1
|
Iqbal MZ, Riaz M, Biedermann T, Klar AS. Breathing new life into tissue engineering: exploring cutting-edge vascularization strategies for skin substitutes. Angiogenesis 2024; 27:587-621. [PMID: 38842751 PMCID: PMC11564345 DOI: 10.1007/s10456-024-09928-6] [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: 07/20/2023] [Accepted: 05/02/2024] [Indexed: 06/07/2024]
Abstract
Tissue-engineered skin substitutes (TESS) emerged as a new therapeutic option to improve skin transplantation. However, establishing an adequate and rapid vascularization in TESS is a critical factor for their clinical application and successful engraftment in patients. Therefore, several methods have been applied to improve the vascularization of skin substitutes including (i) modifying the structural and physicochemical properties of dermal scaffolds; (ii) activating biological scaffolds with growth factor-releasing systems or gene vectors; and (iii) developing prevascularized skin substitutes by loading scaffolds with capillary-forming cells. This review provides a detailed overview of the most recent and important developments in the vascularization strategies for skin substitutes. On the one hand, we present cell-based approaches using stem cells, microvascular fragments, adipose tissue derived stromal vascular fraction, endothelial cells derived from blood and skin as well as other pro-angiogenic stimulation methods. On the other hand, we discuss how distinct 3D bioprinting techniques and microfluidics, miRNA manipulation, cell sheet engineering and photosynthetic scaffolds like GelMA, can enhance skin vascularization for clinical applications. Finally, we summarize and discuss the challenges and prospects of the currently available vascularization techniques that may serve as a steppingstone to a mainstream application of skin tissue engineering.
Collapse
Affiliation(s)
- M Zohaib Iqbal
- Tissue Biology Research Unit, Department of Surgery, University Children's Hospital Zurich, Wagistrasse 12, CH-8952, Zurich, Switzerland
- Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland
- University of Zurich, Zurich, Switzerland
| | - Mahrukh Riaz
- Tissue Biology Research Unit, Department of Surgery, University Children's Hospital Zurich, Wagistrasse 12, CH-8952, Zurich, Switzerland
- Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland
- University of Zurich, Zurich, Switzerland
| | - Thomas Biedermann
- Tissue Biology Research Unit, Department of Surgery, University Children's Hospital Zurich, Wagistrasse 12, CH-8952, Zurich, Switzerland
- Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland
- University of Zurich, Zurich, Switzerland
| | - Agnes S Klar
- Tissue Biology Research Unit, Department of Surgery, University Children's Hospital Zurich, Wagistrasse 12, CH-8952, Zurich, Switzerland.
- Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland.
- University of Zurich, Zurich, Switzerland.
| |
Collapse
|
2
|
Snyder Y, Jana S. Strategies for Development of Synthetic Heart Valve Tissue Engineering Scaffolds. PROGRESS IN MATERIALS SCIENCE 2023; 139:101173. [PMID: 37981978 PMCID: PMC10655624 DOI: 10.1016/j.pmatsci.2023.101173] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2023]
Abstract
The current clinical solutions, including mechanical and bioprosthetic valves for valvular heart diseases, are plagued by coagulation, calcification, nondurability, and the inability to grow with patients. The tissue engineering approach attempts to resolve these shortcomings by producing heart valve scaffolds that may deliver patients a life-long solution. Heart valve scaffolds serve as a three-dimensional support structure made of biocompatible materials that provide adequate porosity for cell infiltration, and nutrient and waste transport, sponsor cell adhesion, proliferation, and differentiation, and allow for extracellular matrix production that together contributes to the generation of functional neotissue. The foundation of successful heart valve tissue engineering is replicating native heart valve architecture, mechanics, and cellular attributes through appropriate biomaterials and scaffold designs. This article reviews biomaterials, the fabrication of heart valve scaffolds, and their in-vitro and in-vivo evaluations applied for heart valve tissue engineering.
Collapse
Affiliation(s)
- Yuriy Snyder
- Department of Bioengineering, University of Missouri, Columbia, MO 65211, USA
| | - Soumen Jana
- Department of Bioengineering, University of Missouri, Columbia, MO 65211, USA
| |
Collapse
|
3
|
Aziz R, Falanga M, Purenovic J, Mancini S, Lamberti P, Guida M. A Review on the Applications of Natural Biodegradable Nano Polymers in Cardiac Tissue Engineering. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:1374. [PMID: 37110959 PMCID: PMC10145986 DOI: 10.3390/nano13081374] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 04/12/2023] [Accepted: 04/13/2023] [Indexed: 06/19/2023]
Abstract
As cardiac diseases, which mostly result in heart failure, are increasing rapidly worldwide, heart transplantation seems the only solution for saving lives. However, this practice is not always possible due to several reasons, such as scarcity of donors, rejection of organs from recipient bodies, or costly medical procedures. In the framework of nanotechnology, nanomaterials greatly contribute to the development of these cardiovascular scaffolds as they provide an easy regeneration of the tissues. Currently, functional nanofibers can be used in the production of stem cells and in the regeneration of cells and tissues. The small size of nanomaterials, however, leads to changes in their chemical and physical characteristics that could alter their interaction and exposure to stem cells with cells and tissues. This article aims to review the naturally occurring biodegradable nanomaterials that are used in cardiovascular tissue engineering for the development of cardiac patches, vessels, and tissues. Moreover, this article also provides an overview of cell sources used for cardiac tissue engineering, explains the anatomy and physiology of the human heart, and explores the regeneration of cardiac cells and the nanofabrication approaches used in cardiac tissue engineering as well as scaffolds.
Collapse
Affiliation(s)
- Rabia Aziz
- Department of Information and Electrical Engineering and Applied Mathematics (DIEM), University of Salerno, 84084 Fisciano, Italy; (M.F.); (S.M.); (P.L.); (M.G.)
- Consiglio Nazionale Delle Ricerche (CNR)-Istituto Officina dei Materiali (IOM), Area Science Park Basovizza S.S. 14-Km. 163, 5-34149 Trieste, Italy
| | - Mariarosaria Falanga
- Department of Information and Electrical Engineering and Applied Mathematics (DIEM), University of Salerno, 84084 Fisciano, Italy; (M.F.); (S.M.); (P.L.); (M.G.)
| | - Jelena Purenovic
- Department of Physics and Materials, Faculty of Sciences at Cacak, University of Kragujevac, 32000 Cacak, Serbia;
| | - Simona Mancini
- Department of Information and Electrical Engineering and Applied Mathematics (DIEM), University of Salerno, 84084 Fisciano, Italy; (M.F.); (S.M.); (P.L.); (M.G.)
| | - Patrizia Lamberti
- Department of Information and Electrical Engineering and Applied Mathematics (DIEM), University of Salerno, 84084 Fisciano, Italy; (M.F.); (S.M.); (P.L.); (M.G.)
- Italian Interuniversity Research Center on Interaction between Electromagnetic Fields and Biosystems (ICEmB), Università Degli Studi di Genova, DITEN, Via all’Opera Pia 11/a, 16145 Genova, Italy
- Interdepartmental Research Centre for Nanomaterials and Nanotechnology at the University of Salerno (NanoMates), Department of Physics, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, Italy
| | - Michele Guida
- Department of Information and Electrical Engineering and Applied Mathematics (DIEM), University of Salerno, 84084 Fisciano, Italy; (M.F.); (S.M.); (P.L.); (M.G.)
- Italian Interuniversity Research Center on Interaction between Electromagnetic Fields and Biosystems (ICEmB), Università Degli Studi di Genova, DITEN, Via all’Opera Pia 11/a, 16145 Genova, Italy
| |
Collapse
|
4
|
Strategies for development of decellularized heart valve scaffolds for tissue engineering. Biomaterials 2022; 288:121675. [DOI: 10.1016/j.biomaterials.2022.121675] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 07/02/2022] [Accepted: 07/06/2022] [Indexed: 01/01/2023]
|
5
|
Marei I, Abu Samaan T, Al-Quradaghi MA, Farah AA, Mahmud SH, Ding H, Triggle CR. 3D Tissue-Engineered Vascular Drug Screening Platforms: Promise and Considerations. Front Cardiovasc Med 2022; 9:847554. [PMID: 35310996 PMCID: PMC8931492 DOI: 10.3389/fcvm.2022.847554] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2022] [Accepted: 02/03/2022] [Indexed: 12/12/2022] Open
Abstract
Despite the efforts devoted to drug discovery and development, the number of new drug approvals have been decreasing. Specifically, cardiovascular developments have been showing amongst the lowest levels of approvals. In addition, concerns over the adverse effects of drugs to the cardiovascular system have been increasing and resulting in failure at the preclinical level as well as withdrawal of drugs post-marketing. Besides factors such as the increased cost of clinical trials and increases in the requirements and the complexity of the regulatory processes, there is also a gap between the currently existing pre-clinical screening methods and the clinical studies in humans. This gap is mainly caused by the lack of complexity in the currently used 2D cell culture-based screening systems, which do not accurately reflect human physiological conditions. Cell-based drug screening is widely accepted and extensively used and can provide an initial indication of the drugs' therapeutic efficacy and potential cytotoxicity. However, in vitro cell-based evaluation could in many instances provide contradictory findings to the in vivo testing in animal models and clinical trials. This drawback is related to the failure of these 2D cell culture systems to recapitulate the human physiological microenvironment in which the cells reside. In the body, cells reside within a complex physiological setting, where they interact with and respond to neighboring cells, extracellular matrix, mechanical stress, blood shear stress, and many other factors. These factors in sum affect the cellular response and the specific pathways that regulate variable vital functions such as proliferation, apoptosis, and differentiation. Although pre-clinical in vivo animal models provide this level of complexity, cross species differences can also cause contradictory results from that seen when the drug enters clinical trials. Thus, there is a need to better mimic human physiological conditions in pre-clinical studies to improve the efficiency of drug screening. A novel approach is to develop 3D tissue engineered miniaturized constructs in vitro that are based on human cells. In this review, we discuss the factors that should be considered to produce a successful vascular construct that is derived from human cells and is both reliable and reproducible.
