1
|
Tornifoglio B, Hughes C, Digeronimo F, Guendouz Y, Johnston RD, Lally C. Imaging the microstructure of the arterial wall - ex vivo to in vivo potential. Acta Biomater 2025; 199:18-34. [PMID: 40348073 DOI: 10.1016/j.actbio.2025.05.022] [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: 12/18/2024] [Revised: 05/01/2025] [Accepted: 05/07/2025] [Indexed: 05/14/2025]
Abstract
Microstructural imaging enables researchers to visualise changes in the arterial wall, allowing for (i) a deeper understanding of the role of specific components in arterial mechanics, (ii) the observation of cellular responses, (iii) insights into pathological alterations in tissue microstructure, and/or (iv) advancements in tissue engineering aimed at replicating healthy native tissue. In this prospective review, we present various imaging modalities spanning from ex vivo to in vivo applications within arterial tissue. The pros, cons, and sensitivities of these modalities are highlighted. By consolidating the latest advancements in microstructural imaging of arterial tissue, the authors aim for this paper to serve as a guide for researchers designing experiments at various stages. Furthermore, the integration of non-invasive, non-destructive imaging techniques into studies provides an additional layer of microstructural information, enhancing scientific findings, improving our understanding of disease, and potentially enabling earlier or more effective diagnostic capabilities. STATEMENT OF SIGNIFICANCE: Imaging the specific microstructural components of the arterial wall provides critical insights into vascular biology, mechanics, and pathology. It enables the visualisation of key structural components and their roles in arterial function, supports the analysis of cell-matrix interactions, and reveals microarchitectural changes associated with disease progression. This level of specificity also informs the design of biomimetic materials and scaffolds in tissue engineering, facilitating the replication of native arterial properties. By synthesising recent developments in microstructural imaging techniques, this paper serves as a reference for investigators designing experiments across a range of vascular research applications. Moreover, the incorporation of non-invasive, non-destructive imaging methods offers a means to acquire detailed microstructural data without compromising tissue integrity. This enhances the interpretability and translational potential of findings, deepens our understanding of vascular disease mechanisms, and may ultimately contribute to the development of earlier and more precise diagnostic approaches.
Collapse
Affiliation(s)
- B Tornifoglio
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Ireland; Discipline of Mechanical, Manufacturing and Biomedical Engineering, School of Engineering, Trinity College Dublin, Ireland.
| | - C Hughes
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Ireland; Discipline of Mechanical, Manufacturing and Biomedical Engineering, School of Engineering, Trinity College Dublin, Ireland
| | - F Digeronimo
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Ireland; Discipline of Mechanical, Manufacturing and Biomedical Engineering, School of Engineering, Trinity College Dublin, Ireland
| | - Y Guendouz
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Ireland; Discipline of Mechanical, Manufacturing and Biomedical Engineering, School of Engineering, Trinity College Dublin, Ireland
| | - R D Johnston
- Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland (RCSI), Dublin, Ireland
| | - C Lally
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Ireland; Discipline of Mechanical, Manufacturing and Biomedical Engineering, School of Engineering, Trinity College Dublin, Ireland; Advanced Materials and Bioengineering Research Centre (AMBER), Royal College of Surgeons in Ireland and Trinity College Dublin, Ireland.
| |
Collapse
|
2
|
Bosch-Rué E, Zhang Q, Truskey GA, Olmos Buitrago J, M Bosch B, Pérez RA. Development of small tissue engineered blood vessels and their clinical and research applications. Biofabrication 2025; 17:032005. [PMID: 40341214 DOI: 10.1088/1758-5090/add626] [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: 07/02/2024] [Accepted: 05/08/2025] [Indexed: 05/10/2025]
Abstract
Since the first tissue engineered blood vessel (TEBV) was developed, different approaches, biomaterial scaffolds and cell sources have been used to obtain an engineered vessel as much similar as native vessels in terms of structure, functionality and mechanical properties. At the same time, diverse needs to obtain a functional TEBV have emerged, such as for blood vessel replacement for cardiovascular diseases (CVDs) to be used as artery bypass, to vascularize tissue engineered constructs, or even to model vascular diseases or drug testing. In this review, after briefly describing the native structure and function of arteries, we will give an overview of different biomaterials, cells and methods that have been used during the last years for the development of small TEBV (1-6 mm diameter). The importance of perfusing the TEBV to acquire functionality and maturation will be also discussed. Finally, we will center the review on TEBV applications beyond their use as vascular graft for CVDs.
Collapse
Affiliation(s)
- Elia Bosch-Rué
- Bioengineering Institute of Technology (BIT), Universitat Internacional de Catalunya (UIC), Sant Cugat del Vallès, 08195 Barcelona, Spain
- Bioengineering Department, Universitat Internacional de Catalunya (UIC), Sant Cugat del Vallès, 08195 Barcelona, Spain
| | - Qiao Zhang
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, United States of America
| | - George A Truskey
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, United States of America
| | - Jenifer Olmos Buitrago
- Bioengineering Institute of Technology (BIT), Universitat Internacional de Catalunya (UIC), Sant Cugat del Vallès, 08195 Barcelona, Spain
- Bioengineering Department, Universitat Internacional de Catalunya (UIC), Sant Cugat del Vallès, 08195 Barcelona, Spain
| | - Begoña M Bosch
- Bioengineering Institute of Technology (BIT), Universitat Internacional de Catalunya (UIC), Sant Cugat del Vallès, 08195 Barcelona, Spain
- Bioengineering Department, Universitat Internacional de Catalunya (UIC), Sant Cugat del Vallès, 08195 Barcelona, Spain
| | - Román A Pérez
- Bioengineering Institute of Technology (BIT), Universitat Internacional de Catalunya (UIC), Sant Cugat del Vallès, 08195 Barcelona, Spain
- Bioengineering Department, Universitat Internacional de Catalunya (UIC), Sant Cugat del Vallès, 08195 Barcelona, Spain
| |
Collapse
|
3
|
Deng D, Zhang Y, Tang B, Zhang Z. Sources and applications of endothelial seed cells: a review. Stem Cell Res Ther 2024; 15:175. [PMID: 38886767 PMCID: PMC11184868 DOI: 10.1186/s13287-024-03773-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Accepted: 05/26/2024] [Indexed: 06/20/2024] Open
Abstract
Endothelial cells (ECs) are widely used as donor cells in tissue engineering, organoid vascularization, and in vitro microvascular model development. ECs are invaluable tools for disease modeling and drug screening in fundamental research. When treating ischemic diseases, EC engraftment facilitates the restoration of damaged blood vessels, enhancing therapeutic outcomes. This article presents a comprehensive overview of the current sources of ECs, which encompass stem/progenitor cells, primary ECs, cell lineage conversion, and ECs derived from other cellular sources, provides insights into their characteristics, potential applications, discusses challenges, and explores strategies to mitigate these issues. The primary aim is to serve as a reference for selecting suitable EC sources for preclinical research and promote the translation of basic research into clinical applications.
Collapse
Affiliation(s)
- Dan Deng
- Department of Cardiovascular Medicine, Center for Circadian Metabolism and Cardiovascular Disease, Southwest Hospital, Army Medical University, Chongqing, China
| | - Yu Zhang
- Department of Cardiovascular Medicine, Center for Circadian Metabolism and Cardiovascular Disease, Southwest Hospital, Army Medical University, Chongqing, China
| | - Bo Tang
- Chongqing International Institute for Immunology, Chongqing, China.
| | - Zhihui Zhang
- Department of Cardiovascular Medicine, Center for Circadian Metabolism and Cardiovascular Disease, Southwest Hospital, Army Medical University, Chongqing, China.
| |
Collapse
|
4
|
Ding H, Hou X, Gao Z, Guo Y, Liao B, Wan J. Challenges and Strategies for Endothelializing Decellularized Small-Diameter Tissue-Engineered Vessel Grafts. Adv Healthc Mater 2024; 13:e2304432. [PMID: 38462702 DOI: 10.1002/adhm.202304432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 02/26/2024] [Indexed: 03/12/2024]
Abstract
Vascular diseases are the leading cause of ischemic necrosis in tissues and organs, necessitating using vascular grafts to restore blood supply. Currently, small vessels for coronary artery bypass grafts are unavailable in clinical settings. Decellularized small-diameter tissue-engineered vessel grafts (SD-TEVGs) hold significant potential. However, they face challenges, as simple implantation of decellularized SD-TEVGs in animals leads to thrombosis and calcification due to incomplete endothelialization. Consequently, research and development focus has shifted toward enhancing the endothelialization process of decellularized SD-TEVGs. This paper reviews preclinical studies involving decellularized SD-TEVGs, highlighting different strategies and their advantages and disadvantages for achieving rapid endothelialization of these vascular grafts. Methods are analyzed to improve the process while addressing potential shortcomings. This paper aims to contribute to the future commercial viability of decellularized SD-TEVGs.
Collapse
Affiliation(s)
- Heng Ding
- Department of Cardiovascular Surgery, The Affiliated Hospital, Southwest Medical University, Metabolic Vascular Diseases Key Laboratory of Sichuan Province, Key Laboratory of cardiovascular remodeling and dysfunction, Luzhou, Sichuan, 646000, China
- Nanjing Medical University, Nanjing, 211166, P. R. China
| | - Xiaojie Hou
- Department of Cardiovascular Surgery and Cardiovascular Surgery Research Laboratory, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Zhen Gao
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100069, China
| | - Yingqiang Guo
- Department of Cardiovascular Surgery and Cardiovascular Surgery Research Laboratory, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Bin Liao
- Department of Cardiovascular Surgery, The Affiliated Hospital, Southwest Medical University, Metabolic Vascular Diseases Key Laboratory of Sichuan Province, Key Laboratory of cardiovascular remodeling and dysfunction, Luzhou, Sichuan, 646000, China
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Juyi Wan
- Department of Cardiovascular Surgery, The Affiliated Hospital, Southwest Medical University, Metabolic Vascular Diseases Key Laboratory of Sichuan Province, Key Laboratory of cardiovascular remodeling and dysfunction, Luzhou, Sichuan, 646000, China
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, 646000, 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
|
Zhang Y, Fardous J, Inoue Y, Doi R, Obata A, Sakai Y, Aishima S, Ijima H. Subcutaneous angiogenesis induced by transdermal delivery of gel-in-oil nanogel dispersion. BIOMATERIALS ADVANCES 2023; 154:213628. [PMID: 37769531 DOI: 10.1016/j.bioadv.2023.213628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 09/01/2023] [Accepted: 09/16/2023] [Indexed: 10/03/2023]
Abstract
Subcutaneous transplantation aims to enhance the growth and functionality of transplanted cells for therapeutic outcomes in tissue engineering. However, the limited subcutaneous vascular network poses a challenge. Conventional methods involve co-transplantation with endothelial cells or angiogenic scaffold implantation, but they have drawbacks like tissue inflammation, compromised endothelial cell functionality, and the risk of repeated scaffold transplantation. Effective techniques are needed to overcome these challenges. This study explores the potential of G/O-NGD, a gel-in-oil nanogel dispersion, as a transdermal carrier of proliferative factors to promote angiogenesis in subcutaneous graft beds before cell transplantation. We observed robust subcutaneous angiogenesis by delivering varying amounts of bFGF using the G/O-NGD emulsion. Quantitative analysis of several parameters confirmed the efficacy of this method for building a subcutaneous vascular network. G/O-NGD is a biodegradable material that facilitates localized transdermal delivery of bFGF while maintaining its activity. The findings of this study have significant implications in both medical and industrial fields.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Chemical Engineering, Faculty of Engineering, Graduate School, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Jannatul Fardous
- Department of Pharmacy, Faculty of Science, Comilla University, Cumilla 3506, Bangladesh
| | - Yuuta Inoue
- Department of Chemical Engineering, Faculty of Engineering, Graduate School, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Ryota Doi
- Department of Chemical Engineering, Faculty of Engineering, Graduate School, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Asami Obata
- Department of Chemical Engineering, Faculty of Engineering, Graduate School, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Yusuke Sakai
- Department of Chemical Engineering, Faculty of Engineering, Graduate School, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Shinichi Aishima
- Department of Scientific Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Hiroyuki Ijima
- Department of Chemical Engineering, Faculty of Engineering, Graduate School, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan.
| |
Collapse
|
7
|
Zhang Y, Zhang C, Li Y, Zhou L, Dan N, Min J, Chen Y, Wang Y. Evolution of biomimetic ECM scaffolds from decellularized tissue matrix for tissue engineering: A comprehensive review. Int J Biol Macromol 2023; 246:125672. [PMID: 37406920 DOI: 10.1016/j.ijbiomac.2023.125672] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 06/18/2023] [Accepted: 07/01/2023] [Indexed: 07/07/2023]
Abstract
Tissue engineering is essentially a technique for imitating nature. Natural tissues are made up of three parts: extracellular matrix (ECM), signaling systems, and cells. Therefore, biomimetic ECM scaffold is one of the best candidates for tissue engineering scaffolds. Among the many scaffold materials of biomimetic ECM structure, decellularized ECM scaffolds (dECMs) obtained from natural ECM after acellular treatment stand out because of their inherent natural components and microenvironment. First, an overview of the family of dECMs is provided. The principle, mechanism, advances, and shortfalls of various decellularization technologies, including physical, chemical, and biochemical methods are then critically discussed. Subsequently, a comprehensive review is provided on recent advances in the versatile applications of dECMs including but not limited to decellularized small intestinal submucosa, dermal matrix, amniotic matrix, tendon, vessel, bladder, heart valves. And detailed examples are also drawn from scientific research and practical work. Furthermore, we outline the underlying development directions of dECMs from the perspective that tissue engineering scaffolds play an important role as an important foothold and fulcrum at the intersection of materials and medicine. As scaffolds that have already found diverse applications, dECMs will continue to present both challenges and exciting opportunities for regenerative medicine and tissue engineering.
Collapse
Affiliation(s)
- Ying Zhang
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Chenyu Zhang
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yuwen Li
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Lingyan Zhou
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Nianhua Dan
- Key Laboratory of Leather Chemistry and Engineering (Sichuan University), Ministry of Education, Chengdu 610065, China; Research Center of Biomedical Engineering, Sichuan University, Chengdu, Sichuan 610065, China
| | - Jie Min
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yining Chen
- Key Laboratory of Leather Chemistry and Engineering (Sichuan University), Ministry of Education, Chengdu 610065, China; Research Center of Biomedical Engineering, Sichuan University, Chengdu, Sichuan 610065, China.
| | - Yunbing Wang
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wang Jiang Road, Chengdu 610065, China
| |
Collapse
|
8
|
Ji J, Xu H, Li C, Luo J. Small-Caliber Tissue-Engineered Vascular Grafts Based on Human-Induced Pluripotent Stem Cells: Progress and Challenges. TISSUE ENGINEERING. PART B, REVIEWS 2023; 29:441-455. [PMID: 36884294 DOI: 10.1089/ten.teb.2023.0005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/09/2023]
Abstract
Small-caliber tissue-engineered vascular grafts (TEVGs, luminal diameter <6 mm) are promising therapies for coronary or peripheral artery bypassing surgeries or emergency treatments of vascular trauma, and a robust seed cell source is required for scalable manufacturing of small-caliber TEVGs with robust mechanical strength and bioactive endothelium in future. Human-induced pluripotent stem cells (hiPSCs) could serve as a robust cell source to derive functional vascular seed cells and potentially lead to generation of immunocompatible engineered vascular tissues. Up to date, this rising field of small-caliber hiPSC-derived TEVG (hiPSC-TEVG) research has received increasing attention and achieved significant progress. Implantable, small-caliber, hiPSC-TEVGs have been generated. These hiPSC-TEVGs displayed rupture pressure and suture retention strength approaching to those of human native saphenous veins, with vessel wall decellularized and luminal surface endothelialized with monolayer of hiPSC-endothelial cells. Meanwhile, a series of challenges remain in this field, including functional maturity of hiPSC-derived vascular cells, poor elastogenesis, suboptimal efficiency of obtaining hiPSC-derived seed cells, and relative low ready availability of hiPSC-TEVGs, which are waiting to be addressed. This review is conceived to introduce representative achievements and challenges in small-caliber TEVG generation using hiPSCs, and encapsulate the potential solution and future directions.
Collapse
Affiliation(s)
- Junyi Ji
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Hongju Xu
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Chen Li
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Jiesi Luo
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| |
Collapse
|
9
|
Liu C, Dai J, Wang X, Hu X. The Influence of Textile Structure Characteristics on the Performance of Artificial Blood Vessels. Polymers (Basel) 2023; 15:3003. [PMID: 37514393 PMCID: PMC10385882 DOI: 10.3390/polym15143003] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 06/26/2023] [Accepted: 06/27/2023] [Indexed: 07/30/2023] Open
Abstract
Cardiovascular disease is a major threat to human health worldwide, and vascular transplantation surgery is a treatment method for this disease. Often, autologous blood vessels cannot meet the needs of surgery. However, allogeneic blood vessels have limited availability or may cause rejection reactions. Therefore, the development of biocompatible artificial blood vessels is needed to solve the problem of donor shortage. Tubular fabrics prepared by textile structures have flexible compliance, which cannot be matched by other structural blood vessels. Therefore, biomedical artificial blood vessels have been widely studied in recent decades up to the present. This article focuses on reviewing four textile methods used, at present, in the manufacture of artificial blood vessels: knitting, weaving, braiding, and electrospinning. The article mainly introduces the particular effects of different structural characteristics possessed by various textile methods on the production of artificial blood vessels, such as compliance, mechanical properties, and pore size. It was concluded that woven blood vessels possess superior mechanical properties and dimensional stability, while the knitted fabrication method facilitates excellent compliance, elasticity, and porosity of blood vessels. Additionally, the study prominently showcases the ease of rebound and compression of braided tubes, as well as the significant biological benefits of electrospinning. Moreover, moderate porosity and good mechanical strength can be achieved by changing the original structural parameters; increasing the floating warp, enlarging the braiding angle, and reducing the fiber fineness and diameter can achieve greater compliance. Furthermore, physical, chemical, or biological methods can be used to further improve the biocompatibility, antibacterial, anti-inflammatory, and endothelialization of blood vessels, thereby improving their functionality. The aim is to provide some guidance for the further development of artificial blood vessels.
Collapse
Affiliation(s)
- Chenxi Liu
- College of Textiles & Clothing, Qingdao University, Qingdao 266000, China
| | - Jieyu Dai
- College of Textiles & Clothing, Qingdao University, Qingdao 266000, China
| | - Xueqin Wang
- College of Textiles & Clothing, Qingdao University, Qingdao 266000, China
| | - Xingyou Hu
- College of Textiles & Clothing, Qingdao University, Qingdao 266000, China
| |
Collapse
|
10
|
Li MX, Wei QQ, Mo HL, Ren Y, Zhang W, Lu HJ, Joung YK. Challenges and advances in materials and fabrication technologies of small-diameter vascular grafts. Biomater Res 2023; 27:58. [PMID: 37291675 PMCID: PMC10251629 DOI: 10.1186/s40824-023-00399-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 05/21/2023] [Indexed: 06/10/2023] Open
Abstract
The arterial occlusive disease is one of the leading causes of cardiovascular diseases, often requiring revascularization. Lack of suitable small-diameter vascular grafts (SDVGs), infection, thrombosis, and intimal hyperplasia associated with synthetic vascular grafts lead to a low success rate of SDVGs (< 6 mm) transplantation in the clinical treatment of cardiovascular diseases. The development of fabrication technology along with vascular tissue engineering and regenerative medicine technology allows biological tissue-engineered vascular grafts to become living grafts, which can integrate, remodel, and repair the host vessels as well as respond to the surrounding mechanical and biochemical stimuli. Hence, they potentially alleviate the shortage of existing vascular grafts. This paper evaluates the current advanced fabrication technologies for SDVGs, including electrospinning, molding, 3D printing, decellularization, and so on. Various characteristics of synthetic polymers and surface modification methods are also introduced. In addition, it also provides interdisciplinary insights into the future of small-diameter prostheses and discusses vital factors and perspectives for developing such prostheses in clinical applications. We propose that the performance of SDVGs can be improved by integrating various technologies in the near future.
Collapse
Affiliation(s)
- Mei-Xian Li
- National and Local Joint Engineering Research Center of Technical Fiber Composites for Safety and Protection, Nantong University, Nantong, 226019, China
- School of Textile and Clothing, Nantong University, Nantong, 226019, China
- Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
| | - Qian-Qi Wei
- Department of Infectious Diseases, General Hospital of Tibet Military Command, Xizang, China
| | - Hui-Lin Mo
- School of Textile and Clothing, Nantong University, Nantong, 226019, China
| | - Yu Ren
- National and Local Joint Engineering Research Center of Technical Fiber Composites for Safety and Protection, Nantong University, Nantong, 226019, China
- School of Textile and Clothing, Nantong University, Nantong, 226019, China
| | - Wei Zhang
- National and Local Joint Engineering Research Center of Technical Fiber Composites for Safety and Protection, Nantong University, Nantong, 226019, China.
- School of Textile and Clothing, Nantong University, Nantong, 226019, China.
| | - Huan-Jun Lu
- Institute of Special Environmental Medicine, Nantong University, Nantong, 226019, China.
| | - Yoon Ki Joung
- Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea.
- Division of Bio-Medical Science and Technology, University of Science and Technology (UST), 217 Gajeong-ro, Yuseong-gu, Daejeon, 34113, Republic of Korea.
| |
Collapse
|
11
|
Balikji J, Mackus M, Garssen J, Hoogbergen MM, Verster JC. Immune Fitness, Migraine, and Headache Complaints in Individuals with Self-Reported Impaired Wound Healing. Int J Gen Med 2023; 16:2245-2253. [PMID: 37293517 PMCID: PMC10246567 DOI: 10.2147/ijgm.s413258] [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: 03/19/2023] [Accepted: 04/25/2023] [Indexed: 06/10/2023] Open
Abstract
Background Having chronic wounds and impaired wound healing are associated with psychological distress. The current study aims to evaluate migraine and headache complaints in young adults with self-reported impaired wound healing. Methods A survey was conducted among N=1935 young adults (83.6% women), 18-30 years old, living in the Netherlands. Wound healing status was verified, immune fitness was assessed using a single-item rating scale, and ID Migraine was completed. In addition, several questions were answered on past year's headache experiences (including frequency, quantity, type, location, and severity). Results In both the control group (p < 0.001) and the IWH group (p = 0.002) immune fitness was significantly lower among those that reported headaches compared to those that reported no headaches. Individuals with self-reported impaired wound healing (IWH) scored significantly higher on the ID Migraine scale, and individuals of the IWH group scored significantly more often positive for migraine (ie, an ID Migraine score ≥2). They reported a younger age of onset of experiencing headaches, and significantly more often reported having a beating or pounding headache than the control group. Compared to the control group, the IWH group reported being significantly more limited in their daily activities compared to the control group. Conclusion Headaches and migraines are more frequently reported by individuals with self-reported impaired wound healing, and their reported immune fitness is significantly poorer compared to healthy controls. These headache and migraine complaints significantly limit them in their daily activities.
Collapse
Affiliation(s)
- Jessica Balikji
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, 3584 CG, the Netherlands
| | - Marlou Mackus
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, 3584 CG, the Netherlands
| | - Johan Garssen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, 3584 CG, the Netherlands
- Division of Plastic Surgery, Catharina Ziekenhuis, Eindhoven, 5623 EJ, the Netherlands
| | - Maarten M Hoogbergen
- Global Centre of Excellence Immunology, Nutricia Danone Research, Utrecht, 3584 CT, the Netherlands
| | - Joris C Verster
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, 3584 CG, the Netherlands
- Centre for Human Psychopharmacology, Swinburne University, Melbourne, VIC, 3122, Australia
| |
Collapse
|
12
|
Messner B, Grab M, Grefen L, Laufer G, Hagl C, König F. Cyclic pressure induced decellularization of porcine descending aortas. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2023; 34:19. [PMID: 37074546 PMCID: PMC10115674 DOI: 10.1007/s10856-023-06723-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 03/28/2023] [Indexed: 05/03/2023]
Abstract
The demand for decellularized xenogeneic tissues used in reconstructive heart surgery has increased over the last decades. Complete decellularization of longer and tubular aortic sections suitable for clinical application has not been achieved so far. The present study aims at analyzing the effect of pressure application on decellularization efficacy of porcine aortas using a device specifically designed for this purpose. Fresh porcine descending aortas of 8 cm length were decellularized using detergents. To increase decellularization efficacy, detergent treatment was combined with pressure application and different treatment schemes. Quantification of penetration depth as well as histological staining, scanning electron microscopy, and tensile strength tests were used to evaluate tissue structure. In general, application of pressure to aortic tissue does neither increase the decellularization success nor the penetration depth of detergents. However, it is of importance from which side of the aorta the pressure is applied. Application of intermittent pressure from the adventitial side does significantly increase the decellularization degree at the intimal side (compared to the reference group), but had no influence on the penetration depth of SDC/SDS at both sides. Although the present setup does not significantly improve the decellularization success of aortas, it is interesting that the application of pressure from the adventitial side leads to improved decellularization of the intimal side. As no adverse effects on tissue structure nor on mechanical properties were observed, optimization of the present protocol may potentially lead to complete decellularization of larger aortic segments.
Collapse
Affiliation(s)
- Barbara Messner
- Department of Cardiac Surgery, Ludwig Maximilians University, Munich, Germany.
- Cardiac Surgery Research Laboratory, Department of Cardiac Surgery, Medical University of Vienna, Vienna, Austria.
| | - Maximilian Grab
- Department of Cardiac Surgery, Ludwig Maximilians University, Munich, Germany
- Chair of Medical Materials and Implants, Technical University Munich, Munich, Germany
| | - Linda Grefen
- Department of Cardiac Surgery, Ludwig Maximilians University, Munich, Germany
| | - Günther Laufer
- Cardiac Surgery Research Laboratory, Department of Cardiac Surgery, Medical University of Vienna, Vienna, Austria
| | - Christian Hagl
- Department of Cardiac Surgery, Ludwig Maximilians University, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner site Munich Heart Alliance, Munich, Germany
| | - Fabian König
- Department of Cardiac Surgery, Ludwig Maximilians University, Munich, Germany
| |
Collapse
|
13
|
Le HT, Mahara A, Nagasaki T, Yamaoka T. Prevention of anastomotic stenosis for decellularized vascular grafts using rapamycin-loaded boronic acid-based hydrogels mimicking the perivascular tissue function. BIOMATERIALS ADVANCES 2023; 147:213324. [PMID: 36796198 DOI: 10.1016/j.bioadv.2023.213324] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 01/23/2023] [Accepted: 01/29/2023] [Indexed: 02/12/2023]
Abstract
Abnormal proliferation of vascular smooth muscle cells (VSMCs) induces graft anastomotic stenosis, resulting in graft failure. Herein, we developed a drug-loaded tissue-adhesive hydrogel as artificial perivascular tissue to suppress VSMCs proliferation. Rapamycin (RPM), an anti-stenosis drug, is selected as the drug model. The hydrogel was composed of poly (3-acrylamidophenylboronic acid-co-acrylamide) (BAAm) and polyvinyl alcohol. Since phenylboronic acid reportedly binds to sialic acid of glycoproteins which is distributed on the tissues, the hydrogel is expected to be adherent to the vascular adventitia. Two hydrogels containing 25 or 50 mg/mL of BAAm (BAVA25 and BAVA50, respectively) were prepared. A decellularized vascular graft with a diameter of <2.5 mm was selected as a graft model. Lap-shear test indicates that both hydrogels adhered to the graft adventitia. In vitro release test indicated that 83 and 73 % of RPM in BAVA25 and BAVA50 hydrogels was released after 24 h, respectively. When VSMCs were cultured with RPM-loaded BAVA hydrogels, their proliferation was suppressed at an earlier stage in RPM-loaded BAVA25 hydrogels compared to RPM-loaded BAVA50 hydrogels. An in vivo preliminary test reveals that the graft coated with RPM-loaded BAVA25 hydrogel shows better graft patency for at least 180 d than the graft coated with RPM-loaded BAVA50 hydrogel or without hydrogel. Our results suggest that RPM-loaded BAVA25 hydrogel with tissue adhesive characteristics has potential to improve decellularized vascular graft patency.
Collapse
Affiliation(s)
- Hue Thi Le
- Department of Biomedical Engineering, National Cerebral and Cardiovascular Center Research Institute, Kishibe Shimmachi, Suita, Osaka 564-8565, Japan
| | - Atsushi Mahara
- Department of Biomedical Engineering, National Cerebral and Cardiovascular Center Research Institute, Kishibe Shimmachi, Suita, Osaka 564-8565, Japan
| | - Takeshi Nagasaki
- Department of Applied Chemistry and Bioengineering, Graduate School of Engineering, Osaka Metropolitan University, 3-3-138 Sugimoto, Sumiyoshi-ku, Osaka 558-8585, Japan
| | - Tetsuji Yamaoka
- Department of Biomedical Engineering, National Cerebral and Cardiovascular Center Research Institute, Kishibe Shimmachi, Suita, Osaka 564-8565, Japan.
| |
Collapse
|
14
|
Afzal Z, Huguet EL. Bioengineering liver tissue by repopulation of decellularised scaffolds. World J Hepatol 2023; 15:151-179. [PMID: 36926238 PMCID: PMC10011915 DOI: 10.4254/wjh.v15.i2.151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 11/22/2022] [Accepted: 02/15/2023] [Indexed: 02/24/2023] Open
Abstract
Liver transplantation is the only curative therapy for end stage liver disease, but is limited by the organ shortage, and is associated with the adverse consequences of immunosuppression. Repopulation of decellularised whole organ scaffolds with appropriate cells of recipient origin offers a theoretically attractive solution, allowing reliable and timely organ sourcing without the need for immunosuppression. Decellularisation methodologies vary widely but seek to address the conflicting objectives of removing the cellular component of tissues whilst keeping the 3D structure of the extra-cellular matrix intact, as well as retaining the instructive cell fate determining biochemicals contained therein. Liver scaffold recellularisation has progressed from small rodent in vitro studies to large animal in vivo perfusion models, using a wide range of cell types including primary cells, cell lines, foetal stem cells, and induced pluripotent stem cells. Within these models, a limited but measurable degree of physiologically significant hepatocyte function has been reported with demonstrable ammonia metabolism in vivo. Biliary repopulation and function have been restricted by challenges relating to the culture and propagations of cholangiocytes, though advances in organoid culture may help address this. Hepatic vasculature repopulation has enabled sustainable blood perfusion in vivo, but with cell types that would limit clinical applications, and which have not been shown to have the specific functions of liver sinusoidal endothelial cells. Minority cell groups such as Kupffer cells and stellate cells have not been repopulated. Bioengineering by repopulation of decellularised scaffolds has significantly progressed, but there remain significant experimental challenges to be addressed before therapeutic applications may be envisaged.
Collapse
Affiliation(s)
- Zeeshan Afzal
- Department of Surgery, Addenbrookes Hospital, NIHR Comprehensive Biomedical Research and Academic Health Sciences Centre; Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, United Kingdom
| | - Emmanuel Laurent Huguet
- Department of Surgery, Addenbrookes Hospital, NIHR Comprehensive Biomedical Research and Academic Health Sciences Centre; Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, United Kingdom
| |
Collapse
|
15
|
Investigation of Cell Adhesion and Cell Viability of the Endothelial and Fibroblast Cells on Electrospun PCL, PLGA and Coaxial Scaffolds for Production of Tissue Engineered Blood Vessel. J Funct Biomater 2022; 13:jfb13040282. [PMID: 36547542 PMCID: PMC9782893 DOI: 10.3390/jfb13040282] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 11/27/2022] [Accepted: 12/04/2022] [Indexed: 12/13/2022] Open
Abstract
Endothelialization of artificial scaffolds is considered an effective strategy for increasing the efficiency of vascular transplantation. This study aimed to compare the biophysical/biocompatible properties of three different biodegradable fibrous scaffolds: Poly (ɛ-caprolactone) (PCL) alone, Poly Lactic-co-Glycolic Acid (PLGA) alone (both processed using Spraybase® electrospinning machine), and Coaxial scaffold where the fiber core and sheath was made of PCL and PLGA, respectively. Scaffold structural morphology was assessed by scanning electron microscope and tensile testing was used to investigate the scaffold tension resistance over time. Biocompatibility studies were carried out with human umbilical vein endothelial cells (HUVEC) and human vascular fibroblasts (HVF) for which cell viability (and cell proliferation over a 4-day period) and cell adhesion to the scaffolds were assessed by cytotoxicity assays and confocal microscopy, respectively. Our results showed that all biodegradable polymeric scaffolds are a reliable host to adhere and promote proliferation in HUVEC and HVF cells. In particular, PLGA membranes performed much better adhesion and enhanced cell proliferation compared to control in the absence of polymers. In addition, we demonstrate here that these biodegradable membranes present improved mechanical properties to construct potential tissue-engineered vascular graft.
Collapse
|
16
|
Jenndahl L, Österberg K, Bogestål Y, Simsa R, Gustafsson-Hedberg T, Stenlund P, Petronis S, Krona A, Fogelstrand P, Strehl R, Håkansson J. Personalized tissue-engineered arteries as vascular graft transplants: A safety study in sheep. Regen Ther 2022; 21:331-341. [PMID: 36110971 PMCID: PMC9463533 DOI: 10.1016/j.reth.2022.08.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 07/04/2022] [Accepted: 08/09/2022] [Indexed: 11/23/2022] Open
Abstract
Patients with cardiovascular disease often need replacement or bypass of a diseased blood vessel. With disadvantages of both autologous blood vessels and synthetic grafts, tissue engineering is emerging as a promising alternative of advanced therapy medicinal products for individualized blood vessels. By reconditioning of a decellularized blood vessel with the recipient’s own peripheral blood, we have been able to prevent rejection without using immunosuppressants and prime grafts for efficient recellularization in vivo. Recently, decellularized veins reconditioned with autologous peripheral blood were shown to be safe and functional in a porcine in vivo study as a potential alternative for vein grafting. In this study, personalized tissue engineered arteries (P-TEA) were developed using the same methodology and evaluated for safety in a sheep in vivo model of carotid artery transplantation. Five personalized arteries were transplanted to carotid arteries and analyzed for safety and patency as well as with histology after four months in vivo. All grafts were fully patent without any occlusion or stenosis. The tissue was well cellularized with a continuous endothelial cell layer covering the luminal surface, revascularized adventitia with capillaries and no sign of rejection or infection. In summary, the results indicate that P-TEA is safe to use and has potential as clinical grafts.
Safety and functionality evaluation of a tissue engineered ATMP in a sheep model of carotid transplantation. Efficient cellularization of a personalized tissue engineered artery in vivo. Personalized tissue engineered artery fully patent after four months in vivo.
Collapse
Affiliation(s)
- Lachmi Jenndahl
- VERIGRAFT AB, Arvid Wallgrensbacke 20, 413 46, Göteborg, Sweden
| | - Klas Österberg
- Sahlgrenska Academy, Institution of Medicine, Department of Molecular and Clinical Medicine, Blå Stråket 5 B Wallenberg Laboratory, 41345 Gothenburg, Sweden
| | - Yalda Bogestål
- RISE Research Institutes of Sweden, Materials and Production, Brinellgatan 4, 504 62 Borås, Sweden
| | - Robin Simsa
- VERIGRAFT AB, Arvid Wallgrensbacke 20, 413 46, Göteborg, Sweden.,Department of Molecular and Clinical Medicine/Wallenberg Laboratory, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| | | | - Patrik Stenlund
- RISE Research Institutes of Sweden, Materials and Production, Brinellgatan 4, 504 62 Borås, Sweden
| | - Sarunas Petronis
- RISE Research Institutes of Sweden, Materials and Production, Brinellgatan 4, 504 62 Borås, Sweden
| | - Annika Krona
- RISE Research Institutes of Sweden, Agriculture and Food, Box 5401, 402 29 Gothenburg, Sweden
| | - Per Fogelstrand
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Raimund Strehl
- VERIGRAFT AB, Arvid Wallgrensbacke 20, 413 46, Göteborg, Sweden
| | - Joakim Håkansson
- RISE Research Institutes of Sweden, Materials and Production, Brinellgatan 4, 504 62 Borås, Sweden.,Gothenburg University, Department of Laboratory Medicine, Institute of Biomedicine, Gothenburg, Sweden
| |
Collapse
|
17
|
Nasiri B, Yi T, Wu Y, Smith RJ, Podder AK, Breuer CK, Andreadis ST. Monocyte Recruitment for Vascular Tissue Regeneration. Adv Healthc Mater 2022; 11:e2200890. [PMID: 36112115 PMCID: PMC9671850 DOI: 10.1002/adhm.202200890] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 09/05/2022] [Indexed: 01/28/2023]
Abstract
A strategy to recruit monocytes (MCs) from blood to regenerate vascular tissue from unseeded (cell-free) tissue engineered vascular grafts is presented. When immobilized on the surface of vascular grafts, the fusion protein, H2R5 can capture blood-derived MC under static or flow conditions in a shear stress dependent manner. The bound MC turns into macrophages (Mϕ) expressing both M1 and M2 phenotype specific genes. When H2R5 functionalized acellular-tissue engineered vessels (A-TEVs) are implanted into the mouse aorta, they remain patent and form a continuous endothelium expressing both endothelial cell (EC) and MC specific proteins. Underneath the EC layer, multiple cells layers are formed coexpressing both smooth muscle cell (SMC) and MC specific markers. Lineage tracing analysis using a novel CX3CR1-confetti mouse model demonstrates that fluorescently labeled MC populates the graft lumen by two and four weeks postimplantation, providing direct evidence in support of MC/Mϕ recruitment to the graft lumen. Given their abundance in the blood, circulating MCs may be a great source of cells that contribute directly to the endothelialization and vascular wall formation of acellular vascular grafts under the right chemical and biomechanical cues.
Collapse
Affiliation(s)
- Bita Nasiri
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, Amherst, NY 14260-4200, USA
| | - Tai Yi
- Nationwide Children’s Hospital, Columbus, Ohio, USA
| | - Yulun Wu
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, Amherst, NY 14260-4200, USA
| | - Randall J. Smith
- Department of Biomedical Engineering, University at Buffalo, The State University of New York, Amherst, NY 14260-4200, USA
| | - Ashis Kumar Podder
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, Amherst, NY 14260-4200, USA
| | | | - Stelios T. Andreadis
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, Amherst, NY 14260-4200, USA
- Department of Biomedical Engineering, University at Buffalo, The State University of New York, Amherst, NY 14260-4200, USA
- New York State Center of Excellence in Bioinformatics and Life Sciences, Buffalo, NY
- Center for Cell, Gene and Tissue Engineering (CGTE), University at Buffalo, The State University of New York, Amherst, NY 14260-4200, USA
| |
Collapse
|
18
|
Gui Y, Qin K, Zhang Y, Bian X, Wang Z, Han D, Peng Y, Yan H, Gao Z. Quercetin improves rapid endothelialization and inflammatory microenvironment in electrospun vascular grafts. Biomed Mater 2022; 17. [DOI: 10.1088/1748-605x/ac9266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 09/15/2022] [Indexed: 11/11/2022]
Abstract
Abstract
There is a great need for small diameter vascular grafts among patients with cardiovascular diseases annually. However, continuous foreign body reactions and fibrosis capsules brought by biomaterials are both prone to poor vascular tissue regeneration. To address this problem, we fabricated a polycaprolactone (PCL) vascular graft incorporated with quercetin (PCL/QCT graft) in this study. In vitro cell assay showed that quercetin reduced the expressions of pro-inflammatory genes of macrophages while increased the expressions of anti-inflammatory genes. Furthermore, in vivo implantation was performed in a rat abdominal aorta replacement model. Upon implantation, the grafts exhibited sustained quercetin release and effectively enhanced the regeneration of vascular tissue. The results revealed that quercetin improved endothelial layer formation along the lumen of the vascular grafts at 4 weeks. Furthermore, the thickness of vascular smooth muscle layers significantly increased in PCL/QCT group compared with PCL group. More importantly, the presence of quercetin stimulated the infiltration of a large amount of M2 phenotype macrophages into the grafts. Collectively, the above data reinforced our hypothesis that the incorporation of quercetin may be in favor of modulating the inflammatory microenvironment and improving vascular tissue regeneration and remodeling in vascular grafts.
Collapse
|
19
|
Chi J, Wang M, Chen J, Hu L, Chen Z, Backman LJ, Zhang W. Topographic Orientation of Scaffolds for Tissue Regeneration: Recent Advances in Biomaterial Design and Applications. Biomimetics (Basel) 2022; 7:131. [PMID: 36134935 PMCID: PMC9496066 DOI: 10.3390/biomimetics7030131] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 08/30/2022] [Accepted: 09/08/2022] [Indexed: 11/17/2022] Open
Abstract
Tissue engineering to develop alternatives for the maintenance, restoration, or enhancement of injured tissues and organs is gaining more and more attention. In tissue engineering, the scaffold used is one of the most critical elements. Its characteristics are expected to mimic the native extracellular matrix and its unique topographical structures. Recently, the topographies of scaffolds have received increasing attention, not least because different topographies, such as aligned and random, have different repair effects on various tissues. In this review, we have focused on various technologies (electrospinning, directional freeze-drying, magnetic freeze-casting, etching, and 3-D printing) to fabricate scaffolds with different topographic orientations, as well as discussed the physicochemical (mechanical properties, porosity, hydrophilicity, and degradation) and biological properties (morphology, distribution, adhesion, proliferation, and migration) of different topographies. Subsequently, we have compiled the effect of scaffold orientation on the regeneration of vessels, skin, neural tissue, bone, articular cartilage, ligaments, tendons, cardiac tissue, corneas, skeletal muscle, and smooth muscle. The compiled information in this review will facilitate the future development of optimal topographical scaffolds for the regeneration of certain tissues. In the majority of tissues, aligned scaffolds are more suitable than random scaffolds for tissue repair and regeneration. The underlying mechanism explaining the various effects of aligned and random orientation might be the differences in "contact guidance", which stimulate certain biological responses in cells.
Collapse
Affiliation(s)
- Jiayu Chi
- School of Medicine, Southeast University, Nanjing 210009, China
| | - Mingyue Wang
- School of Medicine, Southeast University, Nanjing 210009, China
| | - Jialin Chen
- School of Medicine, Southeast University, Nanjing 210009, China
- Jiangsu Key Laboratory for Biomaterials and Devices, Southeast University, Nanjing 210096, China
- China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou 310058, China
| | - Lizhi Hu
- School of Medicine, Southeast University, Nanjing 210009, China
| | - Zhixuan Chen
- School of Medicine, Southeast University, Nanjing 210009, China
| | - Ludvig J. Backman
- Department of Integrative Medical Biology, Anatomy, Umeå University, SE-901 87 Umeå, Sweden
- Department of Community Medicine and Rehabilitation, Physiotherapy, Umeå University, SE-901 87 Umeå, Sweden
| | - Wei Zhang
- School of Medicine, Southeast University, Nanjing 210009, China
- Jiangsu Key Laboratory for Biomaterials and Devices, Southeast University, Nanjing 210096, China
- China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou 310058, China
| |
Collapse
|
20
|
Hashemi G, Dight J, Khosrotehrani K, Sormani L. Melanoma Tumour Vascularization and Tissue-Resident Endothelial Progenitor Cells. Cancers (Basel) 2022; 14:4216. [PMID: 36077754 PMCID: PMC9454996 DOI: 10.3390/cancers14174216] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 08/17/2022] [Accepted: 08/24/2022] [Indexed: 11/20/2022] Open
Abstract
The aggressiveness of solid cancers, such as melanoma, relies on their metastatic potential. It has become evident that this key cause of mortality is largely conferred by the tumour-associated stromal cells, especially endothelial cells. In addition to their essential role in the formation of the tumour vasculature, endothelial cells significantly contribute to the establishment of the tumour microenvironment, thus enabling the dissemination of cancer cells. Melanoma tumour vascularization occurs through diverse biological processes. Vasculogenesis is the formation of de novo blood vessels from endothelial progenitor cells (EPCs), and recent research has shown the role of EPCs in melanoma tumour vascularization. A more detailed understanding of the complex role of EPCs and how they contribute to the abnormal vessel structures in tumours is of importance. Moreover, anti-angiogenic drugs have a limited effect on melanoma tumour vascularization, and the role of these drugs on EPCs remains to be clarified. Overall, targeting cancer vasculature remains a challenge, and the role of anti-angiogenic drugs and combination therapies in melanoma, a focus of this review, is an area of extensive exploration.
Collapse
Affiliation(s)
| | | | - Kiarash Khosrotehrani
- Experimental Dermatology Group, Dermatology Research Centre, The UQ Diamantina Institute, The University of Queensland, Woolloongabba, QLD 4102, Australia
| | - Laura Sormani
- Experimental Dermatology Group, Dermatology Research Centre, The UQ Diamantina Institute, The University of Queensland, Woolloongabba, QLD 4102, Australia
| |
Collapse
|
21
|
Zhang L, Zuo J, Huang S, Chang Q. Endothelial progenitor cells overexpressing Grb2-associated binder 1 for in vitro-constructed tissue-engineered heart valves. KARDIOLOGIIA 2022; 62:38-43. [PMID: 35989628 DOI: 10.18087/cardio.2022.7.n2100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 05/27/2022] [Indexed: 06/15/2023]
Abstract
Aim Endothelial progenitor cells (EPCs) play important roles in heart valve replacement surgery. Up-regulation of Grb2‑associated binder 1 (Gab1) promotes hepatocyte growth factor (HGF) - induced endothelial progenitor cell proliferation and migration. This study aimed to investigate the effects of up-regulation of Gab1 in hepatocyte growth factor-induced EPCs in tissue-engineered heart valves (TEHV).Material and methods Fresh porcine aortic valves were placed in 1 % Triton X-100 and trypsin buffer for decellularization. EPCs in the control group were cultured normally, whereas those in the experimental group were both HGF stimulated and transfected with adenovirus containing the Gab1 gene. Cells in the two groups were seeded onto the decellularized valve scaffolds and cultured for 3 or 7 days. TEHV were analyzed by HE and AB-PAS staining.Results By day 3, the experimental group had formed confluent endothelial monolayers on top of the decellularized valves, on the basis of by HE staining and AB-PAS staining. One week later, the control group showed a imperfect endothelial layer.Conclusion HGF-induced EPCs overexpressing Gab1 can endothelialize the decellularized matrix and create functional TEHV, which may then be preconditioned in a bioreactor before clinical implantation.
Collapse
Affiliation(s)
- Liyu Zhang
- Qingdao University; The Affiliated Hospital of Qingdao University
| | - Jianxin Zuo
- Qingdao University; The Affiliated Hospital of Qingdao University
| | - Siyang Huang
- Qingdao University; The Affiliated Hospital of Qingdao University
| | - Qing Chang
- Qingdao University; The Affiliated Hospital of Qingdao University
| |
Collapse
|
22
|
Xiao W, Chen W, Wang Y, Zhang C, Zhang X, Zhang S, Wu W. Recombinant DTβ4-inspired porous 3D vascular graft enhanced antithrombogenicity and recruited circulating CD93 +/CD34 + cells for endothelialization. SCIENCE ADVANCES 2022; 8:eabn1958. [PMID: 35857526 PMCID: PMC9278867 DOI: 10.1126/sciadv.abn1958] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 05/27/2022] [Indexed: 05/31/2023]
Abstract
Matching material degradation with host remodeling, including endothelialization and muscular remodeling, is important to vascular regeneration. We fabricated 3D PGS-PCL vascular grafts, which presented tunable polymer components, porosity, mechanical strength, and degrading rate. Furthermore, highly porous structures enabled 3D patterning of conjugated heparin-binding peptide, dimeric thymosin β4 (DTβ4), which played key roles in antiplatelets, fibrinogenesis inhibition, and recruiting circulating progenitor cells, thereafter contributed to high patency rate, and unprecedentedly acquired carotid arterial regeneration in rabbit model. Through single-cell RNA sequencing analysis and cell tracing studies, a subset of endothelial progenitor cells, myeloid-derived CD93+/CD34+ cells, was identified as the main contributor to final endothelium regeneration. To conclude, DTβ4-inspired porous 3DVGs present adjustable physical properties, superior anticoagulating, and re-endothelializing potentials, which leads to the regeneration of small-caliber artery, thus offering a promising tool for vessel replacement in clinical applications.
Collapse
Affiliation(s)
- Weiwei Xiao
- Departments of Oral and Maxillofacial Surgery, State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, School of Stomatology, Fourth Military Medical University, Xi’an, China
| | - Wanli Chen
- Departments of Oral and Maxillofacial Surgery, State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, School of Stomatology, Fourth Military Medical University, Xi’an, China
| | - Yinggang Wang
- Departments of Oral and Maxillofacial Surgery, State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, School of Stomatology, Fourth Military Medical University, Xi’an, China
| | - Cun Zhang
- State Key Laboratory of Cancer Biology Biotechnology Center, School of Pharmacy, Fourth Military Medical University, Xi’an, China
| | - Xinchi Zhang
- Departments of Oral and Maxillofacial Surgery, State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, School of Stomatology, Fourth Military Medical University, Xi’an, China
| | - Siqian Zhang
- Departments of Oral and Maxillofacial Surgery, State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, School of Stomatology, Fourth Military Medical University, Xi’an, China
| | - Wei Wu
- Departments of Oral and Maxillofacial Surgery, State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, School of Stomatology, Fourth Military Medical University, Xi’an, China
| |
Collapse
|
23
|
Hong IS. Enhancing Stem Cell-Based Therapeutic Potential by Combining Various Bioengineering Technologies. Front Cell Dev Biol 2022; 10:901661. [PMID: 35865629 PMCID: PMC9294278 DOI: 10.3389/fcell.2022.901661] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 06/17/2022] [Indexed: 12/05/2022] Open
Abstract
Stem cell-based therapeutics have gained tremendous attention in recent years due to their wide range of applications in various degenerative diseases, injuries, and other health-related conditions. Therapeutically effective bone marrow stem cells, cord blood- or adipose tissue-derived mesenchymal stem cells (MSCs), embryonic stem cells (ESCs), and more recently, induced pluripotent stem cells (iPSCs) have been widely reported in many preclinical and clinical studies with some promising results. However, these stem cell-only transplantation strategies are hindered by the harsh microenvironment, limited cell viability, and poor retention of transplanted cells at the sites of injury. In fact, a number of studies have reported that less than 5% of the transplanted cells are retained at the site of injury on the first day after transplantation, suggesting extremely low (<1%) viability of transplanted cells. In this context, 3D porous or fibrous national polymers (collagen, fibrin, hyaluronic acid, and chitosan)-based scaffold with appropriate mechanical features and biocompatibility can be used to overcome various limitations of stem cell-only transplantation by supporting their adhesion, survival, proliferation, and differentiation as well as providing elegant 3-dimensional (3D) tissue microenvironment. Therefore, stem cell-based tissue engineering using natural or synthetic biomimetics provides novel clinical and therapeutic opportunities for a number of degenerative diseases or tissue injury. Here, we summarized recent studies involving various types of stem cell-based tissue-engineering strategies for different degenerative diseases. We also reviewed recent studies for preclinical and clinical use of stem cell-based scaffolds and various optimization strategies.
Collapse
Affiliation(s)
- In-Sun Hong
- Department of Health Sciences and Technology, GAIHST, Gachon University, Seongnam, South Korea
- Department of Molecular Medicine, School of Medicine, Gachon University, Seongnam, South Korea
- *Correspondence: In-Sun Hong,
| |
Collapse
|
24
|
Abstract
Cardiovascular defects, injuries, and degenerative diseases often require surgical intervention and the use of implantable replacement material and conduits. Traditional vascular grafts made of synthetic polymers, animal and cadaveric tissues, or autologous vasculature have been utilized for almost a century with well-characterized outcomes, leaving areas of unmet need for the patients in terms of durability and long-term patency, susceptibility to infection, immunogenicity associated with the risk of rejection, and inflammation and mechanical failure. Research to address these limitations is exploring avenues as diverse as gene therapy, cell therapy, cell reprogramming, and bioengineering of human tissue and replacement organs. Tissue-engineered vascular conduits, either with viable autologous cells or decellularized, are the forefront of technology in cardiovascular reconstruction and offer many benefits over traditional graft materials, particularly in the potential for the implanted material to be adopted and remodeled into host tissue and thus offer safer, more durable performance. This review discusses the key advances and future directions in the field of surgical vascular repair, replacement, and reconstruction, with a focus on the challenges and expected benefits of bioengineering human tissues and blood vessels.
Collapse
Affiliation(s)
- Kaleb M. Naegeli
- Humacyte, Inc, Durham, NC (K.M.N., M.H.K., Y.L., J.W., E.A.H., L.E.N.)
| | - Mehmet H. Kural
- Humacyte, Inc, Durham, NC (K.M.N., M.H.K., Y.L., J.W., E.A.H., L.E.N.)
| | - Yuling Li
- Humacyte, Inc, Durham, NC (K.M.N., M.H.K., Y.L., J.W., E.A.H., L.E.N.)
| | - Juan Wang
- Humacyte, Inc, Durham, NC (K.M.N., M.H.K., Y.L., J.W., E.A.H., L.E.N.)
| | | | - Laura E. Niklason
- Department of Anesthesiology and Biomedical Engineering, Yale University, New Haven, CT (L.E.N.)
| |
Collapse
|
25
|
Hazrati A, Malekpour K, Soudi S, Hashemi SM. Mesenchymal Stromal/Stem Cells and Their Extracellular Vesicles Application in Acute and Chronic Inflammatory Liver Diseases: Emphasizing on the Anti-Fibrotic and Immunomodulatory Mechanisms. Front Immunol 2022; 13:865888. [PMID: 35464407 PMCID: PMC9021384 DOI: 10.3389/fimmu.2022.865888] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Accepted: 03/15/2022] [Indexed: 12/21/2022] Open
Abstract
Various factors, including viral and bacterial infections, autoimmune responses, diabetes, drugs, alcohol abuse, and fat deposition, can damage liver tissue and impair its function. These factors affect the liver tissue and lead to acute and chronic liver damage, and if left untreated, can eventually lead to cirrhosis, fibrosis, and liver carcinoma. The main treatment for these disorders is liver transplantation. Still, given the few tissue donors, problems with tissue rejection, immunosuppression caused by medications taken while receiving tissue, and the high cost of transplantation, liver transplantation have been limited. Therefore, finding alternative treatments that do not have the mentioned problems is significant. Cell therapy is one of the treatments that has received a lot of attention today. Hepatocytes and mesenchymal stromal/stem cells (MSCs) are used in many patients to treat liver-related diseases. In the meantime, the use of mesenchymal stem cells has been studied more than other cells due to their favourable characteristics and has reduced the need for liver transplantation. These cells increase the regeneration and repair of liver tissue through various mechanisms, including migration to the site of liver injury, differentiation into liver cells, production of extracellular vesicles (EVs), secretion of various growth factors, and regulation of the immune system. Notably, cell therapy is not entirely excellent and has problems such as cell rejection, undesirable differentiation, accumulation in unwanted locations, and potential tumorigenesis. Therefore, the application of MSCs derived EVs, including exosomes, can help treat liver disease and prevent its progression. Exosomes can prevent apoptosis and induce proliferation by transferring different cargos to the target cell. In addition, these vesicles have been shown to transport hepatocyte growth factor (HGF) and can promote the hepatocytes'(one of the most important cells in the liver parenchyma) growths.
Collapse
Affiliation(s)
- Ali Hazrati
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Kosar Malekpour
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Sara Soudi
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Seyed Mahmoud Hashemi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
26
|
Peng C, Shao X, Tian X, Li Y, Liu D, Yan C, Han Y. CREG ameliorates embryonic stem cell differentiation into smooth muscle cells by modulation of TGF-β expression. Differentiation 2022; 125:9-17. [DOI: 10.1016/j.diff.2022.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 02/19/2022] [Accepted: 03/14/2022] [Indexed: 11/27/2022]
|
27
|
A Comparative Study of an Anti-Thrombotic Small-Diameter Vascular Graft with Commercially Available e-PTFE Graft in a Porcine Carotid Model. Tissue Eng Regen Med 2022; 19:537-551. [PMID: 35167044 PMCID: PMC9130378 DOI: 10.1007/s13770-021-00422-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 12/09/2021] [Accepted: 12/13/2021] [Indexed: 12/11/2022] Open
Abstract
Background: We have designed a reinforced drug-loaded vascular graft composed of polycaprolactone (PCL) and polydioxanone (PDO) via a combination of electrospinning/3D printing approaches. To evaluate its potential for clinical application, we compared the in vivo blood compatibility and performance of PCL/PDO + 10%DY grafts doped with an antithrombotic drug (dipyridamole) with a commercial expanded polytetrafluoroethylene (e-PTFE) graft in a porcine model. Methods: A total of 10 pigs (weight: 25–35 kg) were used in this study. We made a new 5-mm graft with PCL/PDO composite nanofiber via the electrospinning technique. We simultaneously implanted a commercially available e-PTFE graft (n = 5) and our PCL/PDO + 10%DY graft (n = 5) into the carotid arteries of the pigs. No anticoagulant/antiplatelet agent was administered during the follow-up period, and ultrasonography was performed weekly to confirm the patency of the two grafts in vivo. Four weeks later, we explanted and compared the performance of the two grafts by histological analysis and scanning electron microscopy (SEM). Results: No complications, such as sweating on the graft or significant bleeding from the needle hole site, were seen in the PCL/PDO + 10%DY graft immediately after implantation. Serial ultrasonographic examination and immunohistochemical analysis demonstrated that PCL/PDO + 10%DY grafts showed normal physiological blood flow and minimal lumen reduction, and pulsed synchronously with the native artery at 4 weeks after implantation. However, all e-PTFE grafts occluded within the study period. The luminal surface of the PCL/PDO + 10%DY graft in the transitional zone was fully covered with endothelial cells as observed by SEM. Conclusion: The PCL/PDO + 10%DY graft was well tolerated, and no adverse tissue reaction was observed in porcine carotid models during the short-term follow-up. Colonization of the graft by host endothelial and smooth muscle cells coupled with substantial extracellular matrix production marked the regenerative capability. Thus, this material may be an ideal substitute for vascular reconstruction and bypass surgeries. Long-term observations will be necessary to determine the anti-thrombotic and remodeling potential of this device. Supplementary Information The online version contains supplementary material available at 10.1007/s13770-021-00422-4.
Collapse
|
28
|
Karakaya C, van Asten JGM, Ristori T, Sahlgren CM, Loerakker S. Mechano-regulated cell-cell signaling in the context of cardiovascular tissue engineering. Biomech Model Mechanobiol 2022; 21:5-54. [PMID: 34613528 PMCID: PMC8807458 DOI: 10.1007/s10237-021-01521-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 09/15/2021] [Indexed: 01/18/2023]
Abstract
Cardiovascular tissue engineering (CVTE) aims to create living tissues, with the ability to grow and remodel, as replacements for diseased blood vessels and heart valves. Despite promising results, the (long-term) functionality of these engineered tissues still needs improvement to reach broad clinical application. The functionality of native tissues is ensured by their specific mechanical properties directly arising from tissue organization. We therefore hypothesize that establishing a native-like tissue organization is vital to overcome the limitations of current CVTE approaches. To achieve this aim, a better understanding of the growth and remodeling (G&R) mechanisms of cardiovascular tissues is necessary. Cells are the main mediators of tissue G&R, and their behavior is strongly influenced by both mechanical stimuli and cell-cell signaling. An increasing number of signaling pathways has also been identified as mechanosensitive. As such, they may have a key underlying role in regulating the G&R of tissues in response to mechanical stimuli. A more detailed understanding of mechano-regulated cell-cell signaling may thus be crucial to advance CVTE, as it could inspire new methods to control tissue G&R and improve the organization and functionality of engineered tissues, thereby accelerating clinical translation. In this review, we discuss the organization and biomechanics of native cardiovascular tissues; recent CVTE studies emphasizing the obtained engineered tissue organization; and the interplay between mechanical stimuli, cell behavior, and cell-cell signaling. In addition, we review past contributions of computational models in understanding and predicting mechano-regulated tissue G&R and cell-cell signaling to highlight their potential role in future CVTE strategies.
Collapse
Affiliation(s)
- Cansu Karakaya
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Jordy G M van Asten
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Tommaso Ristori
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Cecilia M Sahlgren
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands
- Faculty of Science and Engineering, Biosciences, Åbo Akademi, Turku, Finland
| | - Sandra Loerakker
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands.
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands.
| |
Collapse
|
29
|
Current Progress in Vascular Engineering and Its Clinical Applications. Cells 2022; 11:cells11030493. [PMID: 35159302 PMCID: PMC8834640 DOI: 10.3390/cells11030493] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Revised: 01/28/2022] [Accepted: 01/29/2022] [Indexed: 02/04/2023] Open
Abstract
Coronary heart disease (CHD) is caused by narrowing or blockage of coronary arteries due to atherosclerosis. Coronary artery bypass grafting (CABG) is widely used for the treatment of severe CHD cases. Although autologous vessels are a preferred choice, healthy autologous vessels are not always available; hence there is a demand for tissue engineered vascular grafts (TEVGs) to be used as alternatives. However, producing clinical grade implantable TEVGs that could healthily survive in the host with long-term patency is still a great challenge. There are additional difficulties in producing small diameter (<6 mm) vascular conduits. As a result, there have not been TEVGs that are commercially available. Properties of vascular scaffolds such as tensile strength, thrombogenicity and immunogenicity are key factors that determine the biocompatibility of TEVGs. The source of vascular cells employed to produce TEVGs is a limiting factor for large-scale productions. Advanced technologies including the combined use of natural and biodegradable synthetic materials for scaffolds in conjunction with the use of mesenchyme stem cells or induced pluripotent stem cells (iPSCs) provide promising solutions for vascular tissue engineering. The aim of this review is to provide an update on various aspects in this field and the current status of TEVG clinical applications.
Collapse
|
30
|
Rodriguez-Soto MA, Suarez Vargas N, Riveros A, Camargo CM, Cruz JC, Sandoval N, Briceño JC. Failure Analysis of TEVG's I: Overcoming the Initial Stages of Blood Material Interaction and Stabilization of the Immune Response. Cells 2021; 10:3140. [PMID: 34831361 PMCID: PMC8625197 DOI: 10.3390/cells10113140] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 10/28/2021] [Accepted: 11/06/2021] [Indexed: 12/16/2022] Open
Abstract
Vascular grafts (VG) are medical devices intended to replace the function of a diseased vessel. Current approaches use non-biodegradable materials that struggle to maintain patency under complex hemodynamic conditions. Even with the current advances in tissue engineering and regenerative medicine with the tissue engineered vascular grafts (TEVGs), the cellular response is not yet close to mimicking the biological function of native vessels, and the understanding of the interactions between cells from the blood and the vascular wall with the material in operative conditions is much needed. These interactions change over time after the implantation of the graft. Here we aim to analyze the current knowledge in bio-molecular interactions between blood components, cells and materials that lead either to an early failure or to the stabilization of the vascular graft before the wall regeneration begins.
Collapse
Affiliation(s)
- Maria A. Rodriguez-Soto
- Department of Biomedical Engineering, Universidad de los Andes, Bogotá 111711, Colombia; (N.S.V.); (A.R.); (C.M.C.); (J.C.C.)
| | - Natalia Suarez Vargas
- Department of Biomedical Engineering, Universidad de los Andes, Bogotá 111711, Colombia; (N.S.V.); (A.R.); (C.M.C.); (J.C.C.)
| | - Alejandra Riveros
- Department of Biomedical Engineering, Universidad de los Andes, Bogotá 111711, Colombia; (N.S.V.); (A.R.); (C.M.C.); (J.C.C.)
| | - Carolina Muñoz Camargo
- Department of Biomedical Engineering, Universidad de los Andes, Bogotá 111711, Colombia; (N.S.V.); (A.R.); (C.M.C.); (J.C.C.)
| | - Juan C. Cruz
- Department of Biomedical Engineering, Universidad de los Andes, Bogotá 111711, Colombia; (N.S.V.); (A.R.); (C.M.C.); (J.C.C.)
| | - Nestor Sandoval
- Department of Congenital Heart Disease and Cardiovascular Surgery, Fundación Cardio Infantil Instituto de Cardiología, Bogotá 111711, Colombia;
| | - Juan C. Briceño
- Department of Biomedical Engineering, Universidad de los Andes, Bogotá 111711, Colombia; (N.S.V.); (A.R.); (C.M.C.); (J.C.C.)
- Department of Research, Fundación Cardio Infantil Instituto de Cardiología, Bogotá 111711, Colombia
| |
Collapse
|
31
|
Fayon A, Menu P, El Omar R. Cellularized small-caliber tissue-engineered vascular grafts: looking for the ultimate gold standard. NPJ Regen Med 2021; 6:46. [PMID: 34385472 PMCID: PMC8361171 DOI: 10.1038/s41536-021-00155-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 07/23/2021] [Indexed: 02/07/2023] Open
Abstract
Due to the lack of efficacy of synthetic vascular substitutes in the replacement of small-caliber arteries, vascular tissue engineering (VTE) has emerged as a promising solution to produce viable small-caliber tissue-engineered vascular grafts (TEVG). Previous studies have shown the importance of a cellular intimal layer at the luminal surface of TEVG to prevent thrombotic events. However, the cellularization of a TEVG seems to be a critical approach to consider in the development of a TEVG. To date, no standard cellularization method or cell type has been established to create the ideal TEVG by promoting its long-term patency and function. In this review, advances in VTE are described and discussed with a particular focus on the construction approaches of cellularized small-caliber TEVGs, the cell types used, as well as their preclinical and clinical applications.
Collapse
Affiliation(s)
- Adrien Fayon
- Université de Lorraine, CNRS, IMoPA, F-54000 Nancy, France
| | - Patrick Menu
- Université de Lorraine, CNRS, IMoPA, F-54000 Nancy, France.
- Université de Lorraine, Faculté de Pharmacie, Nancy, F-54000, France.
| | - Reine El Omar
- Université de Lorraine, CNRS, IMoPA, F-54000 Nancy, France
- Université de Lorraine, Faculté de Pharmacie, Nancy, F-54000, France
| |
Collapse
|
32
|
Heng JW, Yazid MD, Abdul Rahman MR, Sulaiman N. Coatings in Decellularized Vascular Scaffolds for the Establishment of a Functional Endothelium: A Scoping Review of Vascular Graft Refinement. Front Cardiovasc Med 2021; 8:677588. [PMID: 34395554 PMCID: PMC8358320 DOI: 10.3389/fcvm.2021.677588] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 07/06/2021] [Indexed: 12/12/2022] Open
Abstract
Developments in tissue engineering techniques have allowed for the creation of biocompatible, non-immunogenic alternative vascular grafts through the decellularization of existing tissues. With an ever-growing number of patients requiring life-saving vascular bypass grafting surgeries, the production of functional small diameter decellularized vascular scaffolds has never been more important. However, current implementations of small diameter decellularized vascular grafts face numerous clinical challenges attributed to premature graft failure as a consequence of common failure mechanisms such as acute thrombogenesis and intimal hyperplasia resulting from insufficient endothelial coverage on the graft lumen. This review summarizes some of the surface modifying coating agents currently used to improve the re-endothelialization efficiency and endothelial cell persistence in decellularized vascular scaffolds that could be applied in producing a better patency small diameter vascular graft. A comprehensive search yielding 192 publications was conducted in the PubMed, Scopus, Web of Science, and Ovid electronic databases. Careful screening and removal of unrelated publications and duplicate entries resulted in a total of 16 publications, which were discussed in this review. Selected publications demonstrate that the utilization of surface coating agents can induce endothelial cell adhesion, migration, and proliferation therefore leads to increased re-endothelialization efficiency. Unfortunately, the large variance in methodologies complicates comparison of coating effects between studies. Thus far, coating decellularized tissue gave encouraging results. These developments in re-endothelialization could be incorporated in the fabrication of functional, off-the-shelf alternative small diameter vascular scaffolds.
Collapse
Affiliation(s)
- Jun Wei Heng
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Muhammad Dain Yazid
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Mohd Ramzisham Abdul Rahman
- Department of Surgery, Hospital Canselor Tuanku Muhriz, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Nadiah Sulaiman
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| |
Collapse
|
33
|
Håkansson J, Simsa R, Bogestål Y, Jenndahl L, Gustafsson-Hedberg T, Petronis S, Strehl R, Österberg K. Individualized tissue-engineered veins as vascular grafts: A proof of concept study in pig. J Tissue Eng Regen Med 2021; 15:818-830. [PMID: 34318614 DOI: 10.1002/term.3233] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 05/22/2021] [Accepted: 07/22/2021] [Indexed: 11/06/2022]
Abstract
Personalized tissue engineered vascular grafts are a promising advanced therapy medicinal product alternative to autologous or synthetic vascular grafts utilized in blood vessel bypass or replacement surgery. We hypothesized that an individualized tissue engineered vein (P-TEV) would make the body recognize the transplanted blood vessel as autologous, decrease the risk of rejection and thereby avoid lifelong treatment with immune suppressant medication as is standard with allogenic organ transplantation. To individualize blood vessels, we decellularized vena cava from six deceased donor pigs and tested them for cellular removal and histological integrity. A solution with peripheral blood from the recipient pigs was used for individualized reconditioning in a perfusion bioreactor for seven days prior to transplantation. To evaluate safety and functionality of the individualized vascular graft in vivo, we transplanted reconditioned porcine vena cava into six pigs and analyzed histology and patency of the graft at different time points, with three pigs at the final endpoint 4-5 weeks after surgery. Our results showed that the P-TEV was fully patent in all animals, did not induce any occlusion or stenosis formation and we did not find any signs of rejection. The P-TEV showed rapid recellularization in vivo with the luminal surface covered with endothelial cells. In summary, the results indicate that P-TEV is functional and have potential for use as clinical transplant grafts.
Collapse
Affiliation(s)
- Joakim Håkansson
- Chemistry, Biomaterials and Textiles, RISE Research Institutes of Sweden, Borås, Sweden.,Laboratory Medicine, Biomedicine, Gothenburg University, Gothenburg, Sweden
| | | | - Yalda Bogestål
- Chemistry, Biomaterials and Textiles, RISE Research Institutes of Sweden, Borås, Sweden
| | | | | | - Sarunas Petronis
- Chemistry, Biomaterials and Textiles, RISE Research Institutes of Sweden, Borås, Sweden
| | | | - Klas Österberg
- Molecular and Clinical Medicine, Sahlgrenska Academy, Gothenburg, Sweden
| |
Collapse
|
34
|
Zhang Q, Bosch-Rué È, Pérez RA, Truskey GA. Biofabrication of tissue engineering vascular systems. APL Bioeng 2021; 5:021507. [PMID: 33981941 PMCID: PMC8106537 DOI: 10.1063/5.0039628] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 04/02/2021] [Indexed: 12/13/2022] Open
Abstract
Cardiovascular disease (CVD) is the leading cause of death among persons aged 65 and older in the United States and many other developed countries. Tissue engineered vascular systems (TEVS) can serve as grafts for CVD treatment and be used as in vitro model systems to examine the role of various genetic factors during the CVD progressions. Current focus in the field is to fabricate TEVS that more closely resembles the mechanical properties and extracellular matrix environment of native vessels, which depends heavily on the advance in biofabrication techniques and discovery of novel biomaterials. In this review, we outline the mechanical and biological design requirements of TEVS and explore the history and recent advances in biofabrication methods and biomaterials for tissue engineered blood vessels and microvascular systems with special focus on in vitro applications. In vitro applications of TEVS for disease modeling are discussed.
Collapse
Affiliation(s)
- Qiao Zhang
- Department of Biomedical Engineering, Duke University, Durham, North Carolina 27708, USA
| | - Èlia Bosch-Rué
- Bioengineering Institute of Technology (BIT), Universitat Internacional de Catalunya (UIC), Sant Cugat del Vallès 08195, Spain
| | - Román A. Pérez
- Bioengineering Institute of Technology (BIT), Universitat Internacional de Catalunya (UIC), Sant Cugat del Vallès 08195, Spain
| | - George A. Truskey
- Department of Biomedical Engineering, Duke University, Durham, North Carolina 27708, USA
| |
Collapse
|
35
|
Meng X, Xing Y, Li J, Deng C, Li Y, Ren X, Zhang D. Rebuilding the Vascular Network: In vivo and in vitro Approaches. Front Cell Dev Biol 2021; 9:639299. [PMID: 33968926 PMCID: PMC8097043 DOI: 10.3389/fcell.2021.639299] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 03/29/2021] [Indexed: 12/25/2022] Open
Abstract
As the material transportation system of the human body, the vascular network carries the transportation of materials and nutrients. Currently, the construction of functional microvascular networks is an urgent requirement for the development of regenerative medicine and in vitro drug screening systems. How to construct organs with functional blood vessels is the focus and challenge of tissue engineering research. Here in this review article, we first introduced the basic characteristics of blood vessels in the body and the mechanism of angiogenesis in vivo, summarized the current methods of constructing tissue blood vessels in vitro and in vivo, and focused on comparing the functions, applications and advantages of constructing different types of vascular chips to generate blood vessels. Finally, the challenges and opportunities faced by the development of this field were discussed.
Collapse
Affiliation(s)
- Xiangfu Meng
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan, China
| | - Yunhui Xing
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA, United States
| | - Jiawen Li
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Cechuan Deng
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Yifei Li
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Xi Ren
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA, United States
| | - Donghui Zhang
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan, China
| |
Collapse
|
36
|
Melero-Martin JM, Dudley AC, Griffioen AW. Adieu to parting Editor in Chief and pioneering scientist Dr. Joyce Bischoff. Angiogenesis 2021; 24:191-193. [PMID: 33843032 DOI: 10.1007/s10456-021-09786-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Affiliation(s)
- Juan M Melero-Martin
- Department of Cardiac Surgery, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Andrew C Dudley
- Department of Microbiology, Immunology, and Cancer Biology, The University of Virginia, & The Emily Couric Cancer Center, Charlottesville, VA, 22908, USA
| | - Arjan W Griffioen
- Angiogenesis Laboratory, Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.
| |
Collapse
|
37
|
Kobayashi M, Ohara M, Hashimoto Y, Nakamura N, Fujisato T, Kimura T, Kishida A. In vitro evaluation of surface biological properties of decellularized aorta for cardiovascular use. J Mater Chem B 2021; 8:10977-10989. [PMID: 33174886 DOI: 10.1039/d0tb01830a] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The aim of this study was to determine an in vitro evaluation method that could directly predict in vivo performance of decellularized tissue for cardiovascular use. We hypothesized that key factors for in vitro evaluation would be found by in vitro assessment of decellularized aortas that previously showed good performance in vivo, such as high patency. We chose porcine aortas, decellularized using three different decellularization methods: sodium dodecyl-sulfate (SDS), freeze-thawing, and high-hydrostatic pressurization (HHP). Immunohistological staining, a blood clotting test, scanning electron microscopy (SEM) analysis, and recellularization of endothelial cells were used for the in vitro evaluation. There was a significant difference in the remaining extracellular matrix (ECM) components, ECM structure, and the luminal surface structure between the three decellularized aortas, respectively, resulting in differences in the recellularization of endothelial cells. On the other hand, there was no difference observed in the blood clotting test. These results suggested that the blood clotting test could be a key evaluation method for the prediction of in vivo performance. In addition, evaluation of the luminal surface structure and the recellularization experiment should be packaged as an in vitro evaluation because the long-term patency was probably affected. The evaluation approach in this study may be useful to establish regulations and a quality management system for a cardiovascular prosthesis.
Collapse
Affiliation(s)
- Mako Kobayashi
- Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, 2-3-10 Kanda-surugadai, Chiyoda-ku, Tokyo 101-0062, Japan.
| | - Masako Ohara
- Department of Bioscience and Engineering, Shibaura Institute of Technology, 307 Fukasaku, Minuma-ku, Saitama-shi, Saitama 337-8570, Japan
| | - Yoshihide Hashimoto
- Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, 2-3-10 Kanda-surugadai, Chiyoda-ku, Tokyo 101-0062, Japan.
| | - Naoko Nakamura
- Department of Bioscience and Engineering, Shibaura Institute of Technology, 307 Fukasaku, Minuma-ku, Saitama-shi, Saitama 337-8570, Japan
| | - Toshiya Fujisato
- Department of Biomedical Engineering, Osaka Institute of Technology, 5-16-1 Omiya, Asahi-ku, Osaka 535-8585, Japan
| | - Tsuyoshi Kimura
- Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, 2-3-10 Kanda-surugadai, Chiyoda-ku, Tokyo 101-0062, Japan.
| | - Akio Kishida
- Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, 2-3-10 Kanda-surugadai, Chiyoda-ku, Tokyo 101-0062, Japan.
| |
Collapse
|
38
|
Fang S, Ellman DG, Andersen DC. Review: Tissue Engineering of Small-Diameter Vascular Grafts and Their In Vivo Evaluation in Large Animals and Humans. Cells 2021; 10:713. [PMID: 33807009 PMCID: PMC8005053 DOI: 10.3390/cells10030713] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 03/10/2021] [Accepted: 03/15/2021] [Indexed: 12/15/2022] Open
Abstract
To date, a wide range of materials, from synthetic to natural or a mixture of these, has been explored, modified, and examined as small-diameter tissue-engineered vascular grafts (SD-TEVGs) for tissue regeneration either in vitro or in vivo. However, very limited success has been achieved due to mechanical failure, thrombogenicity or intimal hyperplasia, and improvements of the SD-TEVG design are thus required. Here, in vivo studies investigating novel and relative long (10 times of the inner diameter) SD-TEVGs in large animal models and humans are identified and discussed, with emphasis on graft outcome based on model- and graft-related conditions. Only a few types of synthetic polymer-based SD-TEVGs have been evaluated in large-animal models and reflect limited success. However, some polymers, such as polycaprolactone (PCL), show favorable biocompatibility and potential to be further modified and improved in the form of hybrid grafts. Natural polymer- and cell-secreted extracellular matrix (ECM)-based SD-TEVGs tested in large animals still fail due to a weak strength or thrombogenicity. Similarly, native ECM-based SD-TEVGs and in-vitro-developed hybrid SD-TEVGs that contain xenogeneic molecules or matrix seem related to a harmful graft outcome. In contrast, allogeneic native ECM-based SD-TEVGs, in-vitro-developed hybrid SD-TEVGs with allogeneic banked human cells or isolated autologous stem cells, and in-body tissue architecture (IBTA)-based SD-TEVGs seem to be promising for the future, since they are suitable in dimension, mechanical strength, biocompatibility, and availability.
Collapse
Affiliation(s)
- Shu Fang
- Laboratory of Molecular and Cellular Cardiology, Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, J. B. Winsløwsvej 25, 5000 Odense C, Denmark; (D.G.E.); (D.C.A.)
- The Danish Regenerative Center, Odense University Hospital, J. B. Winsløwsvej 4, 5000 Odense C, Denmark
- Institute of Clinical Research, University of Southern Denmark, J. B. Winsløwsvej 19, 5000 Odense C, Denmark
| | - Ditte Gry Ellman
- Laboratory of Molecular and Cellular Cardiology, Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, J. B. Winsløwsvej 25, 5000 Odense C, Denmark; (D.G.E.); (D.C.A.)
- Institute of Clinical Research, University of Southern Denmark, J. B. Winsløwsvej 19, 5000 Odense C, Denmark
| | - Ditte Caroline Andersen
- Laboratory of Molecular and Cellular Cardiology, Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, J. B. Winsløwsvej 25, 5000 Odense C, Denmark; (D.G.E.); (D.C.A.)
- The Danish Regenerative Center, Odense University Hospital, J. B. Winsløwsvej 4, 5000 Odense C, Denmark
- Institute of Clinical Research, University of Southern Denmark, J. B. Winsløwsvej 19, 5000 Odense C, Denmark
| |
Collapse
|
39
|
Vašíček J, Baláži A, Bauer M, Svoradová A, Tirpáková M, Tomka M, Chrenek P. Molecular Profiling and Gene Banking of Rabbit EPCs Derived from Two Biological Sources. Genes (Basel) 2021; 12:genes12030366. [PMID: 33806502 PMCID: PMC7998175 DOI: 10.3390/genes12030366] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 02/27/2021] [Accepted: 03/03/2021] [Indexed: 12/20/2022] Open
Abstract
Endothelial progenitor cells (EPCs) have been broadly studied for several years due to their outstanding regenerative potential. Moreover, these cells might be a valuable source of genetic information for the preservation of endangered animal species. However, a controversy regarding their characterization still exists. The aim of this study was to isolate and compare the rabbit peripheral blood- and bone marrow-derived EPCs with human umbilical vein endothelial cells (HUVECs) in terms of their phenotype and morphology that could be affected by the passage number or cryopreservation as well as to assess their possible neuro-differentiation potential. Briefly, cells were isolated and cultured under standard endothelial conditions until passage 3. The morphological changes during the culture were monitored and each passage was analyzed for the typical phenotype using flow cytometry, quantitative real–time polymerase chain reaction (qPCR) and novel digital droplet PCR (ddPCR), and compared to HUVECs. The neurogenic differentiation was induced using a commercial kit. Rabbit cells were also cryopreserved for at least 3 months and then analyzed after thawing. According to the obtained results, both rabbit EPCs exhibit a spindle-shaped morphology and high proliferation rate. The both cell lines possess same stable phenotype: CD14−CD29+CD31−CD34−CD44+CD45−CD49f+CD73+CD90+CD105+CD133−CD146−CD166+VE-cadherin+VEGFR-2+SSEA-4+MSCA-1−vWF+eNOS+AcLDL+ALDH+vimentin+desmin+α-SMA+, slightly different from HUVECs. Moreover, both induced rabbit EPCs exhibit neuron-like morphological changes and expression of neuronal markers ENO2 and MAP2. In addition, cryopreserved rabbit cells maintained high viability (>85%) and endothelial phenotype after thawing. In conclusion, our findings suggest that cells expanded from the rabbit peripheral blood and bone marrow are of the endothelial origin with a stable marker expression and interesting proliferation and differentiation capacity.
Collapse
Affiliation(s)
- Jaromír Vašíček
- NPPC, Research Institute for Animal Production Nitra, Institute of Farm Animal Genetics and Reproduction, Hlohovecká 2, 951 41 Lužianky, Slovakia; (A.B.); (M.B.); (A.S.); (M.T.)
- Department of Biochemistry and Biotechnology, Faculty of Biotechnology and Food Science, Slovak University of Agriculture in Nitra, Tr. A. Hlinku 2, 949 76 Nitra, Slovakia;
- Correspondence: (J.V.); (P.C.); Tel.: +421-37-654-6600 (J.V.); +421-37-641-4274 (P.C.)
| | - Andrej Baláži
- NPPC, Research Institute for Animal Production Nitra, Institute of Farm Animal Genetics and Reproduction, Hlohovecká 2, 951 41 Lužianky, Slovakia; (A.B.); (M.B.); (A.S.); (M.T.)
| | - Miroslav Bauer
- NPPC, Research Institute for Animal Production Nitra, Institute of Farm Animal Genetics and Reproduction, Hlohovecká 2, 951 41 Lužianky, Slovakia; (A.B.); (M.B.); (A.S.); (M.T.)
- Department of Botany and Genetics, Faculty of Natural Sciences, Constantine the Philosopher University in Nitra, Nábrežie mládeže 91, 949 74 Nitra, Slovakia
| | - Andrea Svoradová
- NPPC, Research Institute for Animal Production Nitra, Institute of Farm Animal Genetics and Reproduction, Hlohovecká 2, 951 41 Lužianky, Slovakia; (A.B.); (M.B.); (A.S.); (M.T.)
| | - Mária Tirpáková
- Department of Biochemistry and Biotechnology, Faculty of Biotechnology and Food Science, Slovak University of Agriculture in Nitra, Tr. A. Hlinku 2, 949 76 Nitra, Slovakia;
- AgroBioTech Research Center, Slovak University of Agriculture in Nitra, Tr. A. Hlinku 2, 949 76 Nitra, Slovakia
| | - Marián Tomka
- NPPC, Research Institute for Animal Production Nitra, Institute of Farm Animal Genetics and Reproduction, Hlohovecká 2, 951 41 Lužianky, Slovakia; (A.B.); (M.B.); (A.S.); (M.T.)
| | - Peter Chrenek
- NPPC, Research Institute for Animal Production Nitra, Institute of Farm Animal Genetics and Reproduction, Hlohovecká 2, 951 41 Lužianky, Slovakia; (A.B.); (M.B.); (A.S.); (M.T.)
- Department of Biochemistry and Biotechnology, Faculty of Biotechnology and Food Science, Slovak University of Agriculture in Nitra, Tr. A. Hlinku 2, 949 76 Nitra, Slovakia;
- Correspondence: (J.V.); (P.C.); Tel.: +421-37-654-6600 (J.V.); +421-37-641-4274 (P.C.)
| |
Collapse
|
40
|
Abdelgawad ME, Desterke C, Uzan G, Naserian S. Single-cell transcriptomic profiling and characterization of endothelial progenitor cells: new approach for finding novel markers. Stem Cell Res Ther 2021; 12:145. [PMID: 33627177 PMCID: PMC7905656 DOI: 10.1186/s13287-021-02185-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 01/24/2021] [Indexed: 12/14/2022] Open
Abstract
Background Endothelial progenitor cells (EPCs) are promising candidates for the cellular therapy of peripheral arterial and cardiovascular diseases. However, hitherto there is no specific marker(s) defining precisely EPCs. Herein, we are proposing a new in silico approach for finding novel EPC markers. Methods We assembled five groups of chosen EPC-related genes/factors using PubMed literature and Gene Ontology databases. This shortened database of EPC factors was fed into publically published transcriptome matrix to compare their expression between endothelial colony-forming cells (ECFCs), HUVECs, and two adult endothelial cell types (ECs) from the skin and adipose tissue. Further, the database was used for functional enrichment on Mouse Phenotype database and protein-protein interaction network analyses. Moreover, we built a digital matrix of healthy donors’ PBMCs (33 thousand single-cell transcriptomes) and analyzed the expression of these EPC factors. Results Transcriptome analyses showed that BMP2, 4, and ephrinB2 were exclusively highly expressed in EPCs; the expression of neuropilin-1 and VEGF-C were significantly higher in EPCs and HUVECs compared with other ECs; Notch 1 was highly expressed in EPCs and skin-ECs; MIR21 was highly expressed in skin-ECs; PECAM-1 was significantly higher in EPCs and adipose ECs. Moreover, functional enrichment of EPC-related genes on Mouse Phenotype and STRING protein database has revealed significant relations between chosen EPC factors and endothelial and vascular functions, development, and morphogenesis, where ephrinB2, BMP2, and BMP4 were highly expressed in EPCs and were connected to abnormal vascular functions. Single-cell RNA-sequencing analyses have revealed that among the EPC-regulated markers in transcriptome analyses, (i) ICAM1 and Endoglin were weekly expressed in the monocyte compartment of the peripheral blood; (ii) CD163 and CD36 were highly expressed in the CD14+ monocyte compartment whereas CSF1R was highly expressed in the CD16+ monocyte compartment, (iii) L-selectin and IL6R were globally expressed in the lymphoid/myeloid compartments, and (iv) interestingly, PLAUR/UPAR and NOTCH2 were highly expressed in both CD14+ and CD16+ monocytic compartments. Conclusions The current study has identified novel EPC markers that could be used for better characterization of EPC subpopulation in adult peripheral blood and subsequent usage of EPCs for various cell therapy and regenerative medicine applications.
Collapse
Affiliation(s)
- Mohamed Essameldin Abdelgawad
- Biochemistry & Molecular Biotechnology Division, Chemistry Department, Faculty of Science; Innovative Cellular Microenvironment Optimization Platform (ICMOP), Helwan University, Cairo, Egypt. .,Inserm UMR-S-MD 1197, Hôpital Paul Brousse - Bâtiment Lavoisier, 12-14 avenue Paul Vaillant Couturier, 94800, Villejuif, France. .,Paris-Saclay University, Villejuif, France.
| | - Christophe Desterke
- Paris-Saclay University, Villejuif, France.,Inserm UMR-S-MD A9, Hôpital Paul Brousse, Villejuif, France
| | - Georges Uzan
- Inserm UMR-S-MD 1197, Hôpital Paul Brousse - Bâtiment Lavoisier, 12-14 avenue Paul Vaillant Couturier, 94800, Villejuif, France.,Paris-Saclay University, Villejuif, France
| | - Sina Naserian
- Inserm UMR-S-MD 1197, Hôpital Paul Brousse - Bâtiment Lavoisier, 12-14 avenue Paul Vaillant Couturier, 94800, Villejuif, France. .,Paris-Saclay University, Villejuif, France. .,CellMedEx, Saint Maur des Fossés, France.
| |
Collapse
|
41
|
Cai Q, Liao W, Xue F, Wang X, Zhou W, Li Y, Zeng W. Selection of different endothelialization modes and different seed cells for tissue-engineered vascular graft. Bioact Mater 2021; 6:2557-2568. [PMID: 33665496 PMCID: PMC7887299 DOI: 10.1016/j.bioactmat.2020.12.021] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 12/09/2020] [Accepted: 12/21/2020] [Indexed: 02/06/2023] Open
Abstract
Tissue-engineered vascular grafts (TEVGs) have enormous potential for vascular replacement therapy. However, thrombosis and intimal hyperplasia are important problems associated with TEVGs especially small diameter TEVGs (<6 mm) after transplantation. Endothelialization of TEVGs is a key point to prevent thrombosis. Here, we discuss different types of endothelialization and different seed cells of tissue-engineered vascular grafts. Meanwhile, endothelial heterogeneity is also discussed. Based on it, we provide a new perspective for selecting suitable types of endothelialization and suitable seed cells to improve the long-term patency rate of tissue-engineered vascular grafts with different diameters and lengths.
The material, diameter and length of tissue-engineered vascular graft are all key factors affecting its long-term patency. Endothelialization strategies should consider the different diameters and lengths of tissue-engineered vascular grafts. Cell heterogeneity and tissue heterogeneity should be considered in the application of seed cells.
Collapse
Affiliation(s)
- Qingjin Cai
- Department of Cell Biology, Third Military Medical University, Chongqing, 400038, China
| | - Wanshan Liao
- Department of Cell Biology, Third Military Medical University, Chongqing, 400038, China
| | - Fangchao Xue
- Department of Cell Biology, Third Military Medical University, Chongqing, 400038, China
| | - Xiaochen Wang
- Department of Cell Biology, Third Military Medical University, Chongqing, 400038, China
| | - Weiming Zhou
- Department of Cell Biology, Third Military Medical University, Chongqing, 400038, China
| | - Yanzhao Li
- State Key Laboratory of Trauma, Burn and Combined Injury, Chongqing, China
| | - Wen Zeng
- Department of Cell Biology, Third Military Medical University, Chongqing, 400038, China.,State Key Laboratory of Trauma, Burn and Combined Injury, Chongqing, China.,Departments of Neurology, Southwest Hospital, Third Military Medical University, Chongqing, China
| |
Collapse
|
42
|
Mallis P, Kostakis A, Stavropoulos-Giokas C, Michalopoulos E. Future Perspectives in Small-Diameter Vascular Graft Engineering. Bioengineering (Basel) 2020; 7:160. [PMID: 33321830 PMCID: PMC7763104 DOI: 10.3390/bioengineering7040160] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 12/04/2020] [Accepted: 12/09/2020] [Indexed: 02/07/2023] Open
Abstract
The increased demands of small-diameter vascular grafts (SDVGs) globally has forced the scientific society to explore alternative strategies utilizing the tissue engineering approaches. Cardiovascular disease (CVD) comprises one of the most lethal groups of non-communicable disorders worldwide. It has been estimated that in Europe, the healthcare cost for the administration of CVD is more than 169 billion €. Common manifestations involve the narrowing or occlusion of blood vessels. The replacement of damaged vessels with autologous grafts represents one of the applied therapeutic approaches in CVD. However, significant drawbacks are accompanying the above procedure; therefore, the exploration of alternative vessel sources must be performed. Engineered SDVGs can be produced through the utilization of non-degradable/degradable and naturally derived materials. Decellularized vessels represent also an alternative valuable source for the development of SDVGs. In this review, a great number of SDVG engineering approaches will be highlighted. Importantly, the state-of-the-art methodologies, which are currently employed, will be comprehensively presented. A discussion summarizing the key marks and the future perspectives of SDVG engineering will be included in this review. Taking into consideration the increased number of patients with CVD, SDVG engineering may assist significantly in cardiovascular reconstructive surgery and, therefore, the overall improvement of patients' life.
Collapse
Affiliation(s)
- Panagiotis Mallis
- Hellenic Cord Blood Bank, Biomedical Research Foundation Academy of Athens, 4 Soranou Ephessiou Street, 115 27 Athens, Greece; (C.S.-G.); (E.M.)
| | - Alkiviadis Kostakis
- Center of Experimental Surgery and Translational Research, Biomedical Research Foundation Academy of Athens, 4 Soranou Ephessiou Street, 115 27 Athens, Greece;
| | - Catherine Stavropoulos-Giokas
- Hellenic Cord Blood Bank, Biomedical Research Foundation Academy of Athens, 4 Soranou Ephessiou Street, 115 27 Athens, Greece; (C.S.-G.); (E.M.)
| | - Efstathios Michalopoulos
- Hellenic Cord Blood Bank, Biomedical Research Foundation Academy of Athens, 4 Soranou Ephessiou Street, 115 27 Athens, Greece; (C.S.-G.); (E.M.)
| |
Collapse
|
43
|
Cellular remodeling of fibrotic conduit as vascular graft. Biomaterials 2020; 268:120565. [PMID: 33310678 DOI: 10.1016/j.biomaterials.2020.120565] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Revised: 11/12/2020] [Accepted: 11/20/2020] [Indexed: 02/07/2023]
Abstract
The replacement of small-diameter arteries remains an unmet clinical need. Here we investigated the cellular remodeling of fibrotic conduits as vascular grafts. The formation of fibrotic conduit around subcutaneously implanted mandrels involved not only fibroblasts but also the trans-differentiation of inflammatory cells such as macrophages into fibroblastic cells, as shown by genetic lineage tracing. When fibrotic conduits were implanted as vascular grafts, the patency was low, and many fibrotic cells were found in neointima. Decellularization and anti-thrombogenic coating of fibrotic conduits produced highly patent autografts that remodeled into neoarteries, offering an effective approach to obtain autografts for clinical therapy. While autografts recruited mostly anti-inflammatory macrophages for constructive remodeling, allogenic DFCs had more T cells and pro-inflammatory macrophages and lower patency. Endothelial progenitors and endothelial migration were observed during endothelialization. Cell infiltration into DFCs was more efficient than decellularized arteries, and infiltrated cells remodeled the matrix and differentiated into smooth muscle cells (SMCs). This work provides insight into the remodeling of fibrotic conduits, autologous DFCs and allogenic DFCs, and will have broad impact on using fibrotic matrix for regenerative engineering.
Collapse
|
44
|
Cheng J, Li J, Cai Z, Xing Y, Wang C, Guo L, Gu Y. Decellularization of porcine carotid arteries using low-concentration sodium dodecyl sulfate. Int J Artif Organs 2020; 44:497-508. [PMID: 33222583 DOI: 10.1177/0391398820975420] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
BACKGROUND The decellularized scaffold is a promising material for producing tissue-engineered vascular grafts (TEVGs) because of its complex, native-like three-dimensional structure and mechanical properties. Sodium dodecyl sulfate (SDS), one of the most commonly used decellularization reagents, appears to be more effective than other detergents for removing cells from dense tissues. The concentrations of SDS used in previous studies and their effects on decellularization are not consistent. METHODS In this study, porcine carotid arteries were decellularized using detergent-based protocols using Triton X-100 followed by SDS at different concentrations and exposing time. Cell removal efficiency and composition were evaluated by histological analysis, and DNA and collagen quantification. Ultrastructure, mechanical properties, pore size distribution, and in vivo biocompatibility of decellularized arteries were also evaluated. RESULTS The DNA content of decellularized scaffolds treated with 0.3% SDS for 72 h or 0.5% SDS for 48 h was significantly less than that treated with 1% SDS for 30 h. There was a significant loss of soluble collagen after treatment with 1% SDS relative to native arteries. The extensive loss of elastin and glycosaminoglycans was observed in decellularized arteries treated with 0.5% SDS or 1% SDS. The basement membrane and biomechanics were also damaged by these two protocols. Moreover, decellularized scaffolds became more porous with many large pores after treatment with 0.3% SDS. CONCLUSION Low-concentration SDS could be a suitable choice for artery decellularization. Decellularized porcine carotid arteries, prepared using Triton X-100 followed by 0.3% SDS, may be a promising biological scaffold for TEVGs.
Collapse
Affiliation(s)
- Jin Cheng
- Department of Vascular Surgery, Xuanwu Hospital, Capital Medical University, Beijing, P.R. China
| | - Ji Li
- Department of Vascular Surgery, Xuanwu Hospital, Capital Medical University, Beijing, P.R. China
| | - Zhiwen Cai
- Department of Vascular Surgery, Xuanwu Hospital, Capital Medical University, Beijing, P.R. China
| | - Yuehao Xing
- Department of Vascular Surgery, Xuanwu Hospital, Capital Medical University, Beijing, P.R. China
| | - Cong Wang
- Department of Vascular Surgery, Xuanwu Hospital, Capital Medical University, Beijing, P.R. China
| | - Lianrui Guo
- Department of Vascular Surgery, Xuanwu Hospital, Capital Medical University, Beijing, P.R. China
| | - Yongquan Gu
- Department of Vascular Surgery, Xuanwu Hospital, Capital Medical University, Beijing, P.R. China
| |
Collapse
|
45
|
Predeina AL, Dukhinova MS, Vinogradov VV. Bioreactivity of decellularized animal, plant, and fungal scaffolds: perspectives for medical applications. J Mater Chem B 2020; 8:10010-10022. [PMID: 33063072 DOI: 10.1039/d0tb01751e] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Numerous biomedical applications imply supportive materials to improve protective, antibacterial, and regenerative abilities upon surgical interventions, oncotherapy, regenerative medicine, and others. With the increasing variability of the possible sources, the materials of natural origin are among the safest and most accessible biomedical tools. Animal, plant, and fungal tissues can further undergo decellularization to improve their biocompatibility. Decellularized scaffolds lack the most reactive cellular material, nuclear and cytoplasmic components, that predominantly trigger immune responses. At the same time, the outstanding initial three-dimensional microarchitecture, biomechanical properties, and general composition of the scaffolds are preserved. These unique features make the scaffolds perfect ready-to-use platforms for various biomedical applications, implying cell growth and functionalization. Decellularized materials can be repopulated with various cells upon request, including epithelial, endothelial, muscle and neuronal cells, and applied for structural and functional biorepair within diverse biological sites, including the skin and musculoskeletal, cardiovascular, and central nervous systems. However, the molecular and cellular mechanisms behind scaffold and host tissue interactions remain not fully understood, which significantly restricts their integration into clinical practice. In this review, we address the essential aspects of decellularization, scaffold preparation techniques, and its biochemical composition and properties, which determine the biocompatibility and immunogenicity of the materials. With the integrated evaluation of the scaffold profile in living systems, decellularized animal, plant, and fungal scaffolds have the potential to become essential instruments for safe and controllable biomedical applications.
Collapse
|
46
|
Engineering an endothelialized, endocrine Neo-Pancreas: Evaluation of islet functionality in an ex vivo model. Acta Biomater 2020; 117:213-225. [PMID: 32949822 DOI: 10.1016/j.actbio.2020.09.022] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 09/08/2020] [Accepted: 09/10/2020] [Indexed: 12/15/2022]
Abstract
Islet-based recellularization of decellularized, repurposed rat livers may form a transplantable Neo-Pancreas. The aim of this study is the establishment of the necessary protocols, the evaluation of the organ structure and the analysis of the islet functionality ex vivo. After perfusion-based decellularization of rat livers, matrices were repopulated with endothelial cells and mesenchymal stromal cells, incubated for 8 days in a perfusion chamber, and finally repopulated on day 9 with intact rodent islets. Integrity and quality of re-endothelialization was assessed by histology and FITC-dextran perfusion assay. Functionality of the islets of Langerhans was determined on day 10 and day 12 via glucose stimulated insulin secretion. Blood gas analysis variables confirmed the stability of the perfusion cultivation. Histological staining showed that cells formed a monolayer inside the intact vascular structure. These findings were confirmed by electron microscopy. Islets infused via the bile duct could histologically be found in the parenchymal space. Adequate insulin secretion after glucose stimulation after 1-day and 3-day cultivation verified islet viability and functionality after the repopulation process. We provide the first proof-of-concept for the functionality of islets of Langerhans engrafted in a decellularized rat liver. Furthermore, a re-endothelialization step was implemented to provide implantability. This technique can serve as a bioengineered platform to generate implantable and functional endocrine Neo-Pancreases.
Collapse
|
47
|
Yang X, Meng Y, Han Z, Ye F, Wei L, Zong C. Mesenchymal stem cell therapy for liver disease: full of chances and challenges. Cell Biosci 2020; 10:123. [PMID: 33117520 PMCID: PMC7590738 DOI: 10.1186/s13578-020-00480-6] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 10/06/2020] [Indexed: 12/13/2022] Open
Abstract
Liver disease is a major health problem that endangers human health worldwide. Currently, whole organ allograft transplantation is the gold standard for the treatment of end-stage liver disease. A shortage of suitable organs, high costs and surgical complications limit the application of liver transplantation. Mesenchymal stem cell therapy has been considered as a promising alternative approach for end-stage liver disease. Some clinical trials have confirmed the effectiveness of MSC therapy for liver disease, but its application has not been promoted and approved. There are still many issues that should be solved prior to using MSC therapy in clinical applications. The types of liver disease that are most suitable for MSC application should be determined, and the preparation and engraftment of MSCs should be standardized. These may be bottlenecks that limit the use of MSCs. We investigated 22 completed and several ongoing clinical trials to discuss these questions from a clinical perspective. We also discussed the important mechanisms by which MSCs play a therapeutic role in liver disease. Finally, we also proposed novel prospective approaches that can improve the therapeutic effect of MSCs.
Collapse
Affiliation(s)
- Xue Yang
- Tumor Immunology and Gene Therapy Center, Shanghai Eastern Hepatobiliary Surgery Hospital, 225 Changhai Road, Shanghai, 200438 China
| | - Yan Meng
- Tumor Immunology and Gene Therapy Center, Shanghai Eastern Hepatobiliary Surgery Hospital, 225 Changhai Road, Shanghai, 200438 China
| | - Zhipeng Han
- Tumor Immunology and Gene Therapy Center, Shanghai Eastern Hepatobiliary Surgery Hospital, 225 Changhai Road, Shanghai, 200438 China
| | - Fei Ye
- Tumor Immunology and Gene Therapy Center, Shanghai Eastern Hepatobiliary Surgery Hospital, 225 Changhai Road, Shanghai, 200438 China
| | - Lixin Wei
- Tumor Immunology and Gene Therapy Center, Shanghai Eastern Hepatobiliary Surgery Hospital, 225 Changhai Road, Shanghai, 200438 China
| | - Chen Zong
- Tumor Immunology and Gene Therapy Center, Shanghai Eastern Hepatobiliary Surgery Hospital, 225 Changhai Road, Shanghai, 200438 China
| |
Collapse
|
48
|
Qian T, Gil DA, Guzman EC, Gastfriend BD, Tweed KE, Palecek SP, Skala MC. Adaptable pulsatile flow generated from stem cell-derived cardiomyocytes using quantitative imaging-based signal transduction. LAB ON A CHIP 2020; 20:3744-3756. [PMID: 33048070 PMCID: PMC7699819 DOI: 10.1039/d0lc00546k] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
Endothelial cells (EC) in vivo are continuously exposed to a mechanical microenvironment from blood flow, and fluidic shear stress plays an important role in EC behavior. New approaches to generate physiologically and pathologically relevant pulsatile flows are needed to understand EC behavior under different shear stress regimes. Here, we demonstrate an adaptable pump (Adapt-Pump) platform for generating pulsatile flows from human pluripotent stem cell-derived cardiac spheroids (CS) via quantitative imaging-based signal transduction. Pulsatile flows generated from the Adapt-Pump system can recapitulate unique CS contraction characteristics, accurately model responses to clinically relevant drugs, and simulate CS contraction changes in response to fluidic mechanical stimulation. We discovered that ECs differentiated under a long QT syndrome derived pathological pulsatile flow exhibit abnormal EC monolayer organization. This Adapt-Pump platform provides a powerful tool for modeling the cardiovascular system and improving our understanding of EC behavior under different mechanical microenvironments.
Collapse
Affiliation(s)
- Tongcheng Qian
- Morgridge Institute for Research, Madison, WI, 53715, USA
| | - Daniel A. Gil
- Morgridge Institute for Research, Madison, WI, 53715, USA
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | | | - Benjamin D. Gastfriend
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Kelsey E. Tweed
- Morgridge Institute for Research, Madison, WI, 53715, USA
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Sean P. Palecek
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Melissa C. Skala
- Morgridge Institute for Research, Madison, WI, 53715, USA
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, 53706, USA
| |
Collapse
|
49
|
Bacci C, Wong V, Barahona V, Merna N. Cardiac and lung endothelial cells in response to fluid shear stress on physiological matrix stiffness and composition. Microcirculation 2020; 28:e12659. [PMID: 32945052 DOI: 10.1111/micc.12659] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 08/17/2020] [Accepted: 09/07/2020] [Indexed: 12/17/2022]
Abstract
OBJECTIVE Preconditioning of endothelial cells from different vascular beds has potential value for re-endothelialization and implantation of engineered tissues. Understanding how substrate stiffness and composition affects tissue-specific cell response to shear stress will aid in successful endothelialization of engineered tissues. We developed a platform to test biomechanical and biochemical stimuli. METHODS A novel polydimethylsiloxane-based parallel plate flow chamber enabled application of laminar fluid shear stress of 2 dynes/cm2 for 12 hours to microvascular cardiac and lung endothelial cells cultured on cardiac and lung-derived extracellular matrix. Optical imaging of cells was used to quantify cell changes in cell alignment. Analysis of integrin expression was performed using flow cytometry. RESULTS Application of fluid shear stress caused the greatest cell alignment in cardiac endothelial cells seeded on polystyrene and lung endothelial cells on polydimethylsiloxane. This resulted in elongation of the lung endothelial cells. αv and β3 integrin expression decreased after application of shear stress in both cell types. CONCLUSION Substrate stiffness plays an important role in regulating tissue-specific endothelial response to shear stress, which may be due to differences in their native microenvironments. Furthermore, cardiac and lung endothelial cell response to shear stress was significantly regulated by the type of coating used.
Collapse
Affiliation(s)
- Cydnee Bacci
- Bioengineering Program, Fred DeMatteis School of Engineering and Applied Sciences, Hofstra University, Hempstead, NY, USA
| | - Vanessa Wong
- Bioengineering Program, Fred DeMatteis School of Engineering and Applied Sciences, Hofstra University, Hempstead, NY, USA
| | - Victor Barahona
- Bioengineering Program, Fred DeMatteis School of Engineering and Applied Sciences, Hofstra University, Hempstead, NY, USA
| | - Nick Merna
- Bioengineering Program, Fred DeMatteis School of Engineering and Applied Sciences, Hofstra University, Hempstead, NY, USA
| |
Collapse
|
50
|
Harper S, Hoff M, Skepper J, Davies S, Huguet E. Portal venous repopulation of decellularised rat liver scaffolds with syngeneic bone marrow stem cells. J Tissue Eng Regen Med 2020; 14:1502-1512. [PMID: 32808475 DOI: 10.1002/term.3117] [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: 11/26/2019] [Revised: 06/22/2020] [Accepted: 07/30/2020] [Indexed: 12/14/2022]
Abstract
Liver transplantation is the only life-saving treatment for end-stage liver failure but is limited by the organ shortage and consequences of immunosuppression. Repopulation of decellularised scaffolds with recipient cells provides a theoretical solution, allowing reliable and timely organ sourcing without the need for immunosuppression. Recellularisation of the vasculature of decellularised liver scaffolds was investigated as an essential prerequisite to the survival of other parenchymal components. Liver decellularisation was carried out by portal vein perfusion using a detergent-based solution. Decellularised scaffolds were placed in a sterile perfusion apparatus consisting of a sealed organ chamber, functioning at 37°C in normal atmospheric conditions. The scaffold was perfused via portal vein with culture medium. A total of 107 primary cultured bone marrow stem cells, selected by plastic adherence, were infused into the scaffold, after which repopulated scaffolds were perfused for up to 30 days. The cultured stem cells were assessed for key marker expression using fluorescence-activated cell sorting (FACS), and recellularised scaffolds were analysed by light, electron and immunofluorescence microscopy. Stem cells were engrafted in portal, sinusoidal and hepatic vein compartments, with cell alignment reminiscent of endothelium. Cell surface marker expression altered following engraftment, from haematopoietic to endothelial phenotype, and engrafted cells expressed sinusoidal endothelial endocytic receptors (mannose, Fc and stabilin receptors). These results represent one step towards complete recellularisation of the liver vasculature and progress towards the objective of generating transplantable neo-organs.
Collapse
Affiliation(s)
- Simon Harper
- Cambridge University, Department of Surgery, Addenbrookes Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Mekhola Hoff
- Cambridge University, Department of Surgery, Addenbrookes Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Jeremy Skepper
- Cambridge Advanced Imaging Centre, University of Cambridge, Cambridge, UK
| | - Susan Davies
- Cambridge University, Department of Histopathology, Addenbrookes Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Emmanuel Huguet
- Cambridge University, Department of Surgery, Addenbrookes Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| |
Collapse
|