1
|
Fan L, Tang Y, Liu J, Liu Y, Xu Y, Liu J, Liu H, Pang W, Guo Y, Yao W, Zhang T, Peng Q, Zhou J. Mechanical Activation of cPLA2 Impedes Fatty Acid β-Oxidation in Vein Grafts. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2411559. [PMID: 39587975 PMCID: PMC11744522 DOI: 10.1002/advs.202411559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 11/03/2024] [Indexed: 11/27/2024]
Abstract
High-magnitude cyclic stretch from arterial blood pressure significantly contributes to the excessive proliferation and migration of vascular smooth muscle cells (VSMCs), leading to neointima formation in vein grafts. However, the molecular mechanisms remain unclear. This study highlights the critical role of cytosolic Phospholipase A2 (cPLA2)/ Yin Yang 1 (YY1)/ carnitine palmitoyltransferase 1b (CPT1B) signaling in coordinating VSMC mechanical activation by inhibiting fatty acid β-oxidation. Metabolomic analysis showed that a 15%-1 Hz arterial cyclic stretch, compared to a 5%-1 Hz venous stretch, increased long-chain fatty acids in VSMCs. cPLA2, identified as a mechanoresponsive molecule, produces excessive arachidonic acid (ArAc) under the 15%-1 Hz stretch, inhibiting CPT1B expression, a key enzyme in fatty acid β-oxidation. ArAc promotes transcription factor YY1 degradation, downregulating CPT1B. Inadequate fatty acid oxidation caused by knockdown of CPT1B or YY1, or etomoxir treatment, increased nuclear membrane tension, orchestrating the activation of cPLA2. Overexpressing CPT1B or inhibiting cPLA2 reduced VSMC proliferation and migration in vein grafts, decreasing neointimal hyperplasia. This study uncovers a novel mechanism in lipid metabolic reprogramming in vein grafts, suggesting a new therapeutic target for vein graft hyperplasia.
Collapse
Affiliation(s)
- Linwei Fan
- Department of Physiology and PathophysiologySchool of Basic Medical SciencesState Key Laboratory of Vascular Homeostasis and RemodelingDepartment of Cardiology and Institute of Vascular MedicinePeking University Third HospitalNational Health Commission Key Laboratory of Cardiovascular Molecular Biology and Regulatory PeptidesBeijing Key Laboratory of Cardiovascular Receptors ResearchPeking UniversityBeijing100191China
| | - Yuanjun Tang
- Department of Physiology and PathophysiologySchool of Basic Medical SciencesState Key Laboratory of Vascular Homeostasis and RemodelingDepartment of Cardiology and Institute of Vascular MedicinePeking University Third HospitalNational Health Commission Key Laboratory of Cardiovascular Molecular Biology and Regulatory PeptidesBeijing Key Laboratory of Cardiovascular Receptors ResearchPeking UniversityBeijing100191China
| | - Jian Liu
- Shenzhen Bay LaboratoryShenzhen518132China
| | - Yueqi Liu
- Department of Physiology and PathophysiologySchool of Basic Medical SciencesState Key Laboratory of Vascular Homeostasis and RemodelingDepartment of Cardiology and Institute of Vascular MedicinePeking University Third HospitalNational Health Commission Key Laboratory of Cardiovascular Molecular Biology and Regulatory PeptidesBeijing Key Laboratory of Cardiovascular Receptors ResearchPeking UniversityBeijing100191China
| | - Yiwei Xu
- Department of Physiology and PathophysiologySchool of Basic Medical SciencesState Key Laboratory of Vascular Homeostasis and RemodelingDepartment of Cardiology and Institute of Vascular MedicinePeking University Third HospitalNational Health Commission Key Laboratory of Cardiovascular Molecular Biology and Regulatory PeptidesBeijing Key Laboratory of Cardiovascular Receptors ResearchPeking UniversityBeijing100191China
| | - Jiayu Liu
- Department of Physiology and PathophysiologySchool of Basic Medical SciencesState Key Laboratory of Vascular Homeostasis and RemodelingDepartment of Cardiology and Institute of Vascular MedicinePeking University Third HospitalNational Health Commission Key Laboratory of Cardiovascular Molecular Biology and Regulatory PeptidesBeijing Key Laboratory of Cardiovascular Receptors ResearchPeking UniversityBeijing100191China
| | - Han Liu
- Department of Physiology and PathophysiologySchool of Basic Medical SciencesState Key Laboratory of Vascular Homeostasis and RemodelingDepartment of Cardiology and Institute of Vascular MedicinePeking University Third HospitalNational Health Commission Key Laboratory of Cardiovascular Molecular Biology and Regulatory PeptidesBeijing Key Laboratory of Cardiovascular Receptors ResearchPeking UniversityBeijing100191China
| | - Wei Pang
- Department of Physiology and PathophysiologySchool of Basic Medical SciencesState Key Laboratory of Vascular Homeostasis and RemodelingDepartment of Cardiology and Institute of Vascular MedicinePeking University Third HospitalNational Health Commission Key Laboratory of Cardiovascular Molecular Biology and Regulatory PeptidesBeijing Key Laboratory of Cardiovascular Receptors ResearchPeking UniversityBeijing100191China
| | - Yuxuan Guo
- Institute of Cardiovascular SciencesSchool of Basic Medical SciencesState Key Laboratory of Vascular Homeostasis and RemodelingBeijing Key Laboratory of Cardiovascular Receptors ResearchPeking UniversityBeijing100191China
| | - Weijuan Yao
- Department of Physiology and PathophysiologySchool of Basic Medical SciencesState Key Laboratory of Vascular Homeostasis and RemodelingDepartment of Cardiology and Institute of Vascular MedicinePeking University Third HospitalNational Health Commission Key Laboratory of Cardiovascular Molecular Biology and Regulatory PeptidesBeijing Key Laboratory of Cardiovascular Receptors ResearchPeking UniversityBeijing100191China
| | - Tao Zhang
- Department of Vascular SurgeryPeking University People's HospitalBeijing100044China
| | - Qin Peng
- Shenzhen Bay LaboratoryShenzhen518132China
| | - Jing Zhou
- Department of Physiology and PathophysiologySchool of Basic Medical SciencesState Key Laboratory of Vascular Homeostasis and RemodelingDepartment of Cardiology and Institute of Vascular MedicinePeking University Third HospitalNational Health Commission Key Laboratory of Cardiovascular Molecular Biology and Regulatory PeptidesBeijing Key Laboratory of Cardiovascular Receptors ResearchPeking UniversityBeijing100191China
| |
Collapse
|
2
|
Zhou Z, Chen W, Cao Y, Abdi R, Tao W. Nanomedicine-based strategies for the treatment of vein graft disease. Nat Rev Cardiol 2024. [DOI: 10.1038/s41569-024-01094-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/27/2024] [Indexed: 01/03/2025]
|
3
|
Haron NA, Ishak MF, Yazid MD, Vijakumaran U, Ibrahim R, Raja Sabudin RZA, Alauddin H, Md Ali NA, Haron H, Ismail MI, Abdul Rahman MR, Sulaiman N. Exploring the Potential of Saphenous Vein Grafts Ex Vivo: A Model for Intimal Hyperplasia and Re-Endothelialization. J Clin Med 2024; 13:4774. [PMID: 39200916 PMCID: PMC11355503 DOI: 10.3390/jcm13164774] [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: 06/20/2024] [Revised: 07/31/2024] [Accepted: 08/10/2024] [Indexed: 09/02/2024] Open
Abstract
Coronary artery bypass grafting (CABG) utilizing saphenous vein grafts (SVGs) stands as a fundamental approach to surgically treating coronary artery disease. However, the long-term success of CABG is often compromised by the development of intimal hyperplasia (IH) and subsequent graft failure. Understanding the mechanisms underlying this pathophysiology is crucial for improving graft patency and patient outcomes. Objectives: This study aims to explore the potential of an ex vivo model utilizing SVG to investigate IH and re-endothelialization. Methods: A thorough histological examination of 15 surplus SVG procured from CABG procedures at Hospital Canselor Tuanku Muhriz, Malaysia, was conducted to establish their baseline characteristics. Results: SVGs exhibited a mean diameter of 2.65 ± 0.93 mm with pre-existing IH averaging 0.42 ± 0.13 mm in thickness, alongside an observable lack of luminal endothelial cell lining. Analysis of extracellular matrix components, including collagen, elastin, and glycosaminoglycans, at baseline and after 7 days of ex vivo culture revealed no significant changes in collagen but demonstrated increased percentages of elastin and glycosaminoglycans. Despite unsuccessful attempts at re-endothelialization with blood outgrowth endothelial cells, the established ex vivo SVG IH model underscores the multifaceted nature of graft functionality and patency, characterized by IH presence, endothelial impairment, and extracellular matrix alterations post-CABG. Conclusions: The optimized ex vivo IH model provides a valuable platform for delving into the underlying mechanisms of IH formation and re-endothelialization of SVG. Further refinements are warranted, yet this model holds promise for future research aimed at enhancing graft durability and outcomes for CAD patients undergoing CABG.
Collapse
Affiliation(s)
- Nur A’tiqah Haron
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia (U.V.)
| | - Mohamad Fikeri Ishak
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia (U.V.)
| | - Muhammad Dain Yazid
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia (U.V.)
| | - Ubashini Vijakumaran
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia (U.V.)
| | - Roszita Ibrahim
- Department of Community Health, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia
| | - Raja Zahratul Azma Raja Sabudin
- Department of Pathology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia
- Department of Diagnostic Laboratory Services, Hospital Canselor Tuanku Muhriz, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia
| | - Hafiza Alauddin
- Department of Pathology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia
- Department of Diagnostic Laboratory Services, Hospital Canselor Tuanku Muhriz, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia
| | - Nur Ayub Md Ali
- Department of Surgery, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia
- Heart and Lung Centre, Hospital Canselor Tuanku Muhriz, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia
| | - Hairulfaizi Haron
- Department of Surgery, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia
- Heart and Lung Centre, Hospital Canselor Tuanku Muhriz, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia
| | - Muhammad Ishamuddin Ismail
- Heart and Lung Centre, Hospital Canselor Tuanku Muhriz, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia
| | - Mohd Ramzisham Abdul Rahman
- Department of Surgery, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia
- Heart and Lung Centre, Hospital Canselor Tuanku Muhriz, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia
| | - Nadiah Sulaiman
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia (U.V.)
| |
Collapse
|
4
|
Laparra-Escareño H, Ortega-Gómez A, Zentella-Dehesa A, Manzo-Merino J, Vergara-Ascencio CA, Antuñano-Blanco MDC, Lopez-Santacruz JR, Montalvo-Jave EE, Anaya-Ayala JE, Lozano-Corona R, Hinojosa CA. The effect of cilostazol on the platelet-derived growth factor-beta/beta isoform reduction on venous hyperplasia in an experimental balloon-induced injury model. Vascular 2024; 32:842-849. [PMID: 36911886 DOI: 10.1177/17085381231162160] [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: 03/14/2023]
Abstract
BACKGROUND Intimal hyperplasia is the response to endothelial injury. Platelet-derived growth factor is released early and favors the formation of intimal hyperplasia. Although multiple treatments, from open surgery to endovascular techniques, have been used they remain controversial. There is currently interest in developing pharmacological strategies to address this pathology. Local vascular inflammation induced by vessel barotrauma generates intimal hyperplasia due to mechanical stress over the venous endothelium. Cilostazol is a selective phosphodiesterase type 3 (PDE3) selective inhibitor with a regulatory effect over intimal hyperplasia. The objective was to investigate cilostazol's role in inhibiting smooth muscle cell proliferation due to changes in the expression and release of PDGF-BB isoform and the effect on developing IH using an experimental model of vascular barotrauma (balloon-induced injury model). METHODS We included 12 New Zealand rabbits. The balloon-induced injury model (BIIM) and experimental group cilostazol (20 mg/kg/day) included 6 rabbits each. Contralateral veins from 6 rabbits used in BIIM model has been taken as control group. We measured and compared the expression of PDGF-BB and the development of IH. A pathologist board chooses a PDGFRα antibody to localized its expression by immunohistochemistry analysis. Subsequently, using an automated immunohistochemical staining machine, the PDGFR expression was evaluated using a Zeiss Primo Star 4 light microscope. RESULTS The measurement obtained in the intimal layer was: 126.12 μm2 in the CG, 232 μm2 in the BIIM group, and 178 μm2 in the EG. A statistically significant difference was observed. Baseline serum concentrations of PDGF-BB in the BIIM group were 0.22 pg/mL. At 12 h 0.42 pg/mL, and 0.17 pg/mL at seven days. In the experimental group, the basal levels were 0.33 pg/mL. With the use of cilostazol, a lower peak was obtained at 12 h (0.08 pg/mL). This difference was statistically significant. CONCLUSIONS Cilostazol induced a significant reduction of IH caused by barotrauma in the venous endothelium, which correlates with decrease in the PDGF-BB in serum. This could be attributed to the pharmacologic effect on PDGFR expression.
Collapse
MESH Headings
- Cilostazol/pharmacology
- Animals
- Hyperplasia
- Disease Models, Animal
- Rabbits
- Vascular System Injuries/pathology
- Vascular System Injuries/metabolism
- Vascular System Injuries/drug therapy
- Becaplermin/pharmacology
- Cell Proliferation/drug effects
- Neointima
- Phosphodiesterase 3 Inhibitors/pharmacology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/metabolism
- Male
- Veins/drug effects
- Veins/metabolism
- Veins/pathology
- Down-Regulation
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/pathology
- Myocytes, Smooth Muscle/metabolism
- Angioplasty, Balloon/adverse effects
- Angioplasty, Balloon/instrumentation
- Protein Isoforms/metabolism
Collapse
Affiliation(s)
- Hugo Laparra-Escareño
- Department of Surgery, Section of Vascular Surgery and Endovascular Therapy, Instituto Nacional de Ciencias Medicas y Nutricion Salvador Zubiran, Ciudad de Mexico, Mexico
| | - Alette Ortega-Gómez
- Laboratory of Translational Medicine, National Institute of Cancerology, Ciudad de Mexico, Mexico
| | - Alejandro Zentella-Dehesa
- Department of Biochemistry, Instituto Nacional de Ciencias Medicas y Nutricion Salvador Zubiran, Ciudad de Mexico, Mexico
| | | | - Carlos Acxel Vergara-Ascencio
- Department of Surgery, Section of Vascular Surgery and Endovascular Therapy, Instituto Nacional de Ciencias Medicas y Nutricion Salvador Zubiran, Ciudad de Mexico, Mexico
| | | | - Jose Roberto Lopez-Santacruz
- School of Veterinary Medicine an Animal Husbandry and Pathology, Universidad Nacional Autonoma de Mexico, Ciudad de Mexico, Mexico
| | | | - Javier E Anaya-Ayala
- Department of Surgery, Section of Vascular Surgery and Endovascular Therapy, Instituto Nacional de Ciencias Medicas y Nutricion Salvador Zubiran, Ciudad de Mexico, Mexico
| | - Rodrigo Lozano-Corona
- Department of Surgery, Section of Vascular Surgery and Endovascular Therapy, Instituto Nacional de Ciencias Medicas y Nutricion Salvador Zubiran, Ciudad de Mexico, Mexico
| | - Carlos A Hinojosa
- Department of Surgery, Section of Vascular Surgery and Endovascular Therapy, Instituto Nacional de Ciencias Medicas y Nutricion Salvador Zubiran, Ciudad de Mexico, Mexico
| |
Collapse
|
5
|
Fan Y, Pei J, Qin Y, Du H, Qu X, Li W, Huang B, Tan J, Liu Y, Li G, Ke M, Xu Y, Zhu C. Construction of tissue-engineered vascular grafts with enhanced patency by integrating heparin, cell-adhesive peptide, and carbon monoxide nanogenerators into acellular blood vessels. Bioact Mater 2024; 34:221-236. [PMID: 38235307 PMCID: PMC10792202 DOI: 10.1016/j.bioactmat.2023.12.015] [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: 10/18/2023] [Revised: 12/11/2023] [Accepted: 12/21/2023] [Indexed: 01/19/2024] Open
Abstract
Small-diameter tissue-engineered vascular grafts (sdTEVGs) have garnered significant attention as a potential treatment modality for vascular bypass grafting and replacement therapy. However, the intimal hyperplasia and thrombosis are two major complications that impair graft patency during transplantation. To address this issue, we fabricated the covalent-organic framework (COF)-based carbon monoxide (CO) nanogenerator-and co-immobilized with LXW-7 peptide and heparin to establish a multifunctional surface on TEVGs constructed from acellular blood vessels for preventing thrombosis and stenosis. The cell-adhesive peptide LXW-7 could capture endothelial-forming cells (EFCs) to promote endothelialization, while the antithrombotic molecule heparin prevented thrombus formation. The reactive oxygen species (ROS)-triggered CO release suppressed the adhesion and activation of macrophages, leading to the reduction of ROS and inflammatory factors. As a result, the endothelial-to-mesenchymal transition (EndMT) triggered by inflammation was restricted, facilitating the maintenance of the homeostasis of the neo-endothelium and preventing pathological remodeling in TEVGs. When transplanted in vivo, these vascular grafts exhibited negligible intimal hyperplasia and remained patent for 3 months. This achievement provided a novel approach for constructing antithrombotic and anti-hyperplastic TEVGs.