Collapse
Affiliation(s)
- Isra Marei
- Department of Pharmacology, Weill Cornell Medicine-Qatar, Doha, Qatar
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
- *Correspondence: Isra Marei
| | - Tala Abu Samaan
- Department of Pharmacology, Weill Cornell Medicine-Qatar, Doha, Qatar
| | | | - Asmaa A. Farah
- Department of Pharmacology, Weill Cornell Medicine-Qatar, Doha, Qatar
| | | | - Hong Ding
- Department of Pharmacology, Weill Cornell Medicine-Qatar, Doha, Qatar
| | - Chris R. Triggle
- Department of Pharmacology, Weill Cornell Medicine-Qatar, Doha, Qatar
- Chris R. Triggle
| |
Collapse
|
6
|
Mehta P, Rasekh M, Patel M, Onaiwu E, Nazari K, Kucuk I, Wilson PB, Arshad MS, Ahmad Z, Chang MW. Recent applications of electrical, centrifugal, and pressurised emerging technologies for fibrous structure engineering in drug delivery, regenerative medicine and theranostics. Adv Drug Deliv Rev 2021; 175:113823. [PMID: 34089777 DOI: 10.1016/j.addr.2021.05.033] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 05/11/2021] [Accepted: 05/31/2021] [Indexed: 12/16/2022]
Abstract
Advancements in technology and material development in recent years has led to significant breakthroughs in the remit of fiber engineering. Conventional methods such as wet spinning, melt spinning, phase separation and template synthesis have been reported to develop fibrous structures for an array of applications. However, these methods have limitations with respect to processing conditions (e.g. high processing temperatures, shear stresses) and production (e.g. non-continuous fibers). The materials that can be processed using these methods are also limited, deterring their use in practical applications. Producing fibrous structures on a nanometer scale, in sync with the advancements in nanotechnology is another challenge met by these conventional methods. In this review we aim to present a brief overview of conventional methods of fiber fabrication and focus on the emerging fiber engineering techniques namely electrospinning, centrifugal spinning and pressurised gyration. This review will discuss the fundamental principles and factors governing each fabrication method and converge on the applications of the resulting spun fibers; specifically, in the drug delivery remit and in regenerative medicine.
Collapse
Affiliation(s)
- Prina Mehta
- Leicester School of Pharmacy, De Montfort University, Leicester LE1 9BH, UK
| | - Manoochehr Rasekh
- College of Engineering, Design and Physical Sciences, Brunel University London, Middlesex UB8 3PH, UK
| | - Mohammed Patel
- Leicester School of Pharmacy, De Montfort University, Leicester LE1 9BH, UK
| | - Ekhoerose Onaiwu
- Leicester School of Pharmacy, De Montfort University, Leicester LE1 9BH, UK
| | - Kazem Nazari
- Leicester School of Pharmacy, De Montfort University, Leicester LE1 9BH, UK
| | - I Kucuk
- Institute of Nanotechnology, Gebze Technical University, 41400 Gebze, Turkey
| | - Philippe B Wilson
- School of Animal, Rural and Environmental Sciences, Nottingham Trent University, Brackenhurst Campus, Southwell NG25 0QF, UK
| | | | - Zeeshan Ahmad
- Leicester School of Pharmacy, De Montfort University, Leicester LE1 9BH, UK
| | - Ming-Wei Chang
- Nanotechnology and Integrated Bioengineering Centre, University of Ulster, Jordanstown Campus, Newtownabbey, Northern Ireland BT37 0QB, UK.
| |
Collapse
|
7
|
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
|
8
|
Chen J, Hendriks M, Chatzis A, Ramasamy SK, Kusumbe AP. Bone Vasculature and Bone Marrow Vascular Niches in Health and Disease. J Bone Miner Res 2020; 35:2103-2120. [PMID: 32845550 DOI: 10.1002/jbmr.4171] [Citation(s) in RCA: 88] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 07/21/2020] [Accepted: 08/05/2020] [Indexed: 12/20/2022]
Abstract
Bone vasculature and bone marrow vascular niches supply oxygen, nutrients, and secrete angiocrine factors required for the survival, maintenance, and self-renewal of stem and progenitor cells. In the skeletal system, vasculature creates nurturing niches for bone and blood-forming stem cells. Blood vessels regulate hematopoiesis and drive bone formation during development, repair, and regeneration. Dysfunctional vascular niches induce skeletal aging, bone diseases, and hematological disorders. Recent cellular and molecular characterization of the bone marrow microenvironment has provided unprecedented insights into the complexity, heterogeneity, and functions of the bone vasculature and vascular niches. The bone vasculature is composed of distinct vessel subtypes that differentially regulate osteogenesis, hematopoiesis, and disease conditions in bones. Further, bone marrow vascular niches supporting stem cells are often complex microenvironments involving multiple different cell populations and vessel subtypes. This review provides an overview of the emerging vascular cell heterogeneity in bone and the new roles of the bone vasculature and associated vascular niches in health and disease. © 2020 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Junyu Chen
- Tissue and Tumor Microenvironments Group, The Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Michelle Hendriks
- Institute of Clinical Sciences, Imperial College London, London, UK
- MRC London Institute of Medical Sciences, Imperial College London, London, UK
| | - Alexandros Chatzis
- Tissue and Tumor Microenvironments Group, The Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Saravana K Ramasamy
- Institute of Clinical Sciences, Imperial College London, London, UK
- MRC London Institute of Medical Sciences, Imperial College London, London, UK
| | - Anjali P Kusumbe
- Tissue and Tumor Microenvironments Group, The Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| |
Collapse
|
9
|
Abstract
The field of Tissue Engineering and Regenerative Medicine has evolved rapidly over the past thirty years. This review will summarize its history, current status and direction through the lens of clinical need, its progress through science in the laboratory and application back into patients. We can take pride in the fact that much effort and progress began with the surgical problems of children and that many surgeons in the pediatric surgical specialties have become pioneers and investigators in this new field of science, engineering, and medicine. Although the field has yet to fulfill its great promise, there have been several examples where a therapy has progressed from the first idea to human application within a short span of time and, in many cases, it has been applied in the surgical care of children.
Collapse
|
10
|
Abstract
Vascularization is a major hurdle in complex tissue and organ engineering. Tissues greater than 200 μm in diameter cannot rely on simple diffusion to obtain nutrients and remove waste. Therefore, an integrated vascular network is required for clinical translation of engineered tissues. Microvessels have been described as <150 μm in diameter, but clinically they are defined as <1 mm. With new advances in super microsurgery, vessels less than 1 mm can be anastomosed to the recipient circulation. However, this technical advancement still relies on the creation of a stable engineered microcirculation that is amenable to surgical manipulation and is readily perfusable. Microvascular engineering lays on the crossroads of microfabrication, microfluidics, and tissue engineering strategies that utilize various cellular constituents. Early research focused on vascularization by co-culture and cellular interactions, with the addition of angiogenic growth factors to promote vascular growth. Since then, multiple strategies have been utilized taking advantage of innovations in additive manufacturing, biomaterials, and cell biology. However, the anatomy and dynamics of native blood vessels has not been consistently replicated. Inconsistent results can be partially attributed to cell sourcing which remains an enigma for microvascular engineering. Variations of endothelial cells, endothelial progenitor cells, and stem cells have all been used for microvascular network fabrication along with various mural cells. As each source offers advantages and disadvantages, there continues to be a lack of consensus. Furthermore, discord may be attributed to incomplete understanding about cell isolation and characterization without considering the microvascular architecture of the desired tissue/organ.
Collapse
|
11
|
Yao T, Chen H, Baker MB, Moroni L. Effects of Fiber Alignment and Coculture with Endothelial Cells on Osteogenic Differentiation of Mesenchymal Stromal Cells. Tissue Eng Part C Methods 2019; 26:11-22. [PMID: 31774033 DOI: 10.1089/ten.tec.2019.0232] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Vascularization is a critical process during bone regeneration. The lack of vascular networks leads to insufficient oxygen and nutrients supply, which compromises the survival of regenerated bone. One strategy for improving the survival and osteogenesis of tissue-engineered bone grafts involves the coculture of endothelial cells (ECs) with mesenchymal stromal cells (MSCs). Moreover, bone regeneration is especially challenging due to its unique structural properties with aligned topographical cues, with which stem cells can interact. Inspired by the aligned fibrillar nanostructures in human cancellous bone, we fabricated polycaprolactone (PCL) electrospun fibers with aligned and random morphology, cocultured human MSCs with human umbilical vein ECs (HUVECs), and finally investigated how these two factors modulate osteogenic differentiation of human MSCs (hMSCs). After optimizing cell ratio, a hMSCs/HUVECs ratio (90:10) was considered to be the best combination for osteogenic differentiation. Coculture results showed that hMSCs and HUVECs adhered to and proliferated well on both scaffolds. The aligned structure of PCL fibers strongly influenced the morphology and orientation of hMSCs and HUVECs; however, fiber alignment was observed to not affect alkaline phosphate (ALP) activity or mineralization of hMSCs compared with random scaffolds. More importantly, cocultured cells on both random and aligned scaffolds had significantly higher ALP activities than monoculture groups, which indicated that coculture with HUVECs provided a larger relative contribution to the osteogenesis of hMSCs compared with fiber alignment. Taken together, we conclude that coculture of hMSCs with ECs is an effective strategy to promote osteogenesis on electrospun scaffolds, and aligned fibers could be introduced to regenerate bone tissues with oriented topography without significant deleterious effects on hMSCs differentiation. This study shows the ability to grow oriented tissue-engineered cocultures with significant increases in osteogenesis over monoculture conditions. Impact statement This work demonstrates an effective method of enhancing osteogenesis of mesenchymal stromal cells on electrospun scaffolds through coculturing with endothelial cells. Furthermore, we provide the optimized conditions for cocultures on electrospun fibrous scaffolds and engineered bone tissues with oriented topography on aligned fibers. This study demonstrates promising findings for growing oriented tissue-engineered cocultures with significant increase in osteogenesis over monoculture conditions.
Collapse
Affiliation(s)
- Tianyu Yao
- Complex Tissue Regeneration Department, MERLN Institute for Technology Inspired Regenerative Medicine, Maastricht University, Maastricht, The Netherlands
| | - Honglin Chen
- Institute for Life Science, School of Medicine, South China University of Technology, Guangzhou, China
| | - Matthew B Baker
- Complex Tissue Regeneration Department, MERLN Institute for Technology Inspired Regenerative Medicine, Maastricht University, Maastricht, The Netherlands
| | - Lorenzo Moroni
- Complex Tissue Regeneration Department, MERLN Institute for Technology Inspired Regenerative Medicine, Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
12
|
Rademakers T, Horvath JM, van Blitterswijk CA, LaPointe VL. Oxygen and nutrient delivery in tissue engineering: Approaches to graft vascularization. J Tissue Eng Regen Med 2019; 13:1815-1829. [PMID: 31310055 PMCID: PMC6852121 DOI: 10.1002/term.2932] [Citation(s) in RCA: 116] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 06/13/2019] [Accepted: 07/01/2019] [Indexed: 12/29/2022]
Abstract
The field of tissue engineering is making great strides in developing replacement tissue grafts for clinical use, marked by the rapid development of novel biomaterials, their improved integration with cells, better-directed growth and differentiation of cells, and improved three-dimensional tissue mass culturing. One major obstacle that remains, however, is the lack of graft vascularization, which in turn renders many grafts to fail upon clinical application. With that, graft vascularization has turned into one of the holy grails of tissue engineering, and for the majority of tissues, it will be imperative to achieve adequate vascularization if tissue graft implantation is to succeed. Many different approaches have been developed to induce or augment graft vascularization, both in vitro and in vivo. In this review, we highlight the importance of vascularization in tissue engineering and outline various approaches inspired by both biology and engineering to achieve and augment graft vascularization.
Collapse
Affiliation(s)
- Timo Rademakers
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityMaastrichtThe Netherlands
| | - Judith M. Horvath
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityMaastrichtThe Netherlands
| | - Clemens A. van Blitterswijk
- Complex Tissue Regeneration, MERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityMaastrichtThe Netherlands
| | - Vanessa L.S. LaPointe
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityMaastrichtThe Netherlands
| |
Collapse
|
13
|
Meivar-Levy I, Zoabi F, Nardini G, Manevitz-Mendelson E, Leichner GS, Zadok O, Gurevich M, Mor E, Dima S, Popescu I, Barzilai A, Ferber S, Greenberger S. The role of the vasculature niche on insulin-producing cells generated by transdifferentiation of adult human liver cells. Stem Cell Res Ther 2019; 10:53. [PMID: 30760321 PMCID: PMC6373031 DOI: 10.1186/s13287-019-1157-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 01/10/2019] [Accepted: 01/27/2019] [Indexed: 02/07/2023] Open
Abstract
Background Insulin-dependent diabetes is a multifactorial disorder that could be theoretically cured by functional pancreatic islets and insulin-producing cell (IPC) implantation. Regenerative medicine approaches include the potential for growing tissues and organs in the laboratory and transplanting them when the body cannot heal itself. However, several obstacles remain to be overcome in order to bring regenerative medicine approach for diabetes closer to its clinical implementation; the cells generated in vitro are typically of heterogenic and immature nature and the site of implantation should be readily vascularized for the implanted cells to survive in vivo. The present study addresses these two limitations by analyzing the effect of co-implanting IPCs with vasculature promoting cells in an accessible site such as subcutaneous. Secondly, it analyzes the effects of reconstituting the in vivo environment in vitro on the maturation and function of insulin-producing cells. Methods IPCs that are generated by the transdifferentiation of human liver cells are exposed to the paracrine effects of endothelial colony-forming cells (ECFCs) and human bone marrow mesenchymal stem cells (MSCs), which are the “building blocks” of the blood vessels. The role of the vasculature on IPC function is analyzed upon subcutaneous implantation in vivo in immune-deficient rodents. The paracrine effects of vasculature on IPC maturation are analyzed in culture. Results Co-implantation of MSCs and ECFCs with IPCs led to doubling the survival rates and a threefold increase in insulin production, in vivo. ECFC and MSC co-culture as well as conditioned media of co-cultures resulted in a significant increased expression of pancreatic-specific genes and an increase in glucose-regulated insulin secretion, compared with IPCs alone. Mechanistically, we demonstrate that ECFC and MSC co-culture increases the expression of CTGF and ACTIVINβα, which play a key role in pancreatic differentiation. Conclusions Vasculature is an important player in generating regenerative medicine approaches for diabetes. Vasculature displays a paracrine effect on the maturation of insulin-producing cells and their survival upon implantation. The reconstitution of the in vivo niche is expected to promote the liver-to-pancreas transdifferentiation and bringing this cell therapy approach closer to its clinical implementation. Electronic supplementary material The online version of this article (10.1186/s13287-019-1157-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Irit Meivar-Levy
- The Sheba Regenerative Medicine, Stem Cell and Tissue Engineering Center, Sheba Medical Center, Tel Hashomer, Israel. .,Dia-Cure, Institute of Medical Scientific Research Acad. Nicolae Cajal, University Titu Maiorescu, Bucharest, Romania.
| | - Fatima Zoabi
- The Sheba Regenerative Medicine, Stem Cell and Tissue Engineering Center, Sheba Medical Center, Tel Hashomer, Israel.,Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Gil Nardini
- Department of Plastic Surgery, Sheba Medical Center, Tel Hashomer, Israel
| | | | - Gil S Leichner
- The Department of Dermatology, Sheba Medical Center, Tel Hashomer, Israel
| | - Oranit Zadok
- The Sheba Regenerative Medicine, Stem Cell and Tissue Engineering Center, Sheba Medical Center, Tel Hashomer, Israel
| | - Michael Gurevich
- The Organ Transplantation Division, Schneider Children Medical Center, Petach Tikvah, Israel
| | - Eytan Mor
- The Organ Transplantation Division, Schneider Children Medical Center, Petach Tikvah, Israel
| | - Simona Dima
- Dia-Cure, Institute of Medical Scientific Research Acad. Nicolae Cajal, University Titu Maiorescu, Bucharest, Romania.,Center of Excellence in Translational Medicine - Fundeni Clinical Institute, Bucharest, Romania.,Center of Digestive Diseases and Liver Transplantation, Fundeni Clinical Institute, Bucharest, Romania
| | - Irinel Popescu
- Dia-Cure, Institute of Medical Scientific Research Acad. Nicolae Cajal, University Titu Maiorescu, Bucharest, Romania.,Center of Excellence in Translational Medicine - Fundeni Clinical Institute, Bucharest, Romania.,Center of Digestive Diseases and Liver Transplantation, Fundeni Clinical Institute, Bucharest, Romania
| | - Aviv Barzilai
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel.,The Department of Dermatology, Sheba Medical Center, Tel Hashomer, Israel
| | - Sarah Ferber
- The Sheba Regenerative Medicine, Stem Cell and Tissue Engineering Center, Sheba Medical Center, Tel Hashomer, Israel.,Dia-Cure, Institute of Medical Scientific Research Acad. Nicolae Cajal, University Titu Maiorescu, Bucharest, Romania.,Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Shoshana Greenberger
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel.,The Department of Dermatology, Sheba Medical Center, Tel Hashomer, Israel
| |
Collapse
|
14
|
Wang K, Lin RZ, Melero-Martin JM. Bioengineering human vascular networks: trends and directions in endothelial and perivascular cell sources. Cell Mol Life Sci 2019; 76:421-439. [PMID: 30315324 PMCID: PMC6349493 DOI: 10.1007/s00018-018-2939-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 10/04/2018] [Accepted: 10/08/2018] [Indexed: 12/13/2022]
Abstract
Tissue engineering holds great promise in regenerative medicine. However, the field of tissue engineering faces a myriad of difficulties. A major challenge is the necessity to integrate vascular networks into bioengineered constructs to enable physiological functions including adequate oxygenation, nutrient delivery, and removal of waste products. The last two decades have seen remarkable progress in our collective effort to bioengineer human-specific vascular networks. Studies have included both in vitro and in vivo investigations, and multiple methodologies have found varying degrees of success. What most approaches to bioengineer human vascular networks have in common, however, is the synergistic use of both (1) endothelial cells (ECs)-the cells used to line the lumen of the vascular structures and (2) perivascular cells-usually used to support EC function and provide perivascular stability to the networks. Here, we have highlighted trends in the use of various cellular sources over the last two decades of vascular network bioengineering research. To this end, we comprehensively reviewed all life science and biomedical publications available at the MEDLINE database up to 2018. Emphasis was put on selective studies that definitively used human ECs and were specifically related to bioengineering vascular networks. To facilitate this analysis, all papers were stratified by publication year and then analyzed according to their use of EC and perivascular cell types. This study provides an illustrating discussion on how each alternative source of cells has come to be used in the field. Our intention was to reveal trends and to provide new insights into the trajectory of vascular network bioengineering with regard to cellular sources.
Collapse
Affiliation(s)
- Kai Wang
- Department of Cardiac Surgery, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Surgery, Harvard Medical School, Boston, MA, 02115, USA
| | - Ruei-Zeng Lin
- Department of Cardiac Surgery, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Surgery, Harvard Medical School, Boston, MA, 02115, USA
| | - Juan M Melero-Martin
- Department of Cardiac Surgery, Boston Children's Hospital, Boston, MA, 02115, USA.
- Department of Surgery, Harvard Medical School, Boston, MA, 02115, USA.
- Harvard Stem Cell Institute, Cambridge, MA, 02138, USA.
| |
Collapse
|
15
|
Bioengineering the innate vasculature of complex organs: what have we learned so far. Curr Opin Organ Transplant 2018; 23:657-663. [DOI: 10.1097/mot.0000000000000577] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
16
|
Dhulekar J, Simionescu A. Challenges in vascular tissue engineering for diabetic patients. Acta Biomater 2018; 70:25-34. [PMID: 29396167 PMCID: PMC5871600 DOI: 10.1016/j.actbio.2018.01.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 01/06/2018] [Accepted: 01/09/2018] [Indexed: 12/16/2022]
Abstract
Hyperglycemia and dyslipidemia coexist in diabetes and result in inflammation, degeneration, and impaired tissue remodeling, processes which are not conducive to the desired integration of tissue engineered products into the surrounding tissues. There are several challenges for vascular tissue engineering such as non-thrombogenicity, adequate burst pressure and compliance, suturability, appropriate remodeling responses, and vasoactivity, but, under diabetic conditions, an additional challenge needs to be considered: the aggressive oxidative environment generated by the high glucose and lipid concentrations that lead to the formation of advanced glycation end products (AGEs) in the vascular wall. Extracellular matrix-based scaffolds have adequate physical properties and are biocompatible, however, these scaffolds are altered in diabetes by the formation AGEs and impaired collagen degradation, consequently increasing vascular wall stiffness. In addition, vascular cells detect and respond to altered stimuli from the matrix by pathological remodeling of the vascular wall. Due to the immunomodulatory effects of mesenchymal stem cells (MSCs), they are frequently used in tissue engineering in order to protect the scaffolds from inflammation. MSCs together with antioxidant treatments of the scaffolds are expected to protect the vascular grafts from diabetes-induced alterations. In conclusion, as one of the most daunting environments that could damage the ECM and its interaction with cells is progressively built in diabetes, we recommend that cells and scaffolds used in vascular tissue engineering for diabetic patients are tested in diabetic animal models, in order to obtain valuable results regarding their resistance to diabetic adversities. STATEMENT OF SIGNIFICANCE Almost 25 million Americans have diabetes, characterized by high levels of blood sugar that binds to tissues and disturbs the function of cardiovascular structures. Therefore, patients with diabetes have a high risk of cardiovascular diseases. Surgery is required to replace diseased arteries with implants, but these fail after 5-10 years because they are made of non-living materials, not resistant to diabetes. New tissue engineering materials are developed, based on the patients' own stem cells, isolated from fat, and added to extracellular matrix-based scaffolds. Our main concern is that diabetes could damage the tissue-like implants. Thus we review studies related to the effect of diabetes on tissue components and recommend antioxidant treatments to increase the resistance of implants to diabetes.
Collapse
|
17
|
Recent Progress in Vascular Tissue-Engineered Blood Vessels. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1064:123-144. [PMID: 30471030 DOI: 10.1007/978-981-13-0445-3_8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cardiovascular disease is the number one cause of death in the U.S and results in the loss of approximately one million lives and more than 400 billion U.S. dollars for treatments every year. Recently, tissue engineered blood vessels have been studied and developed as promising replacements for treatment with autologous veins. Here, we summarize the cell sources and methods to make tissue-engineered blood vessels (TEBVs), the recent progress in TEBV related research, and also the recent progress in TEBV related clinical studies.
Collapse
|
18
|
Generali M, Kehl D, Capulli AK, Parker KK, Hoerstrup SP, Weber B. Comparative analysis of poly-glycolic acid-based hybrid polymer starter matrices for in vitro tissue engineering. Colloids Surf B Biointerfaces 2017; 158:203-212. [DOI: 10.1016/j.colsurfb.2017.06.046] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Revised: 06/23/2017] [Accepted: 06/28/2017] [Indexed: 12/11/2022]
|
19
|
Ravishankar P, Zeballos MA, Balachandran K. Isolation of Endothelial Progenitor Cells from Human Umbilical Cord Blood. J Vis Exp 2017. [PMID: 28994769 DOI: 10.3791/56021] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
The existence of endothelial progenitor cells (EPCs) in peripheral blood and its involvement in vasculogenesis was first reported by Ashara and colleagues1. Later, others documented the existence of similar types of EPCs originating from bone marrow2,3. More recently, Yoder and Ingram showed that EPCs derived from umbilical cord blood had a higher proliferative potential compared to ones isolated from adult peripheral blood4,5,6. Apart from being involved in postnatal vasculogenesis, EPCs have also shown promise as a cell source for creating tissue-engineered vascular and heart valve constructs7,8. Various isolation protocols exist, some of which involve the cell sorting of mononuclear cells (MNCs) derived from the sources mentioned earlier with the help of endothelial and hematopoietic markers, or culturing these MNCs with specialized endothelial growth medium, or a combination of these techniques9. Here, we present a protocol for the isolation and culture of EPCs using specialized endothelial medium supplemented with growth factors, without the use of immunosorting, followed by the characterization of the isolated cells using Western blotting and immunostaining.
Collapse
|
20
|
Peters EB. Endothelial Progenitor Cells for the Vascularization of Engineered Tissues. TISSUE ENGINEERING PART B-REVIEWS 2017; 24:1-24. [PMID: 28548628 DOI: 10.1089/ten.teb.2017.0127] [Citation(s) in RCA: 114] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Self-assembled microvasculature from cocultures of endothelial cells (ECs) and stromal cells has significantly advanced efforts to vascularize engineered tissues by enhancing perfusion rates in vivo and producing investigative platforms for microvascular morphogenesis in vitro. However, to clinically translate prevascularized constructs, the issue of EC source must be resolved. Endothelial progenitor cells (EPCs) can be noninvasively supplied from the recipient through adult peripheral and umbilical cord blood, as well as derived from induced pluripotent stem cells, alleviating antigenicity issues. EPCs can also differentiate into all tissue endothelium, and have demonstrated potential for therapeutic vascularization. Yet, EPCs are not the standard EC choice to vascularize tissue constructs in vitro. Possible reasons include unresolved issues with EPC identity and characterization, as well as uncertainty in the selection of coculture, scaffold, and culture media combinations that promote EPC microvessel formation. This review addresses these issues through a summary of EPC vascular biology and the effects of tissue engineering design parameters upon EPC microvessel formation. Also included are perspectives to integrate EPCs with emerging technologies to produce functional, organotypic vascularized tissues.
Collapse
Affiliation(s)
- Erica B Peters
- Department of Chemical and Biological Engineering, University of Colorado , Boulder, Colorado
| |
Collapse
|
21
|
Ran J, Hu Y, Le H, Chen Y, Zheng Z, Chen X, Yin Z, Yan R, Jin Z, Tang C, Huang J, Gu Y, Xu L, Qian S, Zhang W, Heng BC, Dominique P, Chen W, Wu L, Shen W, Ouyang H. Ectopic tissue engineered ligament with silk collagen scaffold for ACL regeneration: A preliminary study. Acta Biomater 2017; 53:307-317. [PMID: 28213096 DOI: 10.1016/j.actbio.2017.02.027] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Revised: 02/02/2017] [Accepted: 02/13/2017] [Indexed: 12/22/2022]
Abstract
Anterior cruciate ligament (ACL) reconstruction remains a formidable clinical challenge because of the lack of vascularization and adequate cell numbers in the joint cavity. In this study, we developed a novel strategy to mimic the early stage of repair in vivo, which recapitulated extra-articular inflammatory response to facilitate the early ingrowth of blood vessels and cells. A vascularized ectopic tissue engineered ligament (ETEL) with silk collagen scaffold was developed and then transferred to reconstruct the ACL in rabbits without interruption of perfusion. At 2weeks after ACL reconstruction, more well-perfused cells and vessels were found in the regenerated ACL with ETEL, which decreased dramatically at the 4 and 12week time points with collagen deposition and maturation. ACL treated with ETEL exhibited more mature ligament structure and enhanced ligament-bone healing post-reconstructive surgery at 4 and 12weeks, as compared with the control group. In addition, the ETEL group was demonstrated to have higher modulus and stiffness than the control group significantly at 12weeks post-reconstructive surgery. In conclusion, our results demonstrated that the ETEL can provide sufficient vascularity and cellularity during the early stages of healing, and subsequently promote ACL regeneration and ligament-bone healing, suggesting its clinic use as a promising therapeutic modality. STATEMENT OF SIGNIFICANCE Early inflammatory cell infiltration, tissue and vessels ingrowth were significantly higher in the extra-articular implanted scaffolds than theses in the joint cavity. By mimicking the early stages of wound repair, which provided extra-articular inflammatory stimulation to facilitate the early ingrowth of blood vessels and cells, a vascularized ectopic tissue engineered ligament (ETEL) with silk collagen scaffold was constructed by subcutaneous implantation for 2weeks. The fully vascularized TE ligament was then transferred to rebuild ACL without blood perfusion interruption, and was demonstrated to exhibit improved ACL regeneration, bone tunnel healing and mechanical properties.
Collapse
Affiliation(s)
- Jisheng Ran
- Department of Orthopedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China; Orthopaedics Research Institute of Zhejiang University, Hangzhou, People's Republic of China
| | - Yejun Hu
- Department of Orthopedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China; Dr. Li Dak Sum and Yip Yio Chin Center for Stem Cells and Regenerative Medicine, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Huihui Le
- Department of Orthopedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China; Orthopaedics Research Institute of Zhejiang University, Hangzhou, People's Republic of China
| | - Yangwu Chen
- Department of Orthopedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China; Dr. Li Dak Sum and Yip Yio Chin Center for Stem Cells and Regenerative Medicine, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China; Orthopaedics Research Institute of Zhejiang University, Hangzhou, People's Republic of China
| | - Zefeng Zheng
- Department of Orthopedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Xiao Chen
- Dr. Li Dak Sum and Yip Yio Chin Center for Stem Cells and Regenerative Medicine, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China; China Orthopaedic Regenerative Medicine (CORMed), Hangzhou, China
| | - Zi Yin
- Dr. Li Dak Sum and Yip Yio Chin Center for Stem Cells and Regenerative Medicine, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Ruijian Yan
- Department of Orthopedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China; Orthopaedics Research Institute of Zhejiang University, Hangzhou, People's Republic of China
| | - Zhangchu Jin
- Department of Orthopedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China; Dr. Li Dak Sum and Yip Yio Chin Center for Stem Cells and Regenerative Medicine, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China; Orthopaedics Research Institute of Zhejiang University, Hangzhou, People's Republic of China
| | - Chenqi Tang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China; Dr. Li Dak Sum and Yip Yio Chin Center for Stem Cells and Regenerative Medicine, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China; Orthopaedics Research Institute of Zhejiang University, Hangzhou, People's Republic of China
| | - Jiayun Huang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China; Dr. Li Dak Sum and Yip Yio Chin Center for Stem Cells and Regenerative Medicine, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China; Orthopaedics Research Institute of Zhejiang University, Hangzhou, People's Republic of China
| | - Yanjia Gu
- Department of Orthopedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China; Dr. Li Dak Sum and Yip Yio Chin Center for Stem Cells and Regenerative Medicine, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China; Orthopaedics Research Institute of Zhejiang University, Hangzhou, People's Republic of China
| | - Langhai Xu
- Department of Orthopedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China; Orthopaedics Research Institute of Zhejiang University, Hangzhou, People's Republic of China
| | - Shengjun Qian
- Department of Orthopedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China; Orthopaedics Research Institute of Zhejiang University, Hangzhou, People's Republic of China
| | - Wei Zhang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China; Orthopaedics Research Institute of Zhejiang University, Hangzhou, People's Republic of China
| | - Boon Chin Heng
- Department of Endodontology, Faculty of Dentistry, The University of Hong Kong, Pokfulam, Hong Kong
| | | | - Weishan Chen
- Department of Orthopedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China; Orthopaedics Research Institute of Zhejiang University, Hangzhou, People's Republic of China
| | - Lidong Wu
- Department of Orthopedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China; Orthopaedics Research Institute of Zhejiang University, Hangzhou, People's Republic of China
| | - Weiliang Shen
- Department of Orthopedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China; Dr. Li Dak Sum and Yip Yio Chin Center for Stem Cells and Regenerative Medicine, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China; Orthopaedics Research Institute of Zhejiang University, Hangzhou, People's Republic of China; China Orthopaedic Regenerative Medicine (CORMed), Hangzhou, China.
| | - Hongwei Ouyang
- Dr. Li Dak Sum and Yip Yio Chin Center for Stem Cells and Regenerative Medicine, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China; China Orthopaedic Regenerative Medicine (CORMed), Hangzhou, China
| |
Collapse
|
22
|
Comparison of Endothelial Differentiation Capacities of Human and Rat Adipose-Derived Stem Cells. Plast Reconstr Surg 2016; 138:1231-1241. [DOI: 10.1097/prs.0000000000002791] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
23
|
Zhou J, Ding J, Nie B, Hu S, Zhu Z, Chen J, Xu J, Shi J, Dong N. Promotion of adhesion and proliferation of endothelial progenitor cells on decellularized valves by covalent incorporation of RGD peptide and VEGF. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2016; 27:142. [PMID: 27541486 DOI: 10.1007/s10856-016-5750-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Accepted: 07/06/2016] [Indexed: 06/06/2023]
Abstract
Tissue engineered heart valve is a promising alternative to current heart valve surgery, for its capability of growth, repair, and remodeling. However, extensive development is needed to ensure tissue compatibility, durability and antithrombotic potential. This study aims to investigate the biological effects of multi-signal composite material of polyethyl glycol-cross-linked decellularized valve on adhesion and proliferation of endothelial progenitor cells. Group A to E was decellularized valve leaflets, composite material of polyethyl glycol-cross-linked decellularized valves leaflets, vascular endothelial growth factor-composite materials, Arg-Gly-Asp peptide-composite materials and multi-signal modified materials of polyethyl glycol-cross-linked decellularized valve leaflets, respectively. The endothelial progenitor cells were seeded for each group, cell adhesion and proliferation were detected and neo-endothelium antithrombotic function of the multi-signal composite materials was evaluated. At 2, 4, and 8 h after the seeding, the cell numbers and 3H-TdR incorporation in group D were the highest. At 2, 4, and 8 days after the seeding, the cell numbers and 3H-TdR incorporation were significantly higher in groups C, D, and E compared with groups A and B (P < 0.05) and cell numbers and the expression of t-PA and eons in the neo-endothelium were quite similar to those in the human umbilical vein endothelial cells at 2, 4, and 8 days after the seeding. The Arg-Gly-Asp- peptides (a sequential peptide composed of arginine (Arg), glycine (Gly) and aspartic acid (Asp)) and VEGF-conjugated onto the composite material of PEG-crosslinked decellularized valve leaflets synergistically promoted the adhesion and proliferation of endothelial progenitor cells on the composite material, which may help in tissue engineering of heart valves.
Collapse
Affiliation(s)
- Jianliang Zhou
- Department of Cardiothoracic Surgery, the Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China.
| | - Jingli Ding
- Department of Gastroenterology, the Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Bin'en Nie
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Shidong Hu
- Department of Cardiothoracic Surgery, the Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Zhigang Zhu
- Department of Cardiothoracic Surgery, the Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Jia Chen
- Department of Cardiothoracic Surgery, the Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Jianjun Xu
- Department of Cardiothoracic Surgery, the Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Jiawei Shi
- Department of Cardiovascular Surgery, the Union Hospital Affiliated to Tongji Medical College of Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Nianguo Dong
- Department of Cardiovascular Surgery, the Union Hospital Affiliated to Tongji Medical College of Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
24
|
Hsia K, Yao CL, Chen WM, Chen JH, Lee H, Lu JH. Scaffolds and Cell-Based Tissue Engineering for Blood Vessel Therapy. Cells Tissues Organs 2016; 202:281-295. [PMID: 27548610 DOI: 10.1159/000448169] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/04/2016] [Indexed: 11/19/2022] Open
Abstract
The increasing morbidity of cardiovascular diseases in modern society has made it crucial to develop a small-caliber blood vessel. In the absence of appropriate autologous vascular grafts, an alternative prosthesis must be constructed for cardiovascular disease patients. The aim of this article is to describe the advances in making cell-seeded cardiovascular prostheses. It also discusses the combinations of types of scaffolds and cells, especially autologous stem cells, which are suitable for application in tissue-engineered vessels with the favorable properties of mechanical strength, antithrombogenicity, biocompliance, anti-inflammation, fatigue resistance and long-term durability. This article highlights the advancements in cellular tissue-engineered vessels in recent years.
Collapse
|
25
|
Characterization and preparation of bio-tubular scaffolds for fabricating artificial vascular grafts by combining electrospinning and a co-culture system. Macromol Res 2016. [DOI: 10.1007/s13233-016-4017-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
26
|
Laschke MW, Menger MD. Prevascularization in tissue engineering: Current concepts and future directions. Biotechnol Adv 2015; 34:112-21. [PMID: 26674312 DOI: 10.1016/j.biotechadv.2015.12.004] [Citation(s) in RCA: 183] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Revised: 11/16/2015] [Accepted: 12/04/2015] [Indexed: 12/24/2022]
Abstract
The survival of engineered tissue constructs during the initial phase after their implantation depends on the rapid development of an adequate vascularization. This, in turn, is a major prerequisite for the constructs' long-term function. 'Prevascularization' has emerged as a promising concept in tissue engineering, aiming at the generation of a preformed microvasculature in tissue constructs prior to their implantation. This should shorten the time period during which the constructs are avascular and suffer hypoxic conditions. Herein, we provide an overview of current strategies for the generation of preformed microvascular networks within tissue constructs. In vitro approaches use cell seeding, spheroid formation or cell sheet technologies. In situ approaches use the body as a natural bioreactor to induce vascularization by angiogenic ingrowth or flap and arteriovenous (AV)-loop techniques. In future, these strategies may be supplemented by the transplantation of adipose tissue-derived microvascular fragments or the in vitro generation of highly organized microvascular networks by means of sophisticated microscale technologies and microfluidic systems. The further advancement of these prevascularization concepts and their adaptation to individual therapeutic interventions will markedly contribute to a broad implementation of tissue engineering applications into clinical practice.
Collapse
Affiliation(s)
- Matthias W Laschke
- Institute for Clinical & Experimental Surgery, Saarland University, D-66421 Homburg/Saar, Germany.
| | - Michael D Menger
- Institute for Clinical & Experimental Surgery, Saarland University, D-66421 Homburg/Saar, Germany
| |
Collapse
|
27
|
The cerebral cavernous malformation proteins CCM2L and CCM2 prevent the activation of the MAP kinase MEKK3. Proc Natl Acad Sci U S A 2015; 112:14284-9. [PMID: 26540726 DOI: 10.1073/pnas.1510495112] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Three genes, CCM1, CCM2, and CCM3, interact genetically and biochemically and are mutated in cerebral cavernous malformations (CCM). A recently described member of this CCM family of proteins, CCM2-like (CCM2L), has high homology to CCM2. Here we show that its relative expression in different tissues differs from that of CCM2 and, unlike CCM2, the expression of CCM2L in endothelial cells is regulated by density, flow, and statins. In vitro, both CCM2L and CCM2 bind MEKK3 in a complex with CCM1. Both CCM2L and CCM2 interfere with MEKK3 activation and its ability to phosphorylate MEK5, a downstream target. The in vivo relevance of this regulation was investigated in zebrafish. A knockdown of ccm2l and ccm2 in zebrafish leads to a more severe "big heart" and circulation defects compared with loss of function of ccm2 alone, and also leads to substantial body axis abnormalities. Silencing of mekk3 rescues the big heart and body axis phenotype, suggesting cross-talk between the CCM proteins and MEKK3 in vivo. In endothelial cells, CCM2 deletion leads to activation of ERK5 and a transcriptional program that are downstream of MEKK3. These findings suggest that CCM2L and CCM2 cooperate to regulate the activity of MEKK3.
Collapse
|
28
|
Peters EB, Christoforou N, Leong KW, Truskey GA, West JL. Poly(ethylene glycol) Hydrogel Scaffolds Containing Cell-Adhesive and Protease-Sensitive Peptides Support Microvessel Formation by Endothelial Progenitor Cells. Cell Mol Bioeng 2015; 9:38-54. [PMID: 27042236 DOI: 10.1007/s12195-015-0423-6] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The development of stable, functional microvessels remains an important obstacle to overcome for tissue engineered organs and treatment of ischemia. Endothelial progenitor cells (EPCs) are a promising cell source for vascular tissue engineering as they are readily obtainable and carry the potential to differentiate towards all endothelial phenotypes. The aim of this study was to investigate the ability of human umbilical cord blood-derived EPCs to form vessel-like structures within a tissue engineering scaffold material, a cell-adhesive and proteolytically degradable poly(ethylene glycol) (PEG) hydrogel. EPCs in co-culture with angiogenic mural cells were encapsulated in hydrogel scaffolds by mixing with polymeric precursors and using a mild photocrosslinking process to form hydrogels with homogeneously dispersed cells. EPCs formed 3D microvessels networks that were stable for at least 30 days in culture, without the need for supplemental angiogenic growth factors. These 3D EPC microvessels displayed aspects of physiological microvasculature with lumen formation, expression of endothelial cell proteins (connexin 32, VE-cadherin, eNOS), basement membrane formation with collagen IV and laminin, perivascular investment of PDGFR-β and α-SMA positive cells, and EPC quiescence (<1% proliferating cells) by 2 weeks of co-culture. Our findings demonstrate the development of a novel, reductionist system that is well-defined and reproducible for studying progenitor cell-driven microvessel formation.
Collapse
Affiliation(s)
- Erica B Peters
- Fitzpatrick CIEMAS Building, Room 1427, Box 90281, Duke University, Department of Biomedical Engineering, Durham, NC 27708
| | - Nicolas Christoforou
- P.O. Box 127788, Khalifa University, Department of Biomedical Engineering, Abu Dhabi, UAE
| | - Kam W Leong
- 1210 Amsterdam Avenue, Mail Code 8904, Columbia University, Department of Biomedical Engineering, New York, NY 10027
| | - George A Truskey
- Fitzpatrick CIEMAS Building, Room 1427, Box 90281, Duke University, Department of Biomedical Engineering, Durham, NC 27708
| | - Jennifer L West
- Fitzpatrick CIEMAS Building, Room 1427, Box 90281, Duke University, Department of Biomedical Engineering, Durham, NC 27708
| |
Collapse
|
29
|
Williams PA, Silva EA. The Role of Synthetic Extracellular Matrices in Endothelial Progenitor Cell Homing for Treatment of Vascular Disease. Ann Biomed Eng 2015. [DOI: 10.1007/s10439-015-1400-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
|
30
|
Sarker M, Chen X, Schreyer D. Experimental approaches to vascularisation within tissue engineering constructs. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2015; 26:683-734. [DOI: 10.1080/09205063.2015.1059018] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
31
|
Markeson D, Pleat JM, Sharpe JR, Harris AL, Seifalian AM, Watt SM. Scarring, stem cells, scaffolds and skin repair. J Tissue Eng Regen Med 2015; 9:649-68. [PMID: 24668923 DOI: 10.1002/term.1841] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2013] [Revised: 08/09/2013] [Accepted: 09/16/2013] [Indexed: 01/19/2023]
Abstract
The treatment of full thickness skin loss, which can be extensive in the case of large burns, continues to represent a challenging clinical entity. This is due to an on-going inability to produce a suitable tissue engineered substrate that can satisfactorily replicate the epidermal and dermal in vivo niches to fulfil both aesthetic and functional demands. The current gold standard treatment of autologous skin grafting is inadequate because of poor textural durability, scarring and associated contracture, and because of a paucity of donor sites in larger burns. Tissue engineering has seen exponential growth in recent years with a number of 'off-the-shelf' dermal and epidermal substitutes now available. Each has its own limitations. In this review, we examine normal wound repair in relation to stem/progenitor cells that are intimately involved in this process within the dermal niche. Endothelial precursors, in particular, are examined closely and their phenotype, morphology and enrichment from multiple sources are described in an attempt to provide some clarity regarding the controversy surrounding their classification and role in vasculogenesis. We also review the role of the next generation of cellularized scaffolds and smart biomaterials that attempt to improve the revascularisation of artificial grafts, the rate of wound healing and the final cosmetic and functional outcome.
Collapse
Affiliation(s)
- Daniel Markeson
- Stem Cell Research Laboratory, NHS Blood and Transplant, Oxford, UK
- Department of Plastic and Reconstructive Surgery, Stoke Mandeville Hospital, Aylesbury, UK
- Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- University College London Centre for Nanotechnology and Regenerative Medicine, Division of Surgery and Interventional Science, Royal Free Hospital, London, UK
| | - Jonathon M Pleat
- Department of Plastic and Reconstructive Surgery, Stoke Mandeville Hospital, Aylesbury, UK
- Department of Plastic and Reconstructive Surgery, Frenchay Hospital, Bristol, UK
| | - Justin R Sharpe
- Blond McIndoe Research Foundation, Queen Victoria Hospital, East Grinstead, West Sussex, UK
| | - Adrian L Harris
- Molecular Oncology Laboratories, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Alexander M Seifalian
- University College London Centre for Nanotechnology and Regenerative Medicine, Division of Surgery and Interventional Science, Royal Free Hospital, London, UK
| | - Suzanne M Watt
- Stem Cell Research Laboratory, NHS Blood and Transplant, Oxford, UK
- Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
32
|
Kim H, Huang L, Critser PJ, Yang Z, Chan RJ, Wang L, Carlesso N, Voytik-Harbin SL, Bernstein ID, Yoder MC. Notch ligand Delta-like 1 promotes in vivo vasculogenesis in human cord blood-derived endothelial colony forming cells. Cytotherapy 2015; 17:579-92. [PMID: 25559145 DOI: 10.1016/j.jcyt.2014.12.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Revised: 11/06/2014] [Accepted: 12/04/2014] [Indexed: 01/11/2023]
Abstract
BACKGROUND AIMS Human cord blood (CB) is enriched in circulating endothelial colony forming cells (ECFCs) that display high proliferative potential and in vivo vessel forming ability. Because Notch signaling is critical for embryonic blood vessel formation in utero, we hypothesized that Notch pathway activation may enhance cultured ECFC vasculogenic properties in vivo. METHODS In vitro ECFC stimulation with an immobilized chimeric Notch ligand (Delta-like1(ext-IgG)) led to significant increases in the mRNA and protein levels of Notch regulated Hey2 and EphrinB2 that were blocked by treatment with γ-secretase inhibitor addition. However, Notch stimulated preconditioning in vitro failed to enhance ECFC vasculogenesis in vivo. In contrast, in vivo co-implantation of ECFCs with OP9-Delta-like 1 stromal cells that constitutively expressed the Notch ligand delta-like 1 resulted in enhanced Notch activated ECFC-derived increased vessel density and enlarged vessel area in vivo, an effect not induced by OP9 control stromal implantation. RESULTS This Notch activation was associated with diminished apoptosis in the exposed ECFC. CONCLUSIONS We conclude that Notch pathway activation in ECFC in vivo via co-implanted stromal cells expressing delta-like 1 promotes vasculogenesis and augments blood vessel formation via diminishing apoptosis of the implanted ECFC.
Collapse
Affiliation(s)
- Hyojin Kim
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, USA; Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana, USA; Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana, USA; Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Lan Huang
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, USA; Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana, USA; Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Paul J Critser
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, USA; Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Zhenyun Yang
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, USA; Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Rebecca J Chan
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, USA; Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Lin Wang
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, USA; Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana, USA; Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Nadia Carlesso
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, USA; Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana, USA; Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Sherry L Voytik-Harbin
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana, USA
| | | | - Mervin C Yoder
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, USA; Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana, USA; Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana, USA.
| |
Collapse
|
33
|
Piatkowski A, Grieb G, Simons D, Bernhagen J, van der Hulst RR. Endothelial progenitor cells--potential new avenues to improve neoangiogenesis and reendothelialization. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2014; 306:43-81. [PMID: 24016523 DOI: 10.1016/b978-0-12-407694-5.00002-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The term endothelial progenitor cell (EPC) was established more than 10 years ago and is used to refer to a group of circulating cells that display endothelial lineage qualities and are able to home to areas of ischemia or vascular injury and to facilitate the repair of damaged blood vessels or develop new vessels as needed. This chapter reviews the current lineage relationships among all the cells called EPC and will clear the terminology used in EPC research. Furthermore, an overview of the clinical and in vitro research, as well as cytokine and drug interactions and potential EPC applications, is given.
Collapse
Affiliation(s)
- Andrzej Piatkowski
- Department of Plastic Surgery, academisch ziekenhuis Maastricht, MUMC+, Maastricht, The Netherlands.
| | | | | | | | | |
Collapse
|
34
|
Engineering Angiogenesis for Myocardial Infarction Repair: Recent Developments, Challenges, and Future Directions. Cardiovasc Eng Technol 2014. [DOI: 10.1007/s13239-014-0193-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
35
|
Chen YE, Xie C, Yang B. Stem cells for vascular engineering. BIOMATERIALS AND REGENERATIVE MEDICINE 2014:621-639. [DOI: 10.1017/cbo9780511997839.041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
36
|
Cell sheet technology-driven re-epithelialization and neovascularization of skin wounds. Acta Biomater 2014; 10:3145-55. [PMID: 24650971 DOI: 10.1016/j.actbio.2014.03.006] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Revised: 02/12/2014] [Accepted: 03/09/2014] [Indexed: 11/21/2022]
Abstract
Skin regeneration remains a challenge, requiring a well-orchestrated interplay of cell-cell and cell-matrix signalling. Cell sheet (CS) engineering, which has the major advantage of allowing the retrieval of the intact cell layers along with their naturally organized extracellular matrix (ECM), has been poorly explored for the purpose of creating skin substitutes and skin regeneration. This work proposes the use of CS technology to engineer cellular constructs based on human keratinocytes (hKC), key players in wound re-epithelialization, dermal fibroblasts (hDFb), responsible for ECM remodelling, and dermal microvascular endothelial cells (hDMEC), part of the dermal vascular network and modulators of angiogenesis. Homotypic and heterotypic three-dimensional (3-D) CS-based constructs were developed simultaneously to target wound re-vascularization and re-epithelialization. After implantation of the constructs in murine full-thickness wounds, human cells were engrafted into the host wound bed and were present in the neotissue formed up to 14 days post-implantation. Different outcomes were obtained by varying the composition and organization of the 3-D constructs. Both hKC and hDMEC significantly contributed to re-epithelialization by promoting rapid wound closure and early epithelial coverage. Moreover, a significant increase in the density of vessels at day 7 and the incorporation of hDMEC in the neoformed vasculature confirmed its role over neotissue vacularization. As a whole, the obtained results confirmed that the proposed 3-D CS-based constructs provided the necessary cell machinery, when in a specific microenvironment, guiding both re-vascularization and re-epithelialization. Although dependent on the nature of the constructs, the results obtained sustain the hypothesis that different CS-based constructs lead to improved skin healing.
Collapse
|
37
|
Qin D, Long T, Deng J, Zhang Y. Urine-derived stem cells for potential use in bladder repair. Stem Cell Res Ther 2014; 5:69. [PMID: 25157812 PMCID: PMC4055102 DOI: 10.1186/scrt458] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Engineered bladder tissues, created with autologous bladder cells seeded on biodegradable scaffolds, are being developed for use in patients who need cystoplasty. However, in individuals with organ damage from congenital disorders, infection, irradiation, or cancer, abnormal cells obtained by biopsy from the compromised tissue could potentially contaminate the engineered tissue. Thus, an alternative cell source for construction of the neo-organ would be useful. Although other types of stem cells have been investigated, autologous mesenchymal stem cells (MSCs) are most suitable to use in bladder regeneration. These cells are often used as a cell source for bladder repair in three ways - secreting paracrine factors, recruiting resident cells, and trans-differentiation, inducing MSCs to differentiate into bladder smooth muscle cells and urothelial cells. Adult stem cell populations have been demonstrated in bone marrow, fat, muscle, hair follicles, and amniotic fluid. These cells remain an area of intense study, as their potential for therapy may be applicable to bladder disorders. Recently, we have found stem cells in the urine and the cells are highly expandable, and have self-renewal capacity and paracrine properties. As a novel cell source, urine-derived stem cells (USCs) provide advantages for cell therapy and tissue engineering applications in bladder tissue repair because they originate from the urinary tract system. Importantly, USCs can be obtained via a noninvasive, simple, and low-cost approach and induced with high efficiency to differentiate into bladder cells.
Collapse
|
38
|
Joo HJ, Seo HR, Jeong HE, Choi SC, Park JH, Yu CW, Hong SJ, Chung S, Lim DS. Smooth muscle progenitor cells from peripheral blood promote the neovascularization of endothelial colony-forming cells. Biochem Biophys Res Commun 2014; 449:405-11. [PMID: 24858689 DOI: 10.1016/j.bbrc.2014.05.061] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Accepted: 05/15/2014] [Indexed: 11/24/2022]
Abstract
Proangiogenic cell therapy using autologous progenitors is a promising strategy for treating ischemic disease. Considering that neovascularization is a harmonized cellular process that involves both endothelial cells and vascular smooth muscle cells, peripheral blood-originating endothelial colony-forming cells (ECFCs) and smooth muscle progenitor cells (SMPCs), which are similar to mature endothelial cells and vascular smooth muscle cells, could be attractive cellular candidates to achieve therapeutic neovascularization. We successfully induced populations of two different vascular progenitor cells (ECFCs and SMPCs) from adult peripheral blood. Both progenitor cell types expressed endothelial-specific or smooth muscle-specific genes and markers, respectively. In a protein array focused on angiogenic cytokines, SMPCs demonstrated significantly higher expression of bFGF, EGF, TIMP2, ENA78, and TIMP1 compared to ECFCs. Conditioned medium from SMPCs and co-culture with SMPCs revealed that SMPCs promoted cell proliferation, migration, and the in vitro angiogenesis of ECFCs. Finally, co-transplantation of ECFCs and SMPCs induced robust in vivo neovascularization, as well as improved blood perfusion and tissue repair, in a mouse ischemic hindlimb model. Taken together, we have provided the first evidence of a cell therapy strategy for therapeutic neovascularization using two different types of autologous progenitors (ECFCs and SMPCs) derived from adult peripheral blood.
Collapse
Affiliation(s)
- Hyung Joon Joo
- Department of Cardiology, Cardiovascular Center, College of Medicine, Korea University, Seoul, Republic of Korea
| | - Ha-Rim Seo
- Department of Cardiology, Cardiovascular Center, College of Medicine, Korea University, Seoul, Republic of Korea
| | - Hyo Eun Jeong
- Department of Mechanical Engineering, Korea University, Seoul, Republic of Korea
| | - Seung-Cheol Choi
- Department of Cardiology, Cardiovascular Center, College of Medicine, Korea University, Seoul, Republic of Korea
| | - Jae Hyung Park
- Department of Cardiology, Cardiovascular Center, College of Medicine, Korea University, Seoul, Republic of Korea
| | - Cheol Woong Yu
- Department of Cardiology, Cardiovascular Center, College of Medicine, Korea University, Seoul, Republic of Korea
| | - Soon Jun Hong
- Department of Cardiology, Cardiovascular Center, College of Medicine, Korea University, Seoul, Republic of Korea
| | - Seok Chung
- Department of Mechanical Engineering, Korea University, Seoul, Republic of Korea
| | - Do-Sun Lim
- Department of Cardiology, Cardiovascular Center, College of Medicine, Korea University, Seoul, Republic of Korea.
| |
Collapse
|
39
|
Poitevin S, Cussac D, Leroyer AS, Albinet V, Sarlon-Bartoli G, Guillet B, Hubert L, Andrieu-Abadie N, Couderc B, Parini A, Dignat-George F, Sabatier F. Sphingosine kinase 1 expressed by endothelial colony-forming cells has a critical role in their revascularization activity. Cardiovasc Res 2014; 103:121-30. [PMID: 24743591 DOI: 10.1093/cvr/cvu104] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
AIMS Cell therapy based on endothelial colony-forming cells (ECFCs) is a promising option for ischaemic cardiovascular diseases. A better understanding of the mechanisms by which these cells promote revascularization remains a critical challenge to improving their therapeutic potential. We aimed to identify the critical mechanisms involved in the revascularization activity of ECFCs by using the paracrine properties of mesenchymal stem cells (MSC). METHODS AND RESULTS Conditioned medium from human bone marrow-derived MSCs (MSC-CM) increased the angiogenic activity of cord blood ECFCs in vitro (proliferation, migration, and pseudo-tube formation), the survival of ECFCs in mice (Matrigel Plug assay), and the capacity of ECFCs to promote the recovery of blood perfusion in mice with hindlimb ischaemia. Furthermore, the capillary density in ischaemic gastrocnemius muscle was significantly increased in mice transplanted with the ECFCs pre-treated with the MSC-CM. The enhancement of ECFCs activity involved the up-regulation of sphingosine kinase 1 (SphK1) expression and activity. The inhibition of SphK1 in ECFCs by using an inhibitor or a siRNA knockdown of SphK1 prevented the stimulation of the ECFCs induced by the MSC-CM. The improvement of ECFC activity by MSC-CM also involved the up-regulation of sphingosine-1-phosphate receptor 1 (S1P1) and a S1P/S1P1/3-dependent mechanism. Finally, we showed that the stimulation of ECFCs with exogenous S1P increased angiogenesis and promoted blood perfusion in hindlimb ischaemia. CONCLUSION The up-regulation of SphK1 and S1P-dependent pathways is critical for the angiogenic/vasculogenic activity of ECFCs. The identification of this pathway provides attractive targets to optimize cell-based therapy for revascularization in ischaemic diseases.
Collapse
Affiliation(s)
- Stéphane Poitevin
- Aix-Marseille Université, Vascular Research Center of Marseille (VRCM), INSERM UMR-S 1076, Faculté de Pharmacie, 27 Bd Jean Moulin, 13385 Marseille Cedex 05, France
| | - Daniel Cussac
- Institut des Maladies Métaboliques et Cardiovasculaires, INSERM U1048, Université de Toulouse III, 1 Av Jean Poulhès, BP 84225, 31432 Toulouse Cedex 4, France
| | - Aurélie S Leroyer
- Aix-Marseille Université, Vascular Research Center of Marseille (VRCM), INSERM UMR-S 1076, Faculté de Pharmacie, 27 Bd Jean Moulin, 13385 Marseille Cedex 05, France
| | - Virginie Albinet
- Centre de Recherche en Cancérologie, INSERM UMR-1037, Université de Toulouse III, BP 84225, CHU Rangueil, 31432 Toulouse Cedex 4, France
| | - Gabrielle Sarlon-Bartoli
- Aix-Marseille Université, Vascular Research Center of Marseille (VRCM), INSERM UMR-S 1076, Faculté de Pharmacie, 27 Bd Jean Moulin, 13385 Marseille Cedex 05, France
| | - Benjamin Guillet
- Aix-Marseille Université, Vascular Research Center of Marseille (VRCM), INSERM UMR-S 1076, Faculté de Pharmacie, 27 Bd Jean Moulin, 13385 Marseille Cedex 05, France
| | - Lucas Hubert
- Aix-Marseille Université, Vascular Research Center of Marseille (VRCM), INSERM UMR-S 1076, Faculté de Pharmacie, 27 Bd Jean Moulin, 13385 Marseille Cedex 05, France
| | - Nathalie Andrieu-Abadie
- Centre de Recherche en Cancérologie, INSERM UMR-1037, Université de Toulouse III, BP 84225, CHU Rangueil, 31432 Toulouse Cedex 4, France
| | - Bettina Couderc
- EA 4553: Individualisation des Traitements des Cancers Ovariens et ORL, Institut Claudius Regaud, 20-24 rue du Pont St Pierre, 31052 Toulouse Cedex 4, France
| | - Angelo Parini
- Institut des Maladies Métaboliques et Cardiovasculaires, INSERM U1048, Université de Toulouse III, 1 Av Jean Poulhès, BP 84225, 31432 Toulouse Cedex 4, France
| | - Françoise Dignat-George
- Aix-Marseille Université, Vascular Research Center of Marseille (VRCM), INSERM UMR-S 1076, Faculté de Pharmacie, 27 Bd Jean Moulin, 13385 Marseille Cedex 05, France
| | - Florence Sabatier
- Aix-Marseille Université, Vascular Research Center of Marseille (VRCM), INSERM UMR-S 1076, Faculté de Pharmacie, 27 Bd Jean Moulin, 13385 Marseille Cedex 05, France
| |
Collapse
|
40
|
Finosh GT, Jayabalan M. Regenerative therapy and tissue engineering for the treatment of end-stage cardiac failure: new developments and challenges. BIOMATTER 2014; 2:1-14. [PMID: 23507781 DOI: 10.4161/biom.19429] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Regeneration of myocardium through regenerative therapy and tissue engineering is appearing as a prospective treatment modality for patients with end-stage heart failure. Focusing on this area, this review highlights the new developments and challenges in the regeneration of myocardial tissue. The role of various cell sources, calcium ion and cytokine on the functional performance of regenerative therapy is discussed. The evolution of tissue engineering and the role of tissue matrix/scaffold, cell adhesion and vascularisation on tissue engineering of cardiac tissue implant are also discussed.
Collapse
Affiliation(s)
- G T Finosh
- Polymer Science Division, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Kerala, India
| | | |
Collapse
|
41
|
Fernandez CE, Obi-onuoha IC, Wallace CS, Satterwhite LL, Truskey GA, Reichert WM. Late-outgrowth endothelial progenitors from patients with coronary artery disease: endothelialization of confluent stromal cell layers. Acta Biomater 2014; 10:893-900. [PMID: 24140604 DOI: 10.1016/j.actbio.2013.10.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2013] [Revised: 09/16/2013] [Accepted: 10/09/2013] [Indexed: 12/20/2022]
Abstract
Patients with coronary artery disease (CAD) are the primary candidates to receive small-diameter tissue-engineered blood vessels (TEBVs). Peripheral blood derived endothelial progenitor cells (EPCs) from CAD patients (CAD EPCs) represent a minimally invasive source of autologous cells for TEBV endothelialization. We have previously shown that human CAD EPCs are highly proliferative and express many of the hallmarks of mature and healthy endothelial cells; however, their behavior on stromal cells that comprise the media of TEBVs has not yet been evaluated. Primary CAD EPCs or control human aortic endothelial cells (HAECs) were seeded over confluent, quiescent layers of human smooth muscle cells (SMCs) using a direct co-culture model. The percent coverage, adhesion strength, alignment under flow and generation of flow-induced nitric oxide of the seeded CAD EPCs were compared to that of HAECs. The integrin-binding profile of CAD EPCs was also evaluated over a layer of confluent, quiescent SMCs. Direct comparison of our CAD EPC results to analogous co-culture studies with cord blood EPCs show that both types of blood-derived EPCs are viable options for endothelialization of TEBVs.
Collapse
|
42
|
Babczyk P, Conzendorf C, Klose J, Schulze M, Harre K, Tobiasch E. Stem Cells on Biomaterials for Synthetic Grafts to Promote Vascular Healing. J Clin Med 2014; 3:39-87. [PMID: 26237251 PMCID: PMC4449663 DOI: 10.3390/jcm3010039] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2013] [Revised: 10/28/2013] [Accepted: 11/16/2013] [Indexed: 12/25/2022] Open
Abstract
This review is divided into two interconnected parts, namely a biological and a chemical one. The focus of the first part is on the biological background for constructing tissue-engineered vascular grafts to promote vascular healing. Various cell types, such as embryonic, mesenchymal and induced pluripotent stem cells, progenitor cells and endothelial- and smooth muscle cells will be discussed with respect to their specific markers. The in vitro and in vivo models and their potential to treat vascular diseases are also introduced. The chemical part focuses on strategies using either artificial or natural polymers for scaffold fabrication, including decellularized cardiovascular tissue. An overview will be given on scaffold fabrication including conventional methods and nanotechnologies. Special attention is given to 3D network formation via different chemical and physical cross-linking methods. In particular, electron beam treatment is introduced as a method to combine 3D network formation and surface modification. The review includes recently published scientific data and patents which have been registered within the last decade.
Collapse
Affiliation(s)
- Patrick Babczyk
- Department of Natural Science, Bonn-Rhein-Sieg University of Applied Science, Von-Liebig-Street 20, Rheinbach 53359, Germany.
| | - Clelia Conzendorf
- Faculty of Mechanical Engineering/Process Engineering, University of Applied Science Dresden, Friedrich-List-Platz 1, Dresden 01069, Germany.
| | - Jens Klose
- Faculty of Mechanical Engineering/Process Engineering, University of Applied Science Dresden, Friedrich-List-Platz 1, Dresden 01069, Germany.
| | - Margit Schulze
- Department of Natural Science, Bonn-Rhein-Sieg University of Applied Science, Von-Liebig-Street 20, Rheinbach 53359, Germany.
| | - Kathrin Harre
- Faculty of Mechanical Engineering/Process Engineering, University of Applied Science Dresden, Friedrich-List-Platz 1, Dresden 01069, Germany.
| | - Edda Tobiasch
- Department of Natural Science, Bonn-Rhein-Sieg University of Applied Science, Von-Liebig-Street 20, Rheinbach 53359, Germany.
| |
Collapse
|
43
|
Abstract
Tissue engineering aims to create, repair and/or replace tissues and organs by using cells, scaffolds, biologically active molecules and physiologic signals. It is an interdisciplinary field that integrates aspects of engineering, chemistry, biology and medicine. One of the most challenging goals in the field of cardiovascular tissue engineering is the creation of a heart muscle patch. This review describes the principles, achievements and challenges of achieving this ambitious goal of creating contractile heart muscle. In addition, the new strategy of in situ and injectable tissue engineering for myocardial repair and regeneration is presented.
Collapse
Affiliation(s)
- Jonathan Leor
- Sheba-Medical Center, Neufeld Cardiac Research Institute, Tel-Aviv University, Tel-Hashomer 52621, Israel.
| | | | | |
Collapse
|
44
|
Kanitkar M, Jaiswal A, Deshpande R, Bellare J, Kale VP. Enhanced growth of endothelial precursor cells on PCG-matrix facilitates accelerated, fibrosis-free, wound healing: a diabetic mouse model. PLoS One 2013; 8:e69960. [PMID: 23922871 PMCID: PMC3724903 DOI: 10.1371/journal.pone.0069960] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2013] [Accepted: 06/13/2013] [Indexed: 11/19/2022] Open
Abstract
Diabetes mellitus (DM)-induced endothelial progenitor cell (EPC) dysfunction causes impaired wound healing, which can be rescued by delivery of large numbers of 'normal' EPCs onto such wounds. The principal challenges herein are (a) the high number of EPCs required and (b) their sustained delivery onto the wounds. Most of the currently available scaffolds either serve as passive devices for cellular delivery or allow adherence and proliferation, but not both. This clearly indicates that matrices possessing both attributes are 'the need of the day' for efficient healing of diabetic wounds. Therefore, we developed a system that not only allows selective enrichment and expansion of EPCs, but also efficiently delivers them onto the wounds. Murine bone marrow-derived mononuclear cells (MNCs) were seeded onto a PolyCaprolactone-Gelatin (PCG) nano-fiber matrix that offers a combined advantage of strength, biocompatibility wettability; and cultured them in EGM2 to allow EPC growth. The efficacy of the PCG matrix in supporting the EPC growth and delivery was assessed by various in vitro parameters. Its efficacy in diabetic wound healing was assessed by a topical application of the PCG-EPCs onto diabetic wounds. The PCG matrix promoted a high-level attachment of EPCs and enhanced their growth, colony formation, and proliferation without compromising their viability as compared to Poly L-lactic acid (PLLA) and Vitronectin (VN), the matrix and non-matrix controls respectively. The PCG-matrix also allowed a sustained chemotactic migration of EPCs in vitro. The matrix-effected sustained delivery of EPCs onto the diabetic wounds resulted in an enhanced fibrosis-free wound healing as compared to the controls. Our data, thus, highlight the novel therapeutic potential of PCG-EPCs as a combined 'growth and delivery system' to achieve an accelerated fibrosis-free healing of dermal lesions, including diabetic wounds.
