1
|
Jones S, VandenHeuvel S, Luengo Martinez A, Birur R, Burgeson E, Gilbert I, Baker A, Wolf M, Raghavan SA, Rogers S, Cosgriff-Hernandez E. Suspension electrospinning of decellularized extracellular matrix: A new method to preserve bioactivity. Bioact Mater 2024; 41:640-656. [PMID: 39280898 PMCID: PMC11401211 DOI: 10.1016/j.bioactmat.2024.08.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 08/12/2024] [Accepted: 08/14/2024] [Indexed: 09/18/2024] Open
Abstract
Decellularized extracellular matrices (dECM) have strong regenerative potential as tissue engineering scaffolds; however, current clinical options for dECM scaffolds are limited to freeze-drying its native form into sheets. Electrospinning is a versatile scaffold fabrication technique that allows control of macro- and microarchitecture. It remains challenging to electrospin dECM, which has led researchers to either blend it with synthetic materials or use enzymatic digestion to fully solubilize the dECM. Both strategies reduce the innate bioactivity of dECM and limit its regenerative potential. Herein, we developed a new suspension electrospinning method to fabricate a pure dECM fibrous mesh that retains its innate bioactivity. Systematic investigation of suspension parameters was used to identify critical rheological properties required to instill "spinnability," including homogenization, concentration, and particle size. Homogenization enhanced particle interaction to impart the requisite elastic behavior to withstand electrostatic drawing without breaking. A direct correlation between concentration and viscosity was observed that altered fiber morphology; whereas, particle size had minimal impact on suspension properties and fiber morphology. The versatility of this new method was demonstrated by electrospinning dECM with three common decellularization techniques (Abraham, Badylak, Luo) and tissue sources (intestinal submucosa, heart, skin). Bioactivity retention after electrospinning was confirmed using cell proliferation, angiogenesis, and macrophage polarization assays. Collectively, these findings provide a framework for researchers to electrospin dECM for diverse tissue engineering applications.
Collapse
Affiliation(s)
- Sarah Jones
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Sabrina VandenHeuvel
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, 77843, USA
| | - Andres Luengo Martinez
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Ruchi Birur
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Eric Burgeson
- Department of Chemical and Biomolecular Engineering, University of Illinois Urbana-Champaign, Champaign, IL, 61820, USA
| | - Isabelle Gilbert
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Aaron Baker
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Matthew Wolf
- Cancer Biomaterials Engineering Section, Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD, 21702, USA
| | - Shreya A Raghavan
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, 77843, USA
| | - Simon Rogers
- Department of Chemical and Biomolecular Engineering, University of Illinois Urbana-Champaign, Champaign, IL, 61820, USA
| | | |
Collapse
|
2
|
Gorashi RM, Baddour T, Chittle SJ, Vélez NEF, Wenning MA, Anseth KS, Mestroni L, Peña B, Guo P, Aguado BA. Y chromosome linked UTY modulates sex differences in valvular fibroblast methylation in response to nanoscale extracellular matrix cues. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.13.593760. [PMID: 38798394 PMCID: PMC11118428 DOI: 10.1101/2024.05.13.593760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Aortic valve stenosis (AVS) is a progressive disease wherein males more often develop valve calcification relative to females that develop valve fibrosis. Valvular interstitial cells (VICs) aberrantly activate to myofibroblasts during AVS, driving the fibrotic valve phenotype in females. Myofibroblasts further differentiate into osteoblast-like cells and produce calcium nanoparticles, driving valve calcification in males. We hypothesized the lysine demethylase UTY (ubiquitously transcribed tetratricopeptide repeat containing, Y-linked) decreases methylation uniquely in male VICs responding to nanoscale extracellular matrix cues to promote an osteoblast-like cell phenotype. Here, we describe a hydrogel biomaterial cell culture platform to interrogate how nanoscale cues modulate sex-specific methylation states in VICs activating to myofibroblasts and osteoblast-like cells. We found UTY modulates the osteoblast-like cell phenotype in response to nanoscale cues uniquely in male VICs. Overall, we reveal a novel role of UTY in the regulation of calcification processes in males during AVS progression.
Collapse
|
3
|
Wancura M, Nkansah A, Robinson A, Toubbeh S, Talanker M, Jones S, Cosgriff-Hernandez E. PEG-Based Hydrogel Coatings: Design Tools for Biomedical Applications. Ann Biomed Eng 2024; 52:1804-1815. [PMID: 36774427 DOI: 10.1007/s10439-023-03154-9] [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: 11/08/2022] [Accepted: 01/16/2023] [Indexed: 02/13/2023]
Abstract
Device failure due to undesired biological responses remains a substantial roadblock in the development and translation of new devices into clinical care. Polyethylene glycol (PEG)-based hydrogel coatings can be used to confer antifouling properties to medical devices-enabling minimization of biological responses such as bacterial infection, thrombosis, and foreign body reactions. Application of hydrogel coatings to diverse substrates requires careful consideration of multiple material factors. Herein, we report a systematic investigation of two coating methods: (1) traditional photoinitiated hydrogel coatings; (2) diffusion-mediated, redox-initiated hydrogel coatings. The effects of method, substrate, and compositional variables on the resulting hydrogel coating thickness are presented. To expand the redox-based method to include high molecular weight macromers, a mechanistic investigation of the role of cure rate and macromer viscosity was necessary to balance solution infiltration and gelation. Overall, these structure-property relationships provide users with a toolbox for hydrogel coating design for a broad range of medical devices.
Collapse
Affiliation(s)
- Megan Wancura
- Department of Chemistry, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Abbey Nkansah
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Andrew Robinson
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Shireen Toubbeh
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Michael Talanker
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Sarah Jones
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Elizabeth Cosgriff-Hernandez
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, 78712, USA.
- Department of Biomedical Engineering, The University of Texas at Austin, 107 W. Dean Keeton, BME Building, Room 3.503D, Austin, TX, 78712, USA.
| |
Collapse
|
4
|
Robinson A, Nkansah A, Bhat S, Karnik S, Jones S, Fairley A, Leung J, Wancura M, Sacks MS, Dasi LP, Cosgriff-Hernandez E. Hydrogel-polyurethane fiber composites with enhanced microarchitectural control for heart valve replacement. J Biomed Mater Res A 2024; 112:586-599. [PMID: 38018452 DOI: 10.1002/jbm.a.37641] [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: 06/08/2023] [Revised: 09/22/2023] [Accepted: 10/24/2023] [Indexed: 11/30/2023]
Abstract
Polymeric heart valves offer the potential to overcome the limited durability of tissue based bioprosthetic valves and the need for anticoagulant therapy of mechanical valve replacement options. However, developing a single-phase material with requisite biological properties and target mechanical properties remains a challenge. In this study, a composite heart valve material was developed where an electrospun mesh provides tunable mechanical properties and a hydrogel coating confers an antifouling surface for thromboresistance. Key biological responses were evaluated in comparison to glutaraldehyde-fixed pericardium. Platelet and bacterial attachment were reduced by 38% and 98%, respectively, as compared to pericardium that demonstrated the antifouling nature of the hydrogel coating. There was also a notable reduction (59%) in the calcification of the composite material as compared to pericardium. A custom 3D-printed hydrogel coating setup was developed to make valve composites for device-level hemodynamic testing. Regurgitation fraction (9.6 ± 1.8%) and effective orifice area (1.52 ± 0.34 cm2 ) met ISO 5840-2:2021 requirements. Additionally, the mean pressure gradient was comparable to current clinical bioprosthetic heart valves demonstrating preliminary efficacy. Although the hemodynamic properties are promising, it is anticipated that the random microarchitecture will result in suboptimal strain fields and peak stresses that may accelerate leaflet fatigue and degeneration. Previous computational work has demonstrated that bioinspired fiber microarchitectures can improve strain homogeneity of valve materials toward improving durability. To this end, we developed advanced electrospinning methodologies to achieve polyurethane fiber microarchitectures that mimic or exceed the physiological ranges of alignment, tortuosity, and curvilinearity present in the native valve. Control of fiber alignment from a random fiber orientation at a normalized orientation index (NOI) 14.2 ± 6.9% to highly aligned fibers at a NOI of 85.1 ± 1.4%. was achieved through increasing mandrel rotational velocity. Fiber tortuosity and curvilinearity in the range of native valve features were introduced through a post-spinning annealing process and fiber collection on a conical mandrel geometry, respectively. Overall, these studies demonstrate the potential of hydrogel-polyurethane fiber composite as a heart valve material. Future studies will utilize the developed advanced electrospinning methodologies in combination with model-directed fabrication toward optimizing durability as a function of fiber microarchitecture.
Collapse
Affiliation(s)
- Andrew Robinson
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas, USA
| | - Abbey Nkansah
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas, USA
| | - Sanchita Bhat
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Shweta Karnik
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Sarah Jones
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas, USA
| | - Ashauntee Fairley
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas, USA
| | - Jonathan Leung
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas, USA
| | - Megan Wancura
- Department of Chemistry, The University of Texas at Austin, Austin, Texas, USA
| | - Michael S Sacks
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas, USA
- James T. Willerson Center for Cardiovascular Modeling and Simulation, The Oden Institute for Computational Engineering and Sciences, Austin, Texas, USA
- Department of Mechanical Engineering, The University of Texas at Austin, Austin, Texas, USA
| | - Lakshmi Prasad Dasi
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA
| | | |
Collapse
|
5
|
Pedersen DD, Kim S, D'Amore A, Wagner WR. Influence of Polymer Stiffness and Geometric Design on Fluid Mechanics in Tissue-Engineered Pulmonary Valve Scaffolds. Ann Biomed Eng 2024; 52:575-587. [PMID: 37935910 DOI: 10.1007/s10439-023-03401-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 10/29/2023] [Indexed: 11/09/2023]
Abstract
There is still much unknown about the fluid mechanical response to cardiac valve scaffolds, even as their implementation in the clinic is on the horizon. Specifically, while degradable polymer valve scaffolds are currently being tested in the pulmonary valve position, their material and mechanical properties have not been fully elucidated. Optimizing these properties are important determinants not only of acute function, but long-term remodeling prospects. This study aimed to characterize fluid profiles downstream of electrospun valve scaffolds under dynamic pulmonary conditions. Valve scaffold design was changed by either blending poly(carbonate urethane) urea (PCUU) with poly(ε-caprolactone) (PCL) to modulate material stiffness or by changing the geometric design of the valve scaffolds. Specifically, two designs were utilized: one modeled after a clinically used bioprosthetic valve design (termed Mk1 design), and another using a geometrically "optimized" design (termed Mk2) based on anatomical data. Particle image velocimetry results showed that material stiffness only had a mild impact on fluid mechanics, measured by velocity magnitude, vorticity, viscous shear stress, Reynolds shear stress, and turbulent kinetic energy. However, comparing the two geometric designs yielded a much greater impact, with the Mk2 valve groups containing the highest PCUU/PCL ratio demonstrating the overall best performance. This report highlights the easily manipulable design features of polymeric valve scaffolds and demonstrates their relative significance for valve function.
Collapse
Affiliation(s)
- Drake D Pedersen
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Seungil Kim
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Antonio D'Amore
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, USA
- Fondazione Ri.MED, Palermo, Italy
- Clinical and Translational Science Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - William R Wagner
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA.
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA.
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, USA.
- Department of Chemical Engineering, University of Pittsburgh, Pittsburgh, PA, USA.
- Clinical and Translational Science Institute, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
6
|
Pedersen DD, Kim S, D'Amore A, Wagner WR. Cardiac valve scaffold design: Implications of material properties and geometric configuration on performance and mechanics. J Mech Behav Biomed Mater 2023; 146:106043. [PMID: 37531773 DOI: 10.1016/j.jmbbm.2023.106043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 06/07/2023] [Accepted: 07/22/2023] [Indexed: 08/04/2023]
Abstract
Development of tissue engineered scaffolds for cardiac valve replacement is nearing clinical translation. While much work has been done to characterize mechanical behavior of native and bioprosthetic valves, and incorporate those data into models improving valve design, similar work for degradable valve scaffolds is lacking. This is particularly important given the implications mechanics have on short-term survival and long-term remodeling. As such, this study aimed to characterize spatially-resolved strain profiles on the leaflets of degradable polymeric valve scaffolds, manipulating common design features such as material stiffness by blending poly(carbonate urethane)urea with stiffer polymers, and geometric configuration, modeled after either a clinically-used valve design (Mk1 design) or an anatomically "optimized" design (Mk2 design). It was shown that material stiffness plays a significant role in overall valve performance, with the stiffest valve groups showing asymmetric and incomplete opening during systole. However, the geometric configuration had a significantly greater effect on valve performance as well as strain magnitude and distribution. Major findings in the strain maps included systolic strains having overall higher strain magnitudes than diastole, and peak radial-direction strain concentrations in the base region of Mk1 valves during systole, with a significant mitigation of radial strain in Mk2 valves. The high tunability of tissue engineered scaffolds is a great advantage for valve design, and the results reported here indicate that design parameters have significant and unequal impact on valve performance and mechanics.
Collapse
Affiliation(s)
- Drake D Pedersen
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh, PA, USA
| | - Seungil Kim
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, PA, USA; Department of Surgery, University of Pittsburgh, PA, USA
| | - Antonio D'Amore
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh, PA, USA; Department of Surgery, University of Pittsburgh, PA, USA; Fondazione Ri.MED, Palermo, Italy; Clinical and Translational Science Institute, University of Pittsburgh, PA, USA
| | - William R Wagner
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh, PA, USA; Department of Surgery, University of Pittsburgh, PA, USA; Department of Chemical Engineering, University of Pittsburgh, PA, USA; Clinical and Translational Science Institute, University of Pittsburgh, PA, USA.
| |
Collapse
|
7
|
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: 1.0] [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
|
8
|
Łopianiak I, Rzempołuch W, Civelek M, Cicha I, Ciach T, Butruk-Raszeja BA. Multilayered blow-spun vascular prostheses with luminal surfaces in Nano/Micro range: the influence on endothelial cell and platelet adhesion. J Biol Eng 2023; 17:20. [PMID: 36915145 PMCID: PMC10012602 DOI: 10.1186/s13036-023-00337-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 03/05/2023] [Indexed: 03/14/2023] Open
Abstract
BACKGROUND In this study, two types of polyurethane-based cylindrical multilayered grafts with internal diameters ≤ 6 mm were produced by the solution blow spinning (SBS) method. The main aim was to create layered-wall prostheses differing in their luminal surface morphology. Changing the SBS process parameters, i.e. working distance, rotational speed, volume, and concentration of the polymer solution allowed to obtain structures with the required morphologies. The first type of prostheses, termed Nano, possessed nanofibrous luminal surface, and the second type, Micro, presented morphologically diverse luminal surface, with both solid and microfibrous areas. RESULTS The results of mechanical tests confirmed that designed prostheses had high flexibility (Young's modulus value of about 2.5 MPa) and good tensile strength (maximum axial load value of about 60 N), which meet the requirements for vascular prostheses. The influence of the luminal surface morphology on platelet adhesion and the attachment of endothelial cells was investigated. Both surfaces did not cause hemolysis in contact with blood, the percentage of platelet-occupied area for Nano and Micro surfaces was comparable to reference polytetrafluoroethylene (PTFE) surface. However, the change in morphology of surface-adhered platelets between Nano and Micro surfaces was visible, which might suggest differences in their activation level. Endothelial coverage after 1, 3, and 7 days of culture on flat samples (2D model) was higher on Nano prostheses as compared with Micro scaffolds. However, this effect was not seen in 3D culture, where cylindrical prostheses were colonized using magnetic seeding method. CONCLUSIONS We conclude the produced scaffolds meet the material and mechanical requirements for vascular prostheses. However, changing the morphology without changing the chemical modification of the luminal surface is not sufficient to achieve the appropriate effectiveness of endothelialization in the 3D model.
Collapse
Affiliation(s)
- Iwona Łopianiak
- Faculty of Chemical and Process Engineering, Warsaw University of Technology, Waryńskiego 1, 00-645, Warsaw, Poland.,Doctoral School of Warsaw University of Technology, Warsaw University of Technology, Pl. Politechniki 1, 00-661, Warsaw, Poland
| | - Wiktoria Rzempołuch
- Faculty of Chemical and Process Engineering, Warsaw University of Technology, Waryńskiego 1, 00-645, Warsaw, Poland
| | - Mehtap Civelek
- Section of Experimental Oncology Und Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung-Professorship, ENT-Department, Universitätsklinikum, Erlangen, Germany
| | - Iwona Cicha
- Section of Experimental Oncology Und Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung-Professorship, ENT-Department, Universitätsklinikum, Erlangen, Germany
| | - Tomasz Ciach
- Faculty of Chemical and Process Engineering, Warsaw University of Technology, Waryńskiego 1, 00-645, Warsaw, Poland.,Centre for Advanced Materials and Technologies CEZAMAT, Warsaw University of Technology, Poleczki 19, 02-822, Warsaw, Poland
| | - Beata A Butruk-Raszeja
- Faculty of Chemical and Process Engineering, Warsaw University of Technology, Waryńskiego 1, 00-645, Warsaw, Poland.
| |
Collapse
|
9
|
Singh S, Kumar Paswan K, Kumar A, Gupta V, Sonker M, Ashhar Khan M, Kumar A, Shreyash N. Recent Advancements in Polyurethane-based Tissue Engineering. ACS APPLIED BIO MATERIALS 2023; 6:327-348. [PMID: 36719800 DOI: 10.1021/acsabm.2c00788] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
In tissue engineering, polyurethane-based implants have gained significant traction because of their high compatibility and inertness. The implants therefore show fewer side effects and lasts longer. Also, the mechanical properties can be tuned and morphed into a particular shape, owing to which polyurethanes show immense versatility. In the last 3 years, scientists have devised methods to enhance the strength of and induce dynamic properties in polyurethanes, and these developments offer an immense opportunity to use them in tissue engineering. The focus of this review is on applications of polyurethane implants for biomedical application with detailed analysis of hard tissue implants like bone tissues and soft tissues like cartilage, muscles, skeletal tissues, and blood vessels. The synthetic routes for the preparation of scaffolds have been discussed to gain a better understanding of the issues that arise regarding toxicity. The focus here is also on concerns regarding the biocompatibility of the implants, given that the precursors and byproducts are poisonous.
Collapse
Affiliation(s)
- Sukriti Singh
- Department of Chemical and Biochemical Engineering, Rajiv Gandhi Institute of Petroleum Technology, Mubarakpur Mukhatiya, Uttar Pradesh 229304, India
| | - Karan Kumar Paswan
- Department of Chemical and Biochemical Engineering, Rajiv Gandhi Institute of Petroleum Technology, Mubarakpur Mukhatiya, Uttar Pradesh 229304, India
| | - Alok Kumar
- Department of Chemical and Biochemical Engineering, Rajiv Gandhi Institute of Petroleum Technology, Mubarakpur Mukhatiya, Uttar Pradesh 229304, India
| | - Vishwas Gupta
- Department of Petroleum Engineering, Rajiv Gandhi Institute of Petroleum Technology, Mubarakpur Mukhatiya, Uttar Pradesh 229304, India
| | - Muskan Sonker
- Department of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| | - Mohd Ashhar Khan
- Department of Chemical and Biological Engineering, State University of New York at Buffalo, Buffalo, New York 14260, United States
| | - Amrit Kumar
- Indian Oil Corporation Limited, Panipat Refinery, Panipat, Odisha 132140, India
| | - Nehil Shreyash
- Department of Chemical and Biomolecular Engineering, University of Houston, Houston, Texas 77204, United States
| |
Collapse
|
10
|
Chen T, Guo X, Huang Y, Hao W, Deng S, Xu G, Bao J, Xiong Q, Yang W. Bletilla Striata polysaccharide - Waterborne polyurethane hydrogel as a wound dressing. JOURNAL OF BIOMATERIALS SCIENCE, POLYMER EDITION 2022:1-14. [DOI: 10.1080/09205063.2022.2157673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Tianyu Chen
- College of Chemistry and Chemical Engineering, Anhui University, Hefei, China, 230601
| | - Xiaoyan Guo
- College of Chemistry and Chemical Engineering, Anhui University, Hefei, China, 230601
| | - Yiping Huang
- College of Chemistry and Chemical Engineering, Anhui University, Hefei, China, 230601
| | - Wentao Hao
- Anhui Key Laboratory of advanced catalytic materials and reaction engineering, School of Chemistry and Chemical Engineering, Hefei University of Technology, Hefei, China, 230009
| | - Sunyan Deng
- