1
|
Zhang Q, Li H, Zhuang T, Xu L, Wu W, Pi J, Zhu P, Geng L, Duan Y, Xu J, Yue J, Liu X, He C, Chen X, Ruan C, Zhuang S, Liu Z, Wang Y, Zhang L, Liu J, Zhang Y. CCN5 suppresses injury-induced vascular restenosis by inhibiting smooth muscle cell proliferation and facilitating endothelial repair via thymosin β4 and Cd9 pathway. Eur Heart J 2025; 46:1645-1658. [PMID: 39873228 DOI: 10.1093/eurheartj/ehae911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 06/24/2024] [Accepted: 12/10/2024] [Indexed: 01/30/2025] Open
Abstract
BACKGROUND AND AIMS Members of the CCN matricellular protein family are crucial in various biological processes. This study aimed to characterize vascular cell-specific effects of CCN5 on neointimal formation and its role in preventing in-stent restenosis (ISR) after percutaneous coronary intervention (PCI). METHODS Stent-implanted porcine coronary artery RNA-seq and mouse injury-induced femoral artery neointima single-cell RNA sequencing were performed. Plasma CCN5 levels were measured by enzyme-linked immunosorbent assay. Endothelial cell (EC)- and vascular smooth muscle cell (VSMC)-specific CCN5 loss-of-function and gain-of-function mice were generated. Mass spectrometry and co-immunoprecipitation were conducted to identify CCN5 interacting proteins. Additionally, CCN5 recombinant protein (CCN5rp)-coated stents were deployed to evaluate its anti-ISR effects in a porcine model. RESULTS Plasma CCN5 levels were significantly reduced and correlated closely with the degree of restenosis in ISR patients. CCN5 expression was significantly decreased in VSMCs of stent-implanted porcine coronary segments and injured mouse femoral arteries, especially in synthetic VSMCs. In contrast, elevated CCN5 expression was observed in regenerating ECs of injured vessels. Endothelial cell- and VSMC-specific CCN5 deletion mice exhibited exacerbation of injury-induced neointimal hyperplasia, while CCN5 gain-of-function alleviated neointimal formation. Mechanistic studies identified thymosin β4 (Tβ4) as a CCN5 interacting protein in ECs and EC-CCN5 promoted injury repair through Tβ4 cleavage product Ac-SDKP. Also, CCN5rp promoted EC repair to suppress neointimal hyperplasia via interaction with Cd9 extracellular domain. Moreover, implantation with CCN5rp-coated stent significantly increased stent strut coverage with ECs, which suppressed neointimal formation and ultimately alleviated ISR. CONCLUSIONS CCN5 exerts a dual protective effect on ISR by inhibiting VSMC proliferation and facilitating EC repair. CCN5rp-coated stent might be promising in the prevention of ISR after PCI.
Collapse
Affiliation(s)
- Qi Zhang
- State Key Laboratory of Cardiovascular Diseases and Medical Innovation Center, Shanghai Heart Failure Research Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Pudong New District, Shanghai 200120, China
| | - Hongda Li
- State Key Laboratory of Cardiovascular Diseases and Medical Innovation Center, Shanghai Heart Failure Research Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Pudong New District, Shanghai 200120, China
- Department of Cardiovascular Surgery, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Pudong New District, Shanghai 200120, China
| | - Tao Zhuang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Jinshan Hospital, Fudan University, Shanghai 200032, China
| | - Lehua Xu
- Science and Technology Innovation Center, Shanghai East Hospital, The Institute for Biomedical Engineering and Nano Science, Tongji University School of Medicine, Shanghai 200092, China
| | - Wenrun Wu
- State Key Laboratory of Cardiovascular Diseases and Medical Innovation Center, Shanghai Heart Failure Research Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Jingjiang Pi
- State Key Laboratory of Cardiovascular Diseases and Medical Innovation Center, Shanghai Heart Failure Research Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Pudong New District, Shanghai 200120, China
- Research Center for Translational Medicine, Shenzhen Ruipuxun Academy for Stem Cell and Regenerative Medicine, Shenzhen, China
| | - Pengxiong Zhu
- Department of Cardiovascular Surgery, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Pudong New District, Shanghai 200120, China
| | - Liang Geng
- State Key Laboratory of Cardiovascular Diseases and Medical Innovation Center, Shanghai Heart Failure Research Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Pudong New District, Shanghai 200120, China
| | - Yunhao Duan
- State Key Laboratory of Cardiovascular Diseases and Medical Innovation Center, Shanghai Heart Failure Research Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Pudong New District, Shanghai 200120, China
| | - Jianfei Xu
- State Key Laboratory of Cardiovascular Diseases and Medical Innovation Center, Shanghai Heart Failure Research Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Pudong New District, Shanghai 200120, China
| | - Jinnan Yue
- State Key Laboratory of Cardiovascular Diseases and Medical Innovation Center, Shanghai Heart Failure Research Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Xiuxiang Liu
- State Key Laboratory of Cardiovascular Diseases and Medical Innovation Center, Shanghai Heart Failure Research Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Pudong New District, Shanghai 200120, China
| | - Chenlong He
- Science and Technology Innovation Center, Shanghai East Hospital, The Institute for Biomedical Engineering and Nano Science, Tongji University School of Medicine, Shanghai 200092, China
| | - Xiaoli Chen
- State Key Laboratory of Cardiovascular Diseases and Medical Innovation Center, Shanghai Heart Failure Research Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
- Department of Cardiovascular Surgery, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Pudong New District, Shanghai 200120, China
| | - Chengchao Ruan
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Jinshan Hospital, Fudan University, Shanghai 200032, China
| | - Shougang Zhuang
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
- Department of Medicine, Rhode Island Hospital and Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Zhongmin Liu
- State Key Laboratory of Cardiovascular Diseases and Medical Innovation Center, Shanghai Heart Failure Research Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
- Department of Cardiovascular Surgery, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Pudong New District, Shanghai 200120, China
| | - Yilong Wang
- Science and Technology Innovation Center, Shanghai East Hospital, The Institute for Biomedical Engineering and Nano Science, Tongji University School of Medicine, Shanghai 200092, China
| | - Lin Zhang
- State Key Laboratory of Cardiovascular Diseases and Medical Innovation Center, Shanghai Heart Failure Research Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
- Department of Cardiovascular Surgery, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Pudong New District, Shanghai 200120, China
| | - Jie Liu
- State Key Laboratory of Cardiovascular Diseases and Medical Innovation Center, Shanghai Heart Failure Research Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
- Department of Cardiovascular Surgery, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Pudong New District, Shanghai 200120, China
| | - Yuzhen Zhang
- State Key Laboratory of Cardiovascular Diseases and Medical Innovation Center, Shanghai Heart Failure Research Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
- Department of Cardiovascular Surgery, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Pudong New District, Shanghai 200120, China
| |
Collapse
|
2
|
Brown SD, Rodor J, Baker AH. Targeted approach for next-generation coronary stents. Eur Heart J 2025; 46:1659-1661. [PMID: 40037368 PMCID: PMC12046217 DOI: 10.1093/eurheartj/ehaf007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/06/2025] Open
Affiliation(s)
- Simon D Brown
- BHF Centre for Cardiovascular Science, University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Julie Rodor
- BHF Centre for Cardiovascular Science, University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Andrew H Baker
- BHF Centre for Cardiovascular Science, University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
- Department of Pathology and Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
3
|
Meredith E, Schwartz MA. Integrins as Drug Targets in Vascular and Related Diseases. INTERNATIONAL JOURNAL OF DRUG DISCOVERY AND PHARMACOLOGY 2024; 3:100010. [PMID: 39703402 PMCID: PMC11658063 DOI: 10.53941/ijddp.2024.100010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2024]
Abstract
Integrins are transmembrane receptors that, as critical participants in a vast range of pathological processes, are potential therapeutic targets. However, in only a few cases has the promise been realized by drug approval. In this review, we briefly review basic integrin biology and participation in disease, challenges in the development of safe, effective integrin-targeted therapies, and recent advances that may lead to progress.
Collapse
Affiliation(s)
- Emily Meredith
- Yale Cardiovascular Research Center, Department of Internal Medicine (Cardiology), Yale University School of Medicine, New Haven, Connecticut 06511, USA
| | - Martin A. Schwartz
- Yale Cardiovascular Research Center, Department of Internal Medicine (Cardiology), Yale University School of Medicine, New Haven, Connecticut 06511, USA
- Department of Cell Biology, Yale School of Medicine
- Department of Biomedical Engineering, Yale University
| |
Collapse
|
4
|
Yin J, Zhao C, Huang J, Chen C, Lei T, He J, Qiu D. Diabetic conditions promote drug coating degradation but prevent endothelial coverage after stenting. Acta Biomater 2024; 177:189-202. [PMID: 38307481 DOI: 10.1016/j.actbio.2024.01.034] [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: 09/15/2023] [Revised: 01/17/2024] [Accepted: 01/23/2024] [Indexed: 02/04/2024]
Abstract
The endothelialization of drug-eluting stents is delayed after implantation in patients with diabetes. Although numerous factors were implicated in hyperglycemia-induced endothelial dysfunction, the effects of stent drug coating degradation on endothelial dysfunction remains unclear. We hypothesized that diabetic conditions promote drugcoating degradation and enhance antiproliferative agent release, but that the rapid release of these antiproliferative agents inhibits endothelial cell proliferation leading to poor reendothelialization post-stenting. To verify this hypothesis, a dynamic hyperglycemic circulation system was introduced to measure the profile of drugcoating degradation in vitro. Flow cytometry and RNA sequencing were performed to evaluate endothelial cell proliferation. Moreover, a Type 1 diabetic rabbit model was generated and a rescue experiment conducted to evaluate the effects of rapid drugcoating elution on endothelial coverage in vivo. The main findings were as follows: 1) diabetic conditions promoted drugcoating degradation and increased antiproliferative agent release; 2) this increase in antiproliferative agent release inhibited endothelial cell proliferation and delayed endothelial coverage; and 3) strict glycemic control attenuated drugcoating degradation and promoted endothelial coverage post-stenting. This is the first study to illustrate rapid drugcoating degradation and its potential effects on endothelial recovery under diabetic conditions, highlighting the importance of strict glycemic management in patients with diabetes after drug-eluting stent implantation. STATEMENT OF SIGNIFICANCE: Diabetic conditions promote drug coating degradation and increase the release of antiproliferative agents. Rapid drug coating degradation under diabetic conditions inhibits endothelial cell proliferation and delays endothelialization. Strict glycemic control attenuates drug coating degradation and promotes endothelialization.
Collapse
Affiliation(s)
- Jun Yin
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Chunguang Zhao
- Department of Critical Care Medicine, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, No. 87 Xiangya Road, Changsha 410008, Hunan Province, China.
| | - Jiabing Huang
- Department of Cardiology, The Second Affiliated Hospital of Nanchang University, Jiangxi, Nanchang, PR China
| | - Changqing Chen
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
| | - Ting Lei
- State Key Laboratory of Powder Metallurgy, Central South University, Changsha 410083, PR China.
| | - Jiawei He
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
| | - Dongxu Qiu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
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: 11] [Impact Index Per Article: 5.5] [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
|
Hao D, Lin J, Liu R, Pivetti C, Yamashiro K, Schutzman LM, Sageshima J, Kwong M, Bahatyrevich N, Farmer DL, Humphries MD, Lam KS, Panitch A, Wang A. A bio-instructive parylene-based conformal coating suppresses thrombosis and intimal hyperplasia of implantable vascular devices. Bioact Mater 2023; 28:467-479. [PMID: 37408799 PMCID: PMC10318457 DOI: 10.1016/j.bioactmat.2023.06.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 06/17/2023] [Accepted: 06/19/2023] [Indexed: 07/07/2023] Open
Abstract
Implantable vascular devices are widely used in clinical treatments for various vascular diseases. However, current approved clinical implantable vascular devices generally have high failure rates primarily due to their surface lacking inherent functional endothelium. Here, inspired by the pathological mechanisms of vascular device failure and physiological functions of native endothelium, we developed a new generation of bioactive parylene (poly(p-xylylene))-based conformal coating to address these challenges of the vascular devices. This coating used a polyethylene glycol (PEG) linker to introduce an endothelial progenitor cell (EPC) specific binding ligand LXW7 (cGRGDdvc) onto the vascular devices for preventing platelet adhesion and selectively capturing endogenous EPCs. Also, we confirmed the long-term stability and function of this coating in human serum. Using two vascular disease-related large animal models, a porcine carotid artery interposition model and a porcine carotid artery-jugular vein arteriovenous graft model, we demonstrated that this coating enabled rapid generation of self-renewable "living" endothelium on the blood contacting surface of the expanded polytetrafluoroethylene (ePTFE) grafts after implantation. We expect this easy-to-apply conformal coating will present a promising avenue to engineer surface properties of "off-the-shelf" implantable vascular devices for long-lasting performance in the clinical settings.
Collapse
Affiliation(s)
- Dake Hao
- Department of Surgery, School of Medicine, University of California Davis, Sacramento, CA, 95817, United States
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA, 95817, United States
| | - Jonathan Lin
- Department of Surgery, School of Medicine, University of California Davis, Sacramento, CA, 95817, United States
| | - Ruiwu Liu
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California Davis, Sacramento, CA, 95817, United States
| | - Christopher Pivetti
- Department of Surgery, School of Medicine, University of California Davis, Sacramento, CA, 95817, United States
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA, 95817, United States
| | - Kaeli Yamashiro
- Department of Surgery, School of Medicine, University of California Davis, Sacramento, CA, 95817, United States
| | - Linda M. Schutzman
- Department of Surgery, School of Medicine, University of California Davis, Sacramento, CA, 95817, United States
| | - Junichiro Sageshima
- Department of Surgery, School of Medicine, University of California Davis, Sacramento, CA, 95817, United States
| | - Mimmie Kwong
- Department of Surgery, School of Medicine, University of California Davis, Sacramento, CA, 95817, United States
| | - Nataliya Bahatyrevich
- Department of Surgery, School of Medicine, University of California Davis, Sacramento, CA, 95817, United States
| | - Diana L. Farmer
- Department of Surgery, School of Medicine, University of California Davis, Sacramento, CA, 95817, United States
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA, 95817, United States
| | - Misty D. Humphries
- Department of Surgery, School of Medicine, University of California Davis, Sacramento, CA, 95817, United States
| | - Kit S. Lam
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California Davis, Sacramento, CA, 95817, United States
| | - Alyssa Panitch
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30332, United States
- Department of Biomedical Engineering, University of California Davis, Davis, CA, 95616, United States
| | - Aijun Wang
- Department of Surgery, School of Medicine, University of California Davis, Sacramento, CA, 95817, United States
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA, 95817, United States
- Department of Biomedical Engineering, University of California Davis, Davis, CA, 95616, United States
| |
Collapse
|
7
|
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
|
8
|
Yin L, Li X, Wang R, Zeng Y, Zeng Z, Xie T. Recent Research Progress of RGD Peptide–Modified Nanodrug Delivery Systems in Tumor Therapy. Int J Pept Res Ther 2023; 29:53. [DOI: 10.1007/s10989-023-10523-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/20/2023] [Indexed: 01/06/2025]
Abstract
AbstractThere have been great advancements in targeted nanodrug delivery systems for tumor therapy. Liposomes, polymeric nanoparticles, and inorganic nanoparticles are commonly employed as nanocarriers for drug delivery, and it has been found that arginine glycine aspartic acid (RGD) peptides and their derivatives can be used as ligands of integrin receptors to enhance the direct targeting ability. In this paper, we review the recent applications of RGD-modified liposomes, polymeric nanoparticles, and inorganic nanocarriers in cancer diagnosis and treatment, discuss the current challenges and prospects, and examine the progress made by the latest research on RGD peptide–modified nano delivery systems in cancer therapy. In recent years, RGD peptide–modified nanodrug delivery systems have been proven to have great potential in tumor therapy. Finally, we provide an overview of the current limitations and future directions of RGD peptide–modified nano-drug delivery systems for cancer therapy. This review aims to elucidate the contribution of RGD peptide–modified nanodrug delivery systems in the field of tumor therapy.
Collapse
|
9
|
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: 35] [Impact Index Per Article: 11.7] [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
|
10
|
Hernandez JL, Woodrow KA. Medical Applications of Porous Biomaterials: Features of Porosity and Tissue-Specific Implications for Biocompatibility. Adv Healthc Mater 2022; 11:e2102087. [PMID: 35137550 PMCID: PMC9081257 DOI: 10.1002/adhm.202102087] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 12/17/2021] [Indexed: 12/14/2022]
Abstract
Porosity is an important material feature commonly employed in implants and tissue scaffolds. The presence of material voids permits the infiltration of cells, mechanical compliance, and outward diffusion of pharmaceutical agents. Various studies have confirmed that porosity indeed promotes favorable tissue responses, including minimal fibrous encapsulation during the foreign body reaction (FBR). However, increased biofilm formation and calcification is also described to arise due to biomaterial porosity. Additionally, the relevance of host responses like the FBR, infection, calcification, and thrombosis are dependent on tissue location and specific tissue microenvironment. In this review, the features of porous materials and the implications of porosity in the context of medical devices is discussed. Common methods to create porous materials are also discussed, as well as the parameters that are used to tune pore features. Responses toward porous biomaterials are also reviewed, including the various stages of the FBR, hemocompatibility, biofilm formation, and calcification. Finally, these host responses are considered in tissue specific locations including the subcutis, bone, cardiovascular system, brain, eye, and female reproductive tract. The effects of porosity across the various tissues of the body is highlighted and the need to consider the tissue context when engineering biomaterials is emphasized.
Collapse
Affiliation(s)
- Jamie L Hernandez
- Department of Bioengineering, University of Washington, 3720 15th Ave NE, Seattle, WA, 98195, USA
| | - Kim A Woodrow
- Department of Bioengineering, University of Washington, 3720 15th Ave NE, Seattle, WA, 98195, USA
| |
Collapse
|
11
|
Selvakumar PP, Rafuse MS, Johnson R, Tan W. Applying Principles of Regenerative Medicine to Vascular Stent Development. Front Bioeng Biotechnol 2022; 10:826807. [PMID: 35321023 PMCID: PMC8936177 DOI: 10.3389/fbioe.2022.826807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 01/17/2022] [Indexed: 11/16/2022] Open
Abstract
Stents are a widely-used device to treat a variety of cardiovascular diseases. The purpose of this review is to explore the application of regenerative medicine principles into current and future stent designs. This review will cover regeneration-relevant approaches emerging in the current research landscape of stent technology. Regenerative stent technologies include surface engineering of stents with cell secretomes, cell-capture coatings, mimics of endothelial products, surface topography, endothelial growth factors or cell-adhesive peptides, as well as design of bioresorable materials for temporary stent support. These technologies are comparatively analyzed in terms of their regenerative effects, therapeutic effects and challenges faced; their benefits and risks are weighed up for suggestions about future stent developments. This review highlights two unique regenerative features of stent technologies: selective regeneration, which is to selectively grow endothelial cells on a stent but inhibit the proliferation and migration of smooth muscle cells, and stent-assisted regeneration of ischemic tissue injury.
Collapse
Affiliation(s)
| | | | | | - Wei Tan
- University of Colorado Boulder, Boulder, CO, United States
| |
Collapse
|
12
|
Role of Integrins in Modulating Smooth Muscle Cell Plasticity and Vascular Remodeling: From Expression to Therapeutic Implications. Cells 2022; 11:cells11040646. [PMID: 35203297 PMCID: PMC8870356 DOI: 10.3390/cells11040646] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 02/03/2022] [Accepted: 02/11/2022] [Indexed: 02/06/2023] Open
Abstract
Smooth muscle cells (SMCs), present in the media layer of blood vessels, are crucial in maintaining vascular homeostasis. Upon vascular injury, SMCs show a high degree of plasticity, undergo a change from a “contractile” to a “synthetic” phenotype, and play an essential role in the pathophysiology of diseases including atherosclerosis and restenosis. Integrins are cell surface receptors, which are involved in cell-to-cell binding and cell-to-extracellular-matrix interactions. By binding to extracellular matrix components, integrins trigger intracellular signaling and regulate several of the SMC function, including proliferation, migration, and phenotypic switching. Although pharmacological approaches, including antibodies and synthetic peptides, have been effectively utilized to target integrins to limit atherosclerosis and restenosis, none has been commercialized yet. A clear understanding of how integrins modulate SMC biology is essential to facilitate the development of integrin-based interventions to combat atherosclerosis and restenosis. Herein, we highlight the importance of integrins in modulating functional properties of SMCs and their implications for vascular pathology.
Collapse
|
13
|
Wolfe JT, Shradhanjali A, Tefft BJ. Strategies for improving endothelial cell adhesion to blood-contacting medical devices. TISSUE ENGINEERING PART B-REVIEWS 2021; 28:1067-1092. [PMID: 34693761 DOI: 10.1089/ten.teb.2021.0148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The endothelium is a critical mediator of homeostasis on blood-contacting surfaces in the body, serving as a selective barrier to regulate processes such as clotting, immune cell adhesion, and cellular response to fluid shear stress. Implantable cardiovascular devices including stents, vascular grafts, heart valves, and left ventricular assist devices are in direct contact with circulating blood and carry a high risk for platelet activation and thrombosis without a stable endothelial cell (EC) monolayer. Development of a healthy endothelium on the blood-contacting surface of these devices would help ameliorate risks associated with thrombus formation and eliminate the need for long-term anti-platelet or anti-coagulation therapy. Although ECs have been seeded onto or recruited to these blood-contacting surfaces, most ECs are lost upon exposure to shear stress due to circulating blood. Many investigators have attempted to generate a stable EC monolayer by improving EC adhesion using surface modifications, material coatings, nanofiber topology, and modifications to the cells. Despite some success with enhanced EC retention in vitro and in animal models, no studies to date have proven efficacious for routinely creating a stable endothelium in the clinical setting. This review summarizes past and present techniques directed at improving the adhesion of ECs to blood-contacting devices.
Collapse
Affiliation(s)
- Jayne Taylor Wolfe
- Medical College of Wisconsin, 5506, Biomedical Engineering, 8701 Watertown Plank Rd, Milwaukee, Wisconsin, United States, 53226-0509;
| | - Akankshya Shradhanjali
- Medical College of Wisconsin, 5506, Biomedical Engineering, Milwaukee, Wisconsin, United States;
| | - Brandon J Tefft
- Medical College of Wisconsin, 5506, Biomedical Engineering, Milwaukee, Wisconsin, United States;
| |
Collapse
|
14
|
Zhang Y, Xi K, Fu X, Sun H, Wang H, Yu D, Li Z, Ma Y, Liu X, Huang B, Wang J, Li G, Cui J, Li X, Ni S. Versatile metal-phenolic network nanoparticles for multitargeted combination therapy and magnetic resonance tracing in glioblastoma. Biomaterials 2021; 278:121163. [PMID: 34601197 DOI: 10.1016/j.biomaterials.2021.121163] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 09/24/2021] [Accepted: 09/28/2021] [Indexed: 10/20/2022]
Abstract
Glioblastoma multiforme (GBM) is a common malignancy of the central nervous system, but conventional treatments yield unsatisfactory results. Although innovative therapeutic approaches have been developed, they prolong survival by only approximately 5 months. The heterogeneity of GBM renders growth inhibition with a single drug difficult, and exploring combination approaches with multiple targets for the comprehensive treatment of GBM is expected to overcome this limitation. In this study, we designed a biocompatible cRGD/Pt + DOX@GFNPs (RPDGs) nanoformulation to disrupt redox homeostasis in GBM cells and promote the simultaneous occurrence of efficient apoptosis and ferroptosis. Taking advantage of the highly stable Fenton reaction catalytic activity of gallic acid (GA)/Fe2+ nanoparticles in physiological environments, the ability of Pt (IV) to deplete glutathione (GSH) and increase reactive oxygen species (ROS) levels, and the efficient photothermal conversion efficiency of GA/Fe2+ nanoparticles, our synthesized multifunctional and multitargeted RPDGs significantly increased intracellular ROS levels and thus induced ferroptosis. Furthermore, the RPDGs displayed superior photothermal responsiveness and magnetic resonance imaging (MRI) capabilities. These results indicate that RPDGs can not only directly inhibit the growth of tumors but also effectively improve the efficient translocation of conventional chemotherapeutic drugs across the blood-brain barrier, thereby providing a new approach for the comprehensive treatment of GBM.
Collapse
Affiliation(s)
- Yulin Zhang
- Department of Neurosurgery, Qilu Hospital and Institute of Brain and Brain-Inspired Science, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Kaiyan Xi
- Department of Neurosurgery, Qilu Hospital and Institute of Brain and Brain-Inspired Science, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Xiao Fu
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong, 250100, China
| | - Haifeng Sun
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong, 250100, China
| | - Hong Wang
- Radiology Department, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
| | - Dexin Yu
- Radiology Department, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
| | - Zhiwei Li
- Department of Neurosurgery, Qilu Hospital and Institute of Brain and Brain-Inspired Science, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Yuan Ma
- Department of Neurosurgery, Qilu Hospital and Institute of Brain and Brain-Inspired Science, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Xinjie Liu
- Department of Pediatrics, Qilu Hospital, Shandong University, Jinan, Shandong, 250012, China
| | - Bin Huang
- Department of Neurosurgery, Qilu Hospital and Institute of Brain and Brain-Inspired Science, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Jian Wang
- Department of Neurosurgery, Qilu Hospital and Institute of Brain and Brain-Inspired Science, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Gang Li
- Department of Neurosurgery, Qilu Hospital and Institute of Brain and Brain-Inspired Science, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Jiwei Cui
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong, 250100, China
| | - Xingang Li
- Department of Neurosurgery, Qilu Hospital and Institute of Brain and Brain-Inspired Science, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Shilei Ni
- Department of Neurosurgery, Qilu Hospital and Institute of Brain and Brain-Inspired Science, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China.
| |
Collapse
|
15
|
Tsukada J, Mela P, Jinzaki M, Tsukada H, Schmitz-Rode T, Vogt F. Development of In Vitro Endothelialised Stents - Review. Stem Cell Rev Rep 2021; 18:179-197. [PMID: 34403073 DOI: 10.1007/s12015-021-10238-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/06/2021] [Indexed: 01/12/2023]
Abstract
Endovascular treatment is prevalent as a primary treatment for coronary and peripheral arterial diseases. Although the introduction of drug-eluting stents (DES) dramatically reduced the risk of in-stent restenosis, stent thrombosis persists as an issue. Notwithstanding improvements in newer generation DES, they are yet to address the urgent clinical need to abolish the late stent complications that result from in-stent restenosis and are associated with late thrombus formation. These often lead to acute coronary syndromes with high mortality in coronary artery disease and acute limb ischemia with a high risk of limb amputation in peripheral arterial disease. Recently, a significant amount of research has focused on alternative solutions to improve stent biocompatibility by using tissue engineering. There are two types of tissue engineering endothelialisation methods: in vitro and in vivo. To date, commercially available in vivo endothelialised stents have failed to demonstrate antithrombotic or anti-stenosis efficacy in clinical trials. In contrast, the in vitro endothelialisation methods exhibit the advantage of monitoring cell type and growth prior to implantation, enabling better quality control. The present review discusses tissue-engineered candidate stents constructed by distinct in vitro endothelialisation approaches, with a particular focus on fabrication processes, including cell source selection, stent material composition, stent surface modifications, efficacy and safety evidence from in vitro and in vivo studies, and future directions.
Collapse
Affiliation(s)
- Jitsuro Tsukada
- Department of Diagnostic Radiology, Nihon University School of Medicine, 30-1, Oyaguchikamicho, Itabashi-ku, Tokyo, 173-8610, Japan. .,Department of Diagnostic Radiology, Keio University School of Medicine, 35, Shinanomachi, Shinjyuku-ku, Tokyo, 160-8582, Japan.
| | - P Mela
- Department of Mechanical Engineering and Munich School of BioEngineering, Technical University of Munich, Boltzmannstr. 15, Garching, Munich, 85748, Germany
| | - M Jinzaki
- Department of Diagnostic Radiology, Keio University School of Medicine, 35, Shinanomachi, Shinjyuku-ku, Tokyo, 160-8582, Japan
| | - H Tsukada
- Department of Surgery II, Tokyo Women's Medical University, 8-1, Kawada-cho, Shinjuku-ku, Tokyo, 162-8666, Japan
| | - T Schmitz-Rode
- AME - Institute of Applied Medical Engineering, Helmholtz Institute, RWTH Aachen University, Pauwelsstrasse 30, Aachen, 52074, Germany
| | - F Vogt
- Department of Cardiology, University Hospital RWTH Aachen, Pauwelsstrasse 30, Aachen, 52074, Germany
| |
Collapse
|
16
|
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: 1.8] [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
|
17
|
Diaz-Rodriguez S, Rasser C, Mesnier J, Chevallier P, Gallet R, Choqueux C, Even G, Sayah N, Chaubet F, Nicoletti A, Ghaleh B, Feldman LJ, Mantovani D, Caligiuri G. Coronary stent CD31-mimetic coating favours endothelialization and reduces local inflammation and neointimal development in vivo. Eur Heart J 2021; 42:1760-1769. [PMID: 33580685 PMCID: PMC8106951 DOI: 10.1093/eurheartj/ehab027] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 10/12/2020] [Accepted: 01/12/2021] [Indexed: 12/14/2022] Open
Abstract
AIMS The rapid endothelialization of bare metal stents (BMS) is counterbalanced by inflammation-induced neointimal growth. Drug-eluting stents (DES) prevent leukocyte activation but impair endothelialization, delaying effective device integration into arterial walls. Previously, we have shown that engaging the vascular CD31 co-receptor is crucial for endothelial and leukocyte homeostasis and arterial healing. Furthermore, we have shown that a soluble synthetic peptide (known as P8RI) acts like a CD31 agonist. The aim of this study was to evaluate the effect of CD31-mimetic metal stent coating on the in vitro adherence of endothelial cells (ECs) and blood elements and the in vivo strut coverage and neointimal growth. METHODS AND RESULTS We produced Cobalt Chromium discs and stents coated with a CD31-mimetic peptide through two procedures, plasma amination or dip-coating, both yielding comparable results. We found that CD31-mimetic discs significantly reduced the extent of primary human coronary artery EC and blood platelet/leukocyte activation in vitro. In vivo, CD31-mimetic stent properties were compared with those of DES and BMS by coronarography and microscopy at 7 and 28 days post-implantation in pig coronary arteries (n = 9 stents/group/timepoint). Seven days post-implantation, only CD31-mimetic struts were fully endothelialized with no activated platelets/leukocytes. At day 28, neointima development over CD31-mimetic stents was significantly reduced compared to BMS, appearing as a normal arterial media with the absence of thrombosis contrary to DES. CONCLUSION CD31-mimetic coating favours vascular homeostasis and arterial wall healing, preventing in-stent stenosis and thrombosis. Hence, such coatings seem to improve the metal stent biocompatibility.
Collapse
Affiliation(s)
- Sergio Diaz-Rodriguez
- Laboratory for Biomaterials and Bioengineering (CRC-I) Department of Min-Met-Mat Engineering and the CHU de Québec Research Center, Laval University, PLT-1745G, Québec, QC G1V 0A6, Canada
| | - Charlotte Rasser
- Laboratory for Vascular Translational Science, Université de Paris, Inserm U1148, 46 rue Henri HUCHARD, Paris 75018, France
| | - Jules Mesnier
- Laboratory for Vascular Translational Science, Université de Paris, Inserm U1148, 46 rue Henri HUCHARD, Paris 75018, France
| | - Pascale Chevallier
- Laboratory for Biomaterials and Bioengineering (CRC-I) Department of Min-Met-Mat Engineering and the CHU de Québec Research Center, Laval University, PLT-1745G, Québec, QC G1V 0A6, Canada
| | - Romain Gallet
- Institut Mondor de Recherche Biomédicale, école nationale vétérinaire de Maisons-Alfort (ENVA), Institut National de la Santé et de la Recherche Médicale U955, GHU (Groupe Hospitalo-Universitaire) A. Chenevier, Henri Mondor Faculty of Medicine Paris Est, 8 Rue du Général Sarrail, Créteil 94010, France
| | - Christine Choqueux
- Laboratory for Vascular Translational Science, Université de Paris, Inserm U1148, 46 rue Henri HUCHARD, Paris 75018, France
| | - Guillaume Even
- Laboratory for Vascular Translational Science, Université de Paris, Inserm U1148, 46 rue Henri HUCHARD, Paris 75018, France
| | - Neila Sayah
- Laboratory for Vascular Translational Science, Université de Paris, Inserm U1148, 46 rue Henri HUCHARD, Paris 75018, France
| | - Frédéric Chaubet
- Laboratory for Vascular Translational Science, Université de Paris, Inserm U1148, 46 rue Henri HUCHARD, Paris 75018, France
| | - Antonino Nicoletti
- Laboratory for Vascular Translational Science, Université de Paris, Inserm U1148, 46 rue Henri HUCHARD, Paris 75018, France
| | - Bijan Ghaleh
- Institut Mondor de Recherche Biomédicale, école nationale vétérinaire de Maisons-Alfort (ENVA), Institut National de la Santé et de la Recherche Médicale U955, GHU (Groupe Hospitalo-Universitaire) A. Chenevier, Henri Mondor Faculty of Medicine Paris Est, 8 Rue du Général Sarrail, Créteil 94010, France
| | - Laurent J Feldman
- Laboratory for Vascular Translational Science, Université de Paris, Inserm U1148, 46 rue Henri HUCHARD, Paris 75018, France.,Department of Cardiology, Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Paris Nord Val-de-Seine, Site Bichat, 46 rue Henri Huchard, Paris 75018, France
| | - Diego Mantovani
- Laboratory for Biomaterials and Bioengineering (CRC-I) Department of Min-Met-Mat Engineering and the CHU de Québec Research Center, Laval University, PLT-1745G, Québec, QC G1V 0A6, Canada
| | - Giuseppina Caligiuri
- Laboratory for Vascular Translational Science, Université de Paris, Inserm U1148, 46 rue Henri HUCHARD, Paris 75018, France.,Department of Cardiology, Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Paris Nord Val-de-Seine, Site Bichat, 46 rue Henri Huchard, Paris 75018, France
| |
Collapse
|
18
|
Endothelial progenitor cells as the target for cardiovascular disease prediction, personalized prevention, and treatments: progressing beyond the state-of-the-art. EPMA J 2020; 11:629-643. [PMID: 33240451 DOI: 10.1007/s13167-020-00223-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Accepted: 09/11/2020] [Indexed: 02/06/2023]
Abstract
Stimulated by the leading mortalities of cardiovascular diseases (CVDs), various types of cardiovascular biomaterials have been widely investigated in the past few decades. Although great therapeutic effects can be achieved by bare metal stents (BMS) and drug-eluting stents (DES) within months or years, the long-term complications such as late thrombosis and restenosis have limited their further applications. It is well accepted that rapid endothelialization is a promising approach to eliminate these complications. Convincing evidence has shown that endothelial progenitor cells (EPCs) could be mobilized into the damaged vascular sites systemically and achieve endothelial repair in situ, which significantly contributes to the re-endothelialization process. Therefore, how to effectively capture EPCs via specific molecules immobilized on biomaterials is an important point to achieve rapid endothelialization. Further, in the context of predictive, preventive, personalized medicine (PPPM), the abnormal number alteration of EPCs in circulating blood and certain inflammation responses can also serve as important indicators for predicting and preventing early cardiovascular disease. In this contribution, we mainly focused on the following sections: the definition and classification of EPCs, the mechanisms of EPCs in treating CVDs, the potential diagnostic role of EPCs in predicting CVDs, as well as the main strategies for cardiovascular biomaterials to capture EPCs.
Collapse
|
19
|
Tsukada J, Wolf F, Vogt F, Schaaps N, Thoröe-Boveleth S, Keijdener H, Jankowski J, Tsukada H, Jockenhövel S, Jinzaki M, Schmitz-Rode T, Mela P. Development of in vitro endothelialized drug-eluting stent using human peripheral blood-derived endothelial progenitor cells. J Tissue Eng Regen Med 2020; 14:1415-1427. [PMID: 32668066 DOI: 10.1002/term.3107] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 06/05/2020] [Accepted: 07/09/2020] [Indexed: 11/05/2022]
Abstract
We propose in vitro endothelialization of drug-eluting stents (DES) to overcome late stent thrombosis by directly introducing late-outgrowth human endothelial progenitor cells (EPCs) at the target site utilizing abluminal DES. Isolated EPCs were confirmed as late-outgrowth EPCs by flow cytometric analysis. Abluminally paclitaxel-loaded stents were seeded with different cell concentrations and durations to determine optimal seeding conditions, in both uncrimped and crimped configurations. The seeding yield was determined by evaluating the percent coverage of the stent struts' area. The EPC-seeded DES were exposed to arterial shear stress to evaluate the effect of high shear stress on EPCs. To investigate how much paclitaxel elutes during the seeding procedure, a pharmacokinetic analysis was performed. Finally, to validate the proof of concept, EPC-seeded DES were placed on a fibrin matrix with and without smooth muscle cells (SMCs) and cultured for 3 days under perfusion. The seeding procedure resulted in 47% and 26% coverage of the stent surface in uncrimped and crimped conditions, respectively. After the optimal seeding, almost 99% of drug was still available. When EPC-seeded DES were placed on a fibrin matrix and cultured for 3 days, the EPCs confluently covered the stent surface and spread to the surrounding fibrin gel. When EPC-seeded DES were placed on SMC-containing fibrin layers, cells in contact with the struts died. EPCs can be successfully seeded onto DES without losing drug-eluting capability, and EPCs exhibit sufficient proliferative ability. EPC-seeded DES may combine early re-endothelialization ability with the antirestenotic effectiveness of DES.
Collapse
Affiliation(s)
- Jitsuro Tsukada
- Department of Diagnostic Radiology, Keio University School of Medicine, Tokyo, Japan.,Department of Radiology, Nihon University School of Medicine, Tokyo, Japan
| | - Frederic Wolf
- Department of Biohybrid & Medical Textiles (Biotex), AME - Institute of Applied Medical Engineering, Helmholtz Institute, RWTH Aachen University, Aachen, Germany
| | - Felix Vogt
- Department of Cardiology, Pneumology, Angiology and Intensive Care, University Hospital RWTH Aachen, Aachen, Germany
| | - Nicole Schaaps
- Department of Cardiology, Pneumology, Angiology and Intensive Care, University Hospital RWTH Aachen, Aachen, Germany
| | - Sven Thoröe-Boveleth
- Institute for Molecular Cardiovascular Research, University Hospital Aachen, RWTH Aachen University, Aachen, Germany
| | - Hans Keijdener
- Department of Biohybrid & Medical Textiles (Biotex), AME - Institute of Applied Medical Engineering, Helmholtz Institute, RWTH Aachen University, Aachen, Germany
| | - Joachim Jankowski
- Institute for Molecular Cardiovascular Research, University Hospital Aachen, RWTH Aachen University, Aachen, Germany
| | - Hiroko Tsukada
- Department of Surgery II, School of Medicine, Tokyo Women's Medical University, Tokyo, Japan
| | - Stefan Jockenhövel
- Department of Biohybrid & Medical Textiles (Biotex), AME - Institute of Applied Medical Engineering, Helmholtz Institute, RWTH Aachen University, Aachen, Germany
| | - Masahiro Jinzaki
- Department of Diagnostic Radiology, Keio University School of Medicine, Tokyo, Japan
| | - Thomas Schmitz-Rode
- AME - Institute of Applied Medical Engineering, Helmholtz Institute, RWTH Aachen University, Aachen, Germany
| | - Petra Mela
- Department of Biohybrid & Medical Textiles (Biotex), AME - Institute of Applied Medical Engineering, Helmholtz Institute, RWTH Aachen University, Aachen, Germany.,Medical Materials and Implants, Department of Mechanical Engineering and Munich School of BioEngineering, Technical University of Munich, Munich, Germany
| |
Collapse
|
20
|
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: 0.2] [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
|
21
|
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: 0.4] [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
|
22
|
The Potential Impact and Timeline of Engineering on Congenital Interventions. Pediatr Cardiol 2020; 41:522-538. [PMID: 32198587 DOI: 10.1007/s00246-020-02335-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Accepted: 02/22/2020] [Indexed: 10/24/2022]
Abstract
Congenital interventional cardiology has seen rapid growth in recent decades due to the expansion of available medical devices. Percutaneous interventions have become standard of care for many common congenital conditions. Unfortunately, patients with congenital heart disease often require multiple interventions throughout their lifespan. The availability of transcatheter devices that are biodegradable, biocompatible, durable, scalable, and can be delivered in the smallest sized patients will rely on continued advances in engineering. The development pipeline for these devices will require contributions of many individuals in academia and industry including experts in material science and tissue engineering. Advances in tissue engineering, bioresorbable technology, and even new nanotechnologies and nitinol fabrication techniques which may have an impact on the field of transcatheter congenital device in the next decade are summarized in this review. This review highlights recent advances in the engineering of transcatheter-based therapies and discusses future opportunities for engineering of transcatheter devices.
Collapse
|
23
|
Schulz C, Krüger-Genge A, Jung F, Lendlein A. Aptamer supported in vitro endothelialization of poly(ether imide) films. Clin Hemorheol Microcirc 2020; 75:201-217. [PMID: 31985458 DOI: 10.3233/ch-190775] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Implantation of synthetic small-diameter vascular bypass grafts is often associated with an increased risk of failure, due to thrombotic events or late intimal hyperplasia. As one of the causes an insufficient hemocompatibility of the artificial surface is discussed. Endothelialization of synthetic grafts is reported to be a promising strategy for creating a self-renewing and regulative anti-thrombotic graft surface. However, the establishment of a shear resistant cell monolayer is still challenging. In our study, cyto- and immuno-compatible poly(ether imide) (PEI) films were explored as potential biomaterial for cardiovascular applications. Recently, we reported that the initial adherence of primary human umbilical vein endothelial cells (HUVEC) was delayed on PEI-films and about 9 days were needed to establish a confluent and almost shear resistant HUVEC monolayer. To accelerate the initial adherence of HUVEC, the PEI-film surface was functionalized with an aptamer-cRGD peptide based endothelialization supporting system. With this functionalization the initial adherence as well as the shear resistance of HUVEC on PEI-films was considerable improved compared to the unmodified polymer surface. The in vitro results confirm the general applicability of aptamers for an efficient functionalization of substrate surfaces.
Collapse
Affiliation(s)
- Christian Schulz
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Teltow, Germany
| | - Anne Krüger-Genge
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Teltow, Germany
| | - Friedrich Jung
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Teltow, Germany.,Helmholtz Virtual Institute - Multifunctional Biomaterials for Medicine, Teltow and Berlin, Germany
| | - Andreas Lendlein
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Teltow, Germany.,Helmholtz Virtual Institute - Multifunctional Biomaterials for Medicine, Teltow and Berlin, Germany.,Institute of Chemistry, University of Potsdam, Potsdam, Germany
| |
Collapse
|
24
|
Clauder F, Czerniak AS, Friebe S, Mayr SG, Scheinert D, Beck-Sickinger AG. Endothelialization of Titanium Surfaces by Bioinspired Cell Adhesion Peptide Coatings. Bioconjug Chem 2019; 30:2664-2674. [DOI: 10.1021/acs.bioconjchem.9b00573] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Affiliation(s)
- Franziska Clauder
- Institute of Biochemistry, Faculty of Life Sciences, Leipzig University, Brüderstrasse 34, 04103 Leipzig, Germany
| | - Anne Sophie Czerniak
- Institute of Biochemistry, Faculty of Life Sciences, Leipzig University, Brüderstrasse 34, 04103 Leipzig, Germany
| | - Sabrina Friebe
- Leibniz-Institute of Surface Engineering (IOM), Permoserstrasse 15, 04318 Leipzig, Germany
| | - Stefan G. Mayr
- Leibniz-Institute of Surface Engineering (IOM), Permoserstrasse 15, 04318 Leipzig, Germany
| | - Dierk Scheinert
- Department of Angiology, University Hospital Leipzig, Liebigstrasse 20, 04103 Leipzig, Germany
| | - Annette G. Beck-Sickinger
- Institute of Biochemistry, Faculty of Life Sciences, Leipzig University, Brüderstrasse 34, 04103 Leipzig, Germany
| |
Collapse
|
25
|
Elkhodiry MA, Boulanger MD, Bashth O, Tanguay JF, Laroche G, Hoesli CA. Isolating and expanding endothelial progenitor cells from peripheral blood on peptide-functionalized polystyrene surfaces. Biotechnol Bioeng 2019; 116:2598-2609. [PMID: 31286475 DOI: 10.1002/bit.27107] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 06/19/2019] [Accepted: 07/01/2019] [Indexed: 11/11/2022]
Abstract
The expansion of human peripheral blood endothelial progenitor cells to obtain therapeutically relevant endothelial colony-forming cells (ECFCs) has been commonly performed on xeno-derived extracellular matrix proteins. For cellular therapy applications, xeno-free culture conditions are desirable to improve product safety and reduce process variability. We have previously described a novel fluorophore-tagged RGD peptide (RGD-TAMRA) that enhanced the adhesion of mature endothelial cells in vitro. To investigate whether this peptide can replace animal-derived extracellular matrix proteins in the isolation and expansion of ECFCs, peripheral blood mononuclear cells from 22 healthy adult donors were seeded on RGD-TAMRA-modified polystyrene culture surfaces. Endothelial colony formation was significantly enhanced on RGD-TAMRA-modified surfaces compared to the unmodified control. No phenotypic differences were detected between ECFCs obtained on RGD-TAMRA compared to ECFCs obtained on rat-tail collagen-coated surfaces. Compared with collagen-coated surfaces and unmodified surfaces, RGD-TAMRA surfaces promoted ECFC adhesion, cell spreading, and clonal expansion. This study presents a platform that allows for a comprehensive in vitro evaluation of peptide-based biofunctionalization as a promising avenue for ex vivo ECFC expansion.
Collapse
Affiliation(s)
- Mohamed A Elkhodiry
- Department of Chemical Engineering, McGill University, Montréal, Quebec, Canada
| | - Mariève D Boulanger
- Department of Chemical Engineering, McGill University, Montréal, Quebec, Canada
| | - Omar Bashth
- Department of Chemical Engineering, McGill University, Montréal, Quebec, Canada
| | - Jean-François Tanguay
- Coronary Care Unit, Montréal Heart Institute, Montréal, Quebec, Canada.,Department of Medicine, Université de Montréal, Montréal, Quebec, Canada
| | - Gaétan Laroche
- Département de Génie des Mines, des Matériaux et de la Métallurgie, Centre de Recherche du CHU de Québec, Université Laval, Quebec City, Quebec, Canada
| | - Corinne A Hoesli
- Department of Chemical Engineering, McGill University, Montréal, Quebec, Canada
| |
Collapse
|
26
|
Cornelissen A, Vogt FJ. The effects of stenting on coronary endothelium from a molecular biological view: Time for improvement? J Cell Mol Med 2018; 23:39-46. [PMID: 30353645 PMCID: PMC6307786 DOI: 10.1111/jcmm.13936] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 08/27/2018] [Indexed: 12/13/2022] Open
Abstract
Coronary artery stenting following balloon angioplasty represents the gold standard in revascularization of coronary artery stenoses. However, stent deployment as well as percutaneous transluminal coronary angioplasty (PTCA) alone causes severe injury of vascular endothelium. The damaged endothelium is intrinsically repaired by locally derived endothelial cells and by circulating endothelial progenitor cells from the blood, leading to re‐population of the denuded regions within several weeks to months. However, the process of re‐endothelialization is often incomplete or dysfunctional, promoting in‐stent thrombosis and restenosis. The molecular and biomechanical mechanisms that influence the process of re‐endothelialization in stented segments are incompletely understood. Once the endothelium is restored, endothelial function might still be impaired. Several strategies have been followed to improve endothelial function after coronary stenting. In this review, the effects of stenting on coronary endothelium are outlined and current and future strategies to improve endothelial function after stent deployment are discussed.
Collapse
Affiliation(s)
- Anne Cornelissen
- Department of Cardiology, Pneumology, Angiology, and Internal Intensive Medicine, University Hospital Aachen, Aachen, Germany
| | - Felix Jan Vogt
- Department of Cardiology, Pneumology, Angiology, and Internal Intensive Medicine, University Hospital Aachen, Aachen, Germany
| |
Collapse
|
27
|
Hoesli CA, Tremblay C, Juneau PM, Boulanger MD, Beland AV, Ling SD, Gaillet B, Duchesne C, Ruel J, Laroche G, Garnier A. Dynamics of Endothelial Cell Responses to Laminar Shear Stress on Surfaces Functionalized with Fibronectin-Derived Peptides. ACS Biomater Sci Eng 2018; 4:3779-3791. [DOI: 10.1021/acsbiomaterials.8b00774] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Corinne A. Hoesli
- Department of Chemical Engineering, Faculty of Engineering, McGill University, Wong Building, 3610 University Street, Montréal, Québec H3A 0C5, Canada
- PROTEO Research Center, Québec, Canada
| | - Catherine Tremblay
- Département de Génie Mécanique, Faculté des Sciences et de Génie, Université Laval, Pavillon Adrien-Pouliot, 1065 Avenue de la Médecine, Québec, Québec, G1V 0A6, Canada
| | - Pierre-Marc Juneau
- Département de Génie Chimique, Faculté des Sciences et de Génie, Université Laval, Pavillon Adrien-Pouliot, 1065 Avenue de la Médecine, Québec, Québec G1V 0A6, Canada
- PROTEO Research Center, Québec, Canada
| | - Mariève D. Boulanger
- Department of Chemical Engineering, Faculty of Engineering, McGill University, Wong Building, 3610 University Street, Montréal, Québec H3A 0C5, Canada
- Département de Génie Chimique, Faculté des Sciences et de Génie, Université Laval, Pavillon Adrien-Pouliot, 1065 Avenue de la Médecine, Québec, Québec G1V 0A6, Canada
- Centre de Recherche sur les Matériaux Avancés, Département de Génie des Mines, De la Métallurgie et des Matériaux, Université Laval, Québec G1V 0A6,Canada
| | - Ariane V. Beland
- Department of Chemical Engineering, Faculty of Engineering, McGill University, Wong Building, 3610 University Street, Montréal, Québec H3A 0C5, Canada
| | - Si Da Ling
- Department of Chemical Engineering, Faculty of Engineering, McGill University, Wong Building, 3610 University Street, Montréal, Québec H3A 0C5, Canada
| | - Bruno Gaillet
- Département de Génie Chimique, Faculté des Sciences et de Génie, Université Laval, Pavillon Adrien-Pouliot, 1065 Avenue de la Médecine, Québec, Québec G1V 0A6, Canada
- PROTEO Research Center, Québec, Canada
| | - Carl Duchesne
- Département de Génie Chimique, Faculté des Sciences et de Génie, Université Laval, Pavillon Adrien-Pouliot, 1065 Avenue de la Médecine, Québec, Québec G1V 0A6, Canada
| | - Jean Ruel
- Département de Génie Mécanique, Faculté des Sciences et de Génie, Université Laval, Pavillon Adrien-Pouliot, 1065 Avenue de la Médecine, Québec, Québec, G1V 0A6, Canada
| | - Gaétan Laroche
- Centre de Recherche sur les Matériaux Avancés, Département de Génie des Mines, De la Métallurgie et des Matériaux, Université Laval, Québec G1V 0A6,Canada
- Centre de Recherche du CHU de Québec, Hôpital Saint-François d’Assise, 10 rue de l’Espinay, Bureau E0-165Québec, Québec G1L 3L5, Canada
| | - Alain Garnier
- Département de Génie Chimique, Faculté des Sciences et de Génie, Université Laval, Pavillon Adrien-Pouliot, 1065 Avenue de la Médecine, Québec, Québec G1V 0A6, Canada
- PROTEO Research Center, Québec, Canada
| |
Collapse
|
28
|
Yang JX, Pan YY, Wang XX, Qiu YG, Mao W. Endothelial progenitor cells in age-related vascular remodeling. Cell Transplant 2018; 27:786-795. [PMID: 29882417 PMCID: PMC6047273 DOI: 10.1177/0963689718779345] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Accumulating evidence has demonstrated that endothelial progenitor cells (EPCs) could facilitate the reendothelialization of injured arteries by replacing the dysfunctional endothelial cells, thereby suppressing the formation of neointima. Meanwhile, other findings suggest that EPCs may be involved in the pathogenesis of age-related vascular remodeling. This review is presented to summarize the characteristics of EPCs and age-related vascular remodeling. In addition, the role of EPCs in age-related vascular remodeling and possible solutions for improving the therapeutic effects of EPCs in the treatment of age-related diseases are discussed.
Collapse
Affiliation(s)
- Jin-Xiu Yang
- 1 Department of Cardiology, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, P.R. China.,2 Department of Cardiology, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, P.R. China
| | - Yan-Yun Pan
- 1 Department of Cardiology, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, P.R. China
| | - Xing-Xiang Wang
- 2 Department of Cardiology, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, P.R. China
| | - Yuan-Gang Qiu
- 1 Department of Cardiology, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, P.R. China
| | - Wei Mao
- 1 Department of Cardiology, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, P.R. China
| |
Collapse
|
29
|
Huang YH, Xu Q, Shen T, Li JK, Sheng JY, Shi HJ. Prevention of in-stent restenosis with endothelial progenitor cell (EPC) capture stent placement combined with regional EPC transplantation: An atherosclerotic rabbit model. Cardiol J 2018; 26:283-291. [PMID: 29611172 DOI: 10.5603/cj.a2018.0027] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Revised: 12/14/2017] [Accepted: 01/15/2018] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Even with drug-eluting stents, the risk of in-stent restenosis (ISR) remains high. The goal of this study was to investigate the use of an endothelial progenitor cell (EPC) capture stent plus regional EPC transplantation to reduce the ISR rate. METHODS Endothelial progenitor cell capture stents were fabricated using fibrin gel and anti-CD34 plus anti-VEGFR-2 dual antibodies. Twenty male New Zealand white rabbits established as an atherosclerotic model were randomly divided into two groups: group 1 (n = 10), in which EPC capture stents were deployed into the right iliac artery; and group 2 (n = 10), in which sirolimus-eluting stents were placed. In both groups, EPCs were transplanted into target vessels beyond the stents, with outflow blocked. Radiologic-pathologic correlation outcomes were reviewed after 2 months. RESULTS The technical success rate of EPC capture stent placement plus EPC transplantation was 100%. The ISR rate in group 1 was lower than in group 2 (1/10 vs. 4/10; p > 0.05). Minimal luminal diameters were larger in group 1 than in group 2 (computed tomographic angiography, 1.85 ± 0.15 mm vs. 1.50 ± 0.20 mm; duplex ultrasound, 1.90 ± 0.10 mm vs. 1.70 ± 0.30 mm; p > 0.05). Transplanted EPCs were tracked positively only in group 1. Pathologic analysis demonstrated neointimal hyperplasia thickness of 0.21 ± 0.09 mm in group 1 vs. 0.11 ± 0.07 mm in group 2 (p < 0.05). CONCLUSION Endothelial progenitor cell capture stent placement plus local EPC transplant decreases the ISR rate through thrombosis reduction rather than through neointimal hyperplasia inhibition.
Collapse
Affiliation(s)
- You-Hua Huang
- Department of Radiology, Wujin Hospital, Jiangsu University, Changzhou, China
| | - Qiang Xu
- Department of Radiology, Wujin Hospital, Jiangsu University, Changzhou, China
| | - Tao Shen
- Department of Radiology, Wujin Hospital, Jiangsu University, Changzhou, China
| | - Jian-Ke Li
- Department of Radiology, Wujin Hospital, Jiangsu University, Changzhou, China
| | - Jing-Yu Sheng
- Department of Cardiology, Wujin Hospital, Jiangsu University, Changzhou, China
| | - Hong-Jian Shi
- Department of Radiology, Wujin Hospital, Jiangsu University, Changzhou, China.
| |
Collapse
|
30
|
Bose S, Robertson SF, Bandyopadhyay A. Surface modification of biomaterials and biomedical devices using additive manufacturing. Acta Biomater 2018; 66:6-22. [PMID: 29109027 PMCID: PMC5785782 DOI: 10.1016/j.actbio.2017.11.003] [Citation(s) in RCA: 117] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 11/01/2017] [Accepted: 11/02/2017] [Indexed: 12/15/2022]
Abstract
The demand for synthetic biomaterials in medical devices, pharmaceutical products and, tissue replacement applications are growing steadily due to aging population worldwide. The use for patient matched devices is also increasing due to availability and integration of new technologies. Applications of additive manufacturing (AM) or 3D printing (3DP) in biomaterials have also increased significantly over the past decade towards traditional as well as innovative next generation Class I, II and III devices. In this review, we have focused our attention towards the use of AM in surface modified biomaterials to enhance their in vitro and in vivo performances. Specifically, we have discussed the use of AM to deliberately modify the surfaces of different classes of biomaterials with spatial specificity in a single manufacturing process as well as commented on the future outlook towards surface modification using AM. STATEMENT OF SIGNIFICANCE It is widely understood that the success of implanted medical devices depends largely on favorable material-tissue interactions. Additive manufacturing has gained traction as a viable and unique approach to engineered biomaterials, for both bulk and surface properties that improve implant outcomes. This review explores how additive manufacturing techniques have been and can be used to augment the surfaces of biomedical devices for direct clinical applications.
Collapse
Affiliation(s)
- Susmita Bose
- W. M. Keck Biomedical Materials Research Lab, School of Mechanical and Materials Engineering, Washington State University, Pullman, WA 99164, United States.
| | - Samuel Ford Robertson
- W. M. Keck Biomedical Materials Research Lab, School of Mechanical and Materials Engineering, Washington State University, Pullman, WA 99164, United States
| | - Amit Bandyopadhyay
- W. M. Keck Biomedical Materials Research Lab, School of Mechanical and Materials Engineering, Washington State University, Pullman, WA 99164, United States
| |
Collapse
|
31
|
Pacelli S, Basu S, Whitlow J, Chakravarti A, Acosta F, Varshney A, Modaresi S, Berkland C, Paul A. Strategies to develop endogenous stem cell-recruiting bioactive materials for tissue repair and regeneration. Adv Drug Deliv Rev 2017; 120:50-70. [PMID: 28734899 PMCID: PMC5705585 DOI: 10.1016/j.addr.2017.07.011] [Citation(s) in RCA: 118] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2017] [Revised: 07/05/2017] [Accepted: 07/16/2017] [Indexed: 02/07/2023]
Abstract
A leading strategy in tissue engineering is the design of biomimetic scaffolds that stimulate the body's repair mechanisms through the recruitment of endogenous stem cells to sites of injury. Approaches that employ the use of chemoattractant gradients to guide tissue regeneration without external cell sources are favored over traditional cell-based therapies that have limited potential for clinical translation. Following this concept, bioactive scaffolds can be engineered to provide a temporally and spatially controlled release of biological cues, with the possibility to mimic the complex signaling patterns of endogenous tissue regeneration. Another effective way to regulate stem cell activity is to leverage the inherent chemotactic properties of extracellular matrix (ECM)-based materials to build versatile cell-instructive platforms. This review introduces the concept of endogenous stem cell recruitment, and provides a comprehensive overview of the strategies available to achieve effective cardiovascular and bone tissue regeneration.
Collapse
Affiliation(s)
- Settimio Pacelli
- Department of Chemical and Petroleum Engineering, Bioengineering Graduate Program, University of Kansas, Lawrence, KS, USA.
| | - Sayantani Basu
- Department of Chemical and Petroleum Engineering, Bioengineering Graduate Program, University of Kansas, Lawrence, KS, USA.
| | - Jonathan Whitlow
- Department of Chemical and Petroleum Engineering, Bioengineering Graduate Program, University of Kansas, Lawrence, KS, USA.
| | - Aparna Chakravarti
- Department of Chemical and Petroleum Engineering, Bioengineering Graduate Program, University of Kansas, Lawrence, KS, USA.
| | - Francisca Acosta
- Department of Chemical and Petroleum Engineering, Bioengineering Graduate Program, University of Kansas, Lawrence, KS, USA.
| | - Arushi Varshney
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA.
| | - Saman Modaresi
- Department of Chemical and Petroleum Engineering, Bioengineering Graduate Program, University of Kansas, Lawrence, KS, USA.
| | - Cory Berkland
- Department of Chemical and Petroleum Engineering, Bioengineering Graduate Program, University of Kansas, Lawrence, KS, USA; Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, USA.
| | - Arghya Paul
- Department of Chemical and Petroleum Engineering, Bioengineering Graduate Program, University of Kansas, Lawrence, KS, USA.
| |
Collapse
|
32
|
Endothelial Progenitor Cells for Ischemic Stroke: Update on Basic Research and Application. Stem Cells Int 2017; 2017:2193432. [PMID: 28900446 PMCID: PMC5576438 DOI: 10.1155/2017/2193432] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 07/03/2017] [Indexed: 01/14/2023] Open
Abstract
Ischemic stroke is one of the leading causes of human death and disability worldwide. So far, ultra-early thrombolytic therapy is the most effective treatment. However, most patients still live with varying degrees of neurological dysfunction due to its narrow therapeutic time window. It has been confirmed in many studies that endothelial progenitor cells (EPCs), as a kind of adult stem cells, can protect the neurovascular unit by repairing the vascular endothelium and its secretory function, which contribute to the recovery of neurological function after an ischemic stroke. This paper reviews the basic researches and clinical trials of EPCs especially in the field of ischemic stroke and addresses the combination of EPC application with new technologies, including neurovascular intervention, synthetic particles, cytokines, and EPC modification, with the aim of shedding some light on the application of EPCs in treating ischemic stroke in the future.
Collapse
|
33
|
Abstract
As a common etiology for ischemic stroke, atherosclerotic carotid stenosis has been targeted by vascular surgery since 1950s. Compared with carotid endarterectomy, carotid angioplasty and stenting (CAS) is almost similarly efficacious and less invasive. These advantages make CAS an alternative in treating carotid stenosis. However, accumulative evidences suggested that the long-term benefit-risk ratio of CAS may be decreased or even neutralized by the complications related to in-stent restenosis (ISR). Therefore, investigating the mechanisms and identifying the influential factors of ISR are of vital importance for improving the long-term outcomes of CAS. As responses to intrinsic and extrinsic injuries, intimal hyperplasia and vascular smooth muscle cell proliferation have been regarded as the principle mechanisms for ISR development. Due to the lack of consensus-based definition and consistent follow-up protocol, the reported incidences of ISR after CAS varied widely among studies. These variations made the inter-study comparisons of ISR largely illogical. To eliminate restenosis after CAS, both surgery and endovascular procedures have been attempted with promising results. For preventing ISR, drug-eluting stents and antiplatelets have been proposed as potential solutions.
Collapse
Affiliation(s)
- Zhengze Dai
- Department of Neurology, Jinling Hospital, Nanjing Medical University, Nanjing, China
- Department of Neurology, Pukou Hospital, Nanjing, China
| | - Gelin Xu
- Department of Neurology, Jinling Hospital, Nanjing Medical University, Nanjing, China
- Department of Neurology, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| |
Collapse
|
34
|
Mascharak S, Benitez PL, Proctor AC, Madl CM, Hu KH, Dewi RE, Butte MJ, Heilshorn SC. YAP-dependent mechanotransduction is required for proliferation and migration on native-like substrate topography. Biomaterials 2017; 115:155-166. [PMID: 27889666 PMCID: PMC5572766 DOI: 10.1016/j.biomaterials.2016.11.019] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Revised: 11/13/2016] [Accepted: 11/15/2016] [Indexed: 01/02/2023]
Abstract
Native vascular extracellular matrices (vECM) consist of elastic fibers that impart varied topographical properties, yet most in vitro models designed to study the effects of topography on cell behavior are not representative of native architecture. Here, we engineer an electrospun elastin-like protein (ELP) system with independently tunable, vECM-mimetic topography and demonstrate that increasing topographical variation causes loss of endothelial cell-cell junction organization. This loss of VE-cadherin signaling and increased cytoskeletal contractility on more topographically varied ELP substrates in turn promote YAP activation and nuclear translocation, resulting in significantly increased endothelial cell migration and proliferation. Our findings identify YAP as a required signaling factor through which fibrous substrate topography influences cell behavior and highlights topography as a key design parameter for engineered biomaterials.
Collapse
Affiliation(s)
- Shamik Mascharak
- Department of Bioengineering, Stanford University, Stanford, CA, 94305, USA
| | - Patrick L Benitez
- Department of Bioengineering, Stanford University, Stanford, CA, 94305, USA
| | - Amy C Proctor
- Department of Chemical Engineering, Stanford University, Stanford, CA, 94305, USA
| | - Christopher M Madl
- Department of Bioengineering, Stanford University, Stanford, CA, 94305, USA
| | - Kenneth H Hu
- Biophysics Graduate Group, Stanford University, Stanford, CA, 94305, USA
| | - Ruby E Dewi
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, 94305, USA
| | - Manish J Butte
- Department of Pediatrics, Stanford University, Stanford, CA, 94305, USA
| | - Sarah C Heilshorn
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, 94305, USA.
| |
Collapse
|
35
|
Kanie K, Kondo Y, Owaki J, Ikeda Y, Narita Y, Kato R, Honda H. Focused Screening of ECM-Selective Adhesion Peptides on Cellulose-Bound Peptide Microarrays. Bioengineering (Basel) 2016; 3:bioengineering3040031. [PMID: 28952593 PMCID: PMC5597274 DOI: 10.3390/bioengineering3040031] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Revised: 11/14/2016] [Accepted: 11/16/2016] [Indexed: 01/28/2023] Open
Abstract
The coating of surfaces with bio-functional proteins is a promising strategy for the creation of highly biocompatible medical implants. Bio-functional proteins from the extracellular matrix (ECM) provide effective surface functions for controlling cellular behavior. We have previously screened bio-functional tripeptides for feasibility of mass production with the aim of identifying those that are medically useful, such as cell-selective peptides. In this work, we focused on the screening of tripeptides that selectively accumulate collagen type IV (Col IV), an ECM protein that accelerates the re-endothelialization of medical implants. A SPOT peptide microarray was selected for screening owing to its unique cellulose membrane platform, which can mimic fibrous scaffolds used in regenerative medicine. However, since the library size on the SPOT microarray was limited, physicochemical clustering was used to provide broader variation than that of random peptide selection. Using the custom focused microarray of 500 selected peptides, we assayed the relative binding rates of tripeptides to Col IV, collagen type I (Col I), and albumin. We discovered a cluster of Col IV-selective adhesion peptides that exhibit bio-safety with endothelial cells. The results from this study can be used to improve the screening of regeneration-enhancing peptides.
Collapse
Affiliation(s)
- Kei Kanie
- Department of Basic Medicinal Sciences, Graduate School of Pharmaceutical Sciences, Nagoya University, Aichi 464-8601, Japan.
| | - Yuto Kondo
- Department of Biotechnology, Graduate School of Engineering, Nagoya University, Aichi 464-8603, Japan.
| | - Junki Owaki
- Department of Biotechnology, Graduate School of Engineering, Nagoya University, Aichi 464-8603, Japan.
| | - Yurika Ikeda
- Department of Basic Medicinal Sciences, Graduate School of Pharmaceutical Sciences, Nagoya University, Aichi 464-8601, Japan.
| | - Yuji Narita
- Department of Cardiac Surgery, Nagoya University Graduate School of Medicine, Aichi 466-8550, Japan.
| | - Ryuji Kato
- Department of Basic Medicinal Sciences, Graduate School of Pharmaceutical Sciences, Nagoya University, Aichi 464-8601, Japan.
| | - Hiroyuki Honda
- Department of Biotechnology, Graduate School of Engineering, Nagoya University, Aichi 464-8603, Japan.
| |
Collapse
|
36
|
Schulz C, Hecht J, Krüger-Genge A, Kratz K, Jung F, Lendlein A. Generating Aptamers Interacting with Polymeric Surfaces for Biofunctionalization. Macromol Biosci 2016; 16:1776-1791. [PMID: 27689917 DOI: 10.1002/mabi.201600319] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 08/26/2016] [Indexed: 12/23/2022]
Abstract
Common strategies for biofunctionalization of surfaces comprise the immobilization of bioactive molecules used as cell-binding ligands for cell recruitment. Besides covalent binding, multivalent noncovalent physical forces between substrate and ligand are an alternative way to equip surfaces with biomacromolecules. In this study, polymer binding ligands are screened by means of a DNA-based in vitro selection process. As candidate biomaterials poly(ether imide) (PEI), polystyrene, and poly[ethylene-co-(vinyl acetate)] are selected, due to their different chemical structure, but similar macroscopic interface properties, allowing physical interaction with nucleotide bases by varying valences. Multivalent interacting aptamers are successfully enriched by SELEX method and an area-wide surface functionalization is achieved, which can be used for further binding of bioactive molecules. In vitro selection against the polymers result in thymine-dominated aptamer binding motifs. The preferential interaction with thymine is attributed to its chemical structure, connected with a decreased electrostatic repulsion of the π-system and the hydrophobic character maximizing entropy. The aptamer binding stability correlates with available valences for interaction, resulting in a more stable functionalization of PEI.
Collapse
Affiliation(s)
- Christian Schulz
- Institute of Biomaterial Science and Berlin-Brandenburger Centre for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Kantstraße 55, 14513, Teltow, Germany
| | - Jochen Hecht
- Charité - Universitätsmedizin Berlin, Berlin-Brandenburger Centre for Regenerative Therapies, Campus Virchow-Klinikum, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Anne Krüger-Genge
- Institute of Biomaterial Science and Berlin-Brandenburger Centre for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Kantstraße 55, 14513, Teltow, Germany
| | - Karl Kratz
- Institute of Biomaterial Science and Berlin-Brandenburger Centre for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Kantstraße 55, 14513, Teltow, Germany.,Helmholtz Virtual Institute, Multifunctional Biomaterials for Medicine, Kantstraße 55, 14513, Teltow, Germany
| | - Friedrich Jung
- Institute of Biomaterial Science and Berlin-Brandenburger Centre for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Kantstraße 55, 14513, Teltow, Germany.,Helmholtz Virtual Institute, Multifunctional Biomaterials for Medicine, Kantstraße 55, 14513, Teltow, Germany
| | - Andreas Lendlein
- Institute of Biomaterial Science and Berlin-Brandenburger Centre for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Kantstraße 55, 14513, Teltow, Germany.,Helmholtz Virtual Institute, Multifunctional Biomaterials for Medicine, Kantstraße 55, 14513, Teltow, Germany
| |
Collapse
|
37
|
Gu X, Mao Z, Ye SH, Koo Y, Yun Y, Tiasha TR, Shanov V, Wagner WR. Biodegradable, elastomeric coatings with controlled anti-proliferative agent release for magnesium-based cardiovascular stents. Colloids Surf B Biointerfaces 2016; 144:170-179. [DOI: 10.1016/j.colsurfb.2016.03.086] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2015] [Revised: 03/24/2016] [Accepted: 03/31/2016] [Indexed: 01/16/2023]
|
38
|
Wronska MA, O'Connor IB, Tilbury MA, Srivastava A, Wall JG. Adding Functions to Biomaterial Surfaces through Protein Incorporation. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2016; 28:5485-5508. [PMID: 27164952 DOI: 10.1002/adma.201504310] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Revised: 03/16/2016] [Indexed: 06/05/2023]
Abstract
The concept of biomaterials has evolved from one of inert mechanical supports with a long-term, biologically inactive role in the body into complex matrices that exhibit selective cell binding, promote proliferation and matrix production, and may ultimately become replaced by newly generated tissues in vivo. Functionalization of material surfaces with biomolecules is critical to their ability to evade immunorecognition, interact productively with surrounding tissues and extracellular matrix, and avoid bacterial colonization. Antibody molecules and their derived fragments are commonly immobilized on materials to mediate coating with specific cell types in fields such as stent endothelialization and drug delivery. The incorporation of growth factors into biomaterials has found application in promoting and accelerating bone formation in osteogenerative and related applications. Peptides and extracellular matrix proteins can impart biomolecule- and cell-specificities to materials while antimicrobial peptides have found roles in preventing biofilm formation on devices and implants. In this progress report, we detail developments in the use of diverse proteins and peptides to modify the surfaces of hard biomaterials in vivo and in vitro. Chemical approaches to immobilizing active biomolecules are presented, as well as platform technologies for isolation or generation of natural or synthetic molecules suitable for biomaterial functionalization.
