV39H2, but not EHMT2 to protect against neointima formation. Hum Cell 2023; 36:1672-1688. [PMID: 37306883 PMCID: PMC10390615 DOI: 10.1007/s13577-023-00924-4] [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: 01/11/2023] [Accepted: 05/25/2023] [Indexed: 06/13/2023]
Abstract
The behavior of vascular smooth muscle cells (VSMCs) contributes to the formation of neointima. We previously found that EHMT2 suppressed autophagy activation in VSMCs. BRD4770, an inhibitor of EHMT2/G9a, plays a critical role in several kinds of cancers. However, whether and how BRD4770 regulates the behavior of VSMCs remain unknown. In this study, we evaluate the cellular effect of BRD4770 on VSMCs by series of experiments in vivo and ex vivo. We demonstrated that BRD4770 inhibited VSMCs' growth by blockage in G2/M phase in VSMCs. Moreover, our results demonstrated that the inhibition of proliferation was independent on autophagy or EHMT2 suppression which we previous reported. Mechanistically, BRD4770 exhibited an off-target effect from EHMT2 and our further study reveal that the proliferation inhibitory effect by BRD4770 was associated with suppressing on SUV39H2/KTM1B. In vivo, BRD4770 was also verified to rescue VIH. Thus, BRD4770 function as a crucial negative regulator of VSMC proliferation via SUV39H2 and G2/M cell cycle arrest and BRD4770 could be a molecule for the therapy of vascular restenosis.
Collapse
Affiliation(s)
- Tai-Qiang Chen
- Division of Cardiothoracic and Vascular Surgery, Sino-Swiss Heart-Lung Transplantation Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xian Guo
- Division of Cardiothoracic and Vascular Surgery, Sino-Swiss Heart-Lung Transplantation Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bo Huo
- Division of Cardiothoracic and Vascular Surgery, Sino-Swiss Heart-Lung Transplantation Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiao-Xuan Zhong
- Division of Cardiothoracic and Vascular Surgery, Sino-Swiss Heart-Lung Transplantation Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qun-Hui Wang
- Division of Cardiothoracic and Vascular Surgery, Sino-Swiss Heart-Lung Transplantation Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yue Chen
- Division of Cardiothoracic and Vascular Surgery, Sino-Swiss Heart-Lung Transplantation Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xue-Hai Zhu
- Division of Cardiothoracic and Vascular Surgery, Sino-Swiss Heart-Lung Transplantation Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Organ Transplantation, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Minist of Education, Chinese Academy of Medical Sciences, Wuhan, China
| | - Gao-Ke Feng
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ding-Sheng Jiang
- Division of Cardiothoracic and Vascular Surgery, Sino-Swiss Heart-Lung Transplantation Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Organ Transplantation, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Minist of Education, Chinese Academy of Medical Sciences, Wuhan, China
| | - Ze-Min Fang
- Division of Cardiothoracic and Vascular Surgery, Sino-Swiss Heart-Lung Transplantation Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Xiang Wei
- Division of Cardiothoracic and Vascular Surgery, Sino-Swiss Heart-Lung Transplantation Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Key Laboratory of Organ Transplantation, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Minist of Education, Chinese Academy of Medical Sciences, Wuhan, China.
| |
Collapse
|
17
|
Spanjersberg TCF, Oosterhoff LA, Kruitwagen HS, van den Dungen NAM, Vernooij JCM, Asselbergs FW, Mokry M, Spee B, Harakalova M, van Steenbeek FG. Locational memory of macrovessel vascular cells is transcriptionally imprinted. Sci Rep 2023; 13:13028. [PMID: 37563195 PMCID: PMC10415317 DOI: 10.1038/s41598-023-38880-6] [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: 02/17/2023] [Accepted: 07/17/2023] [Indexed: 08/12/2023] Open
Abstract
Vascular pathologies show locational predisposition throughout the body; further insights into the transcriptomics basis of this vascular heterogeneity are needed. We analyzed transcriptomes from cultured endothelial cells and vascular smooth muscle cells from nine adult canine macrovessels: the aorta, coronary artery, vena cava, portal vein, femoral artery, femoral vein, saphenous vein, pulmonary vein, and pulmonary artery. We observed that organ-specific expression patterns persist in vitro, indicating that these genes are not regulated by blood flow or surrounding cell types but are likely fixed in the epigenetic memory. We further demonstrated the preserved location-specific expression of GATA4 protein in cultured cells and in the primary adult vessel. On a functional level, arterial and venous endothelial cells differed in vascular network morphology as the arterial networks maintained a higher complexity. Our findings prompt the rethinking of the extrapolation of results from single-origin endothelial cell systems.
Collapse
Affiliation(s)
- Talitha C F Spanjersberg
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 104, Utrecht, The Netherlands
- Regenerative Medicine Centre Utrecht, University Medical Center Utrecht, Utrecht University, Uppsalalaan 8, Utrecht, The Netherlands
- Division Heart and Lungs, Department of Cardiology, Circulatory Health Research Center, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, Utrecht, The Netherlands
| | - Loes A Oosterhoff
- Regenerative Medicine Centre Utrecht, University Medical Center Utrecht, Utrecht University, Uppsalalaan 8, Utrecht, The Netherlands
| | - Hedwig S Kruitwagen
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 104, Utrecht, The Netherlands
| | - Noortje A M van den Dungen
- Central Diagnostics Laboratory, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Johannes C M Vernooij
- Department of Population Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 7, Utrecht, The Netherlands
| | - Folkert W Asselbergs
- Department of Cardiology, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands
- Health Data Research UK and Institute of Health Informatics, University College London, London, UK
| | - Michal Mokry
- Central Diagnostics Laboratory, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Laboratory of Experimental Cardiology, Department of Cardiology, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| | - Bart Spee
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 104, Utrecht, The Netherlands
- Regenerative Medicine Centre Utrecht, University Medical Center Utrecht, Utrecht University, Uppsalalaan 8, Utrecht, The Netherlands
| | - Magdalena Harakalova
- Regenerative Medicine Centre Utrecht, University Medical Center Utrecht, Utrecht University, Uppsalalaan 8, Utrecht, The Netherlands
- Division Heart and Lungs, Department of Cardiology, Circulatory Health Research Center, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, Utrecht, The Netherlands
| | - Frank G van Steenbeek
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 104, Utrecht, The Netherlands.
- Regenerative Medicine Centre Utrecht, University Medical Center Utrecht, Utrecht University, Uppsalalaan 8, Utrecht, The Netherlands.
- Division Heart and Lungs, Department of Cardiology, Circulatory Health Research Center, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, Utrecht, The Netherlands.
| |
Collapse
|
18
|
Montelione N, Loreni F, Nenna A, Catanese V, Scurto L, Ferrisi C, Jawabra M, Gabellini T, Codispoti FA, Spinelli F, Chello M, Stilo F. Tissue Engineering and Targeted Drug Delivery in Cardiovascular Disease: The Role of Polymer Nanocarrier for Statin Therapy. Biomedicines 2023; 11:798. [PMID: 36979777 PMCID: PMC10045667 DOI: 10.3390/biomedicines11030798] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 02/21/2023] [Accepted: 02/24/2023] [Indexed: 03/09/2023] Open
Abstract
Atherosclerosis-related coronary artery disease (CAD) is the leading cause of mortality and morbidity worldwide. This requires effective primary and secondary prevention in reducing the complications related to CAD; the regression or stabilization of the pathology remains the mainstay of treatment. Statins have proved to be the most effective treatment in reducing adverse effects, but there are limitations related to the administration and achievement of effective doses as well as side effects due to the lack of target-related molecular specificity. The implemented technological steps are polymers and nanoparticles for the administration of statins, as it has been seen how the conjugation of drug delivery systems (DDSs) with statins increases bioavailability by circumventing the hepatic-renal filter and increases the related target specificity, enhancing their action and decreasing side effects. Reduction of endothelial dysfunction, reduced intimal hyperplasia, reduced ischemia-reperfusion injury, cardiac regeneration, positive remodeling in the extracellular matrix, reduced neointimal growth, and increased reendothelialization are all drug-related effects of statins enhanced by binding with DDSs. Recent preclinical studies demonstrate how the effect of statins stimulates the differentiation of endogenous cardiac stem cells. Poly-lactic-co-glycolic acid (PLGA) seems to be the most promising DDS as it succeeds more than the others in enhancing the effect of the bound drug. This review intends to summarize the current evidence on polymers and nanoparticles for statin delivery in the field of cardiovascular disease, trying to shed light on this topic and identify new avenues for future studies.
Collapse
Affiliation(s)
- Nunzio Montelione
- Unit of Vascular Surgery, Università Campus Bio-Medico di Roma, 00128 Rome, Italy
| | - Francesco Loreni
- Unit of Cardiac Surgery, Università Campus Bio-Medico di Roma, 00128 Rome, Italy
| | - Antonio Nenna
- Unit of Cardiac Surgery, Università Campus Bio-Medico di Roma, 00128 Rome, Italy
| | - Vincenzo Catanese
- Unit of Vascular Surgery, Università Campus Bio-Medico di Roma, 00128 Rome, Italy
| | - Lucia Scurto
- Unit of Vascular Surgery, Università Campus Bio-Medico di Roma, 00128 Rome, Italy
| | - Chiara Ferrisi
- Unit of Cardiac Surgery, Università Campus Bio-Medico di Roma, 00128 Rome, Italy
| | - Mohamad Jawabra
- Unit of Cardiac Surgery, Università Campus Bio-Medico di Roma, 00128 Rome, Italy
| | - Teresa Gabellini
- Residency Program of Vascular and Endovascular Surgery, University of Ferrara, 44121 Ferrara, Italy
| | | | - Francesco Spinelli
- Unit of Vascular Surgery, Università Campus Bio-Medico di Roma, 00128 Rome, Italy
| | - Massimo Chello
- Unit of Cardiac Surgery, Università Campus Bio-Medico di Roma, 00128 Rome, Italy
| | - Francesco Stilo
- Unit of Vascular Surgery, Università Campus Bio-Medico di Roma, 00128 Rome, Italy
- Head of Research Unit of Vascular Surgery, Università Campus Bio-Medico di Roma, 00128 Rome, Italy
| |
Collapse
|
19
|
Liu L, Lan X, Chen X, Dai S, Wang Z, Zhao A, Lu L, Huang N, Chen J, Yang P, Liao Y. Multi-functional plant flavonoids regulate pathological microenvironments for vascular stent surface engineering. Acta Biomater 2023; 157:655-669. [PMID: 36436757 DOI: 10.1016/j.actbio.2022.11.035] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 10/23/2022] [Accepted: 11/16/2022] [Indexed: 11/27/2022]
Abstract
In-stent restenosis (ISR) and late thrombosis, usually caused by excessive smooth muscle cell (SMC) proliferation and delayed endothelial layer repair, respectively, are the main risks for the failure of vascular stent implantation. For years, modification of stents with biomolecules that could selectively inhibit SMC proliferation and support endothelial cell (EC) growth had drawn extensive attention. However, the modulatory effect of these biomolecules faces the impact of oxidative stress, inflammation, and hyperlipidemia of the pathological vascular microenvironment, which is caused by the stent implantation injury and atherosclerosis lesions. Here, we modified stents with a natural and multi-functional flavonoid, baicalin (BCL), using poly-dopamine (PDA) coating technology to combat the harmful impact of the pathological microenvironment. Stent with an appropriate BCL immobilization density (approximately 2.03 μg/cm2) successfully supported ECs growth while inhibited SMC proliferation. Furthermore, baicalin-modified surfaces regulated the oxidative stress, inflammation, and high-lipid of the pathological microenvironment to inhibit endothelial dysfunction and the oxidized low-density lipoprotein (ox-LDL)-induced macrophage foam cells formation. In vivo results showed that baicalin-modified stents exhibited significant anti-ISR, anti-inflammatory, and endothelialization-promoting functions. Our study suggests that the multi-functional baicalin with pathological microenvironment-regulation (PMR) effect has potential use in the surface engineering of cardiovascular devices. STATEMENT OF SIGNIFICANCE: Empowering vascular stents with selective modulation of smooth muscle cells and endothelial cells by surface technology has become an important research direction for stent surface engineering. However, stent coatings that can furthermodulate the pathological microenvironment of blood vessels have been rarely reported. In this study, we constructed a multifunctional coating based on a flavonoid, baicalin, which can selectively modulate vascular wall cells and improve the pathological microenvironment. This study may provide a reference for developing advanced vascular stents.