Collapse
Affiliation(s)
- Yonghong Fan
- Department of Anatomy, Engineering Research Center for Organ Intelligent Biological Manufacturing of Chongqing, Key Lab for Biomechanics and Tissue Engineering of Chongqing, Third Military Medical University, Chongqing, 400038, China
- Laboratory of Basic Medicine, The General Hospital of Western Theater Command, Chengdu, 610083, China
- Engineering Research Center of Tissue and Organ Regeneration and Manufacturing, Ministry of Education, Chongqing, 400038, China
| | - Juan Pei
- Department of Anatomy, Engineering Research Center for Organ Intelligent Biological Manufacturing of Chongqing, Key Lab for Biomechanics and Tissue Engineering of Chongqing, Third Military Medical University, Chongqing, 400038, China
| | - Yinhua Qin
- Department of Anatomy, Engineering Research Center for Organ Intelligent Biological Manufacturing of Chongqing, Key Lab for Biomechanics and Tissue Engineering of Chongqing, Third Military Medical University, Chongqing, 400038, China
| | - Huifang Du
- Department of Anatomy, Engineering Research Center for Organ Intelligent Biological Manufacturing of Chongqing, Key Lab for Biomechanics and Tissue Engineering of Chongqing, Third Military Medical University, Chongqing, 400038, China
| | - Xiaohang Qu
- Department of Anatomy, Engineering Research Center for Organ Intelligent Biological Manufacturing of Chongqing, Key Lab for Biomechanics and Tissue Engineering of Chongqing, Third Military Medical University, Chongqing, 400038, China
| | - Wenya Li
- Department of Anatomy, Engineering Research Center for Organ Intelligent Biological Manufacturing of Chongqing, Key Lab for Biomechanics and Tissue Engineering of Chongqing, Third Military Medical University, Chongqing, 400038, China
| | - Boyue Huang
- Department of Anatomy, Engineering Research Center for Organ Intelligent Biological Manufacturing of Chongqing, Key Lab for Biomechanics and Tissue Engineering of Chongqing, Third Military Medical University, Chongqing, 400038, China
| | - Ju Tan
- Department of Anatomy, Engineering Research Center for Organ Intelligent Biological Manufacturing of Chongqing, Key Lab for Biomechanics and Tissue Engineering of Chongqing, Third Military Medical University, Chongqing, 400038, China
| | - Yong Liu
- Department of Anatomy, Engineering Research Center for Organ Intelligent Biological Manufacturing of Chongqing, Key Lab for Biomechanics and Tissue Engineering of Chongqing, Third Military Medical University, Chongqing, 400038, China
| | - Gang Li
- Department of Anatomy, Engineering Research Center for Organ Intelligent Biological Manufacturing of Chongqing, Key Lab for Biomechanics and Tissue Engineering of Chongqing, Third Military Medical University, Chongqing, 400038, China
| | - Ming Ke
- Department of Anatomy, Engineering Research Center for Organ Intelligent Biological Manufacturing of Chongqing, Key Lab for Biomechanics and Tissue Engineering of Chongqing, Third Military Medical University, Chongqing, 400038, China
- Engineering Research Center of Tissue and Organ Regeneration and Manufacturing, Ministry of Education, Chongqing, 400038, China
| | - Youqian Xu
- Department of Anatomy, Engineering Research Center for Organ Intelligent Biological Manufacturing of Chongqing, Key Lab for Biomechanics and Tissue Engineering of Chongqing, Third Military Medical University, Chongqing, 400038, China
- Engineering Research Center of Tissue and Organ Regeneration and Manufacturing, Ministry of Education, Chongqing, 400038, China
| | - Chuhong Zhu
- Department of Anatomy, Engineering Research Center for Organ Intelligent Biological Manufacturing of Chongqing, Key Lab for Biomechanics and Tissue Engineering of Chongqing, Third Military Medical University, Chongqing, 400038, China
- Engineering Research Center of Tissue and Organ Regeneration and Manufacturing, Ministry of Education, Chongqing, 400038, China
- State Key Laboratory of Trauma and Chemical Poisoning, Chongqing, 400038, China
- Department of Plastic and Aesthetic Surgery, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China
| |
Collapse
|
6
|
Russu E, Arbanasi EM, Chirila TV, Muresan AV. Therapeutic strategies based on non-ionizing radiation to prevent venous neointimal hyperplasia: the relevance for stenosed arteriovenous fistula, and the role of vascular compliance. Front Cardiovasc Med 2024; 11:1356671. [PMID: 38374996 PMCID: PMC10875031 DOI: 10.3389/fcvm.2024.1356671] [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: 12/18/2023] [Accepted: 01/23/2024] [Indexed: 02/21/2024] Open
Abstract
We have reviewed the development and current status of therapies based on exposure to non-ionizing radiation (with a photon energy less than 10 eV) aimed at suppressing the venous neointimal hyperplasia, and consequentially at avoiding stenosis in arteriovenous grafts. Due to the drawbacks associated with the medical use of ionizing radiation, prominently the radiation-induced cardiovascular disease, the availability of procedures using non-ionizing radiation is becoming a noteworthy objective for the current research. Further, the focus of the review was the use of such procedures for improving the vascular access function and assuring the clinical success of arteriovenous fistulae in hemodialysis patients. Following a brief discussion of the physical principles underlying radiotherapy, the current methods based on non-ionizing radiation, either in use or under development, were described in detail. There are currently five such techniques, including photodynamic therapy (PDT), far-infrared therapy, photochemical tissue passivation (PTP), Alucent vascular scaffolding, and adventitial photocrosslinking. The last three are contingent on the mechanical stiffening achievable by the exogenous photochemical crosslinking of tissular collagen, a process that leads to the decrease of venous compliance. As there are conflicting opinions on the role of compliance mismatch between arterial and venous conduits in a graft, this aspect was also considered in our review.
Collapse
Affiliation(s)
- Eliza Russu
- Clinic of Vascular Surgery, Mures County Emergency Hospital, Targu Mures, Romania
- Department of Vascular Surgery, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, Targu Mures, Romania
| | - Emil-Marian Arbanasi
- Clinic of Vascular Surgery, Mures County Emergency Hospital, Targu Mures, Romania
- Department of Vascular Surgery, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, Targu Mures, Romania
- Doctoral School of Medicine and Pharmacy, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, Targu Mures, Romania
- Centre for Advanced Medical and Pharmaceutical Research (CCAMF), George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, Targu Mures, Romania
| | - Traian V. Chirila
- Centre for Advanced Medical and Pharmaceutical Research (CCAMF), George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, Targu Mures, Romania
- Queensland Eye Institute, Woolloongabba, QLD, Australia
- Faculty of Medicine, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, Targu Mures, Romania
- School of Chemistry and Physics, Queensland University of Technology, Brisbane, QLD, Australia
- Australian Institute of Bioengineering and Nanotechnology (AIBN), University of Queensland, St Lucia, QLD, Australia
| | - Adrian V. Muresan
- Clinic of Vascular Surgery, Mures County Emergency Hospital, Targu Mures, Romania
- Department of Vascular Surgery, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, Targu Mures, Romania
| |
Collapse
|
7
|
Jiang QL, Xu JY, Yao QP, Jiang R, Xu Q, Zhang BT, Li T, Jiang J. Transfer RNA-derived small RNA tRF-Glu-CTC attenuates neointimal formation via inhibition of fibromodulin. Cell Mol Biol Lett 2024; 29:2. [PMID: 38172726 PMCID: PMC10763295 DOI: 10.1186/s11658-023-00523-z] [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: 08/02/2023] [Accepted: 12/13/2023] [Indexed: 01/05/2024] Open
Abstract
Neointimal hyperplasia is a pathological vascular remodeling caused by abnormal proliferation and migration of subintimal vascular smooth muscle cells (VSMCs) following intimal injury. There is increasing evidence that tRNA-derived small RNA (tsRNA) plays an important role in vascular remodeling. The purpose of this study is to search for tsRNAs signature of neointima formation and to explore their potential functions. The balloon injury model of rat common carotid artery was replicated to induce intimal hyperplasia, and the differentially expressed tsRNAs (DE-tsRNAs) in arteries with intimal hyperplasia were screened by small RNA sequencing and tsRNA library. A total of 24 DE-tsRNAs were found in the vessels with intimal hyperplasia by small RNA sequencing. In vitro, tRF-Glu-CTC inhibited the expression of fibromodulin (FMOD) in VSMCs, which is a negative modulator of TGF-β1 activity. tRF-Glu-CTC also increased VSMC proliferation and migration. In vivo experiments showed that inhibition of tRF-Glu-CTC expression after balloon injury of rat carotid artery can reduce the neointimal area. In conclusion, tRF-Glu-CTC expression is increased after vascular injury and inhibits FMOD expression in VSMCs, which influences neointima formation. On the other hand, reducing the expression of tRF-Glu-CTC after vascular injury may be a potential approach to prevent vascular stenosis.
Collapse
Affiliation(s)
- Qi-Lan Jiang
- Department of Clinical Nutrition, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, China
| | - Jia-Ying Xu
- Department of General Surgery (Thyroid Surgery), The Affiliated Hospital of Southwest Medical University, 25 Taiping Street, Jiangyang District, Luzhou, 646000, Sichuan Province, China
| | - Qing-Ping Yao
- Institute of Mechanobiology and Medical Engineering, School of Life Science and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Rui Jiang
- Department of Urology, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, China
| | - Qin Xu
- Department of General Surgery (Thyroid Surgery), The Affiliated Hospital of Southwest Medical University, 25 Taiping Street, Jiangyang District, Luzhou, 646000, Sichuan Province, China
| | - Bo-Tao Zhang
- Department of General Surgery (Thyroid Surgery), The Affiliated Hospital of Southwest Medical University, 25 Taiping Street, Jiangyang District, Luzhou, 646000, Sichuan Province, China
| | - Tao Li
- Key Laboratory of Medical Electrophysiology of Ministry of Education, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, 646000, Sichuan Province, China.
| | - Jun Jiang
- Department of General Surgery (Thyroid Surgery), The Affiliated Hospital of Southwest Medical University, 25 Taiping Street, Jiangyang District, Luzhou, 646000, Sichuan Province, China.
| |
Collapse
|
8
|
Wang H, Xiao Y, Fang Z, Zhang Y, Yang L, Zhao C, Meng Z, Liu Y, Li C, Han Q, Feng Z. Fabrication and performance evaluation of PLCL-hCOLIII small-diameter vascular grafts crosslinked with procyanidins. Int J Biol Macromol 2023; 251:126293. [PMID: 37591423 DOI: 10.1016/j.ijbiomac.2023.126293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 08/01/2023] [Accepted: 08/10/2023] [Indexed: 08/19/2023]
Abstract
Cardiovascular disease has become one of the main causes of death. It is the common goal of researchers worldwide to develop small-diameter vascular grafts to meet clinical needs. Collagen is a valuable biomaterial that has been used in the preparation of vascular grafts and has shown good results. Recombinant humanized collagen (RHC) has the advantages of clear chemical structure, batch stability, no virus hazard and low immunogenicity compared with animal-derived collagen, which can be developed as vascular materials. In this study, Poly (l-lactide- ε-caprolactone) with l-lactide/ε-caprolactone (PLCL) and type III recombinant humanized collagen (hCOLIII) were selected as raw materials to prepare vascular grafts, which were prepared by the same-nozzle electrospinning apparatus. Meanwhile, procyanidin (PC), a plant polyphenol, was used to cross-link the vascular grafts. The physicochemical properties and biocompatibility of the fabricated vascular grafts were investigated by comparing with glutaraldehyde (GA) crosslinked vascular grafts and pure PLCL grafts. Finally, the performance of PC cross-linked PLCL-hCOLIII vascular grafts were evaluated by rabbit carotid artery transplantation model. The results indicate that the artificial vascular grafts have good cell compatibility, blood compatibility, and anti-calcification performance, and can remain unobstructed after 30 days carotid artery transplantation in rabbits. The grafts also showed inhibitory effects on the proliferation of SMCs and intimal hyperplasia, demonstrating its excellent performance as small diameter vascular grafts.
Collapse
Affiliation(s)
- Han Wang
- School of Materials Science and Engineering, Beijing Institute of Technology, Beijing, 100081, China; National Institute for Food and Drug Control, Beijing 102629, China
| | - Yonghao Xiao
- School of Materials Science and Engineering, Beijing Institute of Technology, Beijing, 100081, China
| | - Zhiping Fang
- School of Materials Science and Engineering, Beijing Institute of Technology, Beijing, 100081, China
| | - Yuanguo Zhang
- Department of Thyroid-Breast-Vascular Surgery, Shanxian Central Hospital, Heze, Shandong 274300, China
| | - Liu Yang
- National Institute for Food and Drug Control, Beijing 102629, China
| | - Chenyu Zhao
- National Institute for Food and Drug Control, Beijing 102629, China
| | - Zhu Meng
- National Institute for Food and Drug Control, Beijing 102629, China
| | - Yu Liu
- National Institute for Food and Drug Control, Beijing 102629, China; Yantai University, Yantai, Shandong 264005, China
| | - Chongchong Li
- National Institute for Food and Drug Control, Beijing 102629, China
| | - Qianqian Han
- National Institute for Food and Drug Control, Beijing 102629, China.
| | - Zengguo Feng
- School of Materials Science and Engineering, Beijing Institute of Technology, Beijing, 100081, China.
| |
Collapse
|
9
|
Ghandakly EC, Tipton AE, Bakaeen FG. Pathophysiology and management of saphenous vein graft disease. Expert Rev Cardiovasc Ther 2023; 21:565-572. [PMID: 37540160 DOI: 10.1080/14779072.2023.2233420] [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: 04/25/2023] [Accepted: 06/29/2023] [Indexed: 08/05/2023]
Abstract
INTRODUCTION The saphenous vein graft (SVG) is the most used conduit in CABG. With standardization of its use as a conduit came an understanding of its accelerated atherosclerosis, known as saphenous vein graft disease (SVGD). Given its extensive use, a review of the pathophysiology and management of SVGD is important as we optimize its use. AREAS COVERED For this review, an extensive literature search was completed to identify and examine the evolution of SVG in CABG, mechanisms driving SVGD, and methods developed to prevent and manage it. This includes a review of relevant major papers and trials in this space. EXPERT OPINION Eras of evolution in SVG usage in CABG include an experimental era, era of SVG dominance in CABG, and the current era of mixed venous and arterial grafting. As SVGD was studied, the mechanisms behind it became more understood, and prevention and management methods were developed. As advances in surgical techniques and pharmacotherapy continue to reduce occurrence and severity of SVGD, long-term patency of SV grafts continues to improve and remain excellent in optimized settings. With continued innovation and improvement in operative techniques, the SVG conduit is and will remain an important player in the field of coronary bypass.