Collapse
Affiliation(s)
- Małgorzata A Wronska
- Microbiology and Center for Research in Medical Devices (CÚRAM), National University of Ireland, Galway, Ireland
| | - Iain B O'Connor
- Microbiology and Center for Research in Medical Devices (CÚRAM), National University of Ireland, Galway, Ireland
| | - Maura A Tilbury
- Microbiology and Center for Research in Medical Devices (CÚRAM), National University of Ireland, Galway, Ireland
| | - Akshay Srivastava
- Microbiology and Center for Research in Medical Devices (CÚRAM), National University of Ireland, Galway, Ireland
| | - J Gerard Wall
- Microbiology and Center for Research in Medical Devices (CÚRAM), National University of Ireland, Galway, Ireland
| |
Collapse
|
39
|
Li Y, McRobb LS, Khachigian LM. Inhibition of intimal thickening after vascular injury with a cocktail of vascular endothelial growth factor and cyclic Arg-Gly-Asp peptide. Int J Cardiol 2016; 220:185-91. [PMID: 27379921 DOI: 10.1016/j.ijcard.2016.06.300] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Revised: 06/10/2016] [Accepted: 06/28/2016] [Indexed: 11/28/2022]
Abstract
BACKGROUND Percutaneous coronary intervention is widely used for the treatment of coronary artery disease; however, significant challenges such as restenosis remain. Key to solving these problems is to inhibit smooth muscle cell activation while enhancing re-endothelialization. Early growth response-1 (Egr-1) is a transcription factor that regulates vascular smooth muscle cell (SMC) proliferation and migration through its control of an array of downstream genes. METHODS A "cocktail" of vascular endothelial growth factor (VEGF)-A, VEGF-D and cyclic RGD was tested for its ability to inhibit neointima formation and accelerate re-endothelialization following balloon injury to carotid arteries of rats. RESULTS In vitro, the cocktail stimulated endothelial cell growth yet inhibited smooth muscle cell growth. In vivo, cocktail-treated injured arteries exhibited reduced intimal thickening by >50% (P<0.05). It increased both re-endothelialization and endothelial nitric oxide synthase (NOS) expression. Cocktail reduced Egr-1 expression, an effect blocked by the NOS inhibitor L-N(G)-nitroarginine methyl ester (L-NAME) that also prevented cocktail inhibition of neointima inhibition. CONCLUSIONS This combination may potentially be useful for the treatment of restenosis with concomitant stimulation of revascularization.
Collapse
Affiliation(s)
- Yue Li
- School of Medical Sciences, University of New South Wales, Sydney, NSW 2052, Australia
| | - Lucinda S McRobb
- School of Medical Sciences, University of New South Wales, Sydney, NSW 2052, Australia; Clinical Medicine, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW 2109, Australia
| | - Levon M Khachigian
- School of Medical Sciences, University of New South Wales, Sydney, NSW 2052, Australia.
| |
Collapse
|
40
|
Fu G, Yu Z, Chen Y, Chen Y, Tian F, Yang X. Direct Adsorption of Anti-CD34 Antibodies on the Nano-Porous Stent Surface to Enhance Endothelialization. ACTA CARDIOLOGICA SINICA 2016; 32:273-80. [PMID: 27274167 DOI: 10.6515/acs20150813a] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
BACKGROUND In-stent restenosis following the insertion of conventional drug-eluting stent has become an extremely serious problem due to coating techniques, with polymer matrices used to bind biological ingredients to the stent surface. However, several studies have indicated that new pro-healing technique could prevent stent thrombosis that can be caused by conventional drug-eluting stents. METHODS A novel method of attaching anti-CD34 antibodies directly on the porous surface of a 316L stainless steel bare metal stent was developed in this study, which achieved both high stability of attached anti-CD34 antibodies on the metal stent surface and high antibody activity for stem cell capture. RESULTS The in vitro and in vivo experimental results indicated that the new stent with directly coupled anti-CD34 antibodies can efficiently enhance stent endothelialization. CONCLUSIONS This study indicates that we have developed a unique method of attaching anti-CD34 antibodies directly on the porous surface of a 316L stainless steel bare metal stent, which provides a novel polymer-free approach for developing pro-healing stents.
Collapse
Affiliation(s)
- Guowei Fu
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052
| | - Zhanjiang Yu
- State Key Laboratory of Natural and Biomimetic Drugs and Department of Chemical Biology, School of Pharmaceutical Sciences, Peking University, Beijing 100191
| | - Yongqiang Chen
- Lepu Medical Technology (Beijing) Co., Ltd, Beijing 100022
| | - Yundai Chen
- Department of Cardiology, The General Hospital of the People's Liberation Army, Beijing 100853, P. R. China
| | - Feng Tian
- Department of Cardiology, The General Hospital of the People's Liberation Army, Beijing 100853, P. R. China
| | - Xiaoda Yang
- State Key Laboratory of Natural and Biomimetic Drugs and Department of Chemical Biology, School of Pharmaceutical Sciences, Peking University, Beijing 100191
| |
Collapse
|
41
|
Simsekyilmaz S, Liehn EA, Weinandy S, Schreiber F, Megens RTA, Theelen W, Smeets R, Jockenhövel S, Gries T, Möller M, Klee D, Weber C, Zernecke A. Targeting In-Stent-Stenosis with RGD- and CXCL1-Coated Mini-Stents in Mice. PLoS One 2016; 11:e0155829. [PMID: 27192172 PMCID: PMC4871500 DOI: 10.1371/journal.pone.0155829] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 05/04/2016] [Indexed: 11/19/2022] Open
Abstract
Atherosclerotic lesions that critically narrow the artery can necessitate an angioplasty and stent implantation. Long-term therapeutic effects, however, are limited by excessive arterial remodeling. We here employed a miniaturized nitinol-stent coated with star-shaped polyethylenglycole (star-PEG), and evaluated its bio-functionalization with RGD and CXCL1 for improving in-stent stenosis after implantation into carotid arteries of mice. Nitinol foils or stents (bare metal) were coated with star-PEG, and bio-functionalized with RGD, or RGD/CXCL1. Cell adhesion to star-PEG-coated nitinol foils was unaltered or reduced, whereas bio-functionalization with RGD but foremost RGD/CXCL1 increased adhesion of early angiogenic outgrowth cells (EOCs) and endothelial cells but not smooth muscle cells when compared with bare metal foils. Stimulation of cells with RGD/CXCL1 furthermore increased the proliferation of EOCs. In vivo, bio-functionalization with RGD/CXCL1 significantly reduced neointima formation and thrombus formation, and increased re-endothelialization in apoE-/- carotid arteries compared with bare-metal nitinol stents, star-PEG-coated stents, and stents bio-functionalized with RGD only. Bio-functionalization of star-PEG-coated nitinol-stents with RGD/CXCL1 reduced in-stent neointima formation. By supporting the adhesion and proliferation of endothelial progenitor cells, RGD/CXCL1 coating of stents may help to accelerate endothelial repair after stent implantation, and thus may harbor the potential to limit the complication of in-stent restenosis in clinical approaches.
Collapse
Affiliation(s)
- Sakine Simsekyilmaz
- Institute for Molecular Cardiovascular Research, University Hospital Aachen, RWTH Aachen University, Aachen, Germany
- Institute of Technical and Macromolecular Chemistry, RWTH Aachen University, Aachen, Germany
- Biochemistry Institute, Justus-Liebig-University, Giessen, Germany
| | - Elisa A. Liehn
- Institute for Molecular Cardiovascular Research, University Hospital Aachen, RWTH Aachen University, Aachen, Germany
- Human Genetic Laboratory, University of Medicine and Pharmacy, Craiova, Romania
- IZKF Aachen, University Hospital Aachen, RWTH Aachen University, Aachen, Germany
| | - Stefan Weinandy
- Institute of Technical and Macromolecular Chemistry, RWTH Aachen University, Aachen, Germany
- Department of Applied Medical Engineering, Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, Aachen, Germany
| | - Fabian Schreiber
- Institute for Textile Technology, RWTH Aachen University, Aachen, Germany
| | - Remco T. A. Megens
- Institute for Cardiovascular Prevention, Ludwig-Maximilians University Munich, Munich, Germany
| | - Wendy Theelen
- Institute for Molecular Cardiovascular Research, University Hospital Aachen, RWTH Aachen University, Aachen, Germany
| | - Ralf Smeets
- Department of Oral and Maxillofacial Surgery, Center of Clinical Neurosciences, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Stefan Jockenhövel
- Department of Applied Medical Engineering, Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, Aachen, Germany
| | - Thomas Gries
- Institute for Textile Technology, RWTH Aachen University, Aachen, Germany
| | - Martin Möller
- Institute of Technical and Macromolecular Chemistry, RWTH Aachen University, Aachen, Germany
| | - Doris Klee
- Institute of Technical and Macromolecular Chemistry, RWTH Aachen University, Aachen, Germany
| | - Christian Weber
- Institute for Cardiovascular Prevention, Ludwig-Maximilians University Munich, Munich, Germany
| | - Alma Zernecke
- Institute for Experimental Biomedicine, University Hospital Würzburg, Würzburg, Germany
| |
Collapse
|
42
|
Le Saux G, Plawinski L, Parrot C, Nlate S, Servant L, Teichmann M, Buffeteau T, Durrieu MC. Surface bound VEGF mimicking peptide maintains endothelial cell proliferation in the absence of soluble VEGF in vitro. J Biomed Mater Res A 2016; 104:1425-36. [PMID: 26845245 DOI: 10.1002/jbm.a.35677] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Revised: 12/23/2015] [Accepted: 02/02/2016] [Indexed: 01/13/2023]
Abstract
Continuous glucose monitoring is an efficient method for the management of diabetes and in limiting the complications induced by large fluctuations in glucose levels. For this, intravascular systems may assist in producing more reliable and accurate devices. However, neovascularization is a key factor to be addressed in improving their biocompatibility. In this scope, the perennial modification of the surface of an implant with the proangiogenic Vascular Endothelial Growth Factor mimic peptide (SVVYGLR peptide sequence) holds great promise. Herein, we report on the preparation of gold substrates presenting the covalently grafted SVVYGLR peptide sequence and their effect on HUVEC behavior. Effective coupling was demonstrated using XPS and PM-IRRAS. The produced surfaces were shown to be beneficial for HUVEC adhesion. Importantly, surface bound SVVYGLR is able to maintain HUVEC proliferation even in the absence of soluble VEGF. © 2016 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 104A: 1425-1436, 2016.
Collapse
Affiliation(s)
| | | | - Camila Parrot
- Equipe Labellisée Contre Le Cancer, F-33607, Pessac, France
- INSERM, U869, ARNA Laboratory, Equipe Labellisée Contre Le Cancer, Bordeaux, F-33076, France
| | - Sylvain Nlate
- University of Bordeaux, CBMN, UMR 5248, Pessac, F-33600, France
| | - Laurent Servant
- University of Bordeaux, ISM, UMR 5255, Talence, F-33400, France
| | - Martin Teichmann
- Equipe Labellisée Contre Le Cancer, F-33607, Pessac, France
- INSERM, U869, ARNA Laboratory, Equipe Labellisée Contre Le Cancer, Bordeaux, F-33076, France
| | | | | |
Collapse
|
43
|
Choi WS, Joung YK, Lee Y, Bae JW, Park HK, Park YH, Park JC, Park KD. Enhanced Patency and Endothelialization of Small-Caliber Vascular Grafts Fabricated by Coimmobilization of Heparin and Cell-Adhesive Peptides. ACS APPLIED MATERIALS & INTERFACES 2016; 8:4336-4346. [PMID: 26824876 DOI: 10.1021/acsami.5b12052] [Citation(s) in RCA: 88] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
The clinical utility of a small-caliber vascular graft is still limited, owing to the occlusion of graft by thrombosis and restenosis. A small-caliber vascular graft (diameter, 2.5 mm) fabricated by electrospinning with a polyurethane (PU) elastomer (Pellethane) and biofunctionalized with heparin and two cell-adhesive peptides, GRGDS and YIGSR, was developed for the purpose of preventing the thrombosis and restenosis through antithrombogenic activities and endothelialization. The vascular grafts showed slightly reduced adhesion of platelets and significantly decreased adsorption of fibrinogen. In vitro studies demonstrated that peptide treatment on a vascular graft enhanced the attachment of human umbilical vein endothelial cells (HUVECs), and the presence of heparin and peptides on the graft significantly increased the proliferation of HUVECs. In vivo implantation of heparin/peptides coimmobilized graft (PU-PEG-Hep/G+Y) and PU (control) grafts was performed using an abdominal aorta rabbit model for 60 days followed by angiographic monitoring and explanting for histological analyses. The patency was significantly higher for the modified PU grafts (71.4%) compared to the PU grafts (46.2%) at 9 weeks after implantation. The nontreated PU grafts showed higher levels of α-SMA expression compared to the modified grafts, and for both samples, the proximal and distal regions expressed higher levels compared to the middle region of the grafts. Moreover, immobilization of heparin and peptides and adequate porous structure were found to play important roles in endothelialization and cellular infiltration. Our results strongly encourage that the development of small-caliber vascular grafts is feasible.
Collapse
Affiliation(s)
- Won Sup Choi
- Department of Molecular Science and Technology, Ajou University , Suwon 443-749, Republic of Korea
| | - Yoon Ki Joung
- Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology , Seoul 136-791, Republic of Korea
| | - Yunki Lee
- Department of Molecular Science and Technology, Ajou University , Suwon 443-749, Republic of Korea
| | - Jin Woo Bae
- Department of Molecular Science and Technology, Ajou University , Suwon 443-749, Republic of Korea
| | | | | | | | - Ki Dong Park
- Department of Molecular Science and Technology, Ajou University , Suwon 443-749, Republic of Korea
| |
Collapse
|
44
|
Endothelial Repair and Regeneration Following Intimal Injury. J Cardiovasc Transl Res 2016; 9:91-101. [DOI: 10.1007/s12265-016-9677-1] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 01/13/2016] [Indexed: 12/19/2022]
|
45
|
Combinational Effect of Cell Adhesion Biomolecules and Their Immobilized Polymer Property to Enhance Cell-Selective Adhesion. INT J POLYM SCI 2016. [DOI: 10.1155/2016/2090985] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Although surface immobilization of medical devices with bioactive molecules is one of the most widely used strategies to improve biocompatibility, the physicochemical properties of the biomaterials significantly impact the activity of the immobilized molecules. Herein we investigate the combinational effects of cell-selective biomolecules and the hydrophobicity/hydrophilicity of the polymeric substrate on selective adhesion of endothelial cells (ECs), fibroblasts (FBs), and smooth muscle cells (SMCs). To control the polymeric substrate, biomolecules are immobilized on thermoresponsive poly(N-isopropylacrylamide-co-2-carboxyisopropylacrylamide) (poly(NIPAAm-co-CIPAAm))-grafted glass surfaces. By switching the molecular conformation of the biomolecule-immobilized polymers, the cell-selective adhesion performances are evaluated. In case of RGDS (Arg-Gly-Asp-Ser) peptide-immobilized surfaces, all cell types adhere well regardless of the surface hydrophobicity. On the other hand, a tri-Arg-immobilized surface exhibits FB-selectivity when the surface is hydrophilic. Additionally, a tri-Ile-immobilized surface exhibits EC-selective cell adhesion when the surface is hydrophobic. We believe that the proposed concept, which is used to investigate the biomolecule-immobilized surface combination, is important to produce new biomaterials, which are highly demanded for medical implants and tissue engineering.
Collapse
|
46
|
Chen H, Zhao Y, Xiong K, Li J, Chen J, Yang P, Huang N. Multifunctional coating based on EPC-specific peptide and phospholipid polymers for potential applications in cardiovascular implants fate. J Mater Chem B 2016; 4:7870-7881. [DOI: 10.1039/c6tb01811d] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Surface biofunctional modification of cardiovascular implants via the conjugation of biomolecules to prevent thrombosis and restenosis formation and to accelerate endothelialization has attracted considerable research interest.
Collapse
Affiliation(s)
- Huiqing Chen
- Key Lab. of Advanced Technology for Materials of Education Ministry
- School of Materials Science and Engineering
- Southwest Jiaotong University
- Chengdu 610031
- P. R. China
| | - Yuancong Zhao
- Key Lab. of Advanced Technology for Materials of Education Ministry
- School of Materials Science and Engineering
- Southwest Jiaotong University
- Chengdu 610031
- P. R. China
| | - Kaiqin Xiong
- Key Lab. of Advanced Technology for Materials of Education Ministry
- School of Materials Science and Engineering
- Southwest Jiaotong University
- Chengdu 610031
- P. R. China
| | - Jingan Li
- Key Lab. of Advanced Technology for Materials of Education Ministry
- School of Materials Science and Engineering
- Southwest Jiaotong University
- Chengdu 610031
- P. R. China
| | - Jiang Chen
- Key Lab. of Advanced Technology for Materials of Education Ministry
- School of Materials Science and Engineering
- Southwest Jiaotong University
- Chengdu 610031
- P. R. China
| | - Ping Yang
- Key Lab. of Advanced Technology for Materials of Education Ministry
- School of Materials Science and Engineering
- Southwest Jiaotong University
- Chengdu 610031
- P. R. China
| | - Nan Huang
- Key Lab. of Advanced Technology for Materials of Education Ministry
- School of Materials Science and Engineering
- Southwest Jiaotong University
- Chengdu 610031
- P. R. China
| |
Collapse
|
47
|
Yu Y, Wise SG, Celermajer DS, Bilek MMM, Ng MKC. Bioengineering stents with proactive biocompatibility. Interv Cardiol 2015. [DOI: 10.2217/ica.15.46] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
48
|
Benvenuto P, Neves MAD, Blaszykowski C, Romaschin A, Chung T, Kim SR, Thompson M. Adlayer-mediated antibody immobilization to stainless steel for potential application to endothelial progenitor cell capture. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2015; 31:5423-5431. [PMID: 25955536 DOI: 10.1021/acs.langmuir.5b00812] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
This work describes the straightforward surface modification of 316L stainless steel with BTS, S-(11-trichlorosilylundecanyl)-benzenethiosulfonate, a thiol-reactive trichlorosilane cross-linker molecule designed to form intermediary coatings with subsequent biofunctionalization capability. The strategy is more specifically exemplified with the immobilization of intact antibodies and their Fab' fragments. Both surface derivatization steps are thoroughly characterized by means of X-ray photoelectron spectroscopy. The antigen binding capability of both types of biofunctionalized surfaces is subsequently assessed by fluorescence microscopy. It was determined that BTS adlayers achieve robust immobilization of both intact and fragmented antibodies, while preserving antigen binding activity. Another key finding was the observation that the Fab' fragment immobilization strategy would constitute a preferential option over that involving intact antibodies in the context of in vivo capture of endothelial progenitor cells in stent applications.
Collapse
Affiliation(s)
- Pasquale Benvenuto
- †Department of Chemistry, University of Toronto, Toronto, Ontario M5S 3H6, Canada
| | - Miguel A D Neves
- †Department of Chemistry, University of Toronto, Toronto, Ontario M5S 3H6, Canada
| | | | - Alexander Romaschin
- §Clinical Biochemistry, St. Michael's Hospital, Toronto, Ontario M5B 1W8, Canada
| | - Timothy Chung
- †Department of Chemistry, University of Toronto, Toronto, Ontario M5S 3H6, Canada
| | - Sa Rang Kim
- †Department of Chemistry, University of Toronto, Toronto, Ontario M5S 3H6, Canada
| | - Michael Thompson
- †Department of Chemistry, University of Toronto, Toronto, Ontario M5S 3H6, Canada
- ‡Econous Systems Inc., Toronto, Ontario M5S 3H6, Canada
| |
Collapse
|
49
|
Hwang CW, Johnston PV, Gerstenblith G, Weiss RG, Tomaselli GF, Bogdan VE, Panigrahi A, Leszczynska A, Xia Z. Stem cell impregnated nanofiber stent sleeve for on-stent production and intravascular delivery of paracrine factors. Biomaterials 2015; 52:318-26. [PMID: 25818438 DOI: 10.1016/j.biomaterials.2015.02.047] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Revised: 02/06/2015] [Accepted: 02/09/2015] [Indexed: 12/24/2022]
Abstract
Stem cell therapies for atherosclerotic diseases are promising, but benefits remain modest with present cell delivery devices in part due to cell washout and immune attack. Many stem cell effects are believed mediated by paracrine factors (PFs) secreted by the stem cells which potentiate tissue repair via activation and enhancement of intrinsic host repair mechanisms We therefore sought to create an "intravascular paracrine factor factory" by harnessing stem cells on a stent using a nanofiber (NF) stent sleeve, and thus providing a sheltered milieu for cells to continuously produce PFs on-stent. The NF sleeve acts as a substrate on which stem cells grow, and as a semi-permeable barrier that protects cells from washout and host immune response while allowing free outward passage of PFs. NF stent sleeves were created by covering stents with electrospun poly-lactic-co-glycolic acid nanofibers and were then uniformly coated with mesenchymal stem cells (MSCs). NF sleeves blocked cell passage but did not hamper MSC attachment or proliferation, and did not alter MSC morphology or surface markers. NF sleeve MSCs continued to secrete PFs that were biologically active and successfully induced tubulogenesis in human endothelial cells. NF stent sleeves seeded with allogeneic MSCs implanted in pigs remained patent at 7 days without thrombotic occlusion or immune rejection. Our results demonstrate the feasibility of creating an intravascular PF factory using a stem cell impregnated NF stent sleeve, and pave the way for animal studies to assess the efficacy of local PF production to treat ischemic artery disease.
Collapse
Affiliation(s)
- Chao-Wei Hwang
- Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Peter V Johnston
- Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Gary Gerstenblith
- Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Robert G Weiss
- Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Gordon F Tomaselli
- Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Virginia E Bogdan
- Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Asmi Panigrahi
- Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Zhiyong Xia
- Research and Exploratory Development Department, Johns Hopkins University Applied Physics Laboratory, Laurel, MD, USA
| |
Collapse
|
50
|
Simsekyilmaz S, Liehn EA, Militaru C, Vogt F. Progress in interventional cardiology: challenges for the future. Thromb Haemost 2015; 113:464-72. [PMID: 25608683 DOI: 10.1160/th14-07-0599] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2014] [Accepted: 12/10/2014] [Indexed: 01/11/2023]
Abstract
Cardiovascular disease is the leading cause of death in the western and developing countries. Percutaneous transluminal coronary interventions have become the most prevalent treatment option for coronary artery disease; however, due to serious complications, such as stent thrombosis and in-stent restenosis (ISR), the efficacy and safety of the procedure remain important issues to address. Strategies to overcome these aspects are under extensive investigation. In this review, we summarise relevant milestones during the time to overcome these limitations of coronary stents, such as the development of polymer-free drug-eluting stents (DES) to avoid pro-inflammatory response due to the polymer coating or the developement of stents with cell-directing drugs to, simultaneously, improve re-endothelialisation and inhibit ISR amongst other techniques most recently developed, which have not fully entered the clinical stage. Also the novel concept of fully biodegradable DES featured by the lack of a permanent foreign body promises to be a beneficial and applicable tool to restore a natural vessel with maintained vasomotion and to enable optional subsequent surgical revascularisation.
Collapse
Affiliation(s)
| | | | | | - Felix Vogt
- Felix Vogt, MD, Department of Cardiology, Pulmonology, Intensive Care and Vascular Medicine, Medical Faculty, RWTH Aachen University, Pauwelsstrasse 30, 52074 Aachen, Germany, Tel.: +49 241 80 35525, Fax: +49 241 80 82716, E-mail:
| |
Collapse
|