Collapse
Affiliation(s)
- Luying Liu
- Institute of Biomaterials and Surface Engineering, Key Lab. for Advanced Technologies of Materials, Ministry of Education, Southwest Jiaotong University, Chengdu 610031, PR China
| | - Xiaorong Lan
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, The Affiliated Stomatological Hospital of Southwest Medical University, Luzhou 646000, China
| | - Xiao Chen
- Institute of Biomaterials and Surface Engineering, Key Lab. for Advanced Technologies of Materials, Ministry of Education, Southwest Jiaotong University, Chengdu 610031, PR China
| | - Sheng Dai
- Institute of Biomaterials and Surface Engineering, Key Lab. for Advanced Technologies of Materials, Ministry of Education, Southwest Jiaotong University, Chengdu 610031, PR China
| | - Zhixing Wang
- Institute of Biomaterials and Surface Engineering, Key Lab. for Advanced Technologies of Materials, Ministry of Education, Southwest Jiaotong University, Chengdu 610031, PR China
| | - Ansha Zhao
- Institute of Biomaterials and Surface Engineering, Key Lab. for Advanced Technologies of Materials, Ministry of Education, Southwest Jiaotong University, Chengdu 610031, PR China
| | - Lei Lu
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou 325027, China
| | - Nan Huang
- Institute of Biomaterials and Surface Engineering, Key Lab. for Advanced Technologies of Materials, Ministry of Education, Southwest Jiaotong University, Chengdu 610031, PR China
| | - Jiang Chen
- Sichuan Key Laboratory for Disease Gene Study, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, No. 32 of the West Second Section of First Ring Road, Chengdu 610031, PR China.
| | - Ping Yang
- Institute of Biomaterials and Surface Engineering, Key Lab. for Advanced Technologies of Materials, Ministry of Education, Southwest Jiaotong University, Chengdu 610031, PR China.
| | - Yuzhen Liao
- Institute of Biomaterials and Surface Engineering, Key Lab. for Advanced Technologies of Materials, Ministry of Education, Southwest Jiaotong University, Chengdu 610031, PR China.
| |
Collapse
|
20
|
Ebrahimi-Nozari T, Imani R, Haghbin-Nazarpak M, Nouri A. Multimodal effects of asymmetric coating of coronary stents by electrospinning and electrophoretic deposition. Int J Pharm 2022; 630:122437. [PMID: 36435505 DOI: 10.1016/j.ijpharm.2022.122437] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Revised: 11/08/2022] [Accepted: 11/20/2022] [Indexed: 11/27/2022]
Abstract
Drug-eluting stents (DESs) are drug-coated vascular implants that inhibit smooth muscle cell proliferation and limit in-stent re-stenosis. However, traditional DESs release a single drug into the blood and cannot cope with complex mechanisms in atherosclerosis and body responses. The present study aimed to develop a novel multimodal stent by fabricating asymmetric coating with electrophoretic deposition and electrospinning. Herein, we use heparin-loaded alginate (Hep/Alg) and atorvastatin calcium-loaded polyurethane (AtvCa/PU) coatings on the stent luminal and abluminal surfaces, respectively. Scanning electron microscopy (SEM) micrographs showed that the alginate coatings had uniformity and thin thickness. Meanwhile, the PU fibers were formed without beads, with an acceptable diameter and suitable mechanical properties. PU nanofiber revealed minimal degradation in a 1-month study. The release of AtvCa and Hep continued for 8 days without a significant initial burst release. None of the stent coatings were cytotoxic or hemolytic, and PU nanofibers supported the survival of human umbilical endothelial cells (HUVEC) with high adhesion and flattened morphologies. The results indicate that electrophoretic deposition and electrospinning have significant potential for achieving asymmetric coating on stents and a promising approach for dual drug release for multimodal effects in vascular stent applications.
Collapse
Affiliation(s)
- Tahoura Ebrahimi-Nozari
- Biomedical Engineering Department, Amirkabir University of Technology, (Tehran Polytechnic), Tehran, Iran
| | - Rana Imani
- Biomedical Engineering Department, Amirkabir University of Technology, (Tehran Polytechnic), Tehran, Iran.
| | - Masoumeh Haghbin-Nazarpak
- New Technologies Research Center (NTRC), Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran.
| | - Alireza Nouri
- Biomedical Engineering Department, Amirkabir University of Technology, (Tehran Polytechnic), Tehran, Iran
| |
Collapse
|
21
|
Ahmed YM, Orfali R, Abdelwahab NS, Hassan HM, Rateb ME, AboulMagd AM. Partial Synthetic PPARƳ Derivative Ameliorates Aorta Injury in Experimental Diabetic Rats Mediated by Activation of miR-126-5p Pi3k/AKT/PDK 1/mTOR Expression. Pharmaceuticals (Basel) 2022; 15:1175. [PMID: 36297290 PMCID: PMC9607084 DOI: 10.3390/ph15101175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 09/17/2022] [Accepted: 09/17/2022] [Indexed: 11/16/2022] Open
Abstract
Type 2 diabetes mellitus (T2D) is a world wild health care issue marked by insulin resistance, a risk factor for the metabolic disorder that exaggerates endothelial dysfunction, increasing the risk of cardiovascular complications. Peroxisome proliferator-activated receptor PPAR) agonists have therapeutically mitigated hyperlipidemia and hyperglycemia in T2D patients. Therefore, we aimed to experimentally investigate the efficacy of newly designed synthetic PPARα/Ƴ partial agonists on a High-Fat Diet (HFD)/streptozotocin (STZ)-induced T2D. Female Wistar rats (200 ± 25 g body weight) were divided into four groups. The experimental groups were fed the HFD for three consecutive weeks before STZ injection (45 mg/kg/i.p) to induce T2D. Standard reference PPARƳ agonist pioglitazone and the partial synthetic PPARƳ (PIO; 20 mg/kg/BW, orally) were administered orally for 2 weeks after 72 h of STZ injection. The aorta tissue was isolated for biological ELISA, qRT-PCR, and Western blotting investigations for vascular inflammatory endothelial mediators endothelin-1 (ET-1), intracellular adhesion molecule 1 (ICAM-1), E-selectin, and anti-inflammatory vasoactive intestinal polypeptide (VIP), as well as microRNA126-5p and p-AKT/p-Pi3k/p-PDK-1/p-mTOR, endothelial Nitric Oxide Synthase (eNOS) immunohistochemical staining all are coupled with and histopathological examination. Our results revealed that HFD/STZ-induced T2D increased fasting blood glucose, ET-1, ICAM-1, E-selectin, and VIP levels, while decreasing the expression of both microRNA126-5p and p-AKT/p-Pi3k/p-PDK-1/p-mTOR phosphorylation. In contrast, the partial synthetic PPARƳ derivative evidenced a vascular alteration significantly more than reference PIO via decreasing (ET-1), ICAM-1, E-selectin, and VIP, along with increased expression of microRNA126-5p and p-AKT/p-Pi3k/p-PDK-1/p-mTOR. In conclusion, the partial synthetic PPARƳ derivative significantly affected HFD/STZ-induced T2D with vascular complications in the rat aorta.
Collapse
Affiliation(s)
- Yasmin M. Ahmed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Nahda University, Beni-Suef 62521, Egypt
| | - Raha Orfali
- Department of Pharmacognosy, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Nada S. Abdelwahab
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Nahda University, Beni-Suef 62521, Egypt
- Department of Pharmaceutical Analytical Chemistry, Faculty of Pharmacy, Beni-Suef University, Beni-Suef 62521, Egypt
| | - Hossam M. Hassan
- Department of Pharmacognosy, Faculty of Pharmacy, Beni-Suef University, Beni-Suef 62521, Egypt
| | - Mostafa E. Rateb
- School of Computing, Engineering & Physical Sciences, University of the West of Scotland, Paisley PA1 2BE, UK
| | - Asmaa M. AboulMagd
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Nahda University, Beni-Suef 62521, Egypt
| |
Collapse
|
22
|
Montelione N, Catanese V, Nenna A, Jawabra M, Verghi E, Loreni F, Nappi F, Lusini M, Mastroianni C, Jiritano F, Serraino GF, Mastroroberto P, Codispoti FA, Chello M, Spinelli F, Stilo F. The Diagnostic Value of Circulating Biomarkers and Role of Drug-Coated Balloons for In-Stent Restenosis in Patients with Peripheral Arterial Disease. Diagnostics (Basel) 2022; 12:diagnostics12092207. [PMID: 36140608 PMCID: PMC9498042 DOI: 10.3390/diagnostics12092207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 09/01/2022] [Accepted: 09/07/2022] [Indexed: 11/16/2022] Open
Abstract
Peripheral arterial disease (PAD) is an increasingly pathological condition that commonly affects the femoropopliteal arteries. The current fashionable treatment is percutaneous transluminal angioplasty (PTA), often with stenting. However, the in-stent restenosis (ISR) rate after the stenting of the femoropopliteal (FP) district remains high. Many techniques have been proposed for the treatment of femoropopliteal ISR, such as intravascular brachytherapy, laser atherectomy, second stenting and drug-coated balloons angioplasty (DCB). DCB showed a significantly lower rate of restenosis and target lesions revascularization (TLR) compared to conventional PTA. However, further studies and multi-center RCTs with dedicated long-term follow-up are needed to verify the true efficiency of this approach. Nowadays, the correlation between PAD and inflammation biomarkers is well known. Multiple studies have shown that proinflammatory markers (such as C-reactive proteins) and the high plasma levels of microRNA could predict the outcomes after stent placement. In particular, circulating microRNA-320a, microRNA-3937, microRNA-642a-3p and microRNA-572 appear to hold promise in diagnosing ISR in patients with PAD, but also as predictors of stent patency. This narrative review intends to summarize the current knowledge on the value of circulating biomarkers as predictors of ISR and to foster the scientific debate on the advantages of using DCB in the treatment of ISR in the FP district.