Collapse
Affiliation(s)
- Elizabeth C Ghandakly
- Department of Thoracic and Cardiovascular Surgery; Heart, Vascular, & Thoracic Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Aaron E Tipton
- Department of Thoracic and Cardiovascular Surgery; Heart, Vascular, & Thoracic Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Faisal G Bakaeen
- Department of Thoracic and Cardiovascular Surgery; Heart, Vascular, & Thoracic Institute, Cleveland Clinic, Cleveland, Ohio, USA
| |
Collapse
|
10
|
Kim JH, Jang EH, Ryu JY, Lee J, Kim JH, Ryu W, Youn YN. Sirolimus-Embedded Silk Microneedle Wrap to Prevent Neointimal Hyperplasia in Vein Graft Model. Int J Mol Sci 2023; 24:ijms24043306. [PMID: 36834717 PMCID: PMC9967879 DOI: 10.3390/ijms24043306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 02/03/2023] [Accepted: 02/06/2023] [Indexed: 02/11/2023] Open
Abstract
We investigated the role of a sirolimus-embedded silk microneedle (MN) wrap as an external vascular device for drug delivery efficacy, inhibition of neointimal hyperplasia, and vascular remodeling. Using dogs, a vein graft model was developed to interpose the carotid or femoral artery with the jugular or femoral vein. The control group contained four dogs with only interposed grafts; the intervention group contained four dogs with vein grafts in which sirolimus-embedded silk-MN wraps were applied. After 12-weeks post-implantation, 15 vein grafts in each group were explanted and analyzed. Vein grafts applied with the rhodamine B-embedded silk-MN wrap showed far higher fluorescent signals than those without the wrap. The diameter of vein grafts in the intervention group decreased or remained stable without dilatation; however, it increased in the control group. The intervention group had femoral vein grafts with a significantly lower mean neointima-to-media ratio, and had vein grafts with an intima layer showing a significantly lower collagen density ratio than the control group. In conclusion, sirolimus-embedded silk-MN wrap in a vein graft model successfully delivered the drug to the intimal layer of the vein grafts. It prevented vein graft dilatation, avoiding shear stress and decreasing wall tension, and it inhibited neointimal hyperplasia.
Collapse
Affiliation(s)
- Jung-Hwan Kim
- Division of Cardiovascular Surgery, Department of Thoracic and Cardiovascular Surgery, Severance Cardiovascular Hospital, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Eui Hwa Jang
- Division of Cardiovascular Surgery, Department of Thoracic and Cardiovascular Surgery, Severance Cardiovascular Hospital, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Ji-Yeon Ryu
- Division of Cardiovascular Surgery, Department of Thoracic and Cardiovascular Surgery, Severance Cardiovascular Hospital, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Jiyong Lee
- School of Mechanical Engineering, Yonsei University, Seoul 03722, Republic of Korea
| | - Jae Ho Kim
- School of Mechanical Engineering, Yonsei University, Seoul 03722, Republic of Korea
| | - Wonhyoung Ryu
- School of Mechanical Engineering, Yonsei University, Seoul 03722, Republic of Korea
| | - Young-Nam Youn
- Division of Cardiovascular Surgery, Department of Thoracic and Cardiovascular Surgery, Severance Cardiovascular Hospital, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
- Correspondence: ; Tel.: +82-2-2228-8487
| |
Collapse
|
11
|
Kanaan R, Medlej-Hashim M, Jounblat R, Pilecki B, Sorensen GL. Microfibrillar-associated protein 4 in health and disease. Matrix Biol 2022; 111:1-25. [DOI: 10.1016/j.matbio.2022.05.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 05/04/2022] [Accepted: 05/24/2022] [Indexed: 10/18/2022]
|
12
|
Failure Analysis of TEVG’s II: Late Failure and Entering the Regeneration Pathway. Cells 2022; 11:cells11060939. [PMID: 35326390 PMCID: PMC8946846 DOI: 10.3390/cells11060939] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 02/03/2022] [Accepted: 03/01/2022] [Indexed: 12/21/2022] Open
Abstract
Tissue-engineered vascular grafts (TEVGs) are a promising alternative to treat vascular disease under complex hemodynamic conditions. However, despite efforts from the tissue engineering and regenerative medicine fields, the interactions between the material and the biological and hemodynamic environment are still to be understood, and optimization of the rational design of vascular grafts is an open challenge. This is of special importance as TEVGs not only have to overcome the surgical requirements upon implantation, they also need to withhold the inflammatory response and sustain remodeling of the tissue. This work aims to analyze and evaluate the bio-molecular interactions and hemodynamic phenomena between blood components, cells and materials that have been reported to be related to the failure of the TEVGs during the regeneration process once the initial stages of preimplantation have been resolved, in order to tailor and refine the needed criteria for the optimal design of TEVGs.
Collapse
|
13
|
Wang K, Gao H, Zhang Y, Yan H, Si J, Mi X, Xia S, Feng X, Liu D, Kong D, Wang T, Ding D. Highly Bright AIE Nanoparticles by Regulating the Substituent of Rhodanine for Precise Early Detection of Atherosclerosis and Drug Screening. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2106994. [PMID: 34921573 DOI: 10.1002/adma.202106994] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 12/15/2021] [Indexed: 06/14/2023]
Abstract
Fluorescent probes capable of precise detection of atherosclerosis (AS) at an early stage and fast assessment of anti-AS drugs in animal level are particularly valuable. Herein, a highly bright aggregation-induced emission (AIE) nanoprobe is introduced by regulating the substituent of rhodanine for early detection of atherosclerotic plaque and screening of anti-AS drugs in a precise, sensitive, and rapid manner. With dicyanomethylene-substituted rhodanine as the electron-withdrawing unit, the AIE luminogen named TPE-T-RCN shows the highest molar extinction coefficient, the largest photoluminescence quantum yield, and the most redshifted absorption/emission spectra simultaneously as compared to the control compounds. The nanoprobes are obtained with an amphiphilic copolymer as the matrix encapsulating TPE-T-RCN molecules, which are further surface functionalized with anti-CD47 antibody for specifically binding to CD47 overexpressed in AS plaques. Such nanoprobes allow efficient recognition of AS plaques at different stages in apolipoprotein E-deficient (apoE-/- ) mice, especially for the recognition of early-stage AS plaques prior to micro-computed tomography (CT) and magnetic resonance imaging (MRI). These features impel to apply the nanoprobes in monitoring the therapeutic effects of anti-AS drugs, providing a powerful tool for anti-AS drug screening. Their potential use in targeted imaging of human carotid plaque is further demonstrated.
Collapse
Affiliation(s)
- Kai Wang
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, Frontiers Science Center for Cell Responses, and State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China
| | - Heqi Gao
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, Frontiers Science Center for Cell Responses, and State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China
| | - Yuwen Zhang
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, Frontiers Science Center for Cell Responses, and State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China
| | - Hongyu Yan
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, Frontiers Science Center for Cell Responses, and State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China
| | - Jianghua Si
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, Frontiers Science Center for Cell Responses, and State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China
| | - Xingyan Mi
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, Frontiers Science Center for Cell Responses, and State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China
| | - Shuang Xia
- Department of Radiology, Tianjin First Central Hospital, Tianjin, 300192, China
| | - Xuequan Feng
- Department of Neurosurgery, Tianjin First Central Hospital, Tianjin, 300192, China
| | - Dingbin Liu
- College of Chemistry, Research Center for Analytical Sciences, State Key Laboratory of Medicinal Chemical Biology, and Tianjin Key Laboratory of Molecular Recognition and Biosensing, Nankai University, Tianjin, 300071, China
| | - Deling Kong
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, Frontiers Science Center for Cell Responses, and State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China
| | - Ting Wang
- Tianjin Key Laboratory of Urban Transport Emission Research, College of Environmental Science and Engineering, Nankai University, Tianjin, 300071, China
| | - Dan Ding
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, Frontiers Science Center for Cell Responses, and State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China
- Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, Tianjin Stomatological Hospital, The Affiliated Stomatological Hospital of Nankai University, Tianjin, 300041, China
| |
Collapse
|
14
|
McQueen LW, Ladak SS, Zakkar M. Acute shear stress and vein graft disease. Int J Biochem Cell Biol 2022; 144:106173. [PMID: 35151879 DOI: 10.1016/j.biocel.2022.106173] [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: 08/19/2021] [Revised: 12/08/2021] [Accepted: 02/08/2022] [Indexed: 11/30/2022]
Abstract
The long saphenous vein is commonly used in cardiac surgery to bypass occluded coronary arteries. Its use is complicated by late stenosis and occlusion due to the development of intimal hyperplasia. It is accepted that intimal hyperplasia is a multifactorial inflammatory process that starts immediately after surgery. The role of acute changes in haemodynamic conditions when the vein is implanted into arterial circulation, especially shear stress, is not fully appreciated. This review provides an overview of intimal hyperplasia and the effect of acute shear stress changes on the activation of pro-inflammatory mediators.
Collapse
Affiliation(s)
- Liam W McQueen
- Department of Cardiovascular Sciences, Clinical Science Wing, University of Leicester, Glenfield Hospital, Leicester, UK
| | - Shameem S Ladak
- Department of Cardiovascular Sciences, Clinical Science Wing, University of Leicester, Glenfield Hospital, Leicester, UK
| | - Mustafa Zakkar
- Department of Cardiovascular Sciences, Clinical Science Wing, University of Leicester, Glenfield Hospital, Leicester, UK.
| |
Collapse
|
15
|
Qiu J, Shu C, Li S, Xiong Q, Wang L, Liu Z, Li X, Zhang W. Radiotherapy inhibits neointimal hyperplasia after artificial vascular replacement through Skp2/P27kip1. JOURNAL OF RADIATION RESEARCH 2022; 63:36-43. [PMID: 34788457 PMCID: PMC8776690 DOI: 10.1093/jrr/rrab089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 08/09/2021] [Indexed: 06/13/2023]
Abstract
We aimed to establish an animal model of abdominal aortic vascular replacement in mongrel dogs to investigate the effect of extracorporeal radiotherapy on the intima. Twenty healthy mongrel dogs were randomly divided into four groups: 5-week control group, 5-week radiotherapy group, 10-week control group and 10-week radiotherapy group. We first performed an artificial vascular replacement of the abdominal aortic segment. The radiotherapy group received external radiotherapy with a dose of 7 Gy for 4 days. The thickness of neointimal hyperplasia, immunoreactivity and expression of proliferation-related factors were detected by hematoxylin and eosin (HE) staining, immunohistochemistry, quantitative real-time polymerase chain reaction (qRT-PCR )and western blotting at 5 and 10 weeks after the reconstruction. The results showed that the intimal thickness of the artificial blood vessel in the 5- and 10-week radiotherapy groups was thinner than that in the control groups by HE staining. The immunoreactivity and expression levels of Skp2, c-Myc and CyclinE1 were significantly decreased in the radiotherapy groups than those in control groups by immunohistochemistry, qRT-PCR and western blotting. On the contrary, immunoreactivity and expression levels of P27kip1 were increased. In conclusion, we discovered that postoperative external radiotherapy significantly decreases the intimal hyperplasia of artificial blood vessels by regulating c-Myc-Skp2-P27-CyclinE1 network.
Collapse
Affiliation(s)
| | - Chang Shu
- Corresponding author. Vascular Surgery Department, The Second XiangYa hospital, Central South University, Changsha 410011, China. Tel: 86-0731-85295132;
| | | | | | | | | | | | | |
Collapse
|
16
|
Zhuang Y, Zhang C, Cheng M, Huang J, Liu Q, Yuan G, Lin K, Yu H. Challenges and strategies for in situ endothelialization and long-term lumen patency of vascular grafts. Bioact Mater 2021; 6:1791-1809. [PMID: 33336112 PMCID: PMC7721596 DOI: 10.1016/j.bioactmat.2020.11.028] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Revised: 11/11/2020] [Accepted: 11/24/2020] [Indexed: 02/08/2023] Open
Abstract
Vascular diseases are the most prevalent cause of ischemic necrosis of tissue and organ, which even result in dysfunction and death. Vascular regeneration or artificial vascular graft, as the conventional treatment modality, has received keen attentions. However, small-diameter (diameter < 4 mm) vascular grafts have a high risk of thrombosis and intimal hyperplasia (IH), which makes long-term lumen patency challengeable. Endothelial cells (ECs) form the inner endothelium layer, and are crucial for anti-coagulation and thrombogenesis. Thus, promoting in situ endothelialization in vascular graft remodeling takes top priority, which requires recruitment of endothelia progenitor cells (EPCs), migration, adhesion, proliferation and activation of EPCs and ECs. Chemotaxis aimed at ligands on EPC surface can be utilized for EPC homing, while nanofibrous structure, biocompatible surface and cell-capturing molecules on graft surface can be applied for cell adhesion. Moreover, cell orientation can be regulated by topography of scaffold, and cell bioactivity can be modulated by growth factors and therapeutic genes. Additionally, surface modification can also reduce thrombogenesis, and some drug release can inhibit IH. Considering the influence of macrophages on ECs and smooth muscle cells (SMCs), scaffolds loaded with drugs that can promote M2 polarization are alternative strategies. In conclusion, the advanced strategies for enhanced long-term lumen patency of vascular grafts are summarized in this review. Strategies for recruitment of EPCs, adhesion, proliferation and activation of EPCs and ECs, anti-thrombogenesis, anti-IH, and immunomodulation are discussed. Ideal vascular grafts with appropriate surface modification, loading and fabrication strategies are required in further studies.
Collapse
Affiliation(s)
- Yu Zhuang
- Department of Oral and Cranio-maxillofacial Surgery, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- National Clinical Research Center for Oral Diseases, Shanghai, 200011, China
- Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, 200011, China
| | - Chenglong Zhang
- Department of Oral and Cranio-maxillofacial Surgery, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- National Clinical Research Center for Oral Diseases, Shanghai, 200011, China
- Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, 200011, China
| | - Mengjia Cheng
- Department of Oral and Cranio-maxillofacial Surgery, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- National Clinical Research Center for Oral Diseases, Shanghai, 200011, China
- Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, 200011, China
| | - Jinyang Huang
- Department of Oral and Cranio-maxillofacial Surgery, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- National Clinical Research Center for Oral Diseases, Shanghai, 200011, China
- Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, 200011, China
| | - Qingcheng Liu
- Department of Oral and Cranio-maxillofacial Surgery, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- National Clinical Research Center for Oral Diseases, Shanghai, 200011, China
- Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, 200011, China
| | - Guangyin Yuan
- National Engineering Research Center of Light Alloy Net Forming & State Key Laboratory of Metal Matrix Composite, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China
| | - Kaili Lin
- Department of Oral and Cranio-maxillofacial Surgery, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- National Clinical Research Center for Oral Diseases, Shanghai, 200011, China
- Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, 200011, China
| | - Hongbo Yu
- Department of Oral and Cranio-maxillofacial Surgery, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- National Clinical Research Center for Oral Diseases, Shanghai, 200011, China
- Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, 200011, China
| |
Collapse
|
17
|
Guo Y, Zhu F, Zhang X, Wu G, Fu P, Yang J. Extracellular signal-regulated kinase inhibition prevents venous adaptive remodeling via regulation of Eph-B4. Vascular 2021; 30:120-129. [PMID: 33706642 DOI: 10.1177/1708538121999854] [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: 11/16/2022]
Abstract
OBJECTIVES Vein graft adaptation (VGA) is a process that vein as a vascular graft conduits in arterial reconstructive surgery; VGA can lead to postoperative vein graft stenosis (VGS) and complications after coronary artery bypass graft and other peripheral artery bypass surgeries. VGA is characterized by vein graft loss the venous features without exhibiting arterial features; furthermore, the activation of ERK inhibited the maintenance of venous properties of the vein graft. We hypothesized that ERK inhibition can affect vein VGS through regulating the expression of EphB4. METHODS Rat vein transplantation model was established using wild-type and EphB4+/- Sprague-Dawley rats. Hematoxylin-eosin, Masson, Verhoeff, actin staining, and immunohistochemistry were applied to observe the structure of the vein grafts. Vascular smooth muscle cells (VSMCs) were isolated from the vein and vein grafts. Western blotting was used to determine the expression of p-ERK1/2 and EphB4, and immunofluorescence was applied to detect the expression and location of EphB4. Cell wound scratch assay and CCK8 assay were used to determine the migration and proliferation of VSMCs. Real-time polymerase chain reaction was used to determine the mRNA expression of EphB4. RESULTS Western blotting in vein sample and vein graft sample detected p-ERK1/2 and ERK1/2 expression in both EphB4+/+ and EphB4+/- rats. The expression of p-ERK was increased in vein graft compared to vein. Immunofluorescence in VSMCs form EphB4+/+ and EphB4+/- rats detected EphB4 expression in both cells, and the expression of EphB4 was increased in VSMCs form EphB4+/+ rats. SCH772984 reduces the proliferation and migration of VSMCs. Inhibition of ERK suppressed the increase of vein graft wall thickness, and the expression of collagen fibers, elastic fibers, and α-actin was decreased. Vein graft from EphB4+/- rats reduces the expression of EphB4, and SCH772984 suppressed the decrease of EphB4 in vivo. Vein graft from EphB4+/- rats increased the expression of EphB4, and SCH772984 suppressed the increase of EphB4 in vivo. CONCLUSIONS The inhibition of ERK1/2 suppressed the process of VGS by decreasing the proliferation of VSMCs. The ERK-inhibitor SCH772984 suppressed the level of VGS by extending the time of EphB4 expression during the process of VGA, thus maintaining the venousization of vein graft. The mechanism may be that the inhibitor SCH772984 suppresses the level of VGS by extending the time of EphB4 expression during the process of VGA. Therefore, our research provides a new target of VGS treatment by inhibiting the expression of ERK1/2 through the process of VGA.
