1
|
Hassanpour M, Salybkov AA, Kobayashi S, Asahara T. Anti-inflammatory Prowess of endothelial progenitor cells in the realm of biology and medicine. NPJ Regen Med 2024; 9:27. [PMID: 39349482 PMCID: PMC11442670 DOI: 10.1038/s41536-024-00365-z] [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: 12/15/2023] [Accepted: 08/23/2024] [Indexed: 10/02/2024] Open
Abstract
Endothelial inflammation plays a crucial role in vascular-related diseases, a leading cause of global mortality. Among various cellular players, endothelial progenitor cells (EPCs) emerge as non-differentiated endothelial cells circulating in the bloodstream. Recent evidence highlights the transformative role of EPCs in shifting from an inflammatory/immunosuppressive crisis to an anti-inflammatory/immunomodulatory response. Despite the importance of these functions, the regulatory mechanisms governing EPC activities and their physiological significance in vascular regenerative medicine remain elusive. Surprisingly, the current literature lacks a comprehensive review of EPCs' effects on inflammatory processes. This narrative review aims to fill this gap by exploring the cutting-edge role of EPCs against inflammation, from molecular intricacies to broader medical perspectives. By examining how EPCs modulate inflammatory responses, we aim to unravel their anti-inflammatory significance in vascular regenerative medicine, deepening insights into EPCs' molecular mechanisms and guiding future therapeutic strategies targeting vascular-related diseases.
Collapse
Affiliation(s)
- Mehdi Hassanpour
- Shonan Research, Institute of Innovative Medicine, Shonan Kamakura General Hospital, Kamakura, Kanagawa, Japan
- Center for Cell therapy & Regenerative Medicine, Shonan Kamakura General Hospital, Kamakura, Kanagawa, Japan
- Kidney Disease and Transplant Center, Shonan Kamakura General Hospital, Kamakura, Kanagawa, Japan
| | - Amankeldi A Salybkov
- Shonan Research, Institute of Innovative Medicine, Shonan Kamakura General Hospital, Kamakura, Kanagawa, Japan
- Center for Cell therapy & Regenerative Medicine, Shonan Kamakura General Hospital, Kamakura, Kanagawa, Japan
- Kidney Disease and Transplant Center, Shonan Kamakura General Hospital, Kamakura, Kanagawa, Japan
| | - Shuzo Kobayashi
- Shonan Research, Institute of Innovative Medicine, Shonan Kamakura General Hospital, Kamakura, Kanagawa, Japan
- Kidney Disease and Transplant Center, Shonan Kamakura General Hospital, Kamakura, Kanagawa, Japan
| | - Takayuki Asahara
- Shonan Research, Institute of Innovative Medicine, Shonan Kamakura General Hospital, Kamakura, Kanagawa, Japan.
- Center for Cell therapy & Regenerative Medicine, Shonan Kamakura General Hospital, Kamakura, Kanagawa, Japan.
| |
Collapse
|
2
|
Kang MK, Heo SH, Yoon JK. In-Stent Re-Endothelialization Strategies: Cells, Extracellular Matrix, and Extracellular Vesicles. TISSUE ENGINEERING. PART B, REVIEWS 2024. [PMID: 39166272 DOI: 10.1089/ten.teb.2024.0178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/22/2024]
Abstract
Arterial stenosis caused by atherosclerosis often requires stent implantation to increase the patency of target artery. However, such external devices often lead to in-stent restenosis due to inadequate re-endothelialization and subsequent inflammatory responses. Therefore, re-endothelialization strategies after stent implantation have been developed to enhance endothelial cell recruitment or to capture circulating endothelial progenitor cells. Notably, recent research indicates that coating stent surfaces with biogenic materials enhances the long-term safety of implantation, markedly diminishing the risk of in-stent restenosis. In this review, we begin by describing the pathophysiology of coronary artery disease and in-stent restenosis. Then, we review the characteristics and materials of existing stents used in clinical practice. Lastly, we explore biogenic materials aimed at accelerating re-endothelialization, including extracellular matrix, cells, and extracellular vesicles. This review helps overcome the limitations of current stents for cardiovascular disease and outlines the next phase of research and development.
Collapse
Affiliation(s)
- Min-Kyu Kang
- Department of Systems Biotechnology, Chung-Ang University, Anseong-Si, Republic of Korea
| | - Seon-Hee Heo
- Department of surgery, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jeong-Kee Yoon
- Department of Systems Biotechnology, Chung-Ang University, Anseong-Si, Republic of Korea
| |
Collapse
|
3
|
Shradhanjali A, Wolfe JT, Tefft BJ. Magnetic Cell Targeting for Cardiovascular Tissue Engineering. TISSUE ENGINEERING. PART B, REVIEWS 2024. [PMID: 39078330 DOI: 10.1089/ten.teb.2024.0103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/31/2024]
Abstract
There is a critical need for novel approaches to translate cell therapy and regenerative medicine to clinical practice. Magnetic cell targeting with site specificity has started to open avenues in these fields as a potential therapeutic platform. Magnetic targeting is gaining popularity in the field of biomedicine due to its ability to concentrate and retain at a target site while minimizing deleterious effects at off-target sites. It is regarded as a relatively straightforward and safe approach for a wide range of therapeutic applications. This review discusses the latest advancements and approaches in magnetic cell targeting using endocytosed and surface-bound magnetic nanoparticles as well as in vivo tracking using magnetic resonance imaging (MRI). The most common form of magnetic nanoparticles is superparamagnetic iron oxide nanoparticles (SPION). The biodegradable and biocompatible properties of these magnetically responsive particles and capacity for rapid endocytosis into cells make them a breakthrough in targeted therapy. This review further discusses specific applications of magnetic targeting approaches in cardiovascular tissue engineering including myocardial regeneration, therapeutic angiogenesis, and endothelialization of implantable cardiovascular devices.
Collapse
Affiliation(s)
- Akankshya Shradhanjali
- Joint Department of Biomedical Engineering, Medical College of Wisconsin & Marquette University, Milwaukee, Wisconsin, USA
| | - Jayne T Wolfe
- Joint Department of Biomedical Engineering, Medical College of Wisconsin & Marquette University, Milwaukee, Wisconsin, USA
| | - Brandon J Tefft
- Joint Department of Biomedical Engineering, Medical College of Wisconsin & Marquette University, Milwaukee, Wisconsin, USA
| |
Collapse
|
4
|
Mahara A, Shirai M, Soni R, Le HT, Shimizu K, Hirano Y, Yamaoka T. Vascular tissue reconstruction by monocyte subpopulations on small-diameter acellular grafts via integrin activation. Mater Today Bio 2023; 23:100847. [PMID: 37953756 PMCID: PMC10632538 DOI: 10.1016/j.mtbio.2023.100847] [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: 06/29/2023] [Revised: 10/17/2023] [Accepted: 10/26/2023] [Indexed: 11/14/2023] Open
Abstract
Although the clinical application of cell-free tissue-engineered vascular grafts (TEVGs) has been proposed, vascular tissue regeneration mechanisms have not been fully clarified. Here, we report that monocyte subpopulations reconstruct vascular-like tissues through integrin signaling. An Arg-Glu-Asp-Val peptide-modified acellular long-bypass graft was used as the TEVG, and tissue regeneration in the graft was evaluated using a cardiopulmonary pump system and porcine transplantation model. In 1 day, the luminal surface of the graft was covered with cells that expressed CD163, CD14, and CD16, which represented the monocyte subpopulation, and they exhibited proliferative and migratory abilities. RNA sequencing showed that captured cells had an immune-related phenotype similar to that of monocytes and strongly expressed cell adhesion-related genes. In vitro angiogenesis assay showed that tube formation of the captured cells occurred via integrin signal activation. After medium- and long-term graft transplantation, the captured cells infiltrated the tunica media layer and constructed vascular with a CD31/CD105-positive layer and an αSMA-positive structure after 3 months. This finding, including multiple early-time observations provides clear evidence that blood-circulating monocytes are directly involved in vascular remodeling.
Collapse
Affiliation(s)
- Atsushi Mahara
- Department of Biomedical Engineering, National Cerebral and Cardiovascular Center Research Institute, Kishibe Shimmachi, Suita Osaka, 564-8565, Japan
| | - Manabu Shirai
- Omics Research Center, National Cerebral and Cardiovascular Center Research Institute, Kishibe Shimmachi, Suita Osaka, 564-8565, Japan
| | - Raghav Soni
- Department of Biomedical Engineering, National Cerebral and Cardiovascular Center Research Institute, Kishibe Shimmachi, Suita Osaka, 564-8565, Japan
| | - Hue Thi Le
- Department of Biomedical Engineering, National Cerebral and Cardiovascular Center Research Institute, Kishibe Shimmachi, Suita Osaka, 564-8565, Japan
| | - Kaito Shimizu
- Faculty of Chemistry, Materials and Bioengineering, Kansai University, 3-3-35 Yamatecho, Suita, Osaka, 565-8680, Japan
| | - Yoshiaki Hirano
- Faculty of Chemistry, Materials and Bioengineering, Kansai University, 3-3-35 Yamatecho, Suita, Osaka, 565-8680, Japan
| | - Tetsuji Yamaoka
- Department of Biomedical Engineering, National Cerebral and Cardiovascular Center Research Institute, Kishibe Shimmachi, Suita Osaka, 564-8565, Japan
| |
Collapse
|
5
|
West-Livingston L, Lim JW, Lee SJ. Translational tissue-engineered vascular grafts: From bench to bedside. Biomaterials 2023; 302:122322. [PMID: 37713761 DOI: 10.1016/j.biomaterials.2023.122322] [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: 04/05/2023] [Revised: 09/01/2023] [Accepted: 09/09/2023] [Indexed: 09/17/2023]
Abstract
Cardiovascular disease is a primary cause of mortality worldwide, and patients often require bypass surgery that utilizes autologous vessels as conduits. However, the limited availability of suitable vessels and the risk of failure and complications have driven the need for alternative solutions. Tissue-engineered vascular grafts (TEVGs) offer a promising solution to these challenges. TEVGs are artificial vascular grafts made of biomaterials and/or vascular cells that can mimic the structure and function of natural blood vessels. The ideal TEVG should possess biocompatibility, biomechanical mechanical properties, and durability for long-term success in vivo. Achieving these characteristics requires a multi-disciplinary approach involving material science, engineering, biology, and clinical translation. Recent advancements in scaffold fabrication have led to the development of TEVGs with improved functional and biomechanical properties. Innovative techniques such as electrospinning, 3D bioprinting, and multi-part microfluidic channel systems have allowed the creation of intricate and customized tubular scaffolds. Nevertheless, multiple obstacles must be overcome to apply these innovations effectively in clinical practice, including the need for standardized preclinical models and cost-effective and scalable manufacturing methods. This review highlights the fundamental approaches required to successfully fabricate functional vascular grafts and the necessary translational methodologies to advance their use in clinical practice.
Collapse
Affiliation(s)
- Lauren West-Livingston
- Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, 27157, USA; Department of Vascular and Endovascular Surgery, Duke University, Durham, NC, 27712, USA
| | - Jae Woong Lim
- Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, 27157, USA; Department of Thoracic and Cardiovascular Surgery, Soonchunhyang University Hospital, Bucheon-Si, Gyeonggi-do, 420-767, Republic of Korea
| | - Sang Jin Lee
- Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, 27157, USA.
| |
Collapse
|
6
|
Schwarz N, Yadegari H. Potentials of Endothelial Colony-Forming Cells: Applications in Hemostasis and Thrombosis Disorders, from Unveiling Disease Pathophysiology to Cell Therapy. Hamostaseologie 2023; 43:325-337. [PMID: 37857295 DOI: 10.1055/a-2101-5936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2023] Open
Abstract
Endothelial colony-forming cells (ECFCs) are endothelial progenitor cells circulating in a limited number in peripheral blood. They can give rise to mature endothelial cells (ECs) and, with intrinsically high proliferative potency, contribute to forming new blood vessels and restoring the damaged endothelium in vivo. ECFCs can be isolated from peripheral blood or umbilical cord and cultured to generate large amounts of autologous ECs in vitro. Upon differentiation in culture, ECFCs are excellent surrogates for mature ECs showing the same phenotypic, genotypic, and functional features. In the last two decades, the ECFCs from various vascular disease patients have been widely used to study the diseases' pathophysiology ex vivo and develop cell-based therapeutic approaches, including vascular regenerative therapy, tissue engineering, and gene therapy. In the current review, we will provide an updated overview of past studies, which have used ECFCs to elucidate the molecular mechanisms underlying the pathogenesis of hemostatic disorders in basic research. Additionally, we summarize preceding studies demonstrating the utility of ECFCs as cellular tools for diagnostic or therapeutic clinical applications in thrombosis and hemostasis.
Collapse
Affiliation(s)
- Nadine Schwarz
- Institute of Experimental Hematology and Transfusion Medicine, University Hospital Bonn, Bonn, Germany
| | - Hamideh Yadegari
- Institute of Experimental Hematology and Transfusion Medicine, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
7
|
Farwa U, Lee HY, Lim H, Park I, Park S, Moon BG, Lee BT. Poly(l-lactide)/polycaprolactone based multifunctional coating to deliver paclitaxel/VEGF and control the degradation rate of magnesium alloy stent. Int J Biol Macromol 2023; 250:126218. [PMID: 37572804 DOI: 10.1016/j.ijbiomac.2023.126218] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 06/26/2023] [Accepted: 08/05/2023] [Indexed: 08/14/2023]
Abstract
Despite significant advancements made in cardiovascular stents, restenosis, thrombosis, biocompatibility, and clinical complications remain a matter of concern. Herein, we report a biodegradable Mg alloy stent with a dual effect of the drug (Paclitaxel) and growth factor (VEGF) release. To mitigate the fast degradation of Mg alloy, inorganic and organic coatings were formed on the alloy surface. The optimized hierarchal sequence of the coating was the first layer consisting of magnesium fluoride, followed by poly(l-lactide) and hydroxyapatite coating, and finally sealed by a polycaprolactone layer (MgC). PLLA and HAp were used to increase the adhesion strength and biocompatibility of the coating. Paclitaxel and VEGF were loaded in the final PCL layer (Mg-C/PTX-VEGF). As compared to bare Mg alloy (28 % weight loss), our MgC system showed (3.1 % weight loss) successful decrease in the degradation rate. Further, the in vitro biocompatibility illustrated the highly biocompatible nature of our drug and growth factor-loaded system. The in vivo results displayed that the drug loading decreased the inflammation and neointimal hyperplasia as indicated by the α-SMA and CD-68 antibody staining. The growth factor helped in the endothelialization which was established by the FLKI and ICAM antibody staining of the tissue.
Collapse
Affiliation(s)
- Ume Farwa
- Institute of Tissue Regeneration, Soonchunhyang University Cheonan, Republic of Korea
| | - Hyun-Yong Lee
- Department of Surgery, Soonchunhyang University Cheonan Hospital, Cheonan, Republic of Korea
| | - Hansung Lim
- Department of Regenerative Medicine, College of Medicine, Soonchunhyang University Cheonan, Republic of Korea
| | - Ihho Park
- Materials Testing & Reliability Division, Korea Institute of Materials Science (KIMS), Changwon, Republic of Korea
| | - Sangho Park
- Cardiology Department, Soonchunhyang University Cheonan Hospital, Cheonan, Republic of Korea
| | - Byoung-Gi Moon
- Department of Magnesium, Advanced Metals Division, Korea Institute of Material Science (KIMS), Changwon, Republic of Korea
| | - Byong-Taek Lee
- Institute of Tissue Regeneration, Soonchunhyang University Cheonan, Republic of Korea; Department of Regenerative Medicine, College of Medicine, Soonchunhyang University Cheonan, Republic of Korea.
| |
Collapse
|
8
|
Chen X, Dong N, Xu X, Zhou Y, Shi J, Qiao W, Hong H. Re-endothelialization of Decellularized Scaffolds With Endothelial Progenitor Cell Capturing Aptamer: A New Strategy for Tissue-Engineered Heart Valve. ASAIO J 2023; 69:885-893. [PMID: 37506117 DOI: 10.1097/mat.0000000000001979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/30/2023] Open
Abstract
Tissue-engineered heart valve (TEHV) is a promising alternative to current heart valve substitute. Decellularized porcine aortic heart valves (DAVs) are the most common scaffolds of TEHV. Hard to endothelialization is one of the disadvantages of DAVs. Therefore, we aimed to immobilize endothelial progenitor cell (EPC)-aptamer onto DAVs for accelerating endothelialization. In this study, three groups of scaffolds were constructed: DAVs, aptamer-immobilized DAVs (aptamer-DAVs), and glutaraldehyde crosslinked DAVs (GA-DAVs). The results of flow cytometry revealed that EPC-aptamer was specific to EPCs and was immobilized onto DAVs. Cells adhesion experiments demonstrated that EPCs adhered more tightly onto aptamer-DAVs group than other two groups of scaffolds. And cell proliferation assay indicated that EPCs seeded onto aptamer-DAVs group grew faster than DAVs group and GA-DAVs group. Moreover, dynamic capture experiment in flow conditions revealed that the number of EPCs captured by aptamer-DAVs group was more than other two groups. In conclusion, aptamer-DAVs could specifically promote adhesion and proliferation of EPCs and had ability to capture EPCs in simulated flow condition. This could promote re-endothelialization of scaffolds.
Collapse
Affiliation(s)
- Xue Chen
- From the Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | | | | | | | | | | | | |
Collapse
|
9
|
Pelliccia F, Zimarino M, Niccoli G, Morrone D, De Luca G, Miraldi F, De Caterina R. In-stent restenosis after percutaneous coronary intervention: emerging knowledge on biological pathways. EUROPEAN HEART JOURNAL OPEN 2023; 3:oead083. [PMID: 37808526 PMCID: PMC10558044 DOI: 10.1093/ehjopen/oead083] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 07/24/2023] [Accepted: 08/15/2023] [Indexed: 10/10/2023]
Abstract
Percutaneous coronary intervention (PCI) has evolved significantly over the past four decades. Since its inception, in-stent restenosis (ISR)-the progressive reduction in vessel lumen diameter after PCI-has emerged as the main complication of the procedure. Although the incidence of ISR has reduced from 30% at 6 months with bare-metal stents to 7% at 4 years with drug-eluting stents (DESs), its occurrence is relevant in absolute terms because of the dimensions of the population treated with PCI. The aim of this review is to summarize the emerging understanding of the biological pathways that underlie ISR. In-stent restenosis is associated with several factors, including patient-related, genetic, anatomic, stent, lesion, and procedural characteristics. Regardless of associated factors, there are common pathophysiological pathways involving molecular phenomena triggered by the mechanical trauma caused by PCI. Such biological pathways are responses to the denudation of the intima during balloon angioplasty and involve inflammation, hypersensitivity reactions, and stem cell mobilization particularly of endothelial progenitor cells (EPCs). The results of these processes are either vessel wall healing or neointimal hyperplasia and/or neo-atherosclerosis. Unravelling the key molecular and signal pathways involved in ISR is crucial to identify appropriate therapeutic strategies aimed at abolishing the 'Achille's heel' of PCI. In this regard, we discuss novel approaches to prevent DES restenosis. Indeed, available evidence suggests that EPC-capturing stents promote rapid stent re-endothelization, which, in turn, has the potential to decrease the risk of stent thrombosis and allow the use of a shorter-duration dual antiplatelet therapy.
Collapse
Affiliation(s)
- Francesco Pelliccia
- Department of Cardiovascular Sciences, University Sapienza, Viale del Policlinico 155, 00161 Rome, Italy
| | - Marco Zimarino
- Department of Neuroscience, Imaging and Clinical Sciences, “G. d’Annunzio” University, Viale Abruzzo, 332, 66100 Chieti, Italy
- Department of Cardiology, “SS. Annunziata Hospital”, ASL 2 Abruzzo, Via dei Vestini, 66100 Chieti, Italy
| | - Giampaolo Niccoli
- Department of Cardiology, University of Parma, Piazzale S. Francesco, 3, 43121 Parma, Italy
| | - Doralisa Morrone
- Department of Surgical, Medical and Molecular Pathology and of Critical Sciences, University of Pisa, Lungarno Antonio Pacinotti 43, 56126 Pisa, Italy
| | - Giuseppe De Luca
- Division of Cardiology, AOU “Policlinico G. Martino”, Department of Clinical and Experimental Medicine, University of Messina, Via Consolare Valeria 1, 98124 Messina, Italy
- Division of Cardiology, IRCCS Hospital Galeazzi-Sant'Ambrogio, Via Cristina Belgioioso 173, 20157 Milan, Italy
| | - Fabio Miraldi
- Department of Cardiovascular Sciences, University Sapienza, Viale del Policlinico 155, 00161 Rome, Italy
| | - Raffaele De Caterina
- Department of Surgical, Medical and Molecular Pathology and of Critical Sciences, University of Pisa, Lungarno Antonio Pacinotti 43, 56126 Pisa, Italy
| |
Collapse
|
10
|
Wu M, Xun M, Chen Y. Adaptation of Vascular Smooth Muscle Cell to Degradable Metal Stent Implantation. ACS Biomater Sci Eng 2023. [PMID: 37364226 DOI: 10.1021/acsbiomaterials.3c00637] [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: 06/28/2023]
Abstract
Iron-, magnesium-, or zinc-based metal vessel stents support vessel expansion at the period early after implantation and degrade away after vascular reconstruction, eliminating the side effects due to the long stay of stent implants in the body and the risks of restenosis and neoatherosclerosis. However, emerging evidence has indicated that their degradation alters the vascular microenvironment and induces adaptive responses of surrounding vessel cells, especially vascular smooth muscle cells (VSMCs). VSMCs are highly flexible cells that actively alter their phenotype in response to the stenting, similarly to what they do during all stages of atherosclerosis pathology, which significantly influences stent performance. This Review discusses how biodegradable metal stents modify vascular conditions and how VSMCs respond to various chemical, biological, and physical signals attributable to stent implantation. The focus is placed on the phenotypic adaptation of VSMCs and the clinical complications, which highlight the importance of VSMC transformation in future stent design.
Collapse
Affiliation(s)
- Meichun Wu
- Hengyang Medical School, University of South China, Hengyang, Hunan 410001, China
- School of Nursing, University of South China, Hengyang, Hunan 410001, China
| | - Min Xun
- Institute of Pharmacy and Pharmacology, School of Pharmaceutical Science, University of South China, Hengyang, Hunan 410001, China
| | - Yuping Chen
- Hengyang Medical School, University of South China, Hengyang, Hunan 410001, China
- Institute of Pharmacy and Pharmacology, School of Pharmaceutical Science, University of South China, Hengyang, Hunan 410001, China
| |
Collapse
|
11
|
Zhang Q, Duncan S, Szulc DA, de Mestral C, Kutryk MJ. Development of a universal, oriented antibody immobilization method to functionalize vascular prostheses for enhanced endothelialization for potential clinical application. J Biol Eng 2023; 17:37. [PMID: 37264409 DOI: 10.1186/s13036-023-00356-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 05/19/2023] [Indexed: 06/03/2023] Open
Abstract
BACKGROUND Thrombosis is a common cause of vascular prosthesis failure. Antibody coating of prostheses to capture circulating endothelial progenitor cells to aid endothelialization on the device surface appears a promising solution to prevent thrombus formation. Compared with random antibody immobilization, oriented antibody coating (OAC) increases antibody-antigen binding capacity and reduces antibody immunogenicity in vivo. Currently, few OAC methods have been documented, with none possessing clinical application potential. RESULTS Dopamine and the linker amino-PEG8-hydrazide-t-boc were successfully deposited on the surface of cobalt chromium (CC) discs, CC stents and expanded polytetrafluoroethylene (ePTFE) grafts under a slightly basic condition. CD34 antibodies were immobilized through the reaction between aldehydes in the Fc region created by oxidation and hydrazides in the linker after t-boc removal. CD34 antibody-coated surfaces were integral and smooth as shown by scanning electron microscopy (SEM), had significantly reduced or no substrate-specific signals as revealed by X-ray photoelectron spectroscopy, were hospitable for HUVEC growth as demonstrated by cell proliferation assay, and specifically bound CD34 + cells as shown by cell binding testing. CD34 antibody coating turned hydrophobic property of ePTFE grafts to hydrophilic. In a porcine carotid artery interposition model, a confluent monolayer of cobblestone-shaped CD31 + endothelial cells on the luminal surface of the CD34 antibody coated ePTFE graft were observed. In contrast, thrombi and fibrin fibers on the bare graft, and sporadic cells on the graft coated by chemicals without antibodies were seen. CONCLUSION A universal, OAC method was developed. Our in vitro and in vivo data suggest that the method can be potentially translated into clinical application, e.g., modifying ePTFE grafts to mitigate their thrombotic propensity and possibly provide for improved long-term patency for small-diameter grafts.
Collapse
Affiliation(s)
- Qiuwang Zhang
- Division of Cardiology, Keenan Research Center for Biomedical Science, St. Michael's Hospital, Unity Health Toronto, University of Toronto, Toronto, ON, Canada.
| | - Sebastian Duncan
- Division of Cardiology, Keenan Research Center for Biomedical Science, St. Michael's Hospital, Unity Health Toronto, University of Toronto, Toronto, ON, Canada
| | - Daniel A Szulc
- Division of Cardiology, Keenan Research Center for Biomedical Science, St. Michael's Hospital, Unity Health Toronto, University of Toronto, Toronto, ON, Canada
| | - Charles de Mestral
- Division of Vascular Surgery, Keenan Research Center for Biomedical Science, St. Michael's Hospital, Unity Health Toronto, Toronto, ON, Canada
- Department of Surgery, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Michael Jb Kutryk
- Division of Cardiology, Keenan Research Center for Biomedical Science, St. Michael's Hospital, Unity Health Toronto, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
12
|
Coronel-Meneses D, Sánchez-Trasviña C, Ratera I, Mayolo-Deloisa K. Strategies for surface coatings of implantable cardiac medical devices. Front Bioeng Biotechnol 2023; 11:1173260. [PMID: 37256118 PMCID: PMC10225971 DOI: 10.3389/fbioe.2023.1173260] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 04/25/2023] [Indexed: 06/01/2023] Open
Abstract
Cardiac medical devices (CMDs) are required when the patient's cardiac capacity or activity is compromised. To guarantee its correct functionality, the building materials in the development of CMDs must focus on several fundamental properties such as strength, stiffness, rigidity, corrosion resistance, etc. The challenge is more significant because CMDs are generally built with at least one metallic and one polymeric part. However, not only the properties of the materials need to be taken into consideration. The biocompatibility of the materials represents one of the major causes of the success of CMDs in the short and long term. Otherwise, the material will lead to several problems of hemocompatibility (e.g., protein adsorption, platelet aggregation, thrombus formation, bacterial infection, and finally, the rejection of the CMDs). To enhance the hemocompatibility of selected materials, surface modification represents a suitable solution. The surface modification involves the attachment of chemical compounds or bioactive compounds to the surface of the material. These coatings interact with the blood and avoid hemocompatibility and infection issues. This work reviews two main topics: 1) the materials employed in developing CMDs and their key characteristics, and 2) the surface modifications reported in the literature, clinical trials, and those that have reached the market. With the aim of providing to the research community, considerations regarding the choice of materials for CMDs, together with the advantages and disadvantages of the surface modifications and the limitations of the studies performed.
Collapse
Affiliation(s)
- David Coronel-Meneses
- Tecnologico de Monterrey, The Institute for Obesity Research, Monterrey, Mexico
- Tecnologico de Monterrey, Escuela de Ingeniería y Ciencias, Centro de Biotecnología-FEMSA, Monterrey, Mexico
| | - Calef Sánchez-Trasviña
- Tecnologico de Monterrey, The Institute for Obesity Research, Monterrey, Mexico
- Tecnologico de Monterrey, Escuela de Ingeniería y Ciencias, Centro de Biotecnología-FEMSA, Monterrey, Mexico
| | - Imma Ratera
- Institute of Materials Science of Barcelona (ICMAB-CSIC), Campus UAB, Bellaterra, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Instituto de Salud Carlos IIIBellaterra, Spain
| | - Karla Mayolo-Deloisa
- Tecnologico de Monterrey, The Institute for Obesity Research, Monterrey, Mexico
- Tecnologico de Monterrey, Escuela de Ingeniería y Ciencias, Centro de Biotecnología-FEMSA, Monterrey, Mexico
- Institute of Materials Science of Barcelona (ICMAB-CSIC), Campus UAB, Bellaterra, Spain
| |
Collapse
|
13
|
Wu Y, Yazdani SK, Bolander JEM, Wagner WD. Syndecan-4 and stromal cell-derived factor-1 alpha functionalized endovascular scaffold facilitates adhesion, spreading and differentiation of endothelial colony forming cells and functions under flow and shear stress conditions. J Biomed Mater Res B Appl Biomater 2023; 111:538-550. [PMID: 36208170 PMCID: PMC10092721 DOI: 10.1002/jbm.b.35170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 08/22/2022] [Accepted: 09/03/2022] [Indexed: 01/25/2023]
Abstract
Acellular vascular scaffolds with capture molecules have shown great promise in recruiting circulating endothelial colony forming cells (ECFCs) to promote in vivo endothelialization. A microenvironment conducive to cell spreading and differentiation following initial cell capture are key to the eventual formation of a functional endothelium. In this study, syndecan-4 and stromal cell-derived factor-1 alpha were used to functionalize an elastomeric biomaterial composed of poly(glycerol sebacate), Silk Fibroin and Type I Collagen, termed PFC, to enhance ECFC-material interaction. Functionalized PFC (fPFC) showed significantly greater ECFCs capture capability under physiological flow. Individual cell spreading area on fPFC (1474 ± 63 μm2 ) was significantly greater than on PFC (1187 ± 54 μm2 ) as early as 2 h, indicating enhanced cell-material interaction. Moreover, fPFC significantly upregulated the expression of endothelial cell specific markers such as platelet endothelial cell adhesion molecule (24-fold) and Von Willebrand Factor (11-fold) compared with tissue culture plastic after 7 days, demonstrating differentiation of ECFCs into endothelial cells. fPFC fabricated as small diameter conduits and tested using a pulsatile blood flow bioreactor were stable and maintained function. The findings suggest that the new surface functionalization strategy proposed here results in an endovascular material with enhanced endothelialization.
Collapse
Affiliation(s)
- Yidi Wu
- Department of Plastic and Reconstructive Surgery, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA.,Biomedical Engineering and Sciences, Virginia Tech - Wake Forest University School, Winston-Salem, North Carolina, USA
| | - Saami K Yazdani
- Department of Engineering, Wake Forest University, Winston-Salem, North Carolina, USA
| | - Johanna Elin Marie Bolander
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina, USA
| | - William D Wagner
- Department of Plastic and Reconstructive Surgery, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA.,Biomedical Engineering and Sciences, Virginia Tech - Wake Forest University School, Winston-Salem, North Carolina, USA.,Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina, USA
| |
Collapse
|
14
|
Targeted Nanoparticles for the Binding of Injured Vascular Endothelium after Percutaneous Coronary Intervention. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27238144. [PMID: 36500236 PMCID: PMC9739478 DOI: 10.3390/molecules27238144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 11/19/2022] [Accepted: 11/20/2022] [Indexed: 11/25/2022]
Abstract
Percutaneous coronary intervention (PCI) is a common procedure for the management of coronary artery obstruction. However, it usually causes vascular wall injury leading to restenosis that limits the long-term success of the PCI endeavor. The ultimate objective of this study was to develop the targeting nanoparticles (NPs) that were destined for the injured subendothelium and attract endothelial progenitor cells (EPCs) to the damaged location for endothelium regeneration. Biodegradable poly(lactic-co-glycolic acid) (PLGA) NPs were conjugated with double targeting moieties, which are glycoprotein Ib alpha chain (GPIbα) and human single-chain antibody variable fragment (HuscFv) specific to the cluster of differentiation 34 (CD34). GPIb is a platelet receptor that interacts with the von Willebrand factor (vWF), highly deposited on the damaged subendothelial surface, while CD34 is a surface marker of EPCs. A candidate anti-CD34 HuscFv was successfully constructed using a phage display biopanning technique. The HuscFv could be purified and showed binding affinity to the CD34-positive cells. The GPIb-conjugated NPs (GPIb-NPs) could target vWF and prevent platelet adherence to vWF in vitro. Furthermore, the HuscFv-conjugated NPs (HuscFv-NPs) could capture CD34-positive cells. The bispecific NPs have high potential to locate at the damaged subendothelial surface and capture EPCs for accelerating the vessel repair.
Collapse
|
15
|
Alferiev IS, Hooshdaran B, Pressly BB, Zoltick PW, Stachelek SJ, Chorny M, Levy RJ, Fishbein I. Intraprocedural endothelial cell seeding of arterial stents via biotin/avidin targeting mitigates in-stent restenosis. Sci Rep 2022; 12:19212. [PMID: 36357462 PMCID: PMC9649779 DOI: 10.1038/s41598-022-23820-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 11/07/2022] [Indexed: 11/12/2022] Open
Abstract
Impaired endothelialization of endovascular stents has been established as a major cause of in-stent restenosis and late stent thrombosis. Attempts to enhance endothelialization of inner stent surfaces by pre-seeding the stents with endothelial cells in vitro prior to implantation are compromised by cell destruction during high-pressure stent deployment. Herein, we report on the novel stent endothelialization strategy of post-deployment seeding of biotin-modified endothelial cells to avidin-functionalized stents. Acquisition of an avidin monolayer on the stent surface was achieved by consecutive treatments of bare metal stents (BMS) with polyallylamine bisphosphonate, an amine-reactive biotinylation reagent and avidin. Biotin-modified endothelial cells retain growth characteristics of normal endothelium and can express reporter transgenes. Under physiological shear conditions, a 50-fold higher number of recirculating biotinylated cells attached to the avidin-modified metal surfaces compared to bare metal counterparts. Delivery of biotinylated endothelial cells to the carotid arterial segment containing the implanted avidin-modified stent in rats results in immediate cell binding to the stent struts and is associated with a 30% reduction of in-stent restenosis in comparison with BMS.
Collapse
Affiliation(s)
- Ivan S Alferiev
- The Children's Hospital of Philadelphia, Philadelphia, PA, USA
- The University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | | | | | | | - Stanley J Stachelek
- The Children's Hospital of Philadelphia, Philadelphia, PA, USA
- The University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Michael Chorny
- The Children's Hospital of Philadelphia, Philadelphia, PA, USA
- The University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Robert J Levy
- The Children's Hospital of Philadelphia, Philadelphia, PA, USA
- The University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Ilia Fishbein
- The Children's Hospital of Philadelphia, Philadelphia, PA, USA.
- The University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
16
|
Zhang Q, Duncan S, Szulc DA, Kutryk MJB. Antibody functionalized intravascular devices combined with genetically engineered endothelial colony-forming cells for targeted drug delivery: a proof-of-concept study. Eur J Pharm Biopharm 2022; 181:218-226. [DOI: 10.1016/j.ejpb.2022.11.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 11/06/2022] [Accepted: 11/14/2022] [Indexed: 11/19/2022]
|
17
|
Marei I, Ahmetaj-Shala B, Triggle CR. Biofunctionalization of cardiovascular stents to induce endothelialization: Implications for in- stent thrombosis in diabetes. Front Pharmacol 2022; 13:982185. [PMID: 36299902 PMCID: PMC9589287 DOI: 10.3389/fphar.2022.982185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 08/31/2022] [Indexed: 11/13/2022] Open
Abstract
Stent thrombosis remains one of the main causes that lead to vascular stent failure in patients undergoing percutaneous coronary intervention (PCI). Type 2 diabetes mellitus is accompanied by endothelial dysfunction and platelet hyperactivity and is associated with suboptimal outcomes following PCI, and an increase in the incidence of late stent thrombosis. Evidence suggests that late stent thrombosis is caused by the delayed and impaired endothelialization of the lumen of the stent. The endothelium has a key role in modulating inflammation and thrombosis and maintaining homeostasis, thus restoring a functional endothelial cell layer is an important target for the prevention of stent thrombosis. Modifications using specific molecules to induce endothelial cell adhesion, proliferation and function can improve stents endothelialization and prevent thrombosis. Blood endothelial progenitor cells (EPCs) represent a potential cell source for the in situ-endothelialization of vascular conduits and stents. We aim in this review to summarize the main biofunctionalization strategies to induce the in-situ endothelialization of coronary artery stents using circulating endothelial stem cells.
Collapse
Affiliation(s)
- Isra Marei
- Department of Pharmacology, Weill Cornell Medicine- Qatar, Doha, Qatar
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
- *Correspondence: Isra Marei, ; Chris R. Triggle,
| | | | - Chris R. Triggle
- Department of Pharmacology, Weill Cornell Medicine- Qatar, Doha, Qatar
- *Correspondence: Isra Marei, ; Chris R. Triggle,
| |
Collapse
|
18
|
Coronary Stenting: Reflections on a 35-Year Journey. Can J Cardiol 2022; 38:S17-S29. [PMID: 34375695 DOI: 10.1016/j.cjca.2021.07.224] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 07/12/2021] [Accepted: 07/14/2021] [Indexed: 01/09/2023] Open
Abstract
Stenting was introduced as a therapy for coronary artery disease 35 years ago, and is currently the most commonly performed minimally invasive procedure globally. Percutaneous coronary revascularization, initially with plain old balloon angioplasty and later with stenting, has dramatically affected the outcomes of acute myocardial infarction and acute coronary syndromes. Coronary stenting is probably the most intensively studied therapy in medicine on the basis of the number of randomized clinical trials for a broad range of indications. Continuous improvements in stent materials, design, and coatings concurrent with procedural innovations have truly been awe-inspiring. The story of stenting is replete with high points and some low points, such as the initial experience with stent thrombosis and restenosis, and the more recent disappointment with bioabsorbable scaffolds. History has shown rapid growth of stent use with expansion of indications followed by contraction of some uses in response to clinical trial evidence in support of bypass surgery or medical therapy. In this review we trace the constantly evolving story of the coronary stent from the earliest experience until the present time. Undoubtedly, future iterations of stent design and materials will continue to move the stent story forward.
Collapse
|
19
|
Douglass M, Garren M, Devine R, Mondal A, Handa H. Bio-inspired hemocompatible surface modifications for biomedical applications. PROGRESS IN MATERIALS SCIENCE 2022; 130:100997. [PMID: 36660552 PMCID: PMC9844968 DOI: 10.1016/j.pmatsci.2022.100997] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
When blood first encounters the artificial surface of a medical device, a complex series of biochemical reactions is triggered, potentially resulting in clinical complications such as embolism/occlusion, inflammation, or device failure. Preventing thrombus formation on the surface of blood-contacting devices is crucial for maintaining device functionality and patient safety. As the number of patients reliant on blood-contacting devices continues to grow, minimizing the risk associated with these devices is vital towards lowering healthcare-associated morbidity and mortality. The current standard clinical practice primarily requires the systemic administration of anticoagulants such as heparin, which can result in serious complications such as post-operative bleeding and heparin-induced thrombocytopenia (HIT). Due to these complications, the administration of antithrombotic agents remains one of the leading causes of clinical drug-related deaths. To reduce the side effects spurred by systemic anticoagulation, researchers have been inspired by the hemocompatibility exhibited by natural phenomena, and thus have begun developing medical-grade surfaces which aim to exhibit total hemocompatibility via biomimicry. This review paper aims to address different bio-inspired surface modifications that increase hemocompatibility, discuss the limitations of each method, and explore the future direction for hemocompatible surface research.
Collapse
Affiliation(s)
- Megan Douglass
- School of Chemical, Materials and Biomedical Engineering, College of Engineering, University of Georgia, Athens, GA, USA
| | - Mark Garren
- School of Chemical, Materials and Biomedical Engineering, College of Engineering, University of Georgia, Athens, GA, USA
| | - Ryan Devine
- School of Chemical, Materials and Biomedical Engineering, College of Engineering, University of Georgia, Athens, GA, USA
| | - Arnab Mondal
- School of Chemical, Materials and Biomedical Engineering, College of Engineering, University of Georgia, Athens, GA, USA
| | - Hitesh Handa
- School of Chemical, Materials and Biomedical Engineering, College of Engineering, University of Georgia, Athens, GA, USA
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, GA, USA
| |
Collapse
|
20
|
Tian Y, Seeto WJ, Páez-Arias MA, Hahn MS, Lipke EA. Endothelial colony forming cell rolling and adhesion supported by peptide-grafted hydrogels. Acta Biomater 2022; 152:74-85. [PMID: 36031035 DOI: 10.1016/j.actbio.2022.08.047] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 07/28/2022] [Accepted: 08/22/2022] [Indexed: 01/13/2023]
Abstract
The aim of this study was to investigate the ability of peptides and peptide combinations to support circulating endothelial colony forming cell (ECFC) rolling and adhesion under shear flow, informing biomaterial design in moving toward rapid cardiovascular device endothelialization. ECFCs have high proliferative capability and can differentiate into endothelial cells, making them a promising cell source for endothelialization. Both single peptides and peptide combinations designed to target integrins α4β1 and α5β1 were coupled to poly(ethylene glycol) hydrogels, and their performance was evaluated by monitoring velocity patterns during the ECFC rolling process, in addition to firm adhesion (capture). Tether percentage and velocity fluctuation, a parameter newly defined here, were found to be valuable in assessing cell rolling velocity patterns and when used in combination were able to predict cell capture. REDV-containing peptides binding integrin α4β1 have been previously shown to reduce ECFC rolling velocity but not to support firm adhesion. This study finds that the performance of REDV-containing peptides in facilitating ECFC dynamic adhesion and capture can be improved by combination with α5β1 integrin-binding peptides, which support ECFC static adhesion. Moreover, when similar in length, the peptide combinations may have synergistic effects in capturing ECFCs. With matching lengths, the peptide combinations including CRRETAWAC(cyclic)+REDV, P_RGDS+KSSP_REDV, and P_RGDS+P_REDV showed high values in both tether percentage and velocity fluctuation and improvement in ECFC capture compared to the single peptides at the shear rate of 20 s-1. These newly identified peptide combinations have the potential to be used as vascular device coatings to recruit ECFCs. STATEMENT OF SIGNIFICANCE: Restoration of functional endothelium following placement of stents and vascular grafts is critical for maintaining long-term patency. Endothelial colony forming cells (ECFCs) circulating in blood flow are a valuable cell source for rapid endothelialization. Here we identify and test novel peptides and peptide combinations that can potentially be used as coatings for vascular devices to support rolling and capture of ECFCs from flow. In addition to the widely used assessment of final ECFC adhesion, we also recorded the rolling process to quantitatively evaluate the interaction between ECFCs and the peptides, obtaining detailed performance of the peptides and gaining insight into effective capture molecule design. Peptide combinations targeting both integrin α4β1 and integrin α5β1 showed the highest percentages of ECFC capture.
Collapse
Affiliation(s)
- Yuan Tian
- Department of Chemical Engineering, Auburn University, 212 Ross Hall, Auburn, AL, 36849, USA
| | - Wen J Seeto
- Department of Chemical Engineering, Auburn University, 212 Ross Hall, Auburn, AL, 36849, USA
| | - Mayra A Páez-Arias
- Department of Chemical Engineering, Auburn University, 212 Ross Hall, Auburn, AL, 36849, USA
| | - Mariah S Hahn
- Biomedical Engineering Department, Rensselaer Polytechnic Institute, Troy, NY, 12180-3590, USA
| | - Elizabeth A Lipke
- Department of Chemical Engineering, Auburn University, 212 Ross Hall, Auburn, AL, 36849, USA.
| |
Collapse
|
21
|
Gori T. Restenosis after Coronary Stent Implantation: Cellular Mechanisms and Potential of Endothelial Progenitor Cells (A Short Guide for the Interventional Cardiologist). Cells 2022; 11:cells11132094. [PMID: 35805178 PMCID: PMC9265311 DOI: 10.3390/cells11132094] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 06/26/2022] [Accepted: 06/28/2022] [Indexed: 02/05/2023] Open
Abstract
Coronary stents are among the most common therapies worldwide. Despite significant improvements in the biocompatibility of these devices throughout the last decades, they are prone, in as many as 10–20% of cases, to short- or long-term failure. In-stent restenosis is a multifactorial process with a complex and incompletely understood pathophysiology in which inflammatory reactions are of central importance. This review provides a short overview for the clinician on the cellular types responsible for restenosis with a focus on the role of endothelial progenitor cells. The mechanisms of restenosis are described, along with the cell-based attempts made to prevent it. While the focus of this review is principally clinical, experimental evidence provides some insight into the potential implications for prevention and therapy of coronary stent restenosis.
Collapse
Affiliation(s)
- Tommaso Gori
- German Center for Cardiac and Vascular Research (DZHK) Standort Rhein-Main, Department of Cardiology, University Medical Center Mainz, 55131 Mainz, Germany
| |
Collapse
|
22
|
Ribeiro DRP, Schmidt MM, Leguisamo N, Cambruzzi E, De Luca G, de Quadros AS. Immunohistochemical characteristics of coronary thrombi in ST-elevation myocardial infarction. AMERICAN HEART JOURNAL PLUS : CARDIOLOGY RESEARCH AND PRACTICE 2022; 18:100175. [PMID: 38559422 PMCID: PMC10978369 DOI: 10.1016/j.ahjo.2022.100175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 06/13/2022] [Accepted: 07/07/2022] [Indexed: 04/04/2024]
Abstract
Background and aims The dynamics and implications of intracoronary thrombus constituency in patients with ST-segment elevation myocardial infarction (STEMI) are not fully understood. We evaluated the expression of CD34, CD61and factor VIII surface markers in thrombi of patients with STEMI and its association with clinical and angiographic characteristics and major adverse cardiovascular events (MACE). Methods Patients presenting with STEMI undergoing aspiration thrombectomy during primary percutaneous coronary intervention (pPCI) were included. Morphological, histopathological and immunohistochemical aspects of thrombi were assessed by two pathologists blinded to clinical variables and outcomes. Results The mean age of the 245 patients included was 58 ± 12 years old, and 70 % were men. Regarding the thrombi microscopic patterns, 61 % were classified as recent, 20 % as lytic and 19 % as organized. There were higher levels of the CD61 index in patients with a history of heart failure. Smokers presented lower CD61 positive cells and CD61 index, but this association did not remain significant after multivariable analysis. There was an inverse correlation between CD61 positive cells and CD61 index with the time from onset of pain to the first medical contact, but no other significant association amongst clinical characteristics and antigenic expression. There was higher expression of the CD61 antigen in patients with in-hospital MACE, but statistical significance was borderline (p = 0.06). Conclusions In this cohort of patients with STEMI, immunohistochemistry of coronary thrombus showed a significantly higher platelet content in patients with previous heart failure and a trend in those with in-hospital MACE. Thrombus' platelet content was inversely related to ischemic time.
Collapse
Affiliation(s)
- Daniel Rios Pinto Ribeiro
- Instituto de Cardiologia do RS/Fundação Universitária de Cardiologia do Rio Grande do Sul – IC/FUC, Brazil
| | - Marcia Moura Schmidt
- Instituto de Cardiologia do RS/Fundação Universitária de Cardiologia do Rio Grande do Sul – IC/FUC, Brazil
| | - Natalia Leguisamo
- Instituto de Cardiologia do RS/Fundação Universitária de Cardiologia do Rio Grande do Sul – IC/FUC, Brazil
| | - Eduardo Cambruzzi
- Instituto de Cardiologia do RS/Fundação Universitária de Cardiologia do Rio Grande do Sul – IC/FUC, Brazil
| | - Giuseppe De Luca
- Division of Clinical and Experimental Cardiology AOU Sassari, University of Sassari, Italy
| | | |
Collapse
|
23
|
Pelliccia F, Zimarino M, De Luca G, Viceconte N, Tanzilli G, De Caterina R. Endothelial Progenitor Cells in Coronary Artery Disease: From Bench to Bedside. Stem Cells Transl Med 2022; 11:451-460. [PMID: 35365823 PMCID: PMC9154346 DOI: 10.1093/stcltm/szac010] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Accepted: 02/04/2022] [Indexed: 11/14/2022] Open
Abstract
Endothelial progenitor cells (EPCs) are a heterogeneous group of cells present in peripheral blood at various stages of endothelial differentiation. EPCs have been extensively investigated in patients with coronary artery disease (CAD), with controversial findings both on their role in atherosclerosis progression and in the process of neointimal growth after a percutaneous coronary intervention (PCI). Despite nearly 2 decades of experimental and clinical investigations, however, the significance of EPCs in clinical practice remains unclear and poorly understood. This review provides an update on the role of EPCs in the most common clinical scenarios that are experienced by cardiologists managing patients with CAD. We here summarize the main findings on the association of EPCs with cardiovascular risk factors, coronary atherosclerosis, and myocardial ischemia. We then discuss the potential effects of EPCs in post-PCI in-stent restenosis, as well as most recent findings with EPC-coated stents. Based on the mounting evidence of the relationship between levels of EPCs and several different adverse cardiovascular events, EPCs are emerging as novel predictive biomarkers of long-term outcomes in patients with CAD.
Collapse
Affiliation(s)
| | - Marco Zimarino
- Institute of Cardiology, “G. d’Annunzio” University, Chieti, Italy
- Cath Lab, SS. Annunziata Hospital, Chieti, Italy
| | - Giuseppe De Luca
- Division of Cardiology, Azienda Ospedaliero-Universitaria Maggiore della Carità, Università del Piemonte Orientale, Novara, Italy
| | - Nicola Viceconte
- Department of Cardiovascular Sciences, Sapienza University, Rome, Italy
| | - Gaetano Tanzilli
- Department of Cardiovascular Sciences, Sapienza University, Rome, Italy
| | | |
Collapse
|
24
|
Attachment of endothelial colony-forming cells onto a surface bearing immobilized anti-CD34 antibodies: Specific CD34 binding versus nonspecific binding. Biointerphases 2022; 17:031003. [PMID: 35589426 DOI: 10.1116/6.0001746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Cardiovascular disease is a leading cause of death worldwide; however, despite substantial advances in medical device surface modifications, no synthetic coatings have so far matched the native endothelium as the optimal hemocompatible surface for blood-contacting implants. A promising strategy for rapid restoration of the endothelium on blood-contacting biomedical devices entails attracting circulating endothelial cells or their progenitors, via immobilized cell-capture molecules; for example, anti-CD34 antibody to attract CD34+ endothelial colony-forming cells (ECFCs). Inherent is the assumption that the cells attracted to the biomaterial surface are bound exclusively via a specific CD34 binding. However, serum proteins might adsorb in-between or on the top of antibody molecules and attract ECFCs via other binding mechanisms. Here, we studied whether a surface with immobilized anti-CD34 antibodies attracts ECFCs via a specific CD34 binding or a nonspecific (non-CD34) binding. To minimize serum protein adsorption, a fouling-resistant layer of hyperbranched polyglycerol (HPG) was used as a "blank slate," onto which anti-CD34 antibodies were immobilized via aldehyde-amine coupling reaction after oxidation of terminal diols to aldehydes. An isotype antibody, mIgG1, was surface-immobilized analogously and was used as the control for antigen-binding specificity. Cell binding was also measured on the HPG hydrogel layer before and after oxidation. The surface analysis methods, x-ray photoelectron spectroscopy and time-of-flight secondary ion mass spectrometry, were used to verify the intended surface chemistries and revealed that the surface coverage of antibodies was sparse, yet the anti-CD34 antibody grafted surface-bound ECFCs very effectively. Moreover, it still captured the ECFCs after BSA passivation. However, cells also attached to oxidized HPG and immobilized mIgG1, though in much lower amounts. While our results confirm the effectiveness of attracting ECFCs via surface-bound anti-CD34 antibodies, our observation of a nonspecific binding component highlights the importance of considering its consequences in future studies.
Collapse
|
25
|
Lu J, Hu X, Yuan T, Cao J, Zhao Y, Xiong C, Li K, Ye X, Xu T, Zhao J. 3D-Printed Poly (P-Dioxanone) Stent for Endovascular Application: In Vitro Evaluations. Polymers (Basel) 2022; 14:polym14091755. [PMID: 35566924 PMCID: PMC9103802 DOI: 10.3390/polym14091755] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 04/13/2022] [Accepted: 04/19/2022] [Indexed: 02/04/2023] Open
Abstract
Rapid formation of innovative, inexpensive, personalized, and quickly reproducible artery bioresorbable stents (BRSs) is significantly important for treating dangerous and sometimes deadly cerebrovascular disorders. It is greatly challenging to give BRSs excellent mechanical properties, biocompatibility, and bioabsorbability. The current BRSs, which are mostly fabricated from poly-l-lactide (PLLA), are usually applied to coronary revascularization but may not be suitable for cerebrovascular revascularization. Here, novel 3D-printed BRSs for cerebrovascular disease enabling anti-stenosis and gradually disappearing after vessel endothelialization are designed and fabricated by combining biocompatible poly (p-dioxanone) (PPDO) and 3D printing technology for the first time. We can control the strut thickness and vessel coverage of BRSs by adjusting the printing parameters to make the size of BRSs suitable for small-diameter vascular use. We added bis-(2,6-diisopropylphenyl) carbodiimide (commercial name: stabaxol®-1) to PPDO to improve its hydrolytic stability without affecting its mechanical properties and biocompatibility. In vitro cell experiments confirmed that endothelial cells can be conveniently seeded and attached to the BRSs and subsequently demonstrated good proliferation ability. Owing to the excellent mechanical properties of the monofilaments fabricated by the PPDO, the 3D-printed BRSs with PPDO monofilaments support desirable flexibility, therefore offering a novel BRS application in the vascular disorders field.
Collapse
Affiliation(s)
- Junlin Lu
- Beijing Tiantan Hospital, Department of Neurosurgery, Capital Medical University, Beijing 100070, China; (J.L.); (Y.Z.)
| | - Xulin Hu
- Clinical Medical College & Affiliated Hospital of Chengdu University, Chengdu University, Chengdu 610081, China; (X.H.); (K.L.)
| | - Tianyu Yuan
- State Key Laboratory for Turbulence and Complex Systems, Department of Mechanics and Engineering Science, College of Engineering, Peking University, Beijing 100871, China;
| | - Jianfei Cao
- School of Materials and Environmental Engineering, Chengdu Technology University, Chengdu 610041, China;
| | - Yuanli Zhao
- Beijing Tiantan Hospital, Department of Neurosurgery, Capital Medical University, Beijing 100070, China; (J.L.); (Y.Z.)
- Beijing Translational Engineering Enter for 3D Printer in Clinical Neuroscience, Beijing 100070, China
| | - Chengdong Xiong
- Chengdu Institute of Organic Chemistry, Chinese Academy of Sciences, Chengdu 610041, China;
| | - Kainan Li
- Clinical Medical College & Affiliated Hospital of Chengdu University, Chengdu University, Chengdu 610081, China; (X.H.); (K.L.)
| | - Xun Ye
- Beijing Tiantan Hospital, Department of Neurosurgery, Capital Medical University, Beijing 100070, China; (J.L.); (Y.Z.)
- Beijing Translational Engineering Enter for 3D Printer in Clinical Neuroscience, Beijing 100070, China
- Correspondence: (X.Y.); (T.X.); (J.Z.)
| | - Tao Xu
- Department of Mechanical Engineering, Tsinghua University, Beijing 100084, China
- Bio-Intelligent Manufacturing and Living Matter Bioprinting Center, Research Institute of Tsinghua University in Shenzhen, Tsinghua University, Shenzhen 518057, China
- East China Institute of Digital Medical Engineering, Shangrao 334000, China
- Correspondence: (X.Y.); (T.X.); (J.Z.)
| | - Jizong Zhao
- Beijing Tiantan Hospital, Department of Neurosurgery, Capital Medical University, Beijing 100070, China; (J.L.); (Y.Z.)
- Beijing Translational Engineering Enter for 3D Printer in Clinical Neuroscience, Beijing 100070, China
- Correspondence: (X.Y.); (T.X.); (J.Z.)
| |
Collapse
|
26
|
Cardioprotective Mechanisms of Interrupted Anesthetic Preconditioning with Sevoflurane in the Setting of Ischemia/Reperfusion Injury in Rats. APPLIED SCIENCES-BASEL 2022. [DOI: 10.3390/app12031476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background: Anesthetic preconditioning (AP) is known to mimic ischemic preconditioning. The purpose of this study was to investigate the effects of an interrupted sevoflurane administration protocol on myocardial ischemia/reperfusion (I/R) injury. Methods: Male Wistar rats (n = 60) were ventilated for 30 min with room air (control group, CG) or with a mixture of air and sevoflurane (1 minimum alveolar concentration—MAC) in 5-min cycles, alternating with 5-min wash-out periods (preconditioned groups). Cytokines implicated in the AP response were measured. An (I/R) lesion was produced immediately after the sham intervention (CG) and preconditioning protocol (early AP group, EAPG) or 24 h after the intervention (late AP group, LAPG). The area of fibrosis, the degree of apoptosis and the number of c-kit+ cells was estimated for each group. Results: Cytokine levels were increased post AP. The area of fibrosis decreased in both EAPG and LAPG compared to the CG (p < 0.0001). When compared to the CG, the degree of apoptosis was reduced in both LAPG (p = 0.006) and EAPG (p = 0.007) and the number of c-kit+ cells was the greatest for the LAPG (p < 0.0001). Conclusions: Sevoflurane preconditioning, using an interrupted anesthesia protocol, is efficient in myocardial protection and could be beneficial to reduce perioperative or periprocedural ischemia in patients with increased cardiovascular risk.
Collapse
|
27
|
Yuan W, Xia D, Wu S, Zheng Y, Guan Z, Rau JV. A review on current research status of the surface modification of Zn-based biodegradable metals. Bioact Mater 2022; 7:192-216. [PMID: 34466727 PMCID: PMC8379348 DOI: 10.1016/j.bioactmat.2021.05.018] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 05/09/2021] [Accepted: 05/09/2021] [Indexed: 12/13/2022] Open
Abstract
Recently, zinc and its alloys have been proposed as promising candidates for biodegradable metals (BMs), owning to their preferable corrosion behavior and acceptable biocompatibility in cardiovascular, bone and gastrointestinal environments, together with Mg-based and Fe-based BMs. However, there is the desire for surface treatment for Zn-based BMs to better control their biodegradation behavior. Firstly, the implantation of some Zn-based BMs in cardiovascular environment exhibited intimal activation with mild inflammation. Secondly, for orthopedic applications, the biodegradation rates of Zn-based BMs are relatively slow, resulting in a long-term retention after fulfilling their mission. Meanwhile, excessive Zn2+ release during degradation will cause in vitro cytotoxicity and in vivo delayed osseointegration. In this review, we firstly summarized the current surface modification methods of Zn-based alloys for the industrial applications. Then we comprehensively summarized the recent progress of biomedical bulk Zn-based BMs as well as the corresponding surface modification strategies. Last but not least, the future perspectives towards the design of surface bio-functionalized coatings on Zn-based BMs for orthopedic and cardiovascular applications were also briefly proposed.
Collapse
Affiliation(s)
- Wei Yuan
- School of Materials Science and Engineering, Peking University, Beijing, 100871, China
| | - Dandan Xia
- School of Materials Science and Engineering, Peking University, Beijing, 100871, China
- National Engineering Laboratory for Digital and Material Technology of Stomatology, National Clinical Research Center for Oral Diseases, Beijing Key Laboratory of Digital Stomatology, National Medical Products Administration Key Laboratory for Dental Materials, Research Center of Engineering and Technology for Digital Dentistry, Ministry of Health, Beijing, 100081, China
| | - Shuilin Wu
- School of Materials Science & Engineering, The Key Laboratory of Advanced Ceramics and Machining Technology by the Ministry of Education of China, Tianjin University, Tianjin, 300072, China
| | - Yufeng Zheng
- School of Materials Science and Engineering, Peking University, Beijing, 100871, China
- National Engineering Laboratory for Digital and Material Technology of Stomatology, National Clinical Research Center for Oral Diseases, Beijing Key Laboratory of Digital Stomatology, National Medical Products Administration Key Laboratory for Dental Materials, Research Center of Engineering and Technology for Digital Dentistry, Ministry of Health, Beijing, 100081, China
| | - Zhenpeng Guan
- Orthopedics Department, Peking University Shougang Hospital, No. 9 Jinyuanzhuang Rd, Shijingshan District, Beijing, 100144, China
| | - Julietta V. Rau
- Istituto di Struttura della Materia, Consiglio Nazionale delle Ricerche (ISM-CNR), Via del Fosso del Cavaliere, 100-00133, Rome, Italy
- Sechenov First Moscow State Medical University, Institute of Pharmacy, Department of Analytical, Physical and Colloid Chemistry, Trubetskaya 8, build. 2, 119991, Moscow, Russia
| |
Collapse
|
28
|
Towards Biohybrid Lung Development-Fibronectin-Coating Bestows Hemocompatibility of Gas Exchange Hollow Fiber Membranes by Improving Flow-Resistant Endothelialization. MEMBRANES 2021; 12:membranes12010035. [PMID: 35054561 PMCID: PMC8779364 DOI: 10.3390/membranes12010035] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 12/22/2021] [Accepted: 12/22/2021] [Indexed: 01/26/2023]
Abstract
To provide an alternative treatment option for patients with end-stage lung disease, we aim for biohybrid lung development (BHL) based on hollow fiber membrane (HFM) technology used in extracorporeal membrane oxygenators. For long-term BHL application, complete hemocompatibility of all blood-contacting surfaces is indispensable and can be achieved by their endothelialization. Indeed, albumin/heparin (AH) coated HFM enables initial endothelialization, but as inexplicable cell loss under flow conditions was seen, we assessed an alternative HFM coating using fibronectin (FN). Therefore, endothelial cell (EC) adherence and viability on both coated HFM were analyzed by fluorescence-based staining. Functional leukocyte and thrombocyte adhesion assays were performed to evaluate hemocompatibility, also in comparison to blood plasma coated HFM as a clinically relevant control. To assess monolayer resistance and EC behavior under clinically relevant flow conditions, a mock circulation setup was established, which also facilitates imitation of lung-disease specific blood gas settings. Besides quantification of flow-associated cell loss, endothelial responses towards external stimuli, like flow exposure or TNFα stimulation, were analyzed by qRT-PCR, focusing on inflammation, thrombus formation and extracellular matrix production. Under static conditions, both coated HFM enabled the generation of a viable, confluent, non-inflammatory and anti-thrombogenic monolayer. However, by means of homogenous FN coating, cell retention and physiologic gene regulation towards an improved hemocompatible-and extracellular matrix producing phenotype, was significantly superior compared to the inhomogeneous AH coating. In summary, our adaptable in-house FN coating secures the endothelial requirements for long-term BHL application and may promote monolayer establishment on all other blood contacting surfaces of the BHL (e.g., cannulae).
Collapse
|
29
|
Kuchinka J, Willems C, Telyshev DV, Groth T. Control of Blood Coagulation by Hemocompatible Material Surfaces-A Review. Bioengineering (Basel) 2021; 8:215. [PMID: 34940368 PMCID: PMC8698751 DOI: 10.3390/bioengineering8120215] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 12/10/2021] [Accepted: 12/12/2021] [Indexed: 11/16/2022] Open
Abstract
Hemocompatibility of biomaterials in contact with the blood of patients is a prerequisite for the short- and long-term applications of medical devices such as cardiovascular stents, artificial heart valves, ventricular assist devices, catheters, blood linings and extracorporeal devices such as artificial kidneys (hemodialysis), extracorporeal membrane oxygenation (ECMO) and cardiopulmonary bypass. Although lower blood compatibility of materials and devices can be handled with systemic anticoagulation, its side effects, such as an increased bleeding risk, make materials that have a better hemocompatibility highly desirable, particularly in long-term applications. This review provides a short overview on the basic mechanisms of blood coagulation including plasmatic coagulation and blood platelets, as well as the activation of the complement system. Furthermore, a survey on concepts for tailoring the blood response of biomaterials to improve the hemocompatibility of medical devices is given which covers different approaches that either inhibit interaction of material surfaces with blood components completely or control the response of the coagulation system, blood platelets and leukocytes.
Collapse
Affiliation(s)
- Janna Kuchinka
- Department Biomedical Materials, Institute of Pharmacy, Martin Luther University Halle-Wittenberg, 06120 Halle (Saale), Germany; (J.K.); (C.W.)
| | - Christian Willems
- Department Biomedical Materials, Institute of Pharmacy, Martin Luther University Halle-Wittenberg, 06120 Halle (Saale), Germany; (J.K.); (C.W.)
| | - Dmitry V. Telyshev
- Institute of Biomedical Systems, National Research University of Electronic Technology, Zelenograd, 124498 Moscow, Russia;
- Laboratory of Biomedical Nanotechnologies, Institute of Bionic Technologies and Engineering, I.M. Sechenov First Moscow State University, 119991 Moscow, Russia
| | - Thomas Groth
- Department Biomedical Materials, Institute of Pharmacy, Martin Luther University Halle-Wittenberg, 06120 Halle (Saale), Germany; (J.K.); (C.W.)
- Laboratory of Biomedical Nanotechnologies, Institute of Bionic Technologies and Engineering, I.M. Sechenov First Moscow State University, 119991 Moscow, Russia
- Interdisciplinary Center of Materials Science, Martin Luther University Halle-Wittenberg, 06120 Halle (Saale), Germany
| |
Collapse
|
30
|
Wang J, Qian HL, Chen SY, Huang WP, Huang DN, Hao HY, Ren KF, Wang YB, Fu GS, Ji J. miR-22 eluting cardiovascular stent based on a self-healable spongy coating inhibits in-stent restenosis. Bioact Mater 2021; 6:4686-4696. [PMID: 34095625 PMCID: PMC8164007 DOI: 10.1016/j.bioactmat.2021.04.037] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 04/09/2021] [Accepted: 04/27/2021] [Indexed: 11/28/2022] Open
Abstract
The in-stent restenosis (IRS) after the percutaneous coronary intervention contributes to the major treatment failure of stent implantation. MicroRNAs have been revealed as powerful gene medicine to regulate endothelial cells (EC) and smooth muscle cells (SMC) in response to vascular injury, providing a promising therapeutic candidate to inhibit IRS. However, the controllable loading and eluting of hydrophilic bioactive microRNAs pose a challenge to current lipophilic stent coatings. Here, we developed a microRNA eluting cardiovascular stent via the self-healing encapsulation process based on an amphipathic poly(ε-caprolactone)-poly(ethylene glycol)-poly(ε-caprolactone) (PCL-PEG-PCL, PCEC) triblock copolymer spongy network. The miR-22 was used as a model microRNA to regulate SMC. The dynamic porous coating realized the uniform and controllable loading of miR-22, reaching the highest dosage of 133 pmol cm-2. We demonstrated that the sustained release of miR-22 dramatically enhanced the contractile phenotype of SMC without interfering with the proliferation of EC, thus leading to the EC dominating growth at an EC/SMC ratio of 5.4. More importantly, the PCEC@miR-22 coated stents showed reduced inflammation, low switching of SMC phenotype, and low secretion of extracellular matrix, which significantly inhibited IRS. This work provides a simple and robust coating platform for the delivery of microRNAs on cardiovascular stent, which may extend to other combination medical devices, and facilitate practical application of bioactive agents in clinics.
Collapse
Affiliation(s)
- Jing Wang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Hong-Lin Qian
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Sheng-Yu Chen
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, 310016, China
| | - Wei-Pin Huang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Dan-Ni Huang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Hong-Ye Hao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Ke-Feng Ren
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, 310016, China
| | - Yun-Bing Wang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, China
| | - Guo-Sheng Fu
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, 310016, China
| | - Jian Ji
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, 310016, China
| |
Collapse
|
31
|
Wang X, Wang R, Jiang L, Xu Q, Guo X. Endothelial repair by stem and progenitor cells. J Mol Cell Cardiol 2021; 163:133-146. [PMID: 34743936 DOI: 10.1016/j.yjmcc.2021.10.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Revised: 10/20/2021] [Accepted: 10/26/2021] [Indexed: 12/19/2022]
Abstract
The integrity of the endothelial barrier is required to maintain vascular homeostasis and fluid balance between the circulatory system and surrounding tissues and to prevent the development of vascular disease. However, the origin of the newly developed endothelial cells is still controversial. Stem and progenitor cells have the potential to differentiate into endothelial cell lines and stimulate vascular regeneration in a paracrine/autocrine fashion. The one source of new endothelial cells was believed to come from the bone marrow, which was challenged by the recent findings. By administration of new techniques, including genetic cell lineage tracing and single cell RNA sequencing, more solid data were obtained that support the concept of stem/progenitor cells for regenerating damaged endothelium. Specifically, it was found that tissue resident endothelial progenitors located in the vessel wall were crucial for endothelial repair. In this review, we summarized the latest advances in stem and progenitor cell research in endothelial regeneration through findings from animal models and discussed clinical data to indicate the future direction of stem cell therapy.
Collapse
Affiliation(s)
- Xuyang Wang
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ruilin Wang
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Liujun Jiang
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qingbo Xu
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Xiaogang Guo
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
32
|
Xiao ST, Kuang CY. Endothelial progenitor cells and coronary artery disease: Current concepts and future research directions. World J Clin Cases 2021; 9:8953-8966. [PMID: 34786379 PMCID: PMC8567528 DOI: 10.12998/wjcc.v9.i30.8953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 05/12/2021] [Accepted: 08/18/2021] [Indexed: 02/06/2023] Open
Abstract
Vascular injury is a frequent pathology in coronary artery disease. To repair the vasculature, scientists have found that endothelial progenitor cells (EPCs) have excellent properties associated with angiogenesis. Over time, research on EPCs has made encouraging progress regardless of pathology or clinical technology. This review focuses on the origins and cell markers of EPCs, and the connection between EPCs and coronary artery disease. In addition, we summarized various studies of EPC-capturing stents and EPC infusion therapy, and aim to learn from past technology to predict the future.
Collapse
Affiliation(s)
- Sen-Tong Xiao
- Department of Cardiovascular Diseases, People’s Hospital Affiliated to Guizhou Medical University, Guiyang 550003, Guizhou Province, China
| | - Chun-Yan Kuang
- Department of Cardiovascular Diseases, Guizhou Provincial People's Hospital, Guiyang 550003, Guizhou Province, China
| |
Collapse
|
33
|
Impact of REDV peptide density and its linker structure on the capture, movement, and adhesion of flowing endothelial progenitor cells in microfluidic devices. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 129:112381. [PMID: 34579900 DOI: 10.1016/j.msec.2021.112381] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 07/29/2021] [Accepted: 08/13/2021] [Indexed: 11/23/2022]
Abstract
Ligand-immobilization to stents and vascular grafts is expected to promote endothelialization by capturing flowing endothelial progenitor cells (EPCs). However, the optimized ligand density and linker structure have not been fully elucidated. Here, we report that flowing EPCs were selectively captured by the REDV peptide conjugated with a short linker. The microchannel surface was modified with the REDV peptide via Gly-Gly-Gly (G3), (Gly-Gly-Gly)3 (G9), and diethylene glycol (diEG) linkers, and the moving velocity and captured ratio were evaluated. On the unmodified microchannels, the moving velocity of the cells exhibited a unimodal distribution similar to the liquid flow. The velocity of the endothelial cells and EPCs on the peptide-immobilized surface indicated a bimodal distribution, and approximately 20 to 30% of cells moved slower than the liquid flow, suggesting that the cells were captured and rolled on the surface. When the immobilized ligand density was lower than 1 molecule/nm2, selective cell capture was observed only in REDV with G3 and diEG linkers, but not in G9 linkers. An in silico study revealed that the G9 linker tends to form a bent structure, and the REDV peptide is oriented to the substrate side. These results indicated that REDV captured the flowing EPC in a sequence-specific manner, and that the short linker was more adequate.
Collapse
|
34
|
A Recombinant Fusion Construct between Human Serum Albumin and NTPDase CD39 Allows Anti-Inflammatory and Anti-Thrombotic Coating of Medical Devices. Pharmaceutics 2021; 13:pharmaceutics13091504. [PMID: 34575580 PMCID: PMC8466136 DOI: 10.3390/pharmaceutics13091504] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 09/15/2021] [Accepted: 09/16/2021] [Indexed: 11/16/2022] Open
Abstract
Medical devices directly exposed to blood are commonly used to treat cardiovascular diseases. However, these devices are associated with inflammatory reactions leading to delayed healing, rejection of foreign material or device-associated thrombus formation. We developed a novel recombinant fusion protein as a new biocompatible coating strategy for medical devices with direct blood contact. We genetically fused human serum albumin (HSA) with ectonucleoside triphosphate diphosphohydrolase-1 (CD39), a promising anti-thrombotic and anti-inflammatory drug candidate. The HSA-CD39 fusion protein is highly functional in degrading ATP and ADP, major pro-inflammatory reagents and platelet agonists. Their enzymatic properties result in the generation of AMP, which is further degraded by CD73 to adenosine, an anti-inflammatory and anti-platelet reagent. HSA-CD39 is functional after lyophilisation, coating and storage of coated materials for up to 8 weeks. HSA-CD39 coating shows promising and stable functionality even after sterilisation and does not hinder endothelialisation of primary human endothelial cells. It shows a high level of haemocompatibility and diminished blood cell adhesion when coated on nitinol stents or polyvinylchloride tubes. In conclusion, we developed a new recombinant fusion protein combining HSA and CD39, and demonstrated that it has potential to reduce thrombotic and inflammatory complications often associated with medical devices directly exposed to blood.
Collapse
|
35
|
Jiang L, Chen T, Sun S, Wang R, Deng J, Lyu L, Wu H, Yang M, Pu X, Du L, Chen Q, Hu Y, Hu X, Zhou Y, Xu Q, Zhang L. Nonbone Marrow CD34 + Cells Are Crucial for Endothelial Repair of Injured Artery. Circ Res 2021; 129:e146-e165. [PMID: 34474592 DOI: 10.1161/circresaha.121.319494] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Liujun Jiang
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China (L. Jiang, T. Chen, S. Sun, R. Wang, J. Deng, L. Lyu, H. Wu, X. Pu, L. Du, Y. Hu, X. Hu, Y. Zhou, Q. Xu)
| | - Ting Chen
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China (L. Jiang, T. Chen, S. Sun, R. Wang, J. Deng, L. Lyu, H. Wu, X. Pu, L. Du, Y. Hu, X. Hu, Y. Zhou, Q. Xu).,Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou, Zhejiang Province, China (T. Chen)
| | - Shasha Sun
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China (L. Jiang, T. Chen, S. Sun, R. Wang, J. Deng, L. Lyu, H. Wu, X. Pu, L. Du, Y. Hu, X. Hu, Y. Zhou, Q. Xu).,Department of Cardiology and Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China. (S. Sun, M. Yang, Q. Chen, L. Zhang)
| | - Ruilin Wang
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China (L. Jiang, T. Chen, S. Sun, R. Wang, J. Deng, L. Lyu, H. Wu, X. Pu, L. Du, Y. Hu, X. Hu, Y. Zhou, Q. Xu)
| | - Jiacheng Deng
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China (L. Jiang, T. Chen, S. Sun, R. Wang, J. Deng, L. Lyu, H. Wu, X. Pu, L. Du, Y. Hu, X. Hu, Y. Zhou, Q. Xu)
| | - Lingxia Lyu
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China (L. Jiang, T. Chen, S. Sun, R. Wang, J. Deng, L. Lyu, H. Wu, X. Pu, L. Du, Y. Hu, X. Hu, Y. Zhou, Q. Xu)
| | - Hong Wu
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China (L. Jiang, T. Chen, S. Sun, R. Wang, J. Deng, L. Lyu, H. Wu, X. Pu, L. Du, Y. Hu, X. Hu, Y. Zhou, Q. Xu)
| | - Mei Yang
- Department of Cardiology and Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China. (S. Sun, M. Yang, Q. Chen, L. Zhang)
| | - Xiangyuan Pu
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China (L. Jiang, T. Chen, S. Sun, R. Wang, J. Deng, L. Lyu, H. Wu, X. Pu, L. Du, Y. Hu, X. Hu, Y. Zhou, Q. Xu)
| | - Luping Du
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China (L. Jiang, T. Chen, S. Sun, R. Wang, J. Deng, L. Lyu, H. Wu, X. Pu, L. Du, Y. Hu, X. Hu, Y. Zhou, Q. Xu)
| | - Qishan Chen
- Department of Cardiology and Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China. (S. Sun, M. Yang, Q. Chen, L. Zhang)
| | - Yanhua Hu
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China (L. Jiang, T. Chen, S. Sun, R. Wang, J. Deng, L. Lyu, H. Wu, X. Pu, L. Du, Y. Hu, X. Hu, Y. Zhou, Q. Xu)
| | - Xiaosheng Hu
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China (L. Jiang, T. Chen, S. Sun, R. Wang, J. Deng, L. Lyu, H. Wu, X. Pu, L. Du, Y. Hu, X. Hu, Y. Zhou, Q. Xu)
| | - Yijiang Zhou
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China (L. Jiang, T. Chen, S. Sun, R. Wang, J. Deng, L. Lyu, H. Wu, X. Pu, L. Du, Y. Hu, X. Hu, Y. Zhou, Q. Xu)
| | - Qingbo Xu
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China (L. Jiang, T. Chen, S. Sun, R. Wang, J. Deng, L. Lyu, H. Wu, X. Pu, L. Du, Y. Hu, X. Hu, Y. Zhou, Q. Xu).,Centre for Clinical Pharmacology, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom (Q. Xu)
| | - Li Zhang
- Department of Cardiology and Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China. (S. Sun, M. Yang, Q. Chen, L. Zhang)
| |
Collapse
|
36
|
Nguyen DT, Smith AF, Jiménez JM. Stent strut streamlining and thickness reduction promote endothelialization. J R Soc Interface 2021; 18:20210023. [PMID: 34404229 PMCID: PMC8371379 DOI: 10.1098/rsif.2021.0023] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 07/22/2021] [Indexed: 12/15/2022] Open
Abstract
Stent thrombosis (ST) carries a high risk of myocardial infarction and death. Lack of endothelial coverage is an important prognostic indicator of ST after stenting. While stent strut thickness is a critical factor in ST, a mechanistic understanding of its effect is limited and the role of haemodynamics is unclear. Endothelialization was tested using a wound-healing assay and five different stent strut models ranging in height between 50 and 150 µm for circular arc (CA) and rectangular (RT) geometries and a control without struts. Under static conditions, all stent strut surfaces were completely endothelialized. Reversing pulsatile disturbed flow caused full endothelialization, except for the stent strut surfaces of the 100 and 150 µm RT geometries, while fully antegrade pulsatile undisturbed flow with a higher mean wall shear stress caused only the control and the 50 µm CA geometries to be fully endothelialized. Modest streamlining and decrease in height of the stent struts improved endothelial coverage of the peri-strut and stent strut surfaces in a haemodynamics dependent manner. This study highlights the impact of the stent strut height (thickness) and geometry (shape) on the local haemodynamics, modulating reendothelialization after stenting, an important factor in reducing the risk of stent thrombosis.
Collapse
Affiliation(s)
- Duy T. Nguyen
- Department of Mechanical and Industrial Engineering, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Alexander F. Smith
- Department of Biomedical Engineering, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Juan M. Jiménez
- Department of Mechanical and Industrial Engineering, University of Massachusetts Amherst, Amherst, MA 01003, USA
- Department of Biomedical Engineering, University of Massachusetts Amherst, Amherst, MA 01003, USA
| |
Collapse
|
37
|
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: 75] [Impact Index Per Article: 25.0] [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
|
38
|
Dal Sasso E, Zamuner A, Filippi A, Romanato F, Palmosi T, Vedovelli L, Gregori D, Gómez Ribelles JL, Russo T, Gloria A, Iop L, Gerosa G, Dettin M. Covalent functionalization of decellularized tissues accelerates endothelialization. Bioact Mater 2021; 6:3851-3864. [PMID: 33937589 PMCID: PMC8065253 DOI: 10.1016/j.bioactmat.2021.04.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 03/31/2021] [Accepted: 04/01/2021] [Indexed: 12/17/2022] Open
Abstract
In the field of tissue regeneration, the lack of a stable endothelial lining may affect the hemocompatibility of both synthetic and biological replacements. These drawbacks might be prevented by specific biomaterial functionalization to induce selective endothelial cell (EC) adhesion. Decellularized bovine pericardia and porcine aortas were selectively functionalized with a REDV tetrapeptide at 10−5 M and 10−6 M working concentrations. The scaffold-bound peptide was quantified and REDV potential EC adhesion enhancement was evaluated in vitro by static seeding of human umbilical vein ECs. The viable cells and MTS production were statistically higher in functionalized tissues than in control. Scaffold histoarchitecture, geometrical features, and mechanical properties were unaffected by peptide anchoring. The selective immobilization of REDV was effective in accelerating ECs adhesion while promoting proliferation in functionalized decellularized tissues intended for blood-contacting applications. Covalent functionalization of the decellularized tissues with REDV peptide accelerates endothelialization. New covalent grafting method not inducing collagen cross-linking. Measurements through two photon miscroscopy allow the quantification of biological matrix bound peptide. The decellularized tissues can be changed by chemical procedures to promote specific cellular behaviour with ECM preservation.
Collapse
Affiliation(s)
- Eleonora Dal Sasso
- Department of Cardiac, Thoracic and Vascular Sciences and Venetian Institute of Molecular Medicine, Padua, Italy
| | - Annj Zamuner
- Department of Industrial Engineering, University of Padua, Padua, Italy.,LIFELAB Program, Consorzio per la Ricerca Sanitaria, CORIS, Veneto Region, Italy
| | - Andrea Filippi
- LIFELAB Program, Consorzio per la Ricerca Sanitaria, CORIS, Veneto Region, Italy.,Department of Physics and Astronomy "G. Galilei", University of Padua, Padua, Italy.,Fondazione Bruno Kessler, Trento, Italy.,Institute of Pediatric Research Città della Speranza, Padua, Italy
| | - Filippo Romanato
- LIFELAB Program, Consorzio per la Ricerca Sanitaria, CORIS, Veneto Region, Italy.,Department of Physics and Astronomy "G. Galilei", University of Padua, Padua, Italy.,Institute of Pediatric Research Città della Speranza, Padua, Italy
| | - Tiziana Palmosi
- Department of Cardiac, Thoracic and Vascular Sciences and Venetian Institute of Molecular Medicine, Padua, Italy
| | - Luca Vedovelli
- Department of Cardiac, Thoracic and Vascular Sciences and Venetian Institute of Molecular Medicine, Padua, Italy
| | - Dario Gregori
- Department of Cardiac, Thoracic and Vascular Sciences and Venetian Institute of Molecular Medicine, Padua, Italy
| | - José Luís Gómez Ribelles
- Center for Biomaterials and Tissue Engineering, CBIT, Universitat Politècnica de València, València, Spain.,Biomedical Research Networking Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Valencia, Spain
| | - Teresa Russo
- Institute of Polymers, Composites and Biomaterials, National Research Council of Italy, Naples, Italy
| | - Antonio Gloria
- Institute of Polymers, Composites and Biomaterials, National Research Council of Italy, Naples, Italy
| | - Laura Iop
- Department of Cardiac, Thoracic and Vascular Sciences and Venetian Institute of Molecular Medicine, Padua, Italy.,LIFELAB Program, Consorzio per la Ricerca Sanitaria, CORIS, Veneto Region, Italy
| | - Gino Gerosa
- Department of Cardiac, Thoracic and Vascular Sciences and Venetian Institute of Molecular Medicine, Padua, Italy.,LIFELAB Program, Consorzio per la Ricerca Sanitaria, CORIS, Veneto Region, Italy
| | - Monica Dettin
- Department of Industrial Engineering, University of Padua, Padua, Italy.,LIFELAB Program, Consorzio per la Ricerca Sanitaria, CORIS, Veneto Region, Italy
| |
Collapse
|
39
|
Wulf K, Arbeiter D, Matschegewski C, Teske M, Huling J, Schmitz KP, Grabow N, Kohse S. Smart releasing electrospun nanofibers—poly: L.lactide fibers as dual drug delivery system for biomedical application. Biomed Mater 2020; 16:015022. [DOI: 10.1088/1748-605x/abbec8] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|
40
|
Bashth OS, Elkhodiry MA, Laroche G, Hoesli CA. Surface grafting of Fc-binding peptides as a simple platform to immobilize and identify antibodies that selectively capture circulating endothelial progenitor cells. Biomater Sci 2020; 8:5465-5475. [PMID: 32902522 DOI: 10.1039/d0bm00650e] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Antibody surface immobilization is a promising strategy to capture cells of interest from circulating fluids in vitro and in vivo. An application of particular interest in vascular interventions is to capture endothelial progenitor cells (EPCs) on the surface of stents to accelerate endothelialization. The clinical impact of EPC capture stents has been limited by the lack of efficient selective cell capture. Here, we describe a simple method to immobilize a variety of immunoglobulin G antibodies through their fragment crystallizable (Fc) regions via surface-conjugated RRGW peptides for cell capture applications. As an EPC capture model, peripheral blood endothelial colony-forming cells suspended in cell culture medium with up to 70% serum were captured by immobilized anti-CD144, anti-CD34 or anti-CD309 antibodies under laminar flow. The endothelial colony-forming cells were successfully enriched from a mixture with peripheral blood mononuclear cells using surfaces with anti-CD309 but not anti-CD45. This antibody immobilization approach holds great promise to engineer vascular biomaterials with improved EPC capture potential. The ease of immobilizing different antibodies using the same Fc-binding peptide surface grafting chemistry renders this platform suitable to screen antibodies that maximize cell capture efficiency and selectivity.
Collapse
Affiliation(s)
- Omar S Bashth
- Department of Chemical Engineering, McGill University, Canada.
| | | | - Gaétan Laroche
- Centre de Recherche du CHU de Québec & Département de Génie des Mines, des Matériaux et de la Métallurgie, Université Laval, Canada
| | - Corinne A Hoesli
- Department of Chemical Engineering, McGill University, Canada. and Department of Biomedical Engineering, McGill University, Canada
| |
Collapse
|
41
|
Wang J, Xue Y, Liu J, Hu M, Zhang H, Ren K, Wang Y, Ji J. Hierarchical Capillary Coating to Biofunctionlize Drug-Eluting Stent for Improving Endothelium Regeneration. RESEARCH (WASHINGTON, D.C.) 2020; 2020:1458090. [PMID: 32885169 PMCID: PMC7455884 DOI: 10.34133/2020/1458090] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Accepted: 07/23/2020] [Indexed: 04/12/2023]
Abstract
The drug-eluting stent (DES) has become one of the most successful and important medical devices for coronary heart disease, but yet suffers from insufficient endothelial cell (EC) growth and intima repair, eventually leading to treatment failure. Although biomacromolecules such as vascular endothelial growth factor (VEGF) would be promising to promote the intima regeneration, combining hydrophilic and vulnerable biomacromolecules with hydrophobic drugs as well as preserving the bioactivity after harsh treatments pose a huge challenge. Here, we report on a design of hierarchical capillary coating, which composes a base solid region and a top microporous region for incorporating rapamycin and VEGF, respectively. The top spongy region can guarantee the efficient, safe, and controllable loading of VEGF up to 1 μg/cm2 in 1 minute, providing a distinctive real-time loading capacity for saving the bioactivity. Based on this, we demonstrate that our rapamycin-VEGF hierarchical coating impressively promoted the competitive growth of endothelial cells over smooth muscle cells (ratio of EC/SMC~25) while relieving the adverse impact of rapamycin to ECs. We further conducted the real-time loading of VEGF on stents and demonstrate that the hierarchical combination of rapamycin and VEGF showed remarkable endothelium regeneration while maintaining a very low level of in-stent restenosis. This work paves an avenue for the combination of both hydrophobic and hydrophilic functional molecules, which should benefit the next generation of DES and may extend applications to diversified combination medical devices.
Collapse
Affiliation(s)
- Jing Wang
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Yunfan Xue
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Jun Liu
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Mi Hu
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - He Zhang
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Kefeng Ren
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Yunbing Wang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| | - Jian Ji
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| |
Collapse
|
42
|
Perrotta F, Perna A, Komici K, Nigro E, Mollica M, D’Agnano V, De Luca A, Guerra G. The State of Art of Regenerative Therapy in Cardiovascular Ischemic Disease: Biology, Signaling Pathways, and Epigenetics of Endothelial Progenitor Cells. Cells 2020; 9:E1886. [PMID: 32796767 PMCID: PMC7465688 DOI: 10.3390/cells9081886] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 07/19/2020] [Accepted: 08/07/2020] [Indexed: 12/18/2022] Open
Abstract
Ischemic heart disease is currently a major cause of mortality and morbidity worldwide. Nevertheless, the actual therapeutic scenario does not target myocardial cell regeneration and consequently, the progression toward the late stage of chronic heart failure is common. Endothelial progenitor cells (EPCs) are bone marrow-derived stem cells that contribute to the homeostasis of the endothelial wall in acute and chronic ischemic disease. Calcium modulation and other molecular pathways (NOTCH, VEGFR, and CXCR4) contribute to EPC proliferation and differentiation. The present review provides a summary of EPC biology with a particular focus on the regulatory pathways of EPCs and describes promising applications for cardiovascular cell therapy.
Collapse
Affiliation(s)
- Fabio Perrotta
- Dipartimento di Medicina e Scienze della Salute “V.Tiberio”, Università del Molise, 86100 Campobasso, Italy; (A.P.); (K.K.); (G.G.)
| | - Angelica Perna
- Dipartimento di Medicina e Scienze della Salute “V.Tiberio”, Università del Molise, 86100 Campobasso, Italy; (A.P.); (K.K.); (G.G.)
| | - Klara Komici
- Dipartimento di Medicina e Scienze della Salute “V.Tiberio”, Università del Molise, 86100 Campobasso, Italy; (A.P.); (K.K.); (G.G.)
| | - Ersilia Nigro
- Dipartimento di Scienze e Tecnologie Ambientali, Biologiche, Farmaceutiche, Università della Campania “Luigi Vanvitelli”, 81100 Caserta, Italy;
- CEINGE-Biotecnologie avanzate, 80145 Naples, Italy
| | - Mariano Mollica
- Dipartimento di Scienze Mediche Traslazionali, Università della Campania “Luigi Vanvitelli”, 80131 Naples, Italy; (M.M.); (V.D.)
| | - Vito D’Agnano
- Dipartimento di Scienze Mediche Traslazionali, Università della Campania “Luigi Vanvitelli”, 80131 Naples, Italy; (M.M.); (V.D.)
| | - Antonio De Luca
- Department of Mental and Physical Health and Preventive Medicine, Section of Human Anatomy, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy;
| | - Germano Guerra
- Dipartimento di Medicina e Scienze della Salute “V.Tiberio”, Università del Molise, 86100 Campobasso, Italy; (A.P.); (K.K.); (G.G.)
| |
Collapse
|
43
|
Badv M, Bayat F, Weitz JI, Didar TF. Single and multi-functional coating strategies for enhancing the biocompatibility and tissue integration of blood-contacting medical implants. Biomaterials 2020; 258:120291. [PMID: 32798745 DOI: 10.1016/j.biomaterials.2020.120291] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 06/27/2020] [Accepted: 08/01/2020] [Indexed: 12/27/2022]
Abstract
Device-associated clot formation and poor tissue integration are ongoing problems with permanent and temporary implantable medical devices. These complications lead to increased rates of mortality and morbidity and impose a burden on healthcare systems. In this review, we outline the current approaches for developing single and multi-functional surface coating techniques that aim to circumvent the limitations associated with existing blood-contacting medical devices. We focus on surface coatings that possess dual hemocompatibility and biofunctionality features and discuss their advantages and shortcomings to providing a biocompatible and biodynamic interface between the medical implant and blood. Lastly, we outline the newly developed surface modification techniques that use lubricant-infused coatings and discuss their unique potential and limitations in mitigating medical device-associated complications.
Collapse
Affiliation(s)
- Maryam Badv
- School of Biomedical Engineering, McMaster University, Hamilton, Ontario, Canada; Department of Mechanical Engineering, McMaster University, Hamilton, Ontario, Canada
| | - Fereshteh Bayat
- School of Biomedical Engineering, McMaster University, Hamilton, Ontario, Canada
| | - Jeffrey I Weitz
- School of Biomedical Engineering, McMaster University, Hamilton, Ontario, Canada; Thrombosis & Atherosclerosis Research Institute (TaARI), Hamilton, Ontario, Canada; Department of Medicine, McMaster University, Hamilton, Ontario, Canada; Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Tohid F Didar
- School of Biomedical Engineering, McMaster University, Hamilton, Ontario, Canada; Department of Mechanical Engineering, McMaster University, Hamilton, Ontario, Canada; Institute for Infectious Disease Research (IIDR), McMaster University, Hamilton, Ontario, Canada.
| |
Collapse
|
44
|
Park KS, Kang SN, Kim DH, Kim HB, Im KS, Park W, Hong YJ, Han DK, Joung YK. Late endothelial progenitor cell-capture stents with CD146 antibody and nanostructure reduce in-stent restenosis and thrombosis. Acta Biomater 2020; 111:91-101. [PMID: 32434081 DOI: 10.1016/j.actbio.2020.05.011] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 05/08/2020] [Accepted: 05/08/2020] [Indexed: 12/13/2022]
Abstract
The restoration of damaged endothelium is promising to reduce side effects, including restenosis and thrombosis, in the stent treatment for vascular diseases. Current technologies based on drug delivery for these complications still do not satisfy patients due to invariant recurrence rate. Recently, even if one approach was applied to clinical trial to develop the firstly commercialized stent employing circulating endothelial progenitor cells (EPCs) in blood vessels, it resulted in failure in clinical trial. Based on instruction of the failed case, we designed an advanced EPC-capture stent covered with anti-CD146 antibody (Ab) immobilized silicone nanofilament (SiNf) for the highly efficient and specific capture of not early but late stage of EPCs. In vitro cell capture test demonstrates enhanced capture efficiency and adhesion morphology of late EPCs on the modified substrate. The modified substrates could capture 8 times more late EPCs and even 3 times more mesenchymal stem cells (MSCs) as compared to unmodified one. A porcine model with high similarity to human reproduced in vivo results ideally translated from in vitro cell capture results. As restenosis indicators, lumen area, neointimal rate and stenosis area for modified stents were reduced at the range of 30-60% as compared to those for bare metal stent (BMS). Fibrin score indicating thrombosis was lowered less than half as comparing to that on BMS. These inspiring results are attributed to ~2-fold increased endothelial coverage, determined by immuno-histological staining. Taken together, the CD146 Ab-armed nanofilamentous stent could show great performance in the reduction of thrombosis and restenosis through re-endothelialization due to highly efficient specific cell capture. STATEMENT OF SIGNIFICANCE: Stents have been developed from simple metal stents to functionalized stents for past decades. However, they have still risks to relapse the occlusion in stented arteries. In this paper, we describe the fabrication and optimization of cell capturing stents to maximize the effective re-endothelialization through the serial coating of silicone nanofilaments and anti-CD146 antibody. The nanofilaments increase the amount of coated antibodies and provide the anchoring points of circulating angiogenic cells for strong focal adhesion. We demonstrate high immobilizing ability of circulating angiogenic cells (endotheliali progenitor cells and mesenchymal stem cells) in vitro under similar shear stress to coronary arteries (15 dyne/cm2). Also, we show accelerating re-endothelialization and the efficient prevention of restenosis in porcine coronary arteries in vivo.
Collapse
Affiliation(s)
- Kwang-Sook Park
- Center for Biomaterials, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea
| | - Sung Nam Kang
- Center for Biomaterials, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea; Department of Biomedical Engineering, Korea University of Science and Technology, Daejeon 34113, Republic of Korea
| | - Dae Hwan Kim
- Center for Biomaterials, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea
| | - Han-Byual Kim
- The Heart Center of Chonnam National University Hospital, Gwangju 61469, Republic of Korea
| | - Kyung Seob Im
- Futuristic Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea
| | - Wooram Park
- Department of Biomedical Science, College of Life Sciences, CHA University, Gyeonggi 13488, Republic of Korea
| | - Young Joon Hong
- The Heart Center of Chonnam National University Hospital, Gwangju 61469, Republic of Korea
| | - Dong Keun Han
- Department of Biomedical Science, College of Life Sciences, CHA University, Gyeonggi 13488, Republic of Korea.
| | - Yoon Ki Joung
- Center for Biomaterials, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea; Department of Biomedical Engineering, Korea University of Science and Technology, Daejeon 34113, Republic of Korea.
| |
Collapse
|
45
|
Assessment of a pro-healing stent in an animal model of early neoatherosclerosis. Sci Rep 2020; 10:8227. [PMID: 32427835 PMCID: PMC7237429 DOI: 10.1038/s41598-020-64940-2] [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: 12/28/2019] [Accepted: 04/23/2020] [Indexed: 12/01/2022] Open
Abstract
Background: Neoatherosclerosis represents an accelerated manifestation of atherosclerosis in nascent neointima after stenting, associated with adverse events. We investigated whether improved reendothelialization using RGD-coated stents results in diminished vascular permeability and reduced foam cell formation compared to standard DES in atherosclerotic rabbits. Methods and Results: Neointimal foam cell formation was induced in rabbits (n = 7). Enhanced endothelial integrity in RGD-coated stents resulted in decreased vascular permeability relative to DES, which was further confirmed by SEM and TEM. Cell culture experiments examined the effect of everolimus on endothelial integrity. Increasing concentrations of everolimus resulted in a dose-dependent decrease of endothelial cell junctions and foam cell transformation of monocytes, confirming the relevance of endothelial integrity in preventing permeability of LDL. Conclusion: Incomplete endothelial integrity was confirmed as a key factor of neointimal foam cell formation following stent implantation. Pro-healing stent coatings may facilitate reendothelialization and reduce the risk of neoatherosclerosis.
Collapse
|
46
|
Royer C, Guay‐Bégin A, Chanseau C, Chevallier P, Bordenave L, Laroche G, Durrieu M. Bioactive micropatterning of biomaterials for induction of endothelial progenitor cell differentiation: Acceleration of in situ endothelialization. J Biomed Mater Res A 2020; 108:1479-1492. [DOI: 10.1002/jbm.a.36918] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 02/24/2020] [Accepted: 03/09/2020] [Indexed: 01/07/2023]
Affiliation(s)
- Caroline Royer
- Univ. BordeauxChimie et Biologie des Membranes et Nano‐Objets (UMR5248 CBMN) Pessac France
- CNRSCBMN UMR5248 Pessac France
- Bordeaux INPCBMN UMR5248 Pessac France
- Laboratoire d'Ingénierie de SurfaceCentre de recherche du CHU de Québec—Université Laval, Hôpital Saint‐François d'Assise Québec Quebec Canada
- Département de génie des minesde la métallurgie et des matériaux, Centre de Recherche sur les Matériaux Avancés Québec Quebec Canada
| | - Andrée‐Anne Guay‐Bégin
- Laboratoire d'Ingénierie de SurfaceCentre de recherche du CHU de Québec—Université Laval, Hôpital Saint‐François d'Assise Québec Quebec Canada
| | | | - Pascale Chevallier
- Laboratoire d'Ingénierie de SurfaceCentre de recherche du CHU de Québec—Université Laval, Hôpital Saint‐François d'Assise Québec Quebec Canada
- Département de génie des minesde la métallurgie et des matériaux, Centre de Recherche sur les Matériaux Avancés Québec Quebec Canada
| | | | - Gaétan Laroche
- Laboratoire d'Ingénierie de SurfaceCentre de recherche du CHU de Québec—Université Laval, Hôpital Saint‐François d'Assise Québec Quebec Canada
- Département de génie des minesde la métallurgie et des matériaux, Centre de Recherche sur les Matériaux Avancés Québec Quebec Canada
| | - Marie‐Christine Durrieu
- Univ. BordeauxChimie et Biologie des Membranes et Nano‐Objets (UMR5248 CBMN) Pessac France
- CNRSCBMN UMR5248 Pessac France
- Bordeaux INPCBMN UMR5248 Pessac France
| |
Collapse
|
47
|
Guo J, Mao L, Yu X, Song C, Zhao X. Design and Biomechanical Analysis of a Novel Retrievable Peripheral Vascular Stent. J Med Device 2020. [DOI: 10.1115/1.4046796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Abstract
Structurally retrievable drug-eluting stents may have valuable clinical applications because they do not leave any foreign materials inside the patient's body. This article presents a novel design of retrievable peripheral vascular stent and the results from biomechanical analysis of its performance. Using the finite element analysis method, principal parameters of the stent were studied. Moreover, to ensure the practicability of the retrieval process, simulation, and in vitro experiments were performed. The retrieval force reached the maximum value when the whole retrievable part had been retrieved. Furthermore, the force was gradually increased during the retrieval process and remained constant after the main part had been retrieved. When the stent was being compressed, the maximum strain of the stent occurred at the connection between the stent's retrieval part and the main body part, at a value of 4%. The index of nonuniformity of the stent was too small to be counted both at the end of the compression and self-expansion processes. With the increase of moment, the bending stiffness (EI) of the stent decreased gradually. After bending moment was applied, the large strain region was mainly located in the stent's main body part rather than the retrieval part. The results of preliminary stent retrieval experiments demonstrated that the stent could be retrieved successfully. This novel retrievable stent displays promising biomechanical performance. The preliminary experiments demonstrated that the stent could be retrieved smoothly from the blood vessels.
Collapse
Affiliation(s)
- Jingzhen Guo
- Shanghai Institute for Minimally Invasive Therapy, School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, 516 Jungong Road, Shanghai 200093, China
| | - Lin Mao
- Shanghai Institute for Minimally Invasive Therapy, School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, 516 Jungong Road, Shanghai 200093, China
| | - Xitong Yu
- Shanghai Institute for Minimally Invasive Therapy, School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, 516 Jungong Road, Shanghai 200093, China
| | - Chengli Song
- Shanghai Institute for Minimally Invasive Therapy, School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, 516 Jungong Road, Shanghai 200093, China
| | - Xue Zhao
- Department of Cardiology, Shanghai Eastern Hepatobiliary Surgery Hospital, 225 Changhai Road, Shanghai 200438, China
| |
Collapse
|
48
|
Lv J, Liu W, Shi G, Zhu F, He X, Zhu Z, Chen H. Human cardiac extracellular matrix-chitosan-gelatin composite scaffold and its endothelialization. Exp Ther Med 2020; 19:1225-1234. [PMID: 32010293 PMCID: PMC6966153 DOI: 10.3892/etm.2019.8349] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 11/08/2019] [Indexed: 12/12/2022] Open
Abstract
The present study developed a cardiac extracellular matrix-chitosan-gelatin (cECM-CG) composite scaffold that can be used as a tissue-engineered heart patch and investigated its endothelialization potential by incorporating CD34+ endothelial progenitor cells (EPCs). The cECM-CG composite scaffold was prepared by blending cardiac extracellular matrix (cECM) with biodegradable chitosan-gelatin (CG). The mixture was lyophilized using vacuum freeze-drying. CD34+ EPCs were isolated and seeded on the scaffolds, and then the endothelialization effect was subsequently investigated. Effects of the scaffolds on CD34+ EPCs survival and proliferation were evaluated by immunofluorescence staining and MTT assay. Cell differentiation into endothelial cells and the influence of the scaffolds on cell differentiation were investigated by reverse transcription-quantitative PCR (RT-qPCR), immunofluorescence staining and tube formation assay. The present results indicated that most cells were removed after decellularization, but the main extracellular matrix components were retained. Scanning electron microscopy imaging illustrated three-dimensional and porous scaffolds. The present results suggested the cECM-CG composite scaffold had a higher water absorption ability compared with the CG scaffold. Additionally, compared with the CG scaffold, the cECM-CG composite scaffold significantly increased cell survival and proliferation, which suggested its non-toxicity and biocompatibility. Furthermore, RT-qPCR, immunofluorescence and tube formation assay results indicated that CD34+ EPCs differentiated into endothelial cells, and the cECM-CG composite scaffold promoted this differentiation process. In conclusion, the present results indicated that the human cECM-CG composite scaffold generated in the present study was a highly porous, biodegradable three-dimensional scaffold which supported endothelialization of seeded CD34+ EPCs. The present results suggested that this cECM-CG composite scaffold may be a promising heart patch for use in heart tissue engineering for congenital heart disease.
Collapse
Affiliation(s)
- Jingjing Lv
- Heart Center, Shanghai Children's Medical Center, Shanghai Jiaotong University, School of Medicine, Shanghai 200127, P.R. China
| | - Wei Liu
- Department of Pediatric Cardiothoracic Surgery, Xinhua Hospital Affiliated to Shanghai Jiaotong University, School of Medicine, Shanghai 200092, P.R. China
| | - Guocheng Shi
- Heart Center, Shanghai Children's Medical Center, Shanghai Jiaotong University, School of Medicine, Shanghai 200127, P.R. China
| | - Fang Zhu
- Heart Center, Shanghai Children's Medical Center, Shanghai Jiaotong University, School of Medicine, Shanghai 200127, P.R. China
| | - Xiaomin He
- Heart Center, Shanghai Children's Medical Center, Shanghai Jiaotong University, School of Medicine, Shanghai 200127, P.R. China
| | - Zhongqun Zhu
- Heart Center, Shanghai Children's Medical Center, Shanghai Jiaotong University, School of Medicine, Shanghai 200127, P.R. China
| | - Huiwen Chen
- Heart Center, Shanghai Children's Medical Center, Shanghai Jiaotong University, School of Medicine, Shanghai 200127, P.R. China
| |
Collapse
|
49
|
Sefton MV, Gorbet MB. Nonthrombogenic Treatments and Strategies. Biomater Sci 2020. [DOI: 10.1016/b978-0-12-816137-1.00035-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
50
|
Badv M, Weitz JI, Didar TF. Lubricant-Infused PET Grafts with Built-In Biofunctional Nanoprobes Attenuate Thrombin Generation and Promote Targeted Binding of Cells. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2019; 15:e1905562. [PMID: 31773877 DOI: 10.1002/smll.201905562] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Revised: 11/02/2019] [Indexed: 05/21/2023]
Abstract
New surface coatings that enhance hemocompatibility and biofunctionality of synthetic vascular grafts such as expanded poly(tetrafluoroethylene) (ePTFE) and poly(ethylene terephthalate) (PET) are urgently needed. Lubricant-infused surfaces prevent nontargeted adhesion and enhance the biocompatibility of blood-contacting surfaces. However, limited success has been made in incorporating biofunctionality onto these surfaces and generating biofunctional lubricant-infused coatings that both prevent nonspecific adhesion and enhance targeted binding of biomolecules remains a challenge. Here, a new generation of fluorosilanized lubricant-infused PET surfaces with built-in biofunctional nanoprobes is reported. These surfaces are synthesized by starting with a self-assembled monolayer of fluorosilane that is partially etched using plasma modification technique, thereby creating a hydroxyl-terminated fluorosilanized PET surface. Simultaneously, silanized nanoprobes are produced by amino-silanizing anti-CD34 antibody in solution and directly coupling the anti-CD34-aminosilane nanoprobes onto the hydroxyl terminated, fluorosilanized PET surface. The PET surfaces are then lubricated, creating fluorosilanized biofunctional lubricant-infused PET substrates. Compared with unmodified PET surfaces, the designed biofunctional lubricant-infused PET surfaces significantly attenuate thrombin generation and blood clot formation and promote targeted binding of endothelial cells from human whole blood.
Collapse
Affiliation(s)
- Maryam Badv
- School of Biomedical Engineering, McMaster University, 1280 Main Street West, L8S 4L8, Hamilton, Ontario, Canada
| | - Jeffrey I Weitz
- School of Biomedical Engineering, McMaster University, 1280 Main Street West, L8S 4L8, Hamilton, Ontario, Canada
- Thrombosis & Atherosclerosis Research Institute, 237 Barton Street East, L8L 2X2, Hamilton, Ontario, Canada
- Department of Medicine, McMaster University, 1280 Main Street West, L8S 4L8, Hamilton, Ontario, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, 1280 Main Street West, L8S 4L8, Hamilton, Ontario, Canada
| | - Tohid F Didar
- School of Biomedical Engineering, McMaster University, 1280 Main Street West, L8S 4L8, Hamilton, Ontario, Canada
- Department of Mechanical Engineering, McMaster University, 1280 Main Street West, L8S 4L8, Hamilton, Ontario, Canada
- Michael G. DeGroote Institute for Infectious Disease Research (IIDR), McMaster University, 1280 Main Street West, L8S 4L8, Hamilton, Ontario, Canada
| |
Collapse
|