Collapse
Affiliation(s)
- Nunzio Montelione
- Vascular Surgery, Università Campus Bio-Medico di Roma, 00128 Rome, Italy
| | - Vincenzo Catanese
- Vascular Surgery, Università Campus Bio-Medico di Roma, 00128 Rome, Italy
- Correspondence:
| | - Antonio Nenna
- Cardiac Surgery, Università Campus Bio-Medico di Roma, 00128 Rome, Italy
| | - Mohamad Jawabra
- Cardiac Surgery, Università Campus Bio-Medico di Roma, 00128 Rome, Italy
| | - Emanuele Verghi
- Cardiac Surgery, Università Campus Bio-Medico di Roma, 00128 Rome, Italy
| | - Francesco Loreni
- Cardiac Surgery, Università Campus Bio-Medico di Roma, 00128 Rome, Italy
| | - Francesco Nappi
- Cardiac Surgery, Centre Cardiologique du Nord de Saint-Denis, 93200 Paris, France
| | - Mario Lusini
- Cardiac Surgery, Università Campus Bio-Medico di Roma, 00128 Rome, Italy
| | - Ciro Mastroianni
- Cardiac Surgery, Università Campus Bio-Medico di Roma, 00128 Rome, Italy
| | - Federica Jiritano
- Cardiovascular Surgery, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy
| | | | | | | | - Massimo Chello
- Cardiac Surgery, Università Campus Bio-Medico di Roma, 00128 Rome, Italy
| | - Francesco Spinelli
- Vascular Surgery, Università Campus Bio-Medico di Roma, 00128 Rome, Italy
| | - Francesco Stilo
- Vascular Surgery, Università Campus Bio-Medico di Roma, 00128 Rome, Italy
| |
Collapse
|
23
|
He Y, Yi X, Zhang Z, Luo H, Li R, Feng X, Fang ZM, Zhu XH, Cheng W, Jiang DS, Zhao F, Wei X. JIB-04, a histone demethylase Jumonji C domain inhibitor, regulates phenotypic switching of vascular smooth muscle cells. Clin Epigenetics 2022; 14:101. [PMID: 35964071 PMCID: PMC9375951 DOI: 10.1186/s13148-022-01321-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 08/05/2022] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Vascular smooth muscle cell (VSMC) phenotype switching is critical for neointima formation, which is the major cause of restenosis after stenting or coronary artery bypass grafting. However, the epigenetic mechanisms regulating phenotype switching of VSMCs, especially histone methylation, are not well understood. As a main component of histone lysine demethylases, Jumonji demethylases might be involved in VSMC phenotype switching and neointima formation. METHODS AND RESULTS A mouse carotid injury model and VSMC proliferation model were constructed to investigate the relationship between histone methylation of H3K36 (downstream target molecule of Jumonji demethylase) and neointima formation. We found that the methylation levels of H3K36 negatively correlated with VSMC proliferation and neointima formation. Next, we revealed that JIB-04 (a pan-inhibitor of the Jumonji demethylase superfamily) could increase the methylation levels of H3K36. Furthermore, we found that JIB-04 obviously inhibited HASMC proliferation, and a cell cycle assay showed that JIB-04 caused G2/M phase arrest in HASMCs by inhibiting the phosphorylation of RB and CDC2 and promoting the phosphorylation of CHK1. Moreover, JIB-04 inhibited the expression of MMP2 to suppress the migration of HASMCs and repressed the expression of contraction-related genes. RNA sequencing analysis showed that the biological processes associated with the cell cycle and autophagy were enriched by using Gene Ontology analysis after HASMCs were treated with JIB-04. Furthermore, we demonstrated that JIB-04 impairs autophagic flux by downregulating STX17 and RAB7 expression to inhibit the fusion of autophagosomes and lysosomes. CONCLUSION JIB-04 suppresses the proliferation, migration, and contractile phenotype of HASMCs by inhibiting autophagic flux, which indicates that JIB-04 is a promising reagent for the treatment of neointima formation.
Collapse
Affiliation(s)
- Yi He
- Division of Cardiothoracic and Vascular Surgery, Sino-Swiss Heart-Lung Transplantation Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, 430030, Hubei, China
| | - Xin Yi
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zihao Zhang
- Division of Cardiothoracic and Vascular Surgery, Sino-Swiss Heart-Lung Transplantation Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, 430030, Hubei, China
| | - Hanshen Luo
- Division of Cardiothoracic and Vascular Surgery, Sino-Swiss Heart-Lung Transplantation Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, 430030, Hubei, China
| | - Rui Li
- Division of Cardiothoracic and Vascular Surgery, Sino-Swiss Heart-Lung Transplantation Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, 430030, Hubei, China
| | - Xin Feng
- Division of Cardiothoracic and Vascular Surgery, Sino-Swiss Heart-Lung Transplantation Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, 430030, Hubei, China
| | - Ze-Min Fang
- Division of Cardiothoracic and Vascular Surgery, Sino-Swiss Heart-Lung Transplantation Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, 430030, Hubei, China
| | - Xue-Hai Zhu
- Division of Cardiothoracic and Vascular Surgery, Sino-Swiss Heart-Lung Transplantation Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, 430030, Hubei, China
- Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation; Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, Hubei, China
| | - Wenlin Cheng
- Department of Cardiology, Zhongnan Hospital of Wuhan University, East Lake Road 169, Wuhan, Hubei, China
- Institute of Myocardial Injury and Repair, Wuhan University, Wuhan, Hubei, China
| | - Ding-Sheng Jiang
- Division of Cardiothoracic and Vascular Surgery, Sino-Swiss Heart-Lung Transplantation Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, 430030, Hubei, China
- Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation; Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, Hubei, China
| | - Fang Zhao
- Department of Cardiology, Zhongnan Hospital of Wuhan University, East Lake Road 169, Wuhan, Hubei, China.
- Institute of Myocardial Injury and Repair, Wuhan University, Wuhan, Hubei, China.
| | - Xiang Wei
- Division of Cardiothoracic and Vascular Surgery, Sino-Swiss Heart-Lung Transplantation Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, 430030, Hubei, China.
- Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation; Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, Hubei, China.
| |
Collapse
|
24
|
Zhang Q, Li F, Ritchie RH, Woodman OL, Zhou X, Qin CX. Novel strategies to promote resolution of inflammation to treat lower extremity artery disease. Curr Opin Pharmacol 2022; 65:102263. [DOI: 10.1016/j.coph.2022.102263] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Revised: 05/24/2022] [Accepted: 05/31/2022] [Indexed: 12/24/2022]
|
25
|
Frolov AV, Shishkova DK, Markova VE, Sinitsky MY, Sinitskaya AV, Poddubnyak AO, Kanonykina AY, Zagorodnikov NI, Grigoriev EV, Kutikhin AG. Paracrine Effects of Conditioned Medium during Its Cross-Addition to Arterial and Venous Endothelial Cells. J EVOL BIOCHEM PHYS+ 2022. [DOI: 10.1134/s0022093022040214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
26
|
Abdulrahman N, Ibrahim M, Joseph JM, Elkoubatry HM, Al-Shamasi AA, Rayan M, Gadeau AP, Ahmed R, Eldassouki H, Hasan A, Mraiche F. Empagliflozin inhibits angiotensin II-induced hypertrophy in H9c2 cardiomyoblasts through inhibition of NHE1 expression. Mol Cell Biochem 2022; 477:1865-1872. [PMID: 35334035 PMCID: PMC9068664 DOI: 10.1007/s11010-022-04411-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 03/10/2022] [Indexed: 02/06/2023]
Abstract
Diabetes mellitus (DM)-induced cardiac morbidities have been the leading cause of death among diabetic patients. Recently, sodium-glucose cotransporter-2 (SGLT-2) inhibitors including empagliflozin (EMPA), which have been approved for the treatment of DM, have gained attention for their cardioprotective effect. The mechanism by which SGLT-2 inhibitors exert their cardioprotective effect remains unclear. Recent studies have suggested that EMPA exerts its cardioprotective effect by inhibiting the Na+/H+ exchanger (NHE), a group of membrane proteins that regulate intracellular pH and cell volume. Increased activity and expression of NHE isoform 1 (NHE1), the predominant isoform expressed in the heart, leads to cardiac hypertrophy. p90 ribosomal s6 kinase (p90 RSK) has been demonstrated to stimulate NHE1 activity. In our study, H9c2 cardiomyoblasts were treated with angiotensin II (ANG) to activate NHE1 and generate a hypertrophic model. We aimed to understand whether EMPA reverses the ANG-induced hypertrophic response and to elucidate the molecular pathway contributing to the cardioprotective effect of EMPA. Our study demonstrated that ANG-induced hypertrophy of H9c2 cardiomyoblasts is accompanied with increased SGLT-1 and NHE1 protein expression, an effect which is prevented in the presence of EMPA. EMPA reduces ANG-induced hypertrophy through the inhibition of SGLT-1 and NHE1 expression.
Collapse
Affiliation(s)
- Nabeel Abdulrahman
- College of Pharmacy, QU Health, Qatar University, P.O. Box 2713, Doha, Qatar
- Biomedical and Pharmaceutical Research Unit, Qatar University, Doha, Qatar
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Meram Ibrahim
- College of Pharmacy, QU Health, Qatar University, P.O. Box 2713, Doha, Qatar
- Biomedical and Pharmaceutical Research Unit, Qatar University, Doha, Qatar
| | - Jensa Mariam Joseph
- College of Pharmacy, QU Health, Qatar University, P.O. Box 2713, Doha, Qatar
- Biomedical and Pharmaceutical Research Unit, Qatar University, Doha, Qatar
| | - Hanan Mahmoud Elkoubatry
- College of Pharmacy, QU Health, Qatar University, P.O. Box 2713, Doha, Qatar
- Biomedical and Pharmaceutical Research Unit, Qatar University, Doha, Qatar
| | - Al-Anood Al-Shamasi
- College of Pharmacy, QU Health, Qatar University, P.O. Box 2713, Doha, Qatar
- Biomedical and Pharmaceutical Research Unit, Qatar University, Doha, Qatar
| | - Menatallah Rayan
- College of Pharmacy, QU Health, Qatar University, P.O. Box 2713, Doha, Qatar
- Biomedical and Pharmaceutical Research Unit, Qatar University, Doha, Qatar
| | | | - Rashid Ahmed
- Department of Mechanical and Industrial Engineering, College of Engineering, Qatar University, 2713, Doha, Qatar
| | - Hussein Eldassouki
- College of Kinesiology, University of Saskatchewan, Saskatoon, SK, Canada
| | - Anwarul Hasan
- Department of Mechanical and Industrial Engineering, College of Engineering, Qatar University, 2713, Doha, Qatar
| | - Fatima Mraiche
- College of Pharmacy, QU Health, Qatar University, P.O. Box 2713, Doha, Qatar.
- Biomedical and Pharmaceutical Research Unit, Qatar University, Doha, Qatar.
| |
Collapse
|
27
|
Fang Y, Lin M, Chen L, Yang C, Liu A. Association between LDL/HDL ratio and in-stent restenosis in patients with acute coronary syndrome after stent implantation. Biomark Med 2022; 16:673-680. [PMID: 35574807 DOI: 10.2217/bmm-2021-1089] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Background: The relationship between LDL/HDL ratio and in-stent restenosis in acute coronary syndrome is unknown. Methods: This observational study recruited 256 patients with acute coronary syndrome who were being followed up by angiography after stenting. The patients were divided into in-stent restenosis (59%) and non-in-stent restenosis (41%) groups. Three stepwise multivariate logistic regression models and area under the curve were conducted to determine the role of LDL/HDL ratio in predicting in-stent restenosis. Results: LDL/HDL ratio was significantly associated with risk of in-stent restenosis (odds ratio ≈ 2.00; p < 0.05 for all) in three models. A good predictive performance of LDL/HDL ratio on in-stent restenosis was found with an area under the curve of 0.74. Conclusion: LDL/HDL ratio was independently associated with the risk of in-stent restenosis in acute coronary syndrome.
Collapse
Affiliation(s)
- Yang Fang
- Department of Cardiology, Benxi Central Hospital, Benxi, Liaoning Province, 117000, China
| | - Min Lin
- Department of Cardiology, Benxi Central Hospital, Benxi, Liaoning Province, 117000, China
| | - Lei Chen
- Department of Cardiology, Benxi Central Hospital, Benxi, Liaoning Province, 117000, China
| | - Chunyan Yang
- Department of Cardiology, Benxi Central Hospital, Benxi, Liaoning Province, 117000, China
| | - Aijun Liu
- Department of Cardiology, Benxi Central Hospital, Benxi, Liaoning Province, 117000, China
| |
Collapse
|
28
|
Spadaccio C, Nenna A, Rose D, Piccirillo F, Nusca A, Grigioni F, Chello M, Vlahakes GJ. The Role of Angiogenesis and Arteriogenesisin Myocardial Infarction and Coronary Revascularization. J Cardiovasc Transl Res 2022; 15:1024-1048. [PMID: 35357670 DOI: 10.1007/s12265-022-10241-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2021] [Accepted: 03/18/2022] [Indexed: 12/25/2022]
Abstract
Surgical myocardial revascularization is associated with long-term survival benefit in patients with multivessel coronary artery disease. However, the exact biological mechanisms underlying the clinical benefits of myocardial revascularization have not been elucidated yet. Angiogenesis and arteriogenesis biologically leading to vascular collateralization are considered one of the endogenous mechanisms to preserve myocardial viability during ischemia, and the presence of coronary collateralization has been regarded as one of the predictors of long-term survival in patients with coronary artery disease (CAD). Some experimental studies and indirect clinical evidence on chronic CAD confirmed an angiogenetic response induced by myocardial revascularization and suggested that revascularization procedures could constitute an angiogenetic trigger per se. In this review, the clinical and basic science evidence regarding arteriogenesis and angiogenesis in both CAD and coronary revascularization is analyzed with the aim to better elucidate their significance in the clinical arena and potential therapeutic use.
Collapse
Affiliation(s)
- Cristiano Spadaccio
- Cardiac Surgery, Massachusetts General Hospital & Harvard Medical School, Boston, USA. .,Cardiac Surgery, Golden Jubilee National Hospital & University of Glasgow, Glasgow, UK.
| | - Antonio Nenna
- Cardiac Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
| | - David Rose
- Cardiac Surgery, Lancashire Cardiac Centre, Blackpool Victoria Hospital, Blackpool, UK
| | | | | | | | - Massimo Chello
- Cardiac Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Gus J Vlahakes
- Cardiac Surgery, Massachusetts General Hospital & Harvard Medical School, Boston, USA
| |
Collapse
|
29
|
Chen S, Coronel R, Hollmann MW, Weber NC, Zuurbier CJ. Direct cardiac effects of SGLT2 inhibitors. Cardiovasc Diabetol 2022; 21:45. [PMID: 35303888 PMCID: PMC8933888 DOI: 10.1186/s12933-022-01480-1] [Citation(s) in RCA: 85] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 03/09/2022] [Indexed: 12/17/2022] Open
Abstract
Sodium-glucose-cotransporter 2 inhibitors (SGLT2is) demonstrate large cardiovascular benefit in both diabetic and non-diabetic, acute and chronic heart failure patients. These inhibitors have on-target (SGLT2 inhibition in the kidney) and off-target effects that likely both contribute to the reported cardiovascular benefit. Here we review the literature on direct effects of SGLT2is on various cardiac cells and derive at an unifying working hypothesis. SGLT2is acutely and directly (1) inhibit cardiac sodium transporters and alter ion homeostasis, (2) reduce inflammation and oxidative stress, (3) influence metabolism, and (4) improve cardiac function. We postulate that cardiac benefit modulated by SGLT2i’s can be commonly attributed to their inhibition of sodium-loaders in the plasma membrane (NHE-1, Nav1.5, SGLT) affecting intracellular sodium-homeostasis (the sodium-interactome), thereby providing a unifying view on the various effects reported in separate studies. The SGLT2is effects are most apparent when cells or hearts are subjected to pathological conditions (reactive oxygen species, inflammation, acidosis, hypoxia, high saturated fatty acids, hypertension, hyperglycemia, and heart failure sympathetic stimulation) that are known to prime these plasmalemmal sodium-loaders. In conclusion, the cardiac sodium-interactome provides a unifying testable working hypothesis and a possible, at least partly, explanation to the clinical benefits of SGLT2is observed in the diseased patient.
Collapse
Affiliation(s)
- Sha Chen
- Department of Anaesthesiology, Laboratory of Experimental Intensive Care and Anaesthesiology (L.E.I.C.A.), Amsterdam UMC, Location Academic Medical Centre (AMC), Amsterdam, University of Amsterdam, Cardiovascular Sciences, Meibergdreef 11, Room M0-129, Amsterdam, Noord-Holland, 1105 AZ, The Netherlands
| | - Ruben Coronel
- Department of Experimental Cardiology, Amsterdam UMC, Location Academic Medical Centre (AMC), Amsterdam,, University of Amsterdam, Cardiovascular Sciences, Amsterdam, The Netherlands
| | - Markus W Hollmann
- Department of Anaesthesiology, Laboratory of Experimental Intensive Care and Anaesthesiology (L.E.I.C.A.), Amsterdam UMC, Location Academic Medical Centre (AMC), Amsterdam, University of Amsterdam, Cardiovascular Sciences, Meibergdreef 11, Room M0-129, Amsterdam, Noord-Holland, 1105 AZ, The Netherlands
| | - Nina C Weber
- Department of Anaesthesiology, Laboratory of Experimental Intensive Care and Anaesthesiology (L.E.I.C.A.), Amsterdam UMC, Location Academic Medical Centre (AMC), Amsterdam, University of Amsterdam, Cardiovascular Sciences, Meibergdreef 11, Room M0-129, Amsterdam, Noord-Holland, 1105 AZ, The Netherlands
| | - Coert J Zuurbier
- Department of Anaesthesiology, Laboratory of Experimental Intensive Care and Anaesthesiology (L.E.I.C.A.), Amsterdam UMC, Location Academic Medical Centre (AMC), Amsterdam, University of Amsterdam, Cardiovascular Sciences, Meibergdreef 11, Room M0-129, Amsterdam, Noord-Holland, 1105 AZ, The Netherlands.
| |
Collapse
|
30
|
Nusca A, Viscusi MM, Piccirillo F, De Filippis A, Nenna A, Spadaccio C, Nappi F, Chello C, Mangiacapra F, Grigioni F, Chello M, Ussia GP. In Stent Neo-Atherosclerosis: Pathophysiology, Clinical Implications, Prevention, and Therapeutic Approaches. Life (Basel) 2022; 12:life12030393. [PMID: 35330144 PMCID: PMC8955389 DOI: 10.3390/life12030393] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 03/02/2022] [Accepted: 03/07/2022] [Indexed: 12/23/2022] Open
Abstract
Despite the dramatic improvements of revascularization therapies occurring in the past decades, a relevant percentage of patients treated with percutaneous coronary intervention (PCI) still develops stent failure due to neo-atherosclerosis (NA). This histopathological phenomenon following stent implantation represents the substrate for late in-stent restenosis (ISR) and late stent thrombosis (ST), with a significant impact on patient’s long-term clinical outcomes. This appears even more remarkable in the setting of drug-eluting stent implantation, where the substantial delay in vascular healing because of the released anti-proliferative agents might increase the occurrence of this complication. Since the underlying pathophysiological mechanisms of NA diverge from native atherosclerosis and early ISR, intra-coronary imaging techniques are crucial for its early detection, providing a proper in vivo assessment of both neo-intimal plaque composition and peri-strut structures. Furthermore, different strategies for NA prevention and treatment have been proposed, including tailored pharmacological therapies as well as specific invasive tools. Considering the increasing population undergoing PCI with drug-eluting stents (DES), this review aims to provide an updated overview of the most recent evidence regarding NA, discussing pathophysiology, contemporary intravascular imaging techniques, and well-established and experimental invasive and pharmacological treatment strategies.
Collapse
Affiliation(s)
- Annunziata Nusca
- Cardiology, Università Campus Bio-Medico di Roma, 00128 Rome, Italy; (A.N.); (M.M.V.); (F.P.); (A.D.F.); (F.M.); (F.G.); (G.P.U.)
| | - Michele Mattia Viscusi
- Cardiology, Università Campus Bio-Medico di Roma, 00128 Rome, Italy; (A.N.); (M.M.V.); (F.P.); (A.D.F.); (F.M.); (F.G.); (G.P.U.)
| | - Francesco Piccirillo
- Cardiology, Università Campus Bio-Medico di Roma, 00128 Rome, Italy; (A.N.); (M.M.V.); (F.P.); (A.D.F.); (F.M.); (F.G.); (G.P.U.)
| | - Aurelio De Filippis
- Cardiology, Università Campus Bio-Medico di Roma, 00128 Rome, Italy; (A.N.); (M.M.V.); (F.P.); (A.D.F.); (F.M.); (F.G.); (G.P.U.)
| | - Antonio Nenna
- Cardiac Surgery, Università Campus Bio-Medico di Roma, 00128 Rome, Italy; (C.C.); (M.C.)
- Correspondence:
| | - Cristiano Spadaccio
- Cardiac Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02115, USA;
| | - Francesco Nappi
- Cardiac Surgery, Centre Cardiologique du Nord de Saint-Denis, 93200 Paris, France;
| | - Camilla Chello
- Cardiac Surgery, Università Campus Bio-Medico di Roma, 00128 Rome, Italy; (C.C.); (M.C.)
| | - Fabio Mangiacapra
- Cardiology, Università Campus Bio-Medico di Roma, 00128 Rome, Italy; (A.N.); (M.M.V.); (F.P.); (A.D.F.); (F.M.); (F.G.); (G.P.U.)
| | - Francesco Grigioni
- Cardiology, Università Campus Bio-Medico di Roma, 00128 Rome, Italy; (A.N.); (M.M.V.); (F.P.); (A.D.F.); (F.M.); (F.G.); (G.P.U.)
| | - Massimo Chello
- Cardiac Surgery, Università Campus Bio-Medico di Roma, 00128 Rome, Italy; (C.C.); (M.C.)
| | - Gian Paolo Ussia
- Cardiology, Università Campus Bio-Medico di Roma, 00128 Rome, Italy; (A.N.); (M.M.V.); (F.P.); (A.D.F.); (F.M.); (F.G.); (G.P.U.)
| |
Collapse
|
31
|
Furtado RHM, Fagundes AA, Oyama K, Zelniker TA, Tang M, Kuder JF, Murphy SA, Hamer A, Wang H, Keech AC, Giugliano RP, Sabatine MS, Bergmark BA. Effect of Evolocumab in Patients With Prior Percutaneous Coronary Intervention. Circ Cardiovasc Interv 2022; 15:e011382. [PMID: 35209731 DOI: 10.1161/circinterventions.121.011382] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
BACKGROUND Patients with prior percutaneous coronary intervention (PCI) are at high residual risk for multiple types of coronary events within and beyond the stented lesion. This risk might be mitigated by more intensive LDL-C (low-density lipoprotein cholesterol)-lowering beyond just with statin therapy. METHODS FOURIER (Further Cardiovascular Outcomes Research With PCSK9 Inhibition in Subjects With Elevated Risk) randomized 27 564 patients with stable atherosclerotic disease on statin to the PCSK9 (proprotein convertase subtilisin-kexin type 9) inhibitor evolocumab or placebo with a median follow-up of 2.2 years. The end points of interest were major adverse cardiovascular events (MACE; a composite of cardiovascular death, myocardial infarction, stroke, unstable angina or coronary revascularization), and major coronary events (a composite of coronary heart death, myocardial infarction, or coronary revascularization). We compared the risk of MACE and the magnitude of relative and absolute risk reductions with evolocumab in patients with and without prior PCI. RESULTS Seventeen thousand seventy-three patients had prior PCI. In the placebo arm, those with prior PCI had higher rates of MACE (13.2% versus 8.3%; hazard ratio [HR]adj 1.61 [95% CI, 1.42-1.84]; P<0.0001) and major coronary events (11.5% versus 6.0%; HRadj, 1.72 [95% CI, 1.49-1.99]; P<0.0001). Relative risk reductions with evolocumab were similar in patients with and without prior PCI (MACE: HR, 0.84 [0.77-0.91] versus HR, 0.88 [0.77-1.01]; Pinteraction 0.51; major coronary events: HR, 0.82 [0.75-0.90] versus HR, 0.88 [0.75-1.04]; Pinteraction 0.42). Absolute risk reductions for MACE were 2.0% versus 0.9% (Pinteraction 0.14) and for major coronary events 2.0% versus 0.7% (Pinteraction 0.045). In those with prior PCI, the effect of evolocumab on coronary revascularization (HR, 0.76 [0.69-0.85]) was directionally consistent across types of revascularization procedures: coronary artery bypass grafting (HR, 0.71 [0.54-0.94]); any PCI (HR, 0.77 [0.69-0.86]); PCI for de novo lesions (HR, 0.76 [0.66-0.88]); and PCI for stent failure or graft lesions (HR, 0.76 [0.63-0.91]). CONCLUSIONS Evolocumab reduces the risk of MACE in patients with prior PCI including the risk of coronary revascularization, with directionally consistent effects across several types of revascularization procedures, including coronary artery bypass grafting and PCI for stent or graft failure. REGISTRATION URL: https://www. CLINICALTRIALS gov; Unique identifier: NCT01764633.
Collapse
Affiliation(s)
- Remo H M Furtado
- Academic Research Organization, Hospital Israelita Albert Einstein, Sao Paulo, Brazil (R.H.M.F.).,Instituto do Coracao (InCor), Hospital das Clinicas da Faculdade de Medicina, Universidade de Sao Paulo, Brazil (R.H.M.F.).,Thrombolysis in Myocardial Infarction (TIMI) Study Group, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (R.H.M.F., A.A.F., K.O., T.A.Z., M.T., J.F.K., S.AM., R.P.G., M.S.S., B.A.B.)
| | - Antônio Aurélio Fagundes
- Thrombolysis in Myocardial Infarction (TIMI) Study Group, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (R.H.M.F., A.A.F., K.O., T.A.Z., M.T., J.F.K., S.AM., R.P.G., M.S.S., B.A.B.)
| | - Kazuma Oyama
- Thrombolysis in Myocardial Infarction (TIMI) Study Group, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (R.H.M.F., A.A.F., K.O., T.A.Z., M.T., J.F.K., S.AM., R.P.G., M.S.S., B.A.B.).,Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan (K.O.)
| | - Thomas A Zelniker
- Thrombolysis in Myocardial Infarction (TIMI) Study Group, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (R.H.M.F., A.A.F., K.O., T.A.Z., M.T., J.F.K., S.AM., R.P.G., M.S.S., B.A.B.).,Division of Cardiology, Vienna General Hospital and Medical University of Vienna, Austria (T.A.Z.)
| | - Minao Tang
- Thrombolysis in Myocardial Infarction (TIMI) Study Group, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (R.H.M.F., A.A.F., K.O., T.A.Z., M.T., J.F.K., S.AM., R.P.G., M.S.S., B.A.B.)
| | - Julia F Kuder
- Thrombolysis in Myocardial Infarction (TIMI) Study Group, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (R.H.M.F., A.A.F., K.O., T.A.Z., M.T., J.F.K., S.AM., R.P.G., M.S.S., B.A.B.)
| | - Sabina A Murphy
- Thrombolysis in Myocardial Infarction (TIMI) Study Group, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (R.H.M.F., A.A.F., K.O., T.A.Z., M.T., J.F.K., S.AM., R.P.G., M.S.S., B.A.B.)
| | - Andrew Hamer
- Cardiol Therapeutics, Oakville, ON, Canada (A.H.)
| | | | - Anthony C Keech
- National Health and Medical Research Council Clinical Trials Centre, Sydney Medical School, University of Sydney, Australia (A.C.K.)
| | - Robert P Giugliano
- Thrombolysis in Myocardial Infarction (TIMI) Study Group, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (R.H.M.F., A.A.F., K.O., T.A.Z., M.T., J.F.K., S.AM., R.P.G., M.S.S., B.A.B.)
| | - Marc S Sabatine
- Thrombolysis in Myocardial Infarction (TIMI) Study Group, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (R.H.M.F., A.A.F., K.O., T.A.Z., M.T., J.F.K., S.AM., R.P.G., M.S.S., B.A.B.)
| | - Brian A Bergmark
- Thrombolysis in Myocardial Infarction (TIMI) Study Group, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (R.H.M.F., A.A.F., K.O., T.A.Z., M.T., J.F.K., S.AM., R.P.G., M.S.S., B.A.B.)
| |
Collapse
|
32
|
Zhang X, Zhou Y, Ye Y, Wu R, Li W, Yao C, Wang S. Human umbilical cord mesenchymal stem cell-derived exosomal microRNA-148a-3p inhibits neointimal hyperplasia by targeting Serpine1. Arch Biochem Biophys 2022; 719:109155. [PMID: 35218720 DOI: 10.1016/j.abb.2022.109155] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 02/17/2022] [Accepted: 02/18/2022] [Indexed: 12/17/2022]
Abstract
BACKGROUND Restenosis is inevitable when patients undergo percutaneous transluminal angioplasty due to neointimal hyperplasia (NIH). Human umbilical cord mesenchymal stem cell-derived exosomes (hucMSC-Exos) have been studied in the field of cardiovascular diseases. However, the effects and mechanisms of hucMSC-Exos on NIH are unclear. We aimed to investigate whether MSC-Exos regulate vascular smooth muscle cell (VSMC) functions to inhibit NIH and explore the underlying mechanisms. METHODS HucMSCs and mouse VSMCs were isolated and characterized by flow cytometry and immunofluorescence. HucMSC-Exos were identified by transmission electron microscopy, nanoparticle tracking analysis and western blots. Exosomes (Exos) were intravenously injected into mice with left common carotid artery ligation, and their effects on NIH were assessed by haematoxylin and eosin (H&E) and immunohistochemistry staining. The effects of hucMSC-Exos on VSMCs were evaluated by Cell Counting Kit-8, scratch wound, Transwell and Western blot assays. MicroRNA sequencing data in the Gene Expression Omnibus and mRNA sequencing results were used to identify potential molecules in hucMSC-Exos and target genes in VSMCs, respectively. We tested the regulatory effect of microRNAs in Exos and target genes in VSMCs using overexpression and knockdown experiments. RESULTS Primary hucMSCs, VSMCs and hucMSC-Exos were isolated and characterized. Administration of hucMSC-Exos suppressed NIH after artery ligation. H&E and immunohistochemistry results showed that hucMSC-Exos decreased the intima and media area and intima/media ratio, increased the contractile phenotype protein SM22a in the media layer and downregulated Serpine1 expression in the carotid artery. Exos were ingested by VSMCs, which inhibited migration and upregulated SM22a expression by suppressing Serpine1 expression in vitro. MiR-148a-3p was enriched in hucMSC-Exos and repressed Serpine1 by targeting its 3' untranslated region. Moreover, exosomal miR-148a-3p suppressed VSMC phenotypic switching and migration by targeting Serpine1. CONCLUSIONS We found that hucMSC-Exos inhibited NIH in a mouse carotid artery ligation model and that the inhibitory effects on VSMC phenotypic switching and migration were mediated by delivery of miR-148a-3p to VSMCs to target Serpine1.
Collapse
Affiliation(s)
- Xiaoyu Zhang
- Division of Vascular Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Yu Zhou
- Division of Vascular Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Yanchen Ye
- Division of Vascular Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Ridong Wu
- Division of Vascular Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Wen Li
- Laboratory of General Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Chen Yao
- Division of Vascular Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China.
| | - Shenming Wang
- Division of Vascular Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China.
| |
Collapse
|
33
|
Wichaiyo S, Saengklub N. Alterations of sodium-hydrogen exchanger 1 function in response to SGLT2 inhibitors: what is the evidence? Heart Fail Rev 2022; 27:1973-1990. [PMID: 35179683 DOI: 10.1007/s10741-022-10220-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/02/2022] [Indexed: 02/07/2023]
Abstract
This review summarizes and describes the current evidence addressing how sodium-glucose cotransporter 2 (SGLT2) inhibitors alter the function of sodium-hydrogen exchanger 1 (NHE-1), in association with their protective effects against adverse cardiovascular events. In the heart, SGLT2 inhibitors modulate the function of NHE-1 (either by direct inhibition or indirect attenuation of protein expression), which promotes cardiac contraction and an enhanced energy supply, in association with improved mitochondrial function, reduced inflammation/oxidative/endoplasmic reticulum stress, and attenuated fibrosis and apoptotic/autophagic cell death. The vasodilating effect of SGLT2 inhibitors has also been proposed due to NHE-1 inhibition. Moreover, platelet-expressed NHE-1 might serve as a target for SGLT2 inhibitors, since these drugs and selective NHE-1 inhibitors produce comparable activity against adenosine diphosphate-stimulated platelet activation. Overall, it is promising that the modulation of the functions of NHE-1 on the heart, blood vessels, and platelets may act as a contributing pathway for the cardiovascular benefits of SGLT2 inhibitors in diabetes and heart failure.
Collapse
Affiliation(s)
- Surasak Wichaiyo
- Department of Pharmacology, Faculty of Pharmacy, Mahidol University, 447 Sri-Ayuthaya Road, Rajathevi, Bangkok, 10400, Thailand. .,Centre of Biopharmaceutical Science for Healthy Ageing, Faculty of Pharmacy, Mahidol University, Bangkok, Thailand.
| | - Nakkawee Saengklub
- Centre of Biopharmaceutical Science for Healthy Ageing, Faculty of Pharmacy, Mahidol University, Bangkok, Thailand.,Department of Physiology, Faculty of Pharmacy, Mahidol University, Bangkok, Thailand
| |
Collapse
|
34
|
Zhao Y, Zeng Q, Li J, Jiang X. Digital Subtraction Angiography Image Features under the Deep Learning Algorithm in Cardiovascular Interventional Treatment and Nursing for Vascular Restenosis. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:5876132. [PMID: 35082913 PMCID: PMC8786521 DOI: 10.1155/2022/5876132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 11/28/2021] [Accepted: 12/21/2021] [Indexed: 11/17/2022]
Abstract
The objective of this study was to explore the application value of digital subtraction angiography (DSA) images optimized by deep learning algorithms in vascular restenosis patients undergoing cardiovascular intervention and their nursing efficacy. In this study, a network model for removing artifacts was constructed based on a deep algorithm. 60 patients with coronary artery restenosis were selected as the research objects, and they were randomly divided into the CTA group guided by CT angiography (CTA) and digital subtraction angiography (DSA) group, with 30 cases in each group. The antiartifact network model constructed based on the depth algorithm was applied to the images of CTA and DSA for experiments. After cardiovascular intervention and clinical pathway nursing intervention, it was found that the diameter stenosis rate in the DSA group decreased from 65.82 ± 12.9% to 4.7 ± 1.3%, and the area stenosis rate decreased from 88.4 ± 14.3% to 5.4 ± 1.7%. During the follow-up period of 3-24 months, 3 out of 46 lesions in the DSA group showed restenosis, so the restenosis rate was 6.5%, which was significantly lower than the 18.4% in the CTA group (P < 0.05). In the DSA group, there was 1 case of bleeding, 0 case of hematoma, 2 cases of urinary retention, and 0 case of hypotension, so the total incidence of adverse reactions was 10%, which was significantly lower than the 30% of the CTA group (P < 0.05). The high-sensitivity C-reactive protein (hs-CRP) levels of the two groups of patients were 3.58 ± 2.02 mg/L and 4.36 ± 3.11 mg/L before surgery and 3.49 ± 2.18 mg/L and 4.57 ± 3.4 mg/L after the surgery. The postoperative hs-CRP level in the CTA group was slightly lower than that before the surgery and the postoperative hs-CRP level in the DSA group was slightly higher than that before the surgery, but they were not statistically significant (P > 0.05). The hs-CRP level of the DSA group before and after the surgery was slightly higher than that of the CTA group, but there was no significant difference (P > 0.05). In summary, the network model based on the deep learning algorithm can remove the artifacts in DSA images and present high-quality clear images, and convolutional neural network (CNN) algorithms had a strong ability to automatically learn features in the field of medical image processing and were worthy of being widely used and popularized. In addition, the DSA-guided intervention can reduce the rate of vascular stenosis in patients, showing low probability of postoperative restenosis and adverse reactions and a good clinical effect.
Collapse
Affiliation(s)
- Yuqin Zhao
- Intracardiac Catheter Room, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, 441021 Hubei, China
| | - Qingting Zeng
- Intracardiac Catheter Room, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, 441021 Hubei, China
| | - Jingjing Li
- Intracardiac Catheter Room, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, 441021 Hubei, China
| | - Xia Jiang
- Intracardiac Catheter Room, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, 441021 Hubei, China
| |
Collapse
|
35
|
Nenna A, Nappi F, Spadaccio C, Greco SM, Pilato M, Stilo F, Montelione N, Catanese V, Lusini M, Spinelli F, Chello M. Hybrid coronary revascularization in multivessel coronary artery disease: a systematic review. Future Cardiol 2022; 18:219-234. [PMID: 35006006 DOI: 10.2217/fca-2020-0244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: Hybrid coronary revascularization (HCR) for multivessel coronary artery disease (CAD) integrates coronary artery bypass grafting (CABG) and percutaneous intervention in a planned revascularization strategy. This systematic review summarizes the state of this art of this technique. Methods: Major databases searched until October 2021. Results: The available literature on HCR includes three randomized trials, ten meta-analysis and 27 retrospective studies. The greatest benefits are observed in patients with low-to-intermediate risk and less complex coronary anatomy; highly complex disease and the presence of risk factors favored conventional CABG in terms of adverse events and survival. Conclusion: HCR is an interesting approach for multivessel CAD but should not be considered a 'one-size-fits-all' procedure. Further studies will specify the subset of patients likely to benefit most from this hybrid approach.
Collapse
Affiliation(s)
- Antonio Nenna
- Cardiovascular surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo 200, Rome 00128, Italy
| | - Francesco Nappi
- Cardiac surgery, Centre Cardiologique du Nord, Rue des Moulins Gémeaux 32, Saint Denis 93200, Paris, France
| | - Cristiano Spadaccio
- Cardiac surgery, Golden Jubilee National Hospital, Agamemnon St, Clydebank G814DY, Glasgow, United Kingdom
| | - Salvatore Matteo Greco
- Cardiovascular surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo 200, Rome 00128, Italy.,Cardiac surgery, ISMETT-IRCCS, Via Ernesto Tricomi 5, Palermo 90127, Italy
| | - Michele Pilato
- Cardiac surgery, ISMETT-IRCCS, Via Ernesto Tricomi 5, Palermo 90127, Italy
| | - Francesco Stilo
- Cardiovascular surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo 200, Rome 00128, Italy
| | - Nunzio Montelione
- Cardiovascular surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo 200, Rome 00128, Italy
| | - Vincenzo Catanese
- Cardiovascular surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo 200, Rome 00128, Italy
| | - Mario Lusini
- Cardiovascular surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo 200, Rome 00128, Italy
| | - Francesco Spinelli
- Cardiovascular surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo 200, Rome 00128, Italy
| | - Massimo Chello
- Cardiovascular surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo 200, Rome 00128, Italy
| |
Collapse
|
36
|
Jiang WW, Xie J, Li XB, Ma CH. Ocotillo-type ginsenosides from the Panax vietnamensis ha et grushv protect H9c2 cardiomyocytes against H 2O 2-induced apoptosis. Pharmacogn Mag 2022. [DOI: 10.4103/pm.pm_253_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
37
|
Yangming-Fan, Jianjun-Ge. Pentoxifylline Prevents Restenosis by Inhibiting Cell Proliferation via p38MAPK Pathway in Rat Vein Graft Model. Cell Transplant 2022; 31:9636897221122999. [PMID: 36066039 PMCID: PMC9459444 DOI: 10.1177/09636897221122999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Coronary artery bypass grafting remains the gold standard in the therapy
of advanced-stage patients. But the vein grafts are prone to
restenosis or failure. Pentoxifylline (PTX) is a methylxanthine
derivative with a function of inhibiting cell proliferation. We thus
applied PTX locally to the vein grafts to study its effect on the
inhibition of graft restenosis using a rat vein graft model.
Morphometric results showed a significant decrease in the thickness of
vein grafts intimal and medial at day 28 after the bypass operation.
Results from Western blot and immunohistochemistry showed that PTX
also significantly reduced the proliferating cell nuclear antigen
(PCNA), alpha-smooth muscle actin (α-SMA) expression, and
phosphorylation of p38 in vein grafts. These results firstly
discovered the positive role of PTX in preventing the vein grafts
restenosis and the mechanism may be inhibition of vascular smooth
muscle cells (VSMCs) proliferation via the p38MAPK pathway.
Collapse
Affiliation(s)
- Yangming-Fan
- Department of Cardiac Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Jianjun-Ge
- Department of Cardiac Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| |
Collapse
|
38
|
Zhou Y, Dai C, Zhang B, Ge J. A Adiponectina Previne a Reestenose pela Inibição da Proliferação Celular em um Modelo de Enxerto Venoso em Ratos. Arq Bras Cardiol 2021; 117:1179-1188. [PMID: 35613174 PMCID: PMC8757157 DOI: 10.36660/abc.20200761] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Accepted: 01/27/2021] [Indexed: 02/08/2023] Open
Abstract
Fundamento: O enxerto de bypass na artéria coronária (CABG) continua a ser eficiente como tratamento para pacientes portadores de doença arterial coronariana; entretanto, o enxerto venoso tende a apresentar reestenose ou oclusão. A adiponectina (ADP) é uma proteína hormonal plasmática com a função de regular a proliferação celular. Objetivo: Foram utilizadas duas doses diferentes da proteína ADP em um modelo de enxerto venoso em ratos para estimular a alteração do enxerto venoso. O objetivo deste estudo foi investigar o efeito da ADP sobre a reestenose em enxerto venoso. Métodos: Veias jugulares autólogas foram implantadas como enxertos interposicionais de carótida pela técnica de anastomose de manga em ratos Sprague Dawley. A adiponectina (2,5 μg e 7,5 μg) foi entregue ao enxerto venoso por bypass de forma perivascular, suspensa em gel Pluronic-F127 a 30%. O grupo tratado apenas com bypass e o grupo tratado com gel veículo carregado apenas com Pluronic funcionaram como controle. Foram feitas comparações com análise de via única de variância e teste post-hoc, com p <0,05 sendo considerado significativo. Resultados: A proliferação celular (índice de PCNA) foi significativamente baixa no grupo tratado com adiponectina em comparação com o grupo de controle e o grupo tratado com o gel veículo na íntima e na adventícia dos enxertos a partir do dia 3 (p <0,01). VCAM-1 e ICAM-1 avaliados por imuno-histoquímica diminuíram significativamente em enxertos venosos tratados com adiponectina na quarta semana (p <0,01). O tratamento de enxertos venosos com gel carregado com adiponectina reduziu a espessura da íntima, da média e da adventícia, em comparação com os enxertos de controle e tratados com gel veículo no dia 28 (p <0,01). Conclusões: Este estudo oferece evidências adicionais do possível papel terapêutico da adiponectina na modulação de lesão vascular e seu reparo.
Collapse
|
39
|
Wakasa S, Matsui Y. Diabetes Mellitus With Left Ventricular Dysfunction - Optimal Indication of Bilateral Internal Thoracic Artery Grafting? Circ J 2021; 85:2002-2003. [PMID: 33994413 DOI: 10.1253/circj.cj-21-0268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Satoru Wakasa
- Department of Cardiovascular and Thoracic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University
| | - Yoshiro Matsui
- Department of Cardiovascular Surgery, Hanaoka Seishu Memorial Hospital
| |
Collapse
|
40
|
Zhao YY, Chen LH, Huang L, Li YZ, Yang C, Zhu Y, Qu SL, Zhang C. Cardiovascular protective effects of GLP-1:A focus on the MAPK signaling pathway. Biochem Cell Biol 2021; 100:9-16. [PMID: 34658256 DOI: 10.1139/bcb-2021-0365] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Cardiovascular and related metabolic diseases are significant global health challenges. Glucagon-like peptide 1 (GLP-1) is a brain-gut peptide secreted by ileal endocrine that is now an established drug target in type 2 diabetes (T2DM). GLP-1 targeting agents have been shown not only to treat T2DM, but also to exert cardiovascular protective effects through regulating multiple signaling pathways. The mitogen-activated protein kinase (MAPK) pathway, a common signal transduction pathway for transmitting extracellular signals to downstream effector molecules, is involved in regulating diverse cell physiological processes, including cell proliferation, differentiation, stress, inflammation, functional synchronization, transformation and apoptosis. The purpose of this review is to highlight the relationship between GLP-1 and cardiovascular disease (CVD), and discuss how GLP-1 exerts cardiovascular protective effects through MAPK signaling pathway. This review also discusses the future challenges in fully characterizing and evaluating the CVD protective effects of GLP-1 receptor agonists (GLP-1RA) at the cellular and molecular level. A better understanding of MAPK signaling pathway that are disregulated in CVD may aid in the design and development of promising GLP-1RA.
Collapse
Affiliation(s)
- Yu-Yan Zhao
- Hengyang Medical College, 34706, Institute of Cardiovascular Disease, Hengyang, China, 421001;
| | - Lin-Hui Chen
- University of South China, 34706, Hengyang, Hunan, China;
| | - Liang Huang
- University of South China, 34706, Hengyang, Hunan, China;
| | - Yong-Zhen Li
- University of South China, 34706, Hengyang, Hunan, China;
| | - Chen Yang
- University of South China, 34706, Hengyang, Hunan, China;
| | - Ying Zhu
- University of South China, 34706, Department of Health Inspection and Quarantine, Hengyang, Hunan, China;
| | - Shun-Lin Qu
- University of South China, 34706, Hengyang, Hunan, China;
| | - Chi Zhang
- University of South China, 34706, Hengyang, Hunan, China, 421001;
| |
Collapse
|
41
|
Wang Y, Tao M, Wei H, Arslan Ahmad M, Ma Y, Mao X, Hao L, Ao Q. PLCL vascular external sheath carrying prednisone for improving patency rate of the vein graft. Tissue Eng Part A 2021; 28:394-404. [PMID: 34605672 DOI: 10.1089/ten.tea.2021.0118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Coronary artery bypass graft surgery (CABG) is an impactful treatment for coronary heart disease. Intimal hyperplasia is the central reason for the restenosis of vein grafts after CABG. The introduction of external vascular sheaths around VGs (VGs) can effectively inhibit intimal hyperplasia and ensure the patency of VGs. In this study, the well-known biodegradable copolymer poly (ε-caprolactone-co-L, L-lactide) (PLCL) was electrospun into high porosity external sheaths. The prednisone loaded in the PLCL sheath was slowly released during the degradation process of PLCL. Under the combined effects of sheath and prednisone, intimal hyperplasia was inhibited. For the cell experiments, all sheaths show low cytotoxicity to L929 cells at different concentrations at different time intervals. The ultrasonography and histological results showed prominent dilation and intimal hyperplasia of VG without sheath after two months of surgery. But there was no dilation in PLCL and PLCLPrednisone groups. Notably, the prednisone-loaded sheath group exhibited efficacy in inhibiting intimal hyperplasia and ensured graft patency.
Collapse
Affiliation(s)
- Yang Wang
- China Medical University, 38019, School of Forensic Medicine, Shenyang, China.,China Medical University, School of Intelligent Medicine, Shenyang, China;
| | - Meihan Tao
- China Medical University, 38019, School of Intelligent Medicine, Shenyang, China;
| | - Huan Wei
- The First Affiliated Hospital of China Medical University, 159407, Shenyang, Liaoning, China;
| | | | - Yizhan Ma
- China Medical University, 38019, School of Intelligent Medicine, Shenyang, China;
| | - Xiaoyan Mao
- China Medical University, 38019, School of Intelligent Medicine, Shenyang, China;
| | - Liang Hao
- China Medical University, School of Forensic Medicine, Shenyang, China;
| | - Qiang Ao
- China Medical University, 38019, School of Intelligent Medicine, Shenyang, China.,Sichuan University, 12530, Chengdu, Sichuan, China;
| |
Collapse
|
42
|
Cui X, Pan G, Chen Y, Guo X, Liu T, Zhang J, Yang X, Cheng M, Gao H, Jiang F. The p53 pathway in vasculature revisited: A therapeutic target for pathological vascular remodeling? Pharmacol Res 2021; 169:105683. [PMID: 34019981 DOI: 10.1016/j.phrs.2021.105683] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 04/26/2021] [Accepted: 05/14/2021] [Indexed: 02/08/2023]
Abstract
Pathological vascular remodeling contributes to the development of restenosis following intraluminal interventions, transplant vasculopathy, and pulmonary arterial hypertension. Activation of the tumor suppressor p53 may counteract vascular remodeling by inhibiting aberrant proliferation of vascular smooth muscle cells and repressing vascular inflammation. In particular, the development of different lines of small-molecule p53 activators ignites the hope of treating remodeling-associated vascular diseases by targeting p53 pharmacologically. In this review, we discuss the relationships between p53 and pathological vascular remodeling, and summarize current experimental data suggesting that drugging the p53 pathway may represent a novel strategy to prevent the development of vascular remodeling.
Collapse
Affiliation(s)
- Xiaopei Cui
- Shandong Key Laboratory of Cardiovascular Proteomics and Department of Geriatric Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China
| | - Guopin Pan
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China; Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang Medical University, Xinxiang, Henan Province, China
| | - Ye Chen
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China
| | - Xiaosun Guo
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China
| | - Tengfei Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China
| | - Jing Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China
| | - Xiaofan Yang
- Department of Pediatrics, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China
| | - Mei Cheng
- Shandong Key Laboratory of Cardiovascular Proteomics and Department of Geriatric Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China
| | - Haiqing Gao
- Shandong Key Laboratory of Cardiovascular Proteomics and Department of Geriatric Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China
| | - Fan Jiang
- Shandong Key Laboratory of Cardiovascular Proteomics and Department of Geriatric Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China.
| |
Collapse
|
43
|
Nenna A, Nappi F, Spadaccio C, Greco SM, Pilato M, Stilo F, Montelione N, Catanese V, Lusini M, Spinelli F, Chello M. Advanced measurements of coronary calcium scores: how does it affect current clinical practice? Future Cardiol 2021; 18:35-41. [PMID: 33885330 DOI: 10.2217/fca-2020-0243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Coronary artery calcium (CAC) scoring has emerged as a marker of the burden of atheromatous disease and has been included in scoring systems. The practice of myocardial revascularization, considering percutaneous procedures or surgical strategies, is dramatically changing over years and the prognostic significance of CAC scoring is gradually being conceived. In this interdisciplinary scenario, vessel specific calcium scoring, mapping of coronary calcification and its integration with functional assessment of coronary artery disease might change the future decisions in the catheterization lab and operative theaters. This article summarizes CAC evaluation techniques and its implications in clinical practice.
Collapse
Affiliation(s)
- Antonio Nenna
- Cardiovascular Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Francesco Nappi
- Cardiac Surgery, Centre Cardiologique du Nord, Saint Denis, Paris, France
| | | | - Salvatore Matteo Greco
- Cardiovascular Surgery, Università Campus Bio-Medico di Roma, Rome, Italy.,Cardiac Surgery, ISMETT-IRCCS, Palermo, Italy
| | | | - Francesco Stilo
- Cardiovascular Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Nunzio Montelione
- Cardiovascular Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Vincenzo Catanese
- Cardiovascular Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Mario Lusini
- Cardiovascular Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Francesco Spinelli
- Cardiovascular Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Massimo Chello
- Cardiovascular Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
| |
Collapse
|
44
|
Polymers and Nanoparticles for Statin Delivery: Current Use and Future Perspectives in Cardiovascular Disease. Polymers (Basel) 2021; 13:polym13050711. [PMID: 33652927 PMCID: PMC7956757 DOI: 10.3390/polym13050711] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 02/20/2021] [Accepted: 02/21/2021] [Indexed: 12/20/2022] Open
Abstract
Atherosclerosis-related coronary artery disease (CAD) is one of the leading sources of mortality and morbidity in the world. Primary and secondary prevention appear crucial to reduce CAD-related complications. In this scenario, statin treatment was shown to be clinically effective in the reduction of adverse events, but systemic administration provides suboptimal results. As an attempt to improve bioavailability and effectiveness, polymers and nanoparticles for statin delivery were recently investigated. Polymers and nanoparticles can help statin delivery and their effects by increasing oral bioavailability or enhancing target-specific interaction, leading to reduced vascular endothelial dysfunction, reduced intimal hyperplasia, reduced ischemia-reperfusion injury, increased cardiac regeneration, positive remodeling in the extracellular matrix, reduced neointimal growth and increased re-endothelization. Moreover, some innovative aspects described in other cardiovascular fields could be translated into the CAD scenario. Recent preclinical studies are underlining the effect of statins in the stimulation and differentiation of endogenous cardiac stem cells, as well as in targeting of local adverse conditions implicated in atherosclerosis, and statin delivery through poly-lactic-co-glycolic acid (PLGA) appears the most promising aspect of current research to enhance drug activity. The present review intends to summarize the current evidence about polymers and nanoparticles for statin delivery in the field of cardiovascular disease, trying to shed light on this topic and identify new avenues for future studies.