Collapse
Affiliation(s)
- Yuanyuan Guo
- Department of Vascular Surgery, Fuwai Yunnan Cardiovascular Hospital, Kunming, China.,Department of Vascular Surgery, Affiliated Cardiovascular Hospital of Kunming Medical University, Kunming, China
| | - Fan Zhu
- Department of Vascular Surgery, Fuwai Yunnan Cardiovascular Hospital, Kunming, China.,Department of Vascular Surgery, Affiliated Cardiovascular Hospital of Kunming Medical University, Kunming, China
| | - Xiong Zhang
- Department of Vascular Surgery, Fuwai Yunnan Cardiovascular Hospital, Kunming, China.,Department of Vascular Surgery, Affiliated Cardiovascular Hospital of Kunming Medical University, Kunming, China
| | - Guangmin Wu
- Department of Vascular Surgery, Fuwai Yunnan Cardiovascular Hospital, Kunming, China.,Department of Vascular Surgery, Affiliated Cardiovascular Hospital of Kunming Medical University, Kunming, China
| | - Pinting Fu
- Department of Vascular Surgery, Fuwai Yunnan Cardiovascular Hospital, Kunming, China.,Department of Vascular Surgery, Affiliated Cardiovascular Hospital of Kunming Medical University, Kunming, China
| | - Jun Yang
- Department of Vascular Surgery, Fuwai Yunnan Cardiovascular Hospital, Kunming, China.,Department of Vascular Surgery, Affiliated Cardiovascular Hospital of Kunming Medical University, Kunming, China
| |
Collapse
|
18
|
Chen H, Wang J, Xie L, Shen YL, Wang HM, Zheng KL, Zhang Q. Correlation between serum cartilage oligomeric matrix protein and major adverse cardiovascular events within 30 days in patients with acute coronary syndrome. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:353. [PMID: 33708980 PMCID: PMC7944313 DOI: 10.21037/atm-21-333] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Background We studied the correlation between cartilage oligomeric matrix protein (COMP) and major adverse cardiovascular events in patients with acute coronary syndrome (ACS) within 30 days. Methods This study included 170 ACS patients who were hospitalized in the Second Affiliated Hospital of Nantong University from August 2017 to April 2019. Serum COMP level was measured at baseline. The enrolled patients were followed up for 30 days and grouped according to the occurrence of major adverse cardiovascular events (MACE) during follow-up. Among the 170 patients, 23 patients had MACE during hospitalization (MACE group), and 147 patients had no MACE (no MACE group). Results The serum COMP levels in the MACE group were significantly higher than those of the non-MACE group [84.85 (51.55, 141.75) vs. 20.65 (9.11, 46.31) ng/mL, respectively, P<0.05]. The area under the receiver operating characteristic (ROC) curve for COMP in predicting the occurrence of MACE within 30 days was 0.839, with a cutoff level of 39.9 ng/mL [95% confidence interval (CI): 0.774–0.890], 86.96% sensitivity, and 72.79% specificity (P<0.0001). Multivariate logistic regression analysis showed that serum COMP could be used as an independent predictor of MACE within 30 days in ACS patients [odds ratio (OR): 1.024, 95% CI: 1.0133–1.0349, P=0.0001]. Conclusions Serum COMP is associated with the short-term prognosis of ACS patients. High serum COMP levels can be used as a predictor of MACE within 30 days in ACS patients.
Collapse
Affiliation(s)
- Hao Chen
- Department of Cardiology, The Second Affiliated Hospital of Nantong University, Nantong, China
| | - Jing Wang
- Department of Cardiology, The Second Affiliated Hospital of Nantong University, Nantong, China
| | - Ling Xie
- Department of Cardiology, The Second Affiliated Hospital of Nantong University, Nantong, China
| | - Ya-Li Shen
- Department of Cardiology, The Second Affiliated Hospital of Nantong University, Nantong, China
| | - Hui-Min Wang
- Department of Cardiology, The Second Affiliated Hospital of Nantong University, Nantong, China
| | - Kou-Long Zheng
- Department of Cardiology, The Second Affiliated Hospital of Nantong University, Nantong, China
| | - Qing Zhang
- Department of Cardiology, The Second Affiliated Hospital of Nantong University, Nantong, China
| |
Collapse
|
19
|
Lei Y, Xu J, Li M, Meng T, Chen M, Yang Y, Li H, Zhuang T, Zuo J. MIA SH3 Domain ER Export Factor 3 Deficiency Prevents Neointimal Formation by Restoring BAT-Like PVAT and Decreasing VSMC Proliferation and Migration. Front Endocrinol (Lausanne) 2021; 12:748216. [PMID: 34858331 PMCID: PMC8631732 DOI: 10.3389/fendo.2021.748216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 09/30/2021] [Indexed: 11/29/2022] Open
Abstract
Abnormal proliferation and migration of vascular smooth muscle cells (VSMCs) and excessive accumulation of dysfunctional PVAT are hallmarks of pathogenesis after angioplasty. Recent genome-wide association studies reveal that single-nucleotide polymorphism (SNP) in MIA3 is associated with atherosclerosis-relevant VSMC phenotypes. However, the role of MIA3 in the vascular remodeling response to injury remains unknown. Here, we found that expression of MIA3 is increased in proliferative VSMCs and knockdown of MIA3 reduces VSMCs proliferation, migration, and inflammation, whereas MIA3 overexpression promoted VSMC migration and proliferation. Moreover, knockdown of MIA3 ameliorates femoral artery wire injury-induced neointimal hyperplasia and increases brown-like perivascular adipocytes. Collectively, the data suggest that MIA3 deficiency prevents neointimal formation by decreasing VSMC proliferation, migration, and inflammation and maintaining BAT-like perivascular adipocytes in PVAT during injury-induced vascular remodeling, which provide a potential therapeutic target for preventing neointimal hyperplasia in proliferative vascular diseases.
Collapse
Affiliation(s)
- Yu Lei
- Department of Geriatrics and Geriatrics Center, Ruijin Hospital, Jiaotong University School of Medicine, Shanghai, China
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
- Jinshan Hospital Centre for Tumor Diagnosis and Therapy, Jinshan Hospital, Fudan University, Shanghai, China
| | - Jianfei Xu
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Mengju Li
- Department of Anesthesiology, Anting Hospital, Jiading District, Shanghai, China
| | - Ting Meng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Meihua Chen
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yongfeng Yang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Hongda Li
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Tao Zhuang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
- Jinshan Hospital Centre for Tumor Diagnosis and Therapy, Jinshan Hospital, Fudan University, Shanghai, China
- *Correspondence: Junli Zuo, ; Tao Zhuang,
| | - Junli Zuo
- Department of Geriatrics and Geriatrics Center, Ruijin Hospital, Jiaotong University School of Medicine, Shanghai, China
- *Correspondence: Junli Zuo, ; Tao Zhuang,
| |
Collapse
|
20
|
miRNA-126-3p carried by human umbilical cord mesenchymal stem cell enhances endothelial function through exosome-mediated mechanisms in vitro and attenuates vein graft neointimal formation in vivo. Stem Cell Res Ther 2020; 11:464. [PMID: 33138861 PMCID: PMC7607661 DOI: 10.1186/s13287-020-01978-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 10/16/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND The aim of this study was to determine whether the combination of MSC implantation with miRNA-126-3p overexpression would further improve the surgical results after vein grafting. METHODS human umbilical cord MSCs (hucMSCs) and human umbilical vein endothelial cells (HUVECs) were isolated from human umbilical cords and characterized by a series of experiments. Lentivirus vector encoding miRNA-126-3p was transfected into hucMSCs and verified by PCR. We analyzed the miRNA-126-3p-hucMSC function in vascular endothelial cells by using a series of co-culture experiments. miRNA-126-3p-hucMSCs-exosomes were separated from cell culture supernatants and identified by WB and TEM. We validated the role of miRNA-126-3p-hucMSCs-exosomes on HUVECs proliferative and migratory and angiogenic activities by using a series of function experiments. We further performed co-culture experiments to detect downstream target genes and signaling pathways of miRNA-126-3p-hucMSCs in HUVECs. We established a rat vein grafting model, CM-Dil-labeled hucMSCs were injected intravenously into rats, and the transplanted cells homing to the vein grafts were detected by fluorescent microscopy. We performed historical and immunohistochemical experiments to exam miRNA-126-3p-hucMSC transplantation on vein graft neointimal formation and reendothelialization in vitro. RESULTS We successfully isolated and identified primary hucMSCs and HUVECs. Primary hucMSCs were transfected with lentiviral vectors carrying miRNA-126-3p at a MOI 75. Co-culture studies indicated that overexpression of miRNA-126-3p in hucMSCs enhanced HUVECs proliferation, migration, and tube formation in vivo. We successfully separated hucMSCs-exosomes and found that miRNA-126-3p-hucMSCs-exosomes can strengthen the proliferative, migratory, and tube formation capacities of HUVECs. Further PCR and WB analysis indicated that, SPRED-1/PIK3R2/AKT/ERK1/2 pathways are involved in this process. In the rat vein arterialization model, reendothelialization analysis showed that transplantation with hucMSCs modified with miRNA-126-3p had a higher reendothelialization of the vein grafts. The subsequent historical and immunohistochemical examination revealed that delivery with miRNA-126-3p overexpressed hucMSCs significantly reduced vein graft intimal hyperplasia in rats. CONCLUSION These results suggest hucMSC-based miRNA-126-3p gene therapy may be a novel option for the treatment of vein graft disease after CABG.
Collapse
|
21
|
Karthika CL, Ahalya S, Radhakrishnan N, Kartha CC, Sumi S. Hemodynamics mediated epigenetic regulators in the pathogenesis of vascular diseases. Mol Cell Biochem 2020; 476:125-143. [PMID: 32844345 DOI: 10.1007/s11010-020-03890-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 08/14/2020] [Indexed: 12/19/2022]
Abstract
Endothelium of blood vessels is continuously exposed to various hemodynamic forces. Flow-mediated epigenetic plasticity regulates vascular endothelial function. Recent studies have highlighted the significant role of mechanosensing-related epigenetics in localized endothelial dysfunction and the regional susceptibility for lesions in vascular diseases. In this article, we review the epigenetic mechanisms such as DNA de/methylation, histone modifications, as well as non-coding RNAs in promoting endothelial dysfunction in major arterial and venous diseases, consequent to hemodynamic alterations. We also discuss the current challenges and future prospects for the use of mechanoepigenetic mediators as biomarkers of early stages of vascular diseases and dysregulated mechanosensing-related epigenetic regulators as therapeutic targets in various vascular diseases.
Collapse
Affiliation(s)
- C L Karthika
- Cardiovascular Diseases and Diabetes Biology, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, 695014, India
| | - S Ahalya
- Cardiovascular Diseases and Diabetes Biology, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, 695014, India
| | - N Radhakrishnan
- St.Thomas Institute of Research on Venous Diseases, Changanassery, Kerala, India
| | - C C Kartha
- Society for Continuing Medical Education & Research (SOCOMER), Kerala Institute of Medical Sciences, Thiruvananthapuram, Kerala, India
| | - S Sumi
- Cardiovascular Diseases and Diabetes Biology, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, 695014, India.
| |
Collapse
|
22
|
Abstract
Revascularization surgeries such as coronary artery bypass grafting (CABG) are sometimes necessary to manage coronary heart disease (CHD). However, more than half of these surgeries fail within 10 years due to the development of intimal hyperplasia (IH) among others. The cytokine transforming growth factor-beta (TGFß) and its signaling components have been found to be upregulated in diseased or injured vessels, and to promote IH after grafting. Interventions that globally inhibit TGFß in CABG have yielded contrasting outcomes in in vitro and in vivo studies including clinical trials. With advances in molecular biology, it becomes clear that TGFß exhibits both protective and damaging roles, and only specific components such as some Smad-dependent TGFß signaling mediate vascular IH. The activin receptor-like kinase (ALK)-mediated Smad-dependent TGFß signaling pathways have been found to be activated in human vascular smooth muscle cells (VSMCs) following injury and in hyperplastic preimplantation vein grafts. It appears that focused targeting of TGFß pathway constitutes a promising therapeutic target to improve the outcome of CABG. This study dissects the role of TGFß pathway in CABG failure, with particular emphasis on the therapeutic potentials of specific targeting of Smad-dependent and ALK-mediated signaling.
Collapse
Affiliation(s)
- Marzuq A Ungogo
- Department of Veterinary Pharmacology and Toxicology, 58989Ahmadu Bello University, Zaria, Nigeria.,Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
23
|
Pyrogallol-Phloroglucinol-6,6-Bieckolon Attenuates Vascular Smooth Muscle Cell Proliferation and Phenotype Switching in Hyperlipidemia through Modulation of Chemokine Receptor 5. Mar Drugs 2020; 18:md18080393. [PMID: 32727125 PMCID: PMC7460451 DOI: 10.3390/md18080393] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 07/21/2020] [Accepted: 07/21/2020] [Indexed: 12/28/2022] Open
Abstract
Hyperlipidemia induces vascular smooth muscle cell (VSMC) proliferation and phenotype switching from contractile to synthetic. This process is involved in arterial remodeling via the chemokine ligand 5 (CCL5)/chemokine receptor 5 (CCR5) pathway. Arterial remodeling is related to atherosclerosis or intimal hyperplasia. The purpose of this study was to evaluate whether pyrogallol-phloroglucinol-6,6-bieckol (PPB) from E. cava reduces VSMC proliferation and phenotype switching via the CCL5/CCR5 pathway. The CCL5/CCR5 expression, VSMC proliferation and phenotypic alterations were evaluated using a cell model of VSMC exposed in hyperlipidemia, and an animal model of mice fed a high-fat-diet (HFD). The expression of CCL5/CCR5 increased in both the cell and animal models of hyperlipidemia. Treatment with PPB decreased CCL5/CCR5 expression in both models. The expression of contractile markers of VSMCs, including alpha-smooth muscle actin (α-SMA), smooth muscle myosin heavy chain (SM-MHC), and smooth muscle protein 22 alpha (SM22α), were decreased by hyperlipidemia and restored after treatment with PPB. The silencing of CCR5 attenuated the effects of PPB treatment. VSMC proliferation and the intima-media thickness of the aortas, increased with HFD and decreased after treatment with PPB. The VSMC proliferation ratio and messenger ribonucleic acid (mRNA) expression of cell cycle regulatory factors increased in the in vitro model and were restored after treatment with PPB. PPB treatment reduced VSMC proliferation and phenotype switching induced by hyperlipidemia through inhibition of the CCL5/CCR5 pathway.
Collapse
|
24
|
Steger CM, Hartmann A, Rieker RJ. Molecular differences between arterial and venous grafts in the first year after coronary artery bypass grafting. Histochem Cell Biol 2020; 154:405-419. [PMID: 32705339 DOI: 10.1007/s00418-020-01896-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/16/2020] [Indexed: 12/27/2022]
Abstract
Despite commonly used for coronary artery bypass surgery, saphenous vein (SV) grafts have significantly lower patency rates in comparison to internal thoracic artery (ITA) grafts, which might be due to the structural characteristics of the vessel wall but also due to differences in oxidative stress adaptation and molecular signaling and regulation. This human post mortem study included a total of 150 human bypass grafts (75 SV grafts and 75 ITA grafts) obtained from 60 patients divided into five groups due to the time period of implantation: group 1: baseline group without grafting; group 2: 1 day; group 3: > 1 day-1 week; group 4: > 1 week-1 month; group 5: > 1 month-1 year. Pieces of 3 mm length were fixed with formaldehyde, dehydrated, wax embedded, cut into sections of 3 µm thickness, and histologically and immunohistochemically examined. Over the whole time period, we observed a lower neointima formation and a better preserved media in ITA grafts with a higher percentage of TNF-α, PDGFR-α, and VEGF-A in nearly all vessel wall layers, a higher amount of MMP-7, MMP-9, EGFR, and bFGF positive cells in SV grafts and a timely different peak not only between ITA and SV grafts but also within the various vessel wall layers of both graft types. Since most of the examined growth factors, growth factor receptors and cytokines are regulated by MAPKs, our results suggest an activation of different pathways in both vessel graft types immediately after bypass grafting.
Collapse
Affiliation(s)
- Christina Maria Steger
- Department of Pathology, Academic Teaching Hospital Feldkirch, Affiliation of the Innsbruck Medical University, Carinagasse 47, 6800, Feldkirch, Austria.
| | - Arndt Hartmann
- Department of Pathology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg, Krankenhausstr. 8-10, 91054, Erlangen, Germany
| | - Ralf Joachim Rieker
- Department of Pathology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg, Krankenhausstr. 8-10, 91054, Erlangen, Germany
| |
Collapse
|
25
|
Liu JT, Liu Z, Chen Y, Qi YX, Yao QP, Jiang ZL. MicroRNA-29a Involvement in Phenotypic Transformation of Venous Smooth Muscle Cells Via Ten-Eleven Translocation Methylcytosinedioxygenase 1 in Response to Mechanical Cyclic Stretch. J Biomech Eng 2020; 142:051009. [PMID: 31513704 DOI: 10.1115/1.4044581] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Indexed: 07/25/2024]
Abstract
Mechanical stimuli play an important role in vein graft restenosis and the abnormal migration and proliferation of vascular smooth muscle cells (VSMCs) are pathological processes contributing to this disorder. Here, based on previous high-throughput sequencing data from vein grafts, miR-29a-3p and its target, the role of Ten-eleven translocation methylcytosinedioxygenase 1 (TET1) in phenotypic transformation of VSMCs induced by mechanical stretch was investigated. Vein grafts were generated by using the "cuff" technique in rats. Deep transcriptome sequencing revealed that the expression of TET1 was significantly decreased, a process confirmed by reverse transcription quantitative real-time polymerase chain reaction (RT-qPCR) analysis. MicroRNA-seq showed that miR-29a-3p was significantly up-regulated, targeting TET1 as predicted by Targetscan. Bioinformatics analysis indicated that the co-expressed genes with TET1 might modulate VSMC contraction. Venous VSMCs exposed to 10%-1.25 Hz cyclic stretch by using the Flexcell system were used to simulate arterial mechanical conditions in vitro. RT-qPCR revealed that mechanical stretch increased the expression of miR-29a-3p at 3 h. Western blot analysis showed that TET1 was significantly decreased, switching contractile VSMCs to cells with a synthetic phenotype. miR-29a-3p mimics (MI) and inhibitor (IN) transfection confirmed the negative impact of miR-29a-3p on TET1. Taken together, results from this investigation demonstrate that mechanical stretch modulates venous VSMC phenotypic transformation via the mediation of the miR-29a-3p/TET1 signaling pathway. miR-29a-3p may have potential clinical implications in the pathogenesis of remodeling of vein graft restenosis.
Collapse
Affiliation(s)
- Ji-Ting Liu
- Institute of Mechanobiology and Medical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Ze Liu
- Institute of Mechanobiology and Medical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yi Chen
- Institute of Mechanobiology and Medical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Ying-Xin Qi
- Institute of Mechanobiology and Medical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Qing-Ping Yao
- Institute of Mechanobiology and Medical Engineering, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, P.O. Box 888, 800 Dongchuan Road Minhang, Shanghai 200240, China
| | - Zong-Lai Jiang
- Institute of Mechanobiology and Medical Engineering, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, P.O. Box 888, 800 Dongchuan Road Minhang, Shanghai 200240, China
| |
Collapse
|
26
|
Qu Q, Pang Y, Zhang C, Liu L, Bi Y. Exosomes derived from human umbilical cord mesenchymal stem cells inhibit vein graft intimal hyperplasia and accelerate reendothelialization by enhancing endothelial function. Stem Cell Res Ther 2020; 11:133. [PMID: 32293542 PMCID: PMC7092460 DOI: 10.1186/s13287-020-01639-1] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 01/05/2020] [Accepted: 03/10/2020] [Indexed: 02/07/2023] Open
Abstract
Background In our previous research, we found that mesenchymal stem cell (MSC) transplantation therapy can inhibit intimal hyperplasia and enhance endothelial function in arterialized vein grafts in rats. However, whether MSC-derived exosomes (MSC-exosomes) can reduce neointimal formation and its possible mechanism is still unclear. Methods The primary human umbilical cord MSCs (hucMSCs) and human umbilical vein endothelial cells (HUVECs) were isolated and characterized by flow cytometry and immunofluorescence. The exosomes derived from hucMSCs (hucMSC-exosomes) were identified by transmission electron microscopy and western blots. hucMSC-exosomes were intravenously injected into a rat model of vein grafting, and its effect on vein grafts reendothelialization and intimal hyperplasia was assessed by physical, histological, immunohistochemistry, and immunofluorescence examinations. The effects of hucMSC-exosomes on endothelial cells were evaluated by integrated experiment, EdU staining, scratch assay, and Transwell assay. The expression levels of key gene and pathways associated with the biological activity of vascular endothelial cells were evaluated following the stimulation of hucMSC-exosomes. Results We successfully isolated and characterized primary hucMSCs and hucMSC-exosomes and primary HUVECs. We verified that the systemic administration of hucMSC-exosomes accelerates reendothelialization and decreases intimal hyperplasia of autologous vein graft in a rat model. We also identified that hucMSC-exosomes can be uptaken by endothelial cells to stimulate cell proliferative and migratory activity in vitro. Furthermore, we detected that vascular endothelial growth factor (VEGF) plays an important part in hucMSC-exosome-mediated proliferation and migration in HUVECs. In addition, we also provided evidence that the signalling pathways of PI3K/AKT and MAPK/ERK1/2 take part in hucMSC-exosome-induced VEGF regulation. Conclusion Our data suggest that hucMSC-exosomes exert a vasculoprotective role in the setting of vein graft disease, which may provide a new clue to protect against vein graft failure in the future.
Collapse
Affiliation(s)
- Qingxi Qu
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, People's Republic of China
| | - Yingxin Pang
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, People's Republic of China
| | - Chunmei Zhang
- Department of Cardiology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, People's Republic of China
| | - Linghong Liu
- Research Center of Stem Cell and Regenerative Medicine, Shandong University, Jinan, 250012, Shandong, People's Republic of China. .,Laboratory of Cryomedicine, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, People's Republic of China.
| | - Yanwen Bi
- Department of Cardiovascular Surgery, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, People's Republic of China.
| |
Collapse
|
27
|
Colucci M, Torreggiani M, Bernardi I, Stangalino S, Catucci D, Esposito V, Sileno G, Esposito C. Smart Flow for the evaluation of the hemodialysis arteriovenous fistula. J Vasc Access 2020; 21:652-657. [PMID: 31894718 DOI: 10.1177/1129729819897171] [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: 11/16/2022] Open
Abstract
BACKGROUND Smart Flow is an innovative tool available on the Carestream Touch Prime Ultrasound machines, which provides automated blood flow measurement and shows the vectors that form the blood flow in the vessel. We compared the use of Smart Flow with traditional Duplex Doppler Ultrasound to evaluate blood flow of arteriovenous fistulas in prevalent hemodialysis patients. METHODS A total of 31 chronic patients on hemodialysis were enrolled. Blood flow was measured on the brachial artery with Smart Flow and duplex Doppler ultrasound. In a subset of 26 patients, a video of the juxta-anastomotic efferent vein was recorded and analyzed to calculate an index of flow turbulence. RESULTS We enrolled 21 males and 10 females aged 68.52 ± 11.64 years at the time of evaluation with an average arteriovenous fistulas vintage of 50.23 ± 47.42 months and followed them up for 18.03 ± 5.18 months. Smart Flow and Duplex Doppler Ultrasound blood flow measurements positively correlated (p < 0.0001) in the same patient but Smart Flow gave higher blood flow values (995.0 vs 730.3 mL/min, p < 0.0001), and the Duplex Doppler Ultrasound blood flow standard deviation was similar to Smart Flow (125.4 vs 114.4 mL/min, p < 0.0001). The time needed to evaluate arteriovenous fistulas with Smart Flow was significantly shorter than Duplex Doppler Ultrasound (67.58 ± 19.89 vs 146.3 ± 26.35 s, p < 0.0001). No correlation was found between blood flow turbulence and the subsequent access failure. CONCLUSION Smart Flow is reliable, reproducible, and faster than traditional duplex ultrasound. However, the additional information given by the Smart Flow technique does not seem to add any further benefits in terms of prediction of the access failure.
Collapse
Affiliation(s)
- Marco Colucci
- Unit of Nephrology and Dialysis, ICS Maugeri s.p.a. SB, Pavia, Italy
| | | | - Irene Bernardi
- Unit of Nephrology and Dialysis, ICS Maugeri s.p.a. SB, Pavia, Italy
| | - Simone Stangalino
- Unit of Nephrology and Dialysis, ICS Maugeri s.p.a. SB, Pavia, Italy
| | - Davide Catucci
- Unit of Nephrology and Dialysis, ICS Maugeri s.p.a. SB, Pavia, Italy
| | - Vittoria Esposito
- Unit of Nephrology and Dialysis, ICS Maugeri s.p.a. SB, Pavia, Italy
| | - Giuseppe Sileno
- Unit of Nephrology and Dialysis, ICS Maugeri s.p.a. SB, Pavia, Italy
| | - Ciro Esposito
- Unit of Nephrology and Dialysis, ICS Maugeri s.p.a. SB, Pavia, Italy.,University of Pavia, Pavia, Italy
| |
Collapse
|
28
|
Jana S. Endothelialization of cardiovascular devices. Acta Biomater 2019; 99:53-71. [PMID: 31454565 DOI: 10.1016/j.actbio.2019.08.042] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 07/19/2019] [Accepted: 08/22/2019] [Indexed: 01/10/2023]
Abstract
Blood-contacting surfaces of cardiovascular devices are not biocompatible for creating an endothelial layer on them. Numerous research studies have mainly sought to modify these surfaces through physical, chemical and biological means to ease early endothelial cell (EC) adhesion, migration and proliferation, and eventually to build an endothelial layer on the surfaces. The first priority for surface modification is inhibition of protein adsorption that leads to inhibition of platelet adhesion to the device surfaces, which may favor EC adhesion. Surface modification through surface texturing, if applicable, can bring some hopeful outcomes in this regard. Surface modifications through chemical and/or biological means may play a significant role in easy endothelialization of cardiovascular devices and inhibit smooth muscle cell proliferation. Cellular engineering of cells relevant to endothelialization can boost the positive outcomes obtained through surface engineering. This review briefly summarizes recent developments and research in early endothelialization of cardiovascular devices. STATEMENT OF SIGNIFICANCE: Endothelialization of cardiovascular implants, including heart valves, vascular stents and vascular grafts is crucial to solve many problems in our health care system. Numerous research efforts have been made to improve endothelialization on the surfaces of cardiovascular implants, mainly through surface modifications in three ways - physically, chemically and biologically. This review is intended to highlight comprehensive research studies to date on surface modifications aiming for early endothelialization on the blood-contacting surfaces of cardiovascular implants. It also discusses future perspectives to help guide endothelialization strategies and inspire further innovations.
Collapse
Affiliation(s)
- Soumen Jana
- Department of Bioengineering, University of Missouri, Columbia, MO 65211, USA.
| |
Collapse
|
29
|
Pachuk CJ, Rushton-Smith SK, Emmert MY. Intraoperative storage of saphenous vein grafts in coronary artery bypass grafting. Expert Rev Med Devices 2019; 16:989-997. [DOI: 10.1080/17434440.2019.1682996] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Affiliation(s)
| | | | - Maximilian Y. Emmert
- Department of Cardiothoracic and Vascular Surgery, German Heart Center Berlin, Berlin, Germany
- Department of Cardiovascular Surgery, Charité-Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
30
|
Uchida D, Saito Y, Kikuchi S, Yoshida Y, Hirata S, Sasajima T, Azuma N. Development of gene therapy with a cyclic adenosine monophosphate response element decoy oligodeoxynucleotide to prevent vascular intimal hyperplasia. J Vasc Surg 2019; 71:229-241. [PMID: 31204215 DOI: 10.1016/j.jvs.2019.02.042] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 02/17/2019] [Indexed: 12/28/2022]
Abstract
OBJECTIVE Intimal hyperplasia (IH) is the main cause of therapeutic failure after vascular and endovascular surgery. However, there is currently no targeted therapy for the treatment of IH. We recently reported that the inhibition of cyclic adenosine monophosphate response element (CRE) binding protein (CREB) activation is important in vein graft IH. We focused on a decoy oligodeoxynucleotide (ODN) therapeutic strategy for suppressing IH as a clinical application. The objective of this study was to confirm the therapeutic effect of a CRE decoy ODN in an animal model as a novel therapy for preventing intimal hyperplasia as the first step of the preclinical study of our strategy. METHODS We designed two phosphorothioate CREs and two scramble decoy ODNs and screened them using a CREB transcription assay to check their ability to bind to a CRE sequence. We chose a CRE decoy ODN with high first-binding ability and transfected it into vascular smooth muscle cells (VSMCs) in vitro. Proliferation and migration were assessed using MTS (3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium) assays and modified Boyden chamber assays. We examined CRE activity using a luciferase reporter gene assay. We assessed the expression of messenger RNAs by quantitative real-time polymerase chain reaction. In a wire-injury mouse model (C57BL6, n = 6), CRE decoy ODN was transfected into the injured vessel wall using an ultrasound-sonoporation method in vivo. Mitogen-activated protein kinase-activated protein kinase 3 (MAPKAPK3) and four and a half LIM domains 5 (FHL5) expression of pregrafting vein remnants were assessed by immunohistologic analyses. RESULTS Compared with scramble decoy ODN, the selected CRE decoy ODN could significantly decrease CRE activity (mean ± standard error of the mean: 0.20 ± 0.03 vs 1.00 ± 0.16, n = 6; P < .05) as shown by a luciferase reporter gene assay, VSMC proliferation (0.73 ± 0.04 vs 0.89 ± 0.02, n = 6; P < .05) and migration (96.4 ± 6.1 vs 311.4 ± 19.1 migrated VSMCs/well, n = 6; P < .05) after 24-hour transfection. The CRE decoy ODN significantly suppressed the formation of IH at injured vessel walls in an animal model, as analyzed by pathologic staining (0.20 ± 0.02 vs 0.56 ± 0.08, area of the intima/area of the artery vs the control after 21 days' transfection, n = 6; P < .05). Furthermore, MAPKAPK3 and FHL5, which are CREB activators, were significantly expressed in pregrafting vein remnants in diabetes mellitus patients. CONCLUSIONS CREB-CRE signaling is an important mechanism of IH formation, and CRE decoy therapy can help preventing IH. This study is the first part of the preclinical study of our strategy.
Collapse
MESH Headings
- Animals
- CREB-Binding Protein/genetics
- CREB-Binding Protein/metabolism
- Cell Movement
- Cell Proliferation
- Cells, Cultured
- Cyclic AMP/metabolism
- Disease Models, Animal
- Humans
- Intracellular Signaling Peptides and Proteins/genetics
- Intracellular Signaling Peptides and Proteins/metabolism
- LIM Domain Proteins/genetics
- LIM Domain Proteins/metabolism
- Male
- Mice, Inbred C57BL
- Muscle, Smooth, Vascular/injuries
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Neointima
- Oligodeoxyribonucleotides/genetics
- Oligodeoxyribonucleotides/metabolism
- Protein Serine-Threonine Kinases/genetics
- Protein Serine-Threonine Kinases/metabolism
- Response Elements/genetics
- Transcription Factors/genetics
- Transcription Factors/metabolism
- Vascular System Injuries/genetics
- Vascular System Injuries/metabolism
- Vascular System Injuries/pathology
- Vascular System Injuries/prevention & control
Collapse
Affiliation(s)
- Daiki Uchida
- Division of Vascular Surgery, Department of Surgery, Asahikawa Medical University, Asahikawa, Hokkaido, Japan
| | - Yukihiro Saito
- Division of Vascular Surgery, Department of Surgery, Asahikawa Medical University, Asahikawa, Hokkaido, Japan.
| | - Shinsuke Kikuchi
- Division of Vascular Surgery, Department of Surgery, Asahikawa Medical University, Asahikawa, Hokkaido, Japan
| | - Yuri Yoshida
- Division of Vascular Surgery, Department of Surgery, Asahikawa Medical University, Asahikawa, Hokkaido, Japan
| | - Satoshi Hirata
- Division of Vascular Surgery, Department of Surgery, Asahikawa Medical University, Asahikawa, Hokkaido, Japan
| | - Tadahiro Sasajima
- Division of Vascular Surgery, Department of Surgery, Asahikawa Medical University, Asahikawa, Hokkaido, Japan
| | - Nobuyoshi Azuma
- Division of Vascular Surgery, Department of Surgery, Asahikawa Medical University, Asahikawa, Hokkaido, Japan
| |
Collapse
|
31
|
One-Stage versus Two-Stage Arteriovenous Loop Reconstructions: An Experience on 103 Cases from a Single Center. Plast Reconstr Surg 2019; 143:912-924. [PMID: 30624338 DOI: 10.1097/prs.0000000000005386] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND The optimal time for flap anastomosis to an arteriovenous loop remains controversial. Whether perforator flaps and axially vascularized muscle or fasciocutaneous flaps lead to comparable outcomes in conjunction with arteriovenous loops has not been investigated. METHODS Medical records from 103 patients undergoing arteriovenous loop reconstruction (76 one-stage and 27 two-stage) between 2007 and 2017 were reviewed. Postoperative outcomes were compared between one- and two-stage arteriovenous loop reconstructions and different types of free flaps. RESULTS Rates of flap thrombosis, major wound complications, and flap failure did not differ significantly between one- and two-stage arteriovenous loop reconstructions (14.47 percent versus 11.11 percent, p = 1.00; 30.26 percent versus 25.93 percent, p = 0.67; and 10.53 percent versus 7.41 percent, p = 1.00). For two-stage arteriovenous loop reconstructions, the time interval between arteriovenous loop placement and flap anastomosis was a predictor for thrombotic events (OR, 1.31; p < 0.05). Anterolateral thigh flaps in conjunction with arteriovenous loops showed higher failure rates (33.33 percent) compared with all other flaps (6.59 percent) (p < 0.05) and combined latissimus dorsi and parascapular flaps (0 percent) (p < 0.05). Thrombosis rates were higher in anterolateral thigh flaps (33.33 percent) compared with all other flaps (10.99 percent; p = 0.056), and combined latissimus dorsi and parascapular flaps (0 percent; p < 0.05). CONCLUSIONS Two-stage arteriovenous loop reconstructions do not lead to increased postoperative complications compared to one-stage arteriovenous loop reconstructions and may be favorable in complicated cases because of shorter operative times. To avoid an increased thrombosis risk, flap anastomosis should not be delayed beyond 10 days in two-stage arteriovenous loop reconstructions. Anterolateral thigh flaps are less suitable for arteriovenous loop reconstructions because of higher complication rates. CLINICAL QUESTION/LEVEL OF EVIDENCE Therapeutic, III.
Collapse
|
32
|
Yu S, Zhang W, Wang L, Li Z, Li Q, Lv M, Liu B, Zhang Y. Effectiveness of Saphenous Vein Y-Grafts in Patients Undergoing Off-Pump Complete Myocardial Revascularization. Med Sci Monit 2019; 25:598-604. [PMID: 30662058 PMCID: PMC6350452 DOI: 10.12659/msm.911981] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Background To evaluate perioperative and mid-term outcomes of saphenous vein Y-grafts in patients with multi-vessel coronary artery disease. Material/Methods Sixty patients who underwent off-pump coronary surgery with Y-graft between 2005 and 2016 were enrolled, including 38 patients with natural Y-graft. Sixty patients with multi-vessel lesions in the same period were randomly selected as a control group. Results A total of 484 conduits were employed. The intraoperative variables were insignificantly different between groups, but Y-graft group compared with control group had more grafts (4.2±0.84 vs. 3.87±0.85) and anastomoses (6.30±1.39 vs. 5.62±1.15). No patient died during coronary artery bypass grafting and no episode of perioperative myocardial infarction was found. Follow-up duration lasted from 1 to 137 (40.0±27.7) months. No significant difference between Y-graft group and control group was found in Kaplan-Meier 3-year survival rate (93.4% vs. 88.0%) or 5-year survival rate (81.4% vs. 88.0%). Conclusions Saphenous vein Y-graft is a feasible and safe revascularization strategy for multi-vessel coronary artery disease patients and brings about satisfactory outcomes.
Collapse
Affiliation(s)
- Shuai Yu
- Key Laboratory of Arrhythmias of the Ministry of Education of China, East Hospital, Tongji University School of Medicine, Shanghai, China (mainland).,Heart Health Center, East Hospital, Tongji University School of Medicine, Shanghai, China (mainland)
| | - Weiran Zhang
- Department of Cardiothoracic Surgery, BenQ Hospital, Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China (mainland)
| | - Luxin Wang
- Key Laboratory of Arrhythmias of the Ministry of Education of China, East Hospital, Tongji University School of Medicine, Shanghai, China (mainland).,Shanghai East Hospital of Clinical Medical College, Nanjing Medical University, Nanjing, Jiangsu, China (mainland)
| | - Zhi Li
- Department of Cardiothoracic Surgery, The First Affiliated Hospital with Nanjing Medical University, Nanjing Medical University, Nanjing, Jiangsu, China (mainland)
| | - Qifan Li
- Department of Cardiothoracic Surgery, The First Affiliated Hospital with Nanjing Medical University, Nanjing Medical University, Nanjing, Jiangsu, China (mainland)
| | - Mengwei Lv
- Shanghai East Hospital of Clinical Medical College, Nanjing Medical University, Nanjing, Jiangsu, China (mainland).,Department of Cardiovascular Surgery, East Hospital, Tongji University School of Medicine, Shanghai, China (mainland)
| | - Ban Liu
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China (mainland)
| | - Yangyang Zhang
- Key Laboratory of Arrhythmias of the Ministry of Education of China, East Hospital, Tongji University School of Medicine, Shanghai, China (mainland).,Department of Cardiovascular Surgery, East Hospital, Tongji University School of Medicine, Shanghai, China (mainland)
| |
Collapse
|
33
|
Zhang Y, Fang Q, Niu K, Gan Z, Yu Q, Gu T. Time-dependently slow-released multiple-drug eluting external sheath for efficient long-term inhibition of saphenous vein graft failure. J Control Release 2019; 293:172-182. [DOI: 10.1016/j.jconrel.2018.12.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2018] [Revised: 11/19/2018] [Accepted: 12/01/2018] [Indexed: 12/22/2022]
|
34
|
Abstract
Surgical interventions on blood vessels bear a risk for intimal hyperplasia and atherosclerosis as a consequence of injury. A specific feature of intimal hyperplasia is the loss of vascular smooth muscle cell (VSMC) differentiation gene expression. We hypothesized that immediate responses following injury induce vascular remodeling. To differentiate injury due to trauma, reperfusion and pressure changes we analyzed vascular responses to carotid artery bypass grafting in mice compared to transient ligation. As a control, the carotid artery was surgically laid open only. In both, bypass or ligation models, the inflammatory responses were transient, peaking after 6h, whereas the loss of VSMC differentiation gene expression persisted. Extended time kinetics showed that transient carotid artery ligation was sufficient to induce a persistent VSMC phenotype change throughout 28 days. Transient arterial ligation in ApoE knockout mice resulted in atherosclerosis in the transiently ligated vascular segment but not on the not-ligated contralateral side. The VSMC phenotype change could not be prevented by anti-TNF antibodies, Sorafenib, Cytosporone B or N-acetylcysteine treatment. Surgical interventions involving hypoxia/reperfusion are sufficient to induce VSMC phenotype changes and vascular remodeling. In situations of a perturbed lipid metabolism this bears the risk to precipitate atherosclerosis.
Collapse
|
35
|
Kenagy RD, Kikuchi S, Evanko SP, Ruiter MS, Piola M, Longchamp A, Pesce M, Soncini M, Deglise S, Fiore GB, Haefliger JA, Schmidt TA, Majesky MW, Sobel M, Wight TN. Versican is differentially regulated in the adventitial and medial layers of human vein grafts. PLoS One 2018; 13:e0204045. [PMID: 30265729 PMCID: PMC6161854 DOI: 10.1371/journal.pone.0204045] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 08/31/2018] [Indexed: 12/13/2022] Open
Abstract
Changes in extracellular matrix proteins may contribute significantly to the adaptation of vein grafts to the arterial circulation. We examined the production and distribution of versican and hyaluronan in intact human vein rings cultured ex vivo, veins perfused ex vivo, and cultured venous adventitial and smooth muscle cells. Immunohistochemistry revealed higher levels of versican in the intima/media compared to the adventitia, and no differences in hyaluronan. In the vasa vasorum, versican and hyaluronan associated with CD34+ progenitor cells. Culturing the vein rings for 14 days revealed increased versican immunostaining of 30–40% in all layers, with no changes in hyaluronan. Changes in versican accumulation appear to result from increased synthesis in the intima/media and decreased degradation in the adventitia as versican transcripts were increased in the intima/media, but unchanged in the adventitia, and versikine (the ADAMTS-mediated cleavage product of versican) was increased in the intima/media, but decreased in the adventitia. In perfused human veins, versican was specifically increased in the intima/media in the presence of venous pressure, but not with arterial pressure. Unexpectedly, cultured adventitial cells express and accumulate more versican and hyaluronan than smooth muscle cells. These data demonstrate a differential regulation of versican and hyaluronan in human venous adventitia vs. intima/media and suggest distinct functions for these extracellular matrix macromolecules in these venous wall compartments during the adaptive response of vein grafts to the arterial circulation.
Collapse
Affiliation(s)
- Richard D. Kenagy
- Center for Cardiovascular Biology, Institute for Stem Cells and Regenerative Medicine, and Department of Surgery, University of Washington, Seattle, WA, United States of America
- * E-mail:
| | - Shinsuke Kikuchi
- Department of Vascular Surgery, Asahikawa Medical University, Asahikawa, Japan
| | - Steve P. Evanko
- Matrix Biology Program, Benaroya Research Institute, Seattle, WA, United States of America
| | - Matthijs S. Ruiter
- Cardiovascular Tissue Engineering Unit—Centro Cardiologico Monzino, IRCCS, Via Parea, 4, Milan, Italy
| | - Marco Piola
- Dipartimento di Elettronica, Informazione e Bioingegneria, Politecnico di Milano, Milan, Italy
| | - Alban Longchamp
- Department of Vascular Surgery, CHUV | Lausanne University Hospital, Lausanne, Switzerland
| | - Maurizio Pesce
- Cardiovascular Tissue Engineering Unit—Centro Cardiologico Monzino, IRCCS, Via Parea, 4, Milan, Italy
| | - Monica Soncini
- Dipartimento di Elettronica, Informazione e Bioingegneria, Politecnico di Milano, Milan, Italy
| | - Sébastien Deglise
- Department of Vascular Surgery, CHUV | Lausanne University Hospital, Lausanne, Switzerland
| | - Gianfranco B. Fiore
- Dipartimento di Elettronica, Informazione e Bioingegneria, Politecnico di Milano, Milan, Italy
| | | | - Tannin A. Schmidt
- Biomedical Engineering Department, School of Dental Medicine, University of Connecticut Health Center, Farmington, CT, United States of America
| | - Mark W. Majesky
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA, United States of America
| | - Michael Sobel
- Division of Vascular Surgery, VA Puget Sound Health Care System, University of Washington, Seattle, WA, United States of America
| | - Thomas N. Wight
- Matrix Biology Program, Benaroya Research Institute, Seattle, WA, United States of America
| |
Collapse
|
36
|
Kwon SH, Li L, Terry CM, Shiu YT, Moos PJ, Milash BA, Cheung AK, Blumenthal DK. Differential gene expression patterns in vein regions susceptible versus resistant to neointimal hyperplasia. Physiol Genomics 2018; 50:615-627. [PMID: 29750603 PMCID: PMC6139633 DOI: 10.1152/physiolgenomics.00082.2017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Revised: 05/04/2018] [Accepted: 05/07/2018] [Indexed: 12/14/2022] Open
Abstract
Arteriovenous hemodialysis graft (AVG) stenosis results in thrombosis and AVG failure, but prevention of stenosis has been unsuccessful due in large part to our limited understanding of the molecular processes involved in neointimal hyperplasia (NH) formation. AVG stenosis develops chiefly as a consequence of highly localized NH formation in the vein-graft anastomosis region. Surprisingly, the vein region just downstream of the vein-graft anastomosis (herein termed proximal vein region) is relatively resistant to NH. We hypothesized that the gene expression profiles of the NH-prone and NH-resistant regions will be different from each other after graft placement, and analysis of their genomic profiles may yield potential therapeutic targets to prevent AVG stenosis. To test this, we evaluated the vein-graft anastomosis (NH-prone) and proximal vein (NH-resistant) regions in a porcine model of AVG stenosis with a porcine microarray. Gene expression changes in these two distinct vein regions, relative to the gene expression in unoperated control veins, were examined at early (5 days) and later (14 days) time points following graft placement. Global genomic changes were much greater in the NH-prone region than in the NH-resistant region at both time points. In the NH-prone region, genes related to regulation of cell proliferation and osteo-/chondrogenic vascular remodeling were most enriched among the significantly upregulated genes, and genes related to smooth muscle phenotype were significantly downregulated. These results provide insights into the spatial and temporal genomic modulation underlying NH formation in AVG and suggest potential therapeutic strategies to prevent and/or limit AVG stenosis.
Collapse
Affiliation(s)
- Sun Hyung Kwon
- Department of Pharmacology and Toxicology, University of Utah , Salt Lake City, Utah
| | - Li Li
- Department of Internal Medicine, Division of Nephrology and Hypertension, University of Utah , Salt Lake City, Utah
| | - Christi M Terry
- Department of Internal Medicine, Division of Nephrology and Hypertension, University of Utah , Salt Lake City, Utah
| | - Yan-Ting Shiu
- Department of Internal Medicine, Division of Nephrology and Hypertension, University of Utah , Salt Lake City, Utah
| | - Philip J Moos
- Department of Pharmacology and Toxicology, University of Utah , Salt Lake City, Utah
| | - Brett A Milash
- Bioinformatics Shared Resource, University of Utah, Huntsman Cancer Institute , Salt Lake City, Utah
| | - Alfred K Cheung
- Department of Internal Medicine, Division of Nephrology and Hypertension, University of Utah , Salt Lake City, Utah
- Medical Service, Veterans Affairs Salt Lake City Healthcare System, Salt Lake City, Utah
- Department of Nephrology, The Second Xiangya Hospital, Central South University , Changsha, Hunan , People's Republic of China
| | - Donald K Blumenthal
- Department of Pharmacology and Toxicology, University of Utah , Salt Lake City, Utah
| |
Collapse
|
37
|
Maga P, Mikolajczyk TP, Partyka L, Siedlinski M, Maga M, Krzanowski M, Malinowski K, Luc K, Nizankowski R, Bhatt DL, Guzik TJ. Involvement of CD8+ T cell subsets in early response to vascular injury in patients with peripheral artery disease in vivo. Clin Immunol 2018; 194:26-33. [PMID: 29936303 DOI: 10.1016/j.clim.2018.06.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 05/20/2018] [Accepted: 06/19/2018] [Indexed: 01/21/2023]
Abstract
AIMS Adaptive immunity is critical in vascular remodelling following arterial injury. We hypothesized that acute changes in T cells at a percutaneous transluminal angioplasty (PTA) site could serve as an index of their potential interaction with the injured vascular wall. METHODS AND RESULTS T cell subsets were characterised in 45 patients with Rutherford 3-4 peripheral artery disease (PAD) undergoing PTA. Direct angioplasty catheter blood sampling was performed before and immediately after the procedure. PTA was associated with an acute reduction of α/β-TcR CD8+ T cells. Further characterisation revealed significant reduction in pro-atherosclerotic CD28nullCD57+ T cells, effector (CD45RA+CCR7-) and effector memory (CD45RA-CCR7-) cells, in addition to cells bearing activation (CD69, CD38) and tissue homing/adhesion markers (CD38, CCR5). CONCLUSIONS The acute reduction observed here is likely due to the adhesion of cells to the injured vascular wall, suggesting that immunosenescent, activated effector CD8+ cells have a role in the early vascular injury immune response following PTA in PAD patients.
Collapse
Affiliation(s)
- Pawel Maga
- Department of Angiology, Jagiellonian University Medical College, Krakow, Poland; Angio-Medcus Angiology Clinic, Krakow, Poland
| | - Tomasz P Mikolajczyk
- Department of Internal and Agricultural Medicine, Jagiellonian University Medical College, Krakow, Poland; Institute of Infection, Immunity and Inflammation, University of Glasgow, UK
| | | | - Mateusz Siedlinski
- Department of Internal and Agricultural Medicine, Jagiellonian University Medical College, Krakow, Poland
| | - Mikolaj Maga
- Department of Angiology, Jagiellonian University Medical College, Krakow, Poland
| | | | - Krzysztof Malinowski
- Institute of Public Health, Faculty of Health Sciences, Jagiellonian University Medical College, Krakow, Poland
| | - Kevin Luc
- Department of Internal and Agricultural Medicine, Jagiellonian University Medical College, Krakow, Poland
| | - Rafal Nizankowski
- Department of Angiology, Jagiellonian University Medical College, Krakow, Poland
| | - Deepak L Bhatt
- Brigham and Women's Hospital Heart & Vascular Center, Harvard Medical School, Boston, MA, USA
| | - Tomasz J Guzik
- Department of Internal and Agricultural Medicine, Jagiellonian University Medical College, Krakow, Poland; Institute of Cardiovascular and Medical Sciences, University of Glasgow, UK.
| |
Collapse
|
38
|
Huang J, Bold M, Rajebi MR. Endovascular retrieval of Greenfield IVC filters 13 and 19 years post placement without major complication. J Radiol Case Rep 2018; 11:15-25. [PMID: 29299094 DOI: 10.3941/jrcr.v11i6.3031] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Inferior vena cava (IVC) filters were first introduced in 1967 by Kazi Mobin-Uddin and later improved by Lazar Greenfield in the 1980s becoming a major component of catastrophic pulmonary embolism prevention. Nevertheless, filters are not entirely harmless. The long term risks include caval thrombosis, visceral penetration, and filters can serve as a nidus for infection. Filter retrieval is often complicated by intimal hyperplasia especially with increased indwelling time. Historically, Greenfield filters in place for longer than 3 weeks were considered permanent due to the risks of retrieval. Herein we present 2 cases of successful retrieval of Greenfield filters 13 and 19 years post implantation.
Collapse
Affiliation(s)
- Junjian Huang
- Department of Radiology, Pennsylvania Hospital, Philadelphia, USA
| | - Michael Bold
- Department of Radiology, Mayo Clinic, Rochester, USA
| | | |
Collapse
|
39
|
Limiting Injury During Saphenous Vein Graft Preparation For Coronary Arterial Bypass Prevents Metabolic Decompensation. Sci Rep 2017; 7:14179. [PMID: 29079734 PMCID: PMC5660200 DOI: 10.1038/s41598-017-13819-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Accepted: 10/02/2017] [Indexed: 02/01/2023] Open
Abstract
Standard harvest and preparation of human saphenous vein (HSV) for autologous coronary and peripheral arterial bypass procedures is associated with injury and increased oxidative stress that negatively affect graft performance. In this study we investigated the global metabolomic profiles of HSV before (unprepared; UP) and after standard vein graft preparation (AP). AP-HSV showed impaired vasomotor function that was associated with increased oxidative stress, phospholipid hydrolysis and energy depletion that are characteristic of mechanical and chemical injury. A porcine model (PSV) was utilized to validate these metabolomic changes in HSV and to determine the efficacy of an improved preparation technique (OP) using pressure-regulated distension, a non-toxic vein marker, and graft storage in buffered PlasmaLyte solution in limiting metabolic decompensation due to graft preparation. Deficits in vasomotor function and metabolic signature observed in AP-PSV could be largely mitigated with the OP procedure. These findings suggest that simple strategies aimed at reducing injury during graft harvest and preparation represents a straightforward and viable strategy to preserve conduit function and possibly improve graft patency.
Collapse
|
40
|
Activation and inflammation of the venous endothelium in vein graft disease. Atherosclerosis 2017; 265:266-274. [PMID: 28865843 DOI: 10.1016/j.atherosclerosis.2017.08.023] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Revised: 07/19/2017] [Accepted: 08/23/2017] [Indexed: 01/13/2023]
Abstract
The long saphenous vein is the most commonly used conduit in coronary artery bypass graft (CABG) surgery when bypassing multiple diseased arteries; however, its use is complicated by the development of vascular inflammation, intimal hyperplasia and accelerated atherosclerosis leading to compromised graft efficacy. Despite refinement of surgical techniques to improve graft patency, late vein graft failure remains a significant problem. Moreover, there is a lack of pharmacological interventions proven to be effective in the treatment of late vein graft failure. A greater understanding of the molecular nature of the disease and the interactions between endothelial and smooth muscle cells as a result of alterations in local haemodynamics may assist with designing future beneficial pharmacological interventions. Venous endothelial cells (ECs) are physiologically adapted to chronic low shear stress; however, once the graft is implanted into the arterial circulation, they become suddenly exposed to acute high levels of shear stress. A small number of in vitro and ex vivo studies have demonstrated that acute high shear stress is associated with the activation of a pro-inflammatory profile in saphenous vein ECs, which may be mediated by mitogen-activated protein kinase (MAPK) and nuclear factor-κB (NF-κB) signalling pathways. The impact of acute changes in shear stress on venous ECs and the role of ECs in the development of intimal hyperplasia remains incomplete and is the subject of this review.
Collapse
|
41
|
Gao A, Hang R, Li W, Zhang W, Li P, Wang G, Bai L, Yu XF, Wang H, Tong L, Chu PK. Linker-free covalent immobilization of heparin, SDF-1α, and CD47 on PTFE surface for antithrombogenicity, endothelialization and anti-inflammation. Biomaterials 2017; 140:201-211. [DOI: 10.1016/j.biomaterials.2017.06.023] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 06/17/2017] [Accepted: 06/18/2017] [Indexed: 01/20/2023]
|
42
|
Guerciotti B, Vergara C, Ippolito S, Quarteroni A, Antona C, Scrofani R. A computational fluid–structure interaction analysis of coronary Y-grafts. Med Eng Phys 2017; 47:117-127. [DOI: 10.1016/j.medengphy.2017.05.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 04/12/2017] [Accepted: 05/16/2017] [Indexed: 10/19/2022]
|
43
|
Hess CN, Norgren L, Ansel GM, Capell WH, Fletcher JP, Fowkes FGR, Gottsäter A, Hitos K, Jaff MR, Nordanstig J, Hiatt WR. A Structured Review of Antithrombotic Therapy in Peripheral Artery Disease With a Focus on Revascularization. Circulation 2017. [DOI: 10.1161/circulationaha.117.024469] [Citation(s) in RCA: 106] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Connie N. Hess
- From Department of Medicine, Division of Cardiology, University of Colorado School of Medicine, Aurora (C.N.H., W.R.H.); CPC Clinical Research, Aurora, CO (C.N.H., W.H.C., W.R.H.); Department of Surgery, Faculty of Medicine and Health, Örebro University, Sweden (L.N.); Ohio Health, Columbus (G.M.A.); Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Colorado School of Medicine, Aurora (W.H.C.); University of Sydney, Westmead Hospital, Australia (J.P.F.)
| | - Lars Norgren
- From Department of Medicine, Division of Cardiology, University of Colorado School of Medicine, Aurora (C.N.H., W.R.H.); CPC Clinical Research, Aurora, CO (C.N.H., W.H.C., W.R.H.); Department of Surgery, Faculty of Medicine and Health, Örebro University, Sweden (L.N.); Ohio Health, Columbus (G.M.A.); Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Colorado School of Medicine, Aurora (W.H.C.); University of Sydney, Westmead Hospital, Australia (J.P.F.)
| | - Gary M. Ansel
- From Department of Medicine, Division of Cardiology, University of Colorado School of Medicine, Aurora (C.N.H., W.R.H.); CPC Clinical Research, Aurora, CO (C.N.H., W.H.C., W.R.H.); Department of Surgery, Faculty of Medicine and Health, Örebro University, Sweden (L.N.); Ohio Health, Columbus (G.M.A.); Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Colorado School of Medicine, Aurora (W.H.C.); University of Sydney, Westmead Hospital, Australia (J.P.F.)
| | - Warren H. Capell
- From Department of Medicine, Division of Cardiology, University of Colorado School of Medicine, Aurora (C.N.H., W.R.H.); CPC Clinical Research, Aurora, CO (C.N.H., W.H.C., W.R.H.); Department of Surgery, Faculty of Medicine and Health, Örebro University, Sweden (L.N.); Ohio Health, Columbus (G.M.A.); Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Colorado School of Medicine, Aurora (W.H.C.); University of Sydney, Westmead Hospital, Australia (J.P.F.)
| | - John P. Fletcher
- From Department of Medicine, Division of Cardiology, University of Colorado School of Medicine, Aurora (C.N.H., W.R.H.); CPC Clinical Research, Aurora, CO (C.N.H., W.H.C., W.R.H.); Department of Surgery, Faculty of Medicine and Health, Örebro University, Sweden (L.N.); Ohio Health, Columbus (G.M.A.); Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Colorado School of Medicine, Aurora (W.H.C.); University of Sydney, Westmead Hospital, Australia (J.P.F.)
| | - F. Gerry R. Fowkes
- From Department of Medicine, Division of Cardiology, University of Colorado School of Medicine, Aurora (C.N.H., W.R.H.); CPC Clinical Research, Aurora, CO (C.N.H., W.H.C., W.R.H.); Department of Surgery, Faculty of Medicine and Health, Örebro University, Sweden (L.N.); Ohio Health, Columbus (G.M.A.); Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Colorado School of Medicine, Aurora (W.H.C.); University of Sydney, Westmead Hospital, Australia (J.P.F.)
| | - Anders Gottsäter
- From Department of Medicine, Division of Cardiology, University of Colorado School of Medicine, Aurora (C.N.H., W.R.H.); CPC Clinical Research, Aurora, CO (C.N.H., W.H.C., W.R.H.); Department of Surgery, Faculty of Medicine and Health, Örebro University, Sweden (L.N.); Ohio Health, Columbus (G.M.A.); Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Colorado School of Medicine, Aurora (W.H.C.); University of Sydney, Westmead Hospital, Australia (J.P.F.)
| | - Kerry Hitos
- From Department of Medicine, Division of Cardiology, University of Colorado School of Medicine, Aurora (C.N.H., W.R.H.); CPC Clinical Research, Aurora, CO (C.N.H., W.H.C., W.R.H.); Department of Surgery, Faculty of Medicine and Health, Örebro University, Sweden (L.N.); Ohio Health, Columbus (G.M.A.); Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Colorado School of Medicine, Aurora (W.H.C.); University of Sydney, Westmead Hospital, Australia (J.P.F.)
| | - Michael R. Jaff
- From Department of Medicine, Division of Cardiology, University of Colorado School of Medicine, Aurora (C.N.H., W.R.H.); CPC Clinical Research, Aurora, CO (C.N.H., W.H.C., W.R.H.); Department of Surgery, Faculty of Medicine and Health, Örebro University, Sweden (L.N.); Ohio Health, Columbus (G.M.A.); Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Colorado School of Medicine, Aurora (W.H.C.); University of Sydney, Westmead Hospital, Australia (J.P.F.)
| | - Joakim Nordanstig
- From Department of Medicine, Division of Cardiology, University of Colorado School of Medicine, Aurora (C.N.H., W.R.H.); CPC Clinical Research, Aurora, CO (C.N.H., W.H.C., W.R.H.); Department of Surgery, Faculty of Medicine and Health, Örebro University, Sweden (L.N.); Ohio Health, Columbus (G.M.A.); Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Colorado School of Medicine, Aurora (W.H.C.); University of Sydney, Westmead Hospital, Australia (J.P.F.)
| | - William R. Hiatt
- From Department of Medicine, Division of Cardiology, University of Colorado School of Medicine, Aurora (C.N.H., W.R.H.); CPC Clinical Research, Aurora, CO (C.N.H., W.H.C., W.R.H.); Department of Surgery, Faculty of Medicine and Health, Örebro University, Sweden (L.N.); Ohio Health, Columbus (G.M.A.); Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Colorado School of Medicine, Aurora (W.H.C.); University of Sydney, Westmead Hospital, Australia (J.P.F.)
| |
Collapse
|
44
|
Shokri M, Moradpour R, Shafaroudi MM, Rezaei N, Tabary SZ. Comparing the Effects of Krebs Plus Verapamil Solution on Endothelial Function of Harvested Human Greater Saphenous Vein with Heparinized Blood, an Invitro Study. Med Arch 2017; 71:188-192. [PMID: 28974831 PMCID: PMC5585787 DOI: 10.5455/medarh.2017.71.188-192] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2017] [Accepted: 06/15/2017] [Indexed: 12/16/2022] Open
Abstract
INTRODUCTION Integrity of the great saphenous vein (GSV) endothelium is the most important key element for long-term patency rate of grafts in coronary artery bypass graft (CABG). Storage solutions play an important role in maintaining viability of vein endothelium. Diminished nitric oxide (NO) because of endothelial dysfunction may facilitate vascular inflammation and formation of atherosclerotic plaque. AIM So, we decided to find a reasonable alternative preservative solution instead of heparinized blood (HB) by measuring NO concentration with Griess assay. MATERIAL AND METHOD SVG samples were obtained from 54 patients undergoing elective CABG. 3 mm rings were stored in solutions: heparinized blood (HB), Krebs (K), Krebs + Propranolol (K+P) 6.66 g/l, Krebs + Adrenaline (K+A) 200 µl/l, and Krebs + Verapamil (K+V) 200 µl/l for 30, 45, 60 and 90 min. Nitrite concentration was measured by Griess assay at 540 nm. H&E staining was performed for histologic test. Statistical analysis was performed using SPSS (V16). Results were expressed as (Means ± SE) followed by One-Way ANOVA for finding best preservative solution. Repeated measurement test was used to investigate best time. In all analysis, (P<0.05) was considered significant. RESULTS Average concentration of NO in (K+V) compare with HB (1st control), K (2nd control), (K+A) and (K+P) showed higher rate in all times from 30 to 90 min (16.55±1.85:) and in (K+A, K+P) compare with (HB) and (K) there was no statistically significant difference in the same times. Comparing the average concentration of (NO) between (HB) and (K) showed no significant difference (K+V>HB=K=K+A=K+P). Also, our investigations showed that NO concentration in (K+V) has the highest rate in time 90 min (10.07±0.56, p=0.002):. More than 50 percent of endothelial cells stay normal in (K+V) compare with other solutions. CONCLUSION It seems that (K+V) is the best solution for the maintenance of normal physiology of SVGs endothelial cells. The most appropriate SVGs endothelial function is within 90 minutes after harvesting.
Collapse
Affiliation(s)
- Mitra Shokri
- Department of Anatomy and Cell Biology, Cell and Molecoular Research Center (CMRC), Medical Faculty, Mazandaran University of Medical Sciences, Sari, Iran
| | | | - Majid Malekzade Shafaroudi
- Department of Anatomy and Cell Biology, Cell and Molecoular Research Center (CMRC), Medical Faculty, Mazandaran University of Medical Sciences, Sari, Iran
| | - Nourollah Rezaei
- Department of Anatomy and Cell Biology, Cell and Molecoular Research Center (CMRC), Medical Faculty, Mazandaran University of Medical Sciences, Sari, Iran
| | - Shervin Ziabakhsh Tabary
- Department of Cardiac Surgery, Mazandaran Heart Center, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
45
|
Ultrasound Vector Flow Imaging – could be a new tool in evaluation of arteriovenous fistulas for hemodialysis? J Vasc Access 2017; 18:284-289. [DOI: 10.5301/jva.5000721] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/18/2017] [Indexed: 02/06/2023] Open
Abstract
Introduction We report the use of a new ultrasound technique to evaluate the axial and lateral components of a complex flow in the arteriovenous fistula (AVF). Vector Flow Imaging (VFI) allows to identify different components of the flow in every direction, even orthogonal to the flow streamline, represented by many single vectors. VFI could help to identify flow alterations in AVF, probably responsible for its malfunction. Methods From February to June 2016, 14 consecutive patients with upper-limb AVF were examined with a Resona 7 (Mindray, Shenzhen, China) ultrasound scanner equipped with VFI. An analysis of mean velocity, angular direction and mean number of vectors impacting the vessel wall was carried out. We also identified main flow patterns present in the arterial side, into the venous aneurysm and in correspondence of significant stenosis. Results A disturbed flow with the presence of vectors directed against the vessel walls was found in 9/14 patients (64.28%): in correspondence of the iuxta-anastomotic venous side (4/9; 44.4%), into the venous aneurysmal tracts (3/9; 33.3%) and in concomitance of stenosis (2/9; 22.2%). The mean velocity of the vectors was around 20-25 cm/s, except in presence of stenosis, where the velocities were much higher (45-50 cm/s). The vectors directed against the vessel walls presented high angle attack (from 45° to 90°, with a median angular deviation 65°). Conclusions VFI was confirmed to be an innovative and intuitive imaging technology to study the flow complexity in the arteriovenous fistulas.
Collapse
|
46
|
Cunnane CV, Cunnane EM, Walsh MT. A Review of the Hemodynamic Factors Believed to Contribute to Vascular Access Dysfunction. Cardiovasc Eng Technol 2017; 8:280-294. [DOI: 10.1007/s13239-017-0307-0] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Accepted: 05/09/2017] [Indexed: 11/29/2022]
|
47
|
Yeh YH, Chang SH, Chen SY, Wen CJ, Wei FC, Tang R, Achilefu S, Wun TC, Chen WJ. Bolus injections of novel thrombogenic site-targeted fusion proteins comprising annexin-V and Kunitz protease inhibitors attenuate intimal hyperplasia after balloon angioplasty. Int J Cardiol 2017; 240:339-346. [PMID: 28433556 DOI: 10.1016/j.ijcard.2017.03.150] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2016] [Revised: 02/26/2017] [Accepted: 03/27/2017] [Indexed: 11/13/2022]
Abstract
BACKGROUND Systemic administrations of conventional antithrombotics reduce neointima formation after angioplasty in experimental animals. However, clinical translation of these results has not been successful due to high risk for bleeding. OBJECTIVES We sought to determine whether novel annexin-V (ANV)-Kunitz protease inhibitor fusion proteins, TAP-ANV and ANV-6L15, can specifically target to vascular injury site and limit neointima formation without inducing systemic hypo-coagulation in a rat carotid artery balloon angioplasty injury model. METHODS Near infrared imaging was carried out after balloon-injury and injection of fluorescent ANV or ANV-6L15 to examine their bio-distributions. For peri-procedure treatment, TAP-ANV or ANV-6L15 was administered as i.v. boluses 3 times: 30-minutes before balloon-injury, immediate after procedure, and 120-minutes post-balloon-injury. For extended treatment, additional i.v. bolus injection was given on day-2, day-3 and every other day thereafter. Carotid arteries were collected on day-7 and day-14 for analysis. Blood was collected for measurement of clotting parameters. RESULTS Near infrared imaging and immunochemistry showed that fluorescent ANV and ANV-6L15 specifically localized to injured carotid artery and significant amount of ANV-6L15 remained bound to the injured artery after 24-h. Peri-procedure injections of TAP-ANV or ANV-6L15 resulted in decrease of intima/media ratio by 56%. Extended injections of both yielded similar results. Both decreased the expression of PCNA on day-7 and increased the expression calponin on day-14 in the intima post-balloon-injury. CONCLUSIONS TAP-ANV and ANV-6L15 can specifically localize to balloon injured carotid arteries after i.v. bolus injections, resulting in substantial attenuation of intimal hyperplasia without inducing a state of systemic hypo-coagulation.
Collapse
Affiliation(s)
- Yung-Hsin Yeh
- Cardiovascular Division, Chang-Gung Memorial Hospital, Chang-Gung University College of Medicine, Taoyuan, Taiwan.
| | - Shang-Hung Chang
- Cardiovascular Division, Chang-Gung Memorial Hospital, Chang-Gung University College of Medicine, Taoyuan, Taiwan
| | - Shin-Yu Chen
- Cardiovascular Division, Chang-Gung Memorial Hospital, Chang-Gung University College of Medicine, Taoyuan, Taiwan
| | - Chih-Jen Wen
- College of Medicine, Chang-Gung University, Center for Vascularized Composite Allotransplantation, Chang-Gung Medical Foundation, Department of Plastic and Reconstructive Surgery and Center for Vascularized Composite Allotransplantation, Chang-Gung Memorial Hospital, Taoyuan, Taiwan
| | - Fu-Chan Wei
- College of Medicine, Chang-Gung University, Center for Vascularized Composite Allotransplantation, Chang-Gung Medical Foundation, Department of Plastic and Reconstructive Surgery and Center for Vascularized Composite Allotransplantation, Chang-Gung Memorial Hospital, Taoyuan, Taiwan
| | - Rui Tang
- Department of Radiology, Washington University Medical School, St Louis, MO 63110, USA
| | - Sam Achilefu
- Department of Radiology, Washington University Medical School, St Louis, MO 63110, USA
| | - Tze-Chein Wun
- EVAS Therapeutics, LLC, 613 Huntley Heights Drive, Ballwin, MO 63021, USA
| | - Wei-Jan Chen
- Cardiovascular Division, Chang-Gung Memorial Hospital, Chang-Gung University College of Medicine, Taoyuan, Taiwan.
| |
Collapse
|
48
|
Extracellular Vesicles Derived from Adipose Mesenchymal Stem Cells Regulate the Phenotype of Smooth Muscle Cells to Limit Intimal Hyperplasia. Cardiovasc Drugs Ther 2017; 30:111-8. [PMID: 26650931 DOI: 10.1007/s10557-015-6630-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
PURPOSE Extracellular vesicles (EVs) derived from mesenchymal stem cells (MSCs) play important roles in the reduction of inflammation in multiple disease models. However, their role in vein graft (VG) remodeling is undefined. We aimed to investigate the effect of EVs from adipose MSCs (ADMSC-EVs) on VG intimal hyperplasia and to explore the possible mechanisms. METHODS After generation and characterization of control-EVs and ADMSC-EVs in vitro, we investigated their effect on the proliferation and migration of vascular smooth muscle cells (VSMCs) in vitro. Next, we established a mouse model of VG transplantation. Mice underwent surgery and received control-EVs or ADMSC-EVs by intraperitoneal injection every other day for 20 days. VG remodeling was evaluated after 4 weeks. We also assessed the effect of ADMSC-EVs on macrophage migration and inflammatory cytokine expression. RESULTS Significant inhibitory effects of ADMSC-EVs on in vitro VSMC proliferation (p < 0.05) and migration (p < 0.05) were observed compared with control-EVs. The extent of intimal hyperplasia was significantly decreased in ADMSC-EV-treated mice compared with control-EV-treated mice (26 ± 8.4 vs. 45 ± 9.0 μm, p < 0.05). A reduced presence of macrophages was observed in ADMSC-EV-treated mice (p < 0.05). Significantly decreased expression of inflammatory cytokines interleukin (IL)-6 and monocyte chemoattractant protein-1 (MCP-1) was also found in the ADMSC-EV-treated group (both p < 0.05). In addition, phosphorylation of Akt, Erk1/2, and p38 in VGs was decreased in the ADMSC-EV-treated group. CONCLUSIONS We demonstrated that ADMSC-EVs exert an inhibitory effect on VG neointima formation by regulating VSMC proliferation and migration, macrophage migration, inflammatory cytokine expression, and the related signaling pathways.
Collapse
|
49
|
Ganesan MK, Finsterwalder R, Leb H, Resch U, Neumüller K, de Martin R, Petzelbauer P. Three-Dimensional Coculture Model to Analyze the Cross Talk Between Endothelial and Smooth Muscle Cells. Tissue Eng Part C Methods 2017; 23:38-49. [PMID: 27923320 PMCID: PMC5240006 DOI: 10.1089/ten.tec.2016.0299] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Accepted: 11/28/2016] [Indexed: 12/21/2022] Open
Abstract
The response of blood vessels to physiological and pathological stimuli partly depends on the cross talk between endothelial cells (EC) lining the luminal side and smooth muscle cells (SMC) building the inner part of the vascular wall. Thus, the in vitro analysis of the pathophysiology of blood vessels requires coculture systems of EC and SMC. We have developed and validated a modified three-dimensional sandwich coculture (3D SW-CC) of EC and SMC using open μ-Slides with a thin glass bottom allowing direct imaging. The culture dish comprises an intermediate plate to minimize the meniscus resulting in homogenous cell distribution. Human umbilical artery SMC were sandwiched between coatings of rat tail collagen I. Following SMC quiescence, human umbilical vein EC were seeded on top of SMC and cultivated until confluence. By day 7, EC had formed a confluent monolayer and continuous vascular endothelial (VE)-cadherin-positive cell/cell contacts. Below, spindle-shaped SMC had formed parallel bundles and showed increased calponin expression compared to day 1. EC and SMC were interspaced by a matrix consisting of laminin, collagen IV, and perlecan. Basal messenger RNA (mRNA) expression levels of E-selectin, angiopoietin-1, calponin, and intercellular adhesion molecule 1 (ICAM-1) of the 3D SW-CC was comparable to that of a freshly isolated mouse inferior vena cava. Addition of tumor necrosis factor alpha (TNF α) to the 3D SW-CC induced E-selectin and ICAM-1 mRNA and protein induction, comparable to the EC and SMC monolayers. In contrast, the addition of activated platelets induced a significantly delayed but more pronounced activation in the 3D SW-CC compared to EC and SMC monolayers. Thus, this 3D SW-CC permits analyzing the cross talk between EC and SMC that mediate cellular quiescence as well as the response to complex activation signals.
Collapse
Affiliation(s)
- Minu Karthika Ganesan
- Skin and Endothelium Research Division (SERD), Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Richard Finsterwalder
- Skin and Endothelium Research Division (SERD), Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Heide Leb
- Skin and Endothelium Research Division (SERD), Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Ulrike Resch
- Department of Vascular Biology and Thrombosis Research, Medical University of Vienna, Vienna, Austria
| | - Karin Neumüller
- Skin and Endothelium Research Division (SERD), Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Rainer de Martin
- Department of Vascular Biology and Thrombosis Research, Medical University of Vienna, Vienna, Austria
| | - Peter Petzelbauer
- Skin and Endothelium Research Division (SERD), Department of Dermatology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
50
|
Boire TC, Balikov DA, Lee Y, Guth CM, Cheung-Flynn J, Sung HJ. Biomaterial-Based Approaches to Address Vein Graft and Hemodialysis Access Failures. Macromol Rapid Commun 2016; 37:1860-1880. [PMID: 27673474 PMCID: PMC5156561 DOI: 10.1002/marc.201600412] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 08/15/2016] [Indexed: 12/19/2022]
Abstract
Veins used as grafts in heart bypass or as access points in hemodialysis exhibit high failure rates, thereby causing significant morbidity and mortality for patients. Interventional or revisional surgeries required to correct these failures have been met with limited success and exorbitant costs, particularly for the US Centers for Medicare & Medicaid Services. Vein stenosis or occlusion leading to failure is primarily the result of neointimal hyperplasia. Systemic therapies have achieved little long-term success, indicating the need for more localized, sustained, biomaterial-based solutions. Numerous studies have demonstrated the ability of external stents to reduce neointimal hyperplasia. However, successful results from animal models have failed to translate to the clinic thus far, and no external stent is currently approved for use in the US to prevent vein graft or hemodialysis access failures. This review discusses current progress in the field, design considerations, and future perspectives for biomaterial-based external stents. More comparative studies iteratively modulating biomaterial and biomaterial-drug approaches are critical in addressing mechanistic knowledge gaps associated with external stent application to the arteriovenous environment. Addressing these gaps will ultimately lead to more viable solutions that prevent vein graft and hemodialysis access failures.
Collapse
Affiliation(s)
- Timothy C Boire
- Department of Biomedical Engineering, Vanderbilt University, 37235, Nashville, TN, USA
| | - Daniel A Balikov
- Department of Biomedical Engineering, Vanderbilt University, 37235, Nashville, TN, USA
| | - Yunki Lee
- Department of Biomedical Engineering, Vanderbilt University, 37235, Nashville, TN, USA
| | - Christy M Guth
- Division of Vascular Surgery, Department of Surgery, Vanderbilt University Medical Center, Nashville, TN, 37235, USA
| | - Joyce Cheung-Flynn
- Division of Vascular Surgery, Department of Surgery, Vanderbilt University Medical Center, Nashville, TN, 37235, USA
| | - Hak-Joon Sung
- Department of Biomedical Engineering, Vanderbilt University, 37235, Nashville, TN, USA
- Severance Biomedical Science Institute, College of Medicine, Yonsei University, Seoul, 120-752, Republic of Korea
| |
Collapse
|