Collapse
Affiliation(s)
- Meghana Kanitkar
- National Centre for Cell Science, NCCS Complex, University of Pune Campus, Ganeshkhind, Pune, Maharashtra, India
| | - Amit Jaiswal
- Department of Chemical Engineering, Indian Institute of Technology-Bombay, Powai, Mumbai, Maharashtra, India
| | - Rucha Deshpande
- National Centre for Cell Science, NCCS Complex, University of Pune Campus, Ganeshkhind, Pune, Maharashtra, India
| | - Jayesh Bellare
- Department of Chemical Engineering, Indian Institute of Technology-Bombay, Powai, Mumbai, Maharashtra, India
| | - Vaijayanti P. Kale
- National Centre for Cell Science, NCCS Complex, University of Pune Campus, Ganeshkhind, Pune, Maharashtra, India
- * E-mail:
| |
Collapse
|
45
|
Czirok A. Endothelial cell motility, coordination and pattern formation during vasculogenesis. WILEY INTERDISCIPLINARY REVIEWS-SYSTEMS BIOLOGY AND MEDICINE 2013; 5:587-602. [PMID: 23857825 DOI: 10.1002/wsbm.1233] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Revised: 05/24/2013] [Accepted: 05/28/2013] [Indexed: 01/13/2023]
Abstract
How vascular networks assemble is a fundamental problem of developmental biology that also has medical importance. To explain the organizational principles behind vascular patterning, we must understand how can tissue level structures be controlled through cell behavior patterns like motility and adhesion that, in turn, are determined by biochemical signal transduction processes? We discuss the various ideas that have been proposed as mechanisms for vascular network assembly: cell motility guided by extracellular matrix alignment (contact guidance), chemotaxis guided by paracrine and autocrine morphogens, and multicellular sprouting guided by cell-cell contacts. All of these processes yield emergent patterns, thus endothelial cells can form an interconnected structure autonomously, without guidance from an external pre-pattern.
Collapse
Affiliation(s)
- Andras Czirok
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS, USA; Department of Biological Physics, Eötvös Loránd University, Budapest, Hungary
| |
Collapse
|
46
|
Roura S, Gálvez-Montón C, Bayes-Genis A. The challenges for cardiac vascular precursor cell therapy: lessons from a very elusive precursor. J Vasc Res 2013; 50:304-23. [PMID: 23860201 DOI: 10.1159/000353294] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2013] [Accepted: 05/01/2013] [Indexed: 11/19/2022] Open
Abstract
There is compelling evidence that cardiovascular disorders arise and/or progress due mainly to endothelial dysfunction. Novel therapeutic strategies aim to generate new myocardial tissue using cells with regenerative potential, either alone or in combination with biomaterials, cytokines and advanced monitoring devices. Among the human adult progenitor cells used in such methods, those historically termed 'endothelial progenitor cells' show promise for vascular growth and repair. Asahara et al. [Science 1997;275:964-967] initially described putative endothelial cell precursors in 1997. Subsequently, distinct cell populations termed endothelial colony-forming units-Hill, circulating angiogenic cells and endothelial colony-forming cells were identified that varied in terms of phenotype, vascular homeostasis contribution and purity. Notably, most of these cells are not genuine vascular precursor cells belonging to the endothelial lineage. This review provides a broad overview of the main properties of the endothelium, focusing on the basis governing its growth and repair. We discuss efforts to identify true vascular precursors, a matter of debate for the past 15 years, as well as recent methodological advances in identifying new hierarchies of more homogeneous, clonogenic and proliferative vascular endothelial-lineage precursors. Consideration of these issues provides insights that may help develop more effective therapies against human diseases that involve vascular deficits.
Collapse
Affiliation(s)
- Santiago Roura
- ICREC Research Program, Health Research Institute Germans Trias i Pujol-IGTP, University Hospital Germans Trias i Pujol, Badalona, Spain.
| | | | | |
Collapse
|
47
|
Petrovic V, Zivkovic P, Petrovic D, Stefanovic V. Craniofacial bone tissue engineering. Oral Surg Oral Med Oral Pathol Oral Radiol 2013; 114:e1-9. [PMID: 22862985 DOI: 10.1016/j.oooo.2012.02.030] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2011] [Revised: 01/18/2012] [Accepted: 02/29/2012] [Indexed: 12/17/2022]
Abstract
There are numerous conditions, such as trauma, cancer, congenital malformations, and progressive deforming skeletal diseases, that can compromise the function and architectonics of bones of craniofacial region. The need to develop new approaches for treatment of these disorders arises from the fact that conventional therapeutic strategies face many obstacles and limitations. The use of tissue engineering in regeneration of craniofacial bone structures is a very promising possibility and a great challenge for researchers and practitioners. Developments in stem cell biology and engineering have led to the discovery of different stem cell populations and biodegradable materials with suitable properties. This review summarizes the current achievements in tissue engineering of craniofacial bone, temporomandibular joint, and periodontal ligament.
Collapse
Affiliation(s)
- Vladimir Petrovic
- Department of Histology, Stem Cells Laboratory, University School of Medicine, Nis, Serbia
| | | | | | | |
Collapse
|
48
|
Blood outgrowth endothelial cells increase tumor growth rates and modify tumor physiology: relevance for therapeutic targeting. Cancers (Basel) 2013; 5:205-17. [PMID: 24216704 PMCID: PMC3730307 DOI: 10.3390/cancers5010205] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2013] [Revised: 01/26/2013] [Accepted: 02/06/2013] [Indexed: 11/28/2022] Open
Abstract
Endothelial cell precursors from human peripheral blood have been shown to home to areas of neovascularization and may assist tumor growth by increasing or fortifying blood vessel growth. In the present study, the influence of these cells on tumor growth and physiology was investigated and the role of these cells as a therapeutic target or in determining treatment sensitivity was tested. After isolation from human blood and expansion in vitro, actively growing cells with verified endothelial phenotype (Blood Outgrowth Endothelial Cell, BOEC) were injected i.v. into tumor bearing mice for three consecutive days. The growth rate was significantly enhanced in relatively small RERF human lung tumors (i.e., less than 150 mm3) grown in immunocompromised mice by an average of 1.5-fold while it had no effect when injections were given to animals bearing larger tumors. There were no signs of toxicity or unwanted systemic effects. We also observed evidence of increased perfusion, vessel number, response to 15 Gy radiation and oxygenation in RERF tumors of animals injected with BOECs compared to control tumors. In addition, FSaII murine fibrosarcoma tumors were found to grow faster upon injection of BOECs. When FSaII tumors were subjected to a partial thermal ablation treatment using high intensity focused ultrasound (HIFU) there was consistently elevated detection of fluorescently labeled and i.v. injected endothelial precursors in the tumor when analyzed with optical imaging and/or histological preparations. Importantly, we also observed that BOECs treated with the novel anti-angiogenic peptide anginex in-vitro, show decreased proliferation and increased sensitivity to radiation. In vivo, the normal increase in FSaII tumor growth induced by injected BOECs was blunted by the addition of anginex treatment. It appears that endothelial precursors may significantly contribute to tumor vessel growth, tumor progression and/or repair of tumor damage and may improve the oxygenation and subsequent radiation response of tumors. We surmise that these cells are preferentially stimulated to divide in the tumor microenvironment, thereby inducing the significant increase in tumor growth observed and that the use of injected BOECs could be a viable approach to modulate the tumor microenvironment for therapeutic gain. Conversely, agents or approaches to block their recruitment and integration of BOECs into primary or metastatic lesions may be an effective way to restrain cancer progression before or after other treatments are applied.
Collapse
|
49
|
Trends in tissue engineering for blood vessels. J Biomed Biotechnol 2012; 2012:956345. [PMID: 23251085 PMCID: PMC3518873 DOI: 10.1155/2012/956345] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2012] [Accepted: 09/25/2012] [Indexed: 11/18/2022] Open
Abstract
Over the years, cardiovascular diseases continue to increase and affect not only human health but also the economic stability worldwide. The advancement in tissue engineering is contributing a lot in dealing with this immediate need of alleviating human health. Blood vessel diseases are considered as major cardiovascular health problems. Although blood vessel transplantation is the most convenient treatment, it has been delimited due to scarcity of donors and the patient's conditions. However, tissue-engineered blood vessels are promising alternatives as mode of treatment for blood vessel defects. The purpose of this paper is to show the importance of the advancement on biofabrication technology for treatment of soft tissue defects particularly for vascular tissues. This will also provide an overview and update on the current status of tissue reconstruction especially from autologous stem cells, scaffolds, and scaffold-free cellular transplantable constructs. The discussion of this paper will be focused on the historical view of cardiovascular tissue engineering and stem cell biology. The representative studies featured in this paper are limited within the last decade in order to trace the trend and evolution of techniques for blood vessel tissue engineering.
Collapse
|
50
|
Baiguera S, Ribatti D. Endothelialization approaches for viable engineered tissues. Angiogenesis 2012; 16:1-14. [PMID: 23010872 DOI: 10.1007/s10456-012-9307-8] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2012] [Accepted: 09/15/2012] [Indexed: 12/21/2022]
Abstract
One of the main limitation in obtaining thick, 3-dimensional viable engineered constructs is the inability to provide a sufficient and functional blood vessel system essential for the in vitro survival and the in vivo integration of the construct. Different strategies have been proposed to simulate the ingrowth of new blood vessels into engineered tissue, such as the use of growth factors, fabrication scaffold technologies, in vivo prevascularization and cell-based strategies, and it has been demonstrated that endothelial cells play a central role in the neovascularization process and in the control of blood vessel function. In particular, different "environmental" settings (origin, presence of supporting cells, biomaterial surface, presence of hemodynamic forces) strongly influence endothelial cell function, angiogenic potential and the in vivo formation of durable vessels. This review provides an overview of the different techniques developed so far for the vascularization of tissue-engineered constructs (with their advantages and pitfalls), focusing the attention on the recent development in the cell-based vascularization strategy and the in vivo applications.
Collapse
Affiliation(s)
- Silvia Baiguera
- BIOAIRLab, European Center for Thoracic Surgery, University Hospital Careggi, Florence, Italy.
| | | |
Collapse
|