Collapse
|
45
|
Nappi F, Nenna A, Larobina D, Martuscelli G, Singh SSA, Chello M, Ambrosio L. The Use of Bioactive Polymers for Intervention and Tissue Engineering: The New Frontier for Cardiovascular Therapy. Polymers (Basel) 2021; 13:446. [PMID: 33573282 PMCID: PMC7866823 DOI: 10.3390/polym13030446] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 01/19/2021] [Accepted: 01/25/2021] [Indexed: 12/28/2022] Open
Abstract
Coronary heart disease remains one of the leading causes of death in most countries. Healthcare improvements have seen a shift in the presentation of disease with a reducing number of ST-segment elevation myocardial infarctions (STEMIs), largely due to earlier reperfusion strategies such as percutaneous coronary intervention (PCI). Stents have revolutionized the care of these patients, but the long-term effects of these devices have been brought to the fore. The conceptual and technologic evolution of these devices from bare-metal stents led to the creation and wide application of drug-eluting stents; further research introduced the idea of polymer-based resorbable stents. We look at the evolution of stents and the multiple advantages and disadvantages offered by each of the different polymers used to make stents in order to identify what the stent of the future may consist of whilst highlighting properties that are beneficial to the patient alongside the role of the surgeon, the cardiologist, engineers, chemists, and biophysicists in creating the ideal stent.
Collapse
Affiliation(s)
- Francesco Nappi
- Department of Cardiac Surgery, Centre Cardiologique du Nord de Saint-Denis, 93200 Paris, France
| | - Antonio Nenna
- Department of Cardiovascular Surgery, Università Campus Bio-Medico di Roma, 00128 Rome, Italy; (A.N.); (M.C.)
| | - Domenico Larobina
- Institute for Polymers, Composites and Biomaterials, National Research Council of Italy, 06128 Rome, Italy; (D.L.); (L.A.)
| | - Giorgia Martuscelli
- Multidisciplinary Department of Medical-Surgical and Dental Specialties, University of Campania Luigi Vanvitelli, 81100 Naples, Italy;
| | | | - Massimo Chello
- Department of Cardiovascular Surgery, Università Campus Bio-Medico di Roma, 00128 Rome, Italy; (A.N.); (M.C.)
| | - Luigi Ambrosio
- Institute for Polymers, Composites and Biomaterials, National Research Council of Italy, 06128 Rome, Italy; (D.L.); (L.A.)
| |
Collapse
|
46
|
Effect of Statins on Platelet Activation and Function: From Molecular Pathways to Clinical Effects. BIOMED RESEARCH INTERNATIONAL 2021; 2021:6661847. [PMID: 33564680 PMCID: PMC7850835 DOI: 10.1155/2021/6661847] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 01/09/2021] [Accepted: 01/12/2021] [Indexed: 12/11/2022]
Abstract
Purpose Statins are a class of drugs widely used in clinical practice for their lipid-lowering and pleiotropic effects. In recent years, a correlation between statins and platelet function has been unveiled in the literature that might introduce new therapeutic indications for this class of drugs. This review is aimed at summarizing the mechanisms underlying statin-platelet interaction in the cardiologic scenario and building the basis for future in-depth studies. Methods We conducted a literature search through PubMed, Embase, EBSCO, Cochrane Database of Systematic Reviews, and Web of Science from their inception to June 2020. Results Many pathways could explain the interaction between statins and platelets, but the specific effect depends on the specific compound. Some could be mediated by enzymes that allow the entry of drugs into the cell (OATP2B1) and others by enzymes that mediate their activation (PLA2, MAPK, TAX2, PPARs, AKT, and COX-1), recruitment and adhesion (LOX-1, CD36, and CD40L), or apoptosis (BCL2). Statins also appear to have a synergistic effect with aspirin and low molecular weight heparins. Surprisingly, they seem to have an antagonistic effect with clopidogrel. Conclusion There are many pathways potentially responsible for the interactions between statins and platelets. Their effect appears to be closely related, and each single effect can be barely measured. Also, the same compound might have complex downstream signaling with potentially opposite effects, i.e., beneficial or deleterious. The multiple clinical implications that can be derived as a result of this interaction, however, represent an excellent reason to develop future in-depth studies.
Collapse
|
47
|
Das L, Kumari TK, Renuka K, Rajeev R, Raju N. Microanatomy of Left Internal Thoracic Artery and Left Anterior Descending Artery: A Comparative Study. NATIONAL JOURNAL OF CLINICAL ANATOMY 2021. [DOI: 10.4103/njca.njca_30_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
48
|
Bujak K, Lejawa M, Gąsior M, Osadnik T. The CTGF gene -945 G/C polymorphism is associated with target lesion revascularization for in-stent restenosis. Exp Mol Pathol 2020; 118:104598. [PMID: 33358742 DOI: 10.1016/j.yexmp.2020.104598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 12/11/2020] [Accepted: 12/16/2020] [Indexed: 10/22/2022]
Abstract
BACKGROUND AND AIMS Previous studies have shown that transforming growth factor β (TGF-β) and vascular endothelial growth factor A (VEGF-A) pathways are involved in the in-stent restenosis (ISR) process. The present study aimed to assess the relationship between single-nucleotide polymorphisms (SNPs) in genes encoding downstream proteins of TGF-β and VEGF-A pathways and the risk of target lesion revascularization (TLR) for in-stent restenosis. METHODS A total of 657 patients (with 781 treated lesions) who underwent percutaneous coronary intervention (PCI) with stent implantation at our center between 2007 and 2012 and completed a 4-year follow-up for clinically-driven TLR, were included. SNPs in CTGF (rs6918698), TGFBR2 (rs2228048), SMAD3 (rs17293632), KDR (rs2071559), CCL2 (rs1024610) were genotyped using TaqMan assay. RESULTS Major allele carriers of CTGF gene -945 G/C polymorphism (rs6918698) were significantly less likely to underwent clinically-driven TLR during follow-up than minor allele carriers. After adjustment for clinical, angiographic, and procedural covariates, CTGF polymorphism was significantly associated with TLR, and minor allele (C) carriers had nearly two times higher risk of developing ISR requiring TLR (HR of 1.93, 95%CI 1.15-3.24) compared to patients with major (GG) genotype. No significant relationship was found between other analyzed polymorphisms and cumulative incidence of TLR at 4-years. CONCLUSIONS Our results suggest that functional -945 G/C polymorphism in the gene encoding connective tissue growth factor is associated with the need for TLR in patients who underwent PCI for stable coronary artery disease.
Collapse
Affiliation(s)
- Kamil Bujak
- 3rd Department of Cardiology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Katowice, Poland.
| | - Mateusz Lejawa
- Kardio-Med Silesia, Zabrze, Poland; Department of Pharmacology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Katowice, Poland
| | - Mariusz Gąsior
- 3rd Department of Cardiology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Katowice, Poland
| | - Tadeusz Osadnik
- Department of Pharmacology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Katowice, Poland; 2nd Department of Cardiology and Angiology, Silesian Center for Heart Diseases, Zabrze, Poland
| |
Collapse
|
49
|
Liu Z, Wu C, Zou X, Shen W, Yang J, Zhang X, Hu X, Wang H, Liao Y, Jing T. Exosomes derived from mesenchymal stem cells inhibit neointimal hyperplasia by activating the Erk1/2 signalling pathway in rats. Stem Cell Res Ther 2020; 11:220. [PMID: 32513275 PMCID: PMC7278178 DOI: 10.1186/s13287-020-01676-w] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 03/17/2020] [Accepted: 04/13/2020] [Indexed: 01/29/2023] Open
Abstract
Background Restenosis is a serious problem in patients who have undergone percutaneous transluminal angioplasty. Endothelial injury resulting from surgery can lead to endothelial dysfunction and neointimal formation by inducing aberrant proliferation and migration of vascular smooth muscle cells. Exosomes secreted by mesenchymal stem cells have been a hot topic in cardioprotective research. However, to date, exosomes derived from mesenchymal stem cells (MSC-Exo) have rarely been reported in association with restenosis after artery injury. The aim of this study was to investigate whether MSC-Exo inhibit neointimal hyperplasia in a rat model of carotid artery balloon-induced injury and, if so, to explore the underlying mechanisms. Methods Characterization of MSC-Exo immunophenotypes was performed by electron microscopy, nanoparticle tracking analysis and western blot assays. To investigate whether MSC-Exo inhibited neointimal hyperplasia, rats were intravenously injected with normal saline or MSC-Exo after carotid artery balloon-induced injury. Haematoxylin-eosin staining was performed to examine the intimal and media areas. Evans blue dye staining was performed to examine re-endothelialization. Moreover, immunohistochemistry and immunofluorescence were performed to examine the expression of CD31, vWF and α-SMA. To further investigate the involvement of MSC-Exo-induced re-endothelialization, the underlying mechanisms were studied by cell counting kit-8, cell scratch, immunofluorescence and western blot assays. Results Our data showed that MSC-Exo were ingested by endothelial cells and that systemic injection of MSC-Exo suppressed neointimal hyperplasia after artery injury. The Evans blue staining results showed that MSC-Exo could accelerate re-endothelialization compared to the saline group. The immunofluorescence and immunohistochemistry results showed that MSC-Exo upregulated the expression of CD31 and vWF but downregulated the expression of α-SMA. Furthermore, MSC-Exo mechanistically facilitated proliferation and migration by activating the Erk1/2 signalling pathway. The western blot results showed that MSC-Exo upregulated the expression of PCNA, Cyclin D1, Vimentin, MMP2 and MMP9 compared to that in the control group. Interestingly, an Erk1/2 inhibitor reversed the expression of the above proteins. Conclusion Our data suggest that MSC-Exo can inhibit neointimal hyperplasia after carotid artery injury by accelerating re-endothelialization, which is accompanied by activation of the Erk1/2 signalling pathway. Importantly, our study provides a novel cell-free approach for the treatment of restenosis diseases after intervention.
Collapse
Affiliation(s)
- Zhihui Liu
- Department of Cardiology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, China.,State Key Laboratory of Silkworm Genome Biology, The Institute of Sericulture and Systems Biology, Southwest University, Chongqing, China
| | - Chao Wu
- State Key Laboratory of Silkworm Genome Biology, The Institute of Sericulture and Systems Biology, Southwest University, Chongqing, China.,Department of Thoracic Surgery, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Xinliang Zou
- Department of Cardiology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Weiming Shen
- Laboratory of Integrative Medicine, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jiacai Yang
- The Institute of Burn Research, South-West Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Xiaorong Zhang
- The Institute of Burn Research, South-West Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Xiaohong Hu
- The Institute of Burn Research, South-West Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Haidong Wang
- Department of Thoracic Surgery, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Yi Liao
- Department of Thoracic Surgery, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Tao Jing
- Department of Cardiology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, China.
| |
Collapse
|
50
|
Maleknia M, Ansari N, Haybar H, Maniati M, Saki N. Inflammatory Growth Factors and In-Stent Restenosis: Effect of Cytokines and Growth Factors. ACTA ACUST UNITED AC 2020. [DOI: 10.1007/s42399-020-00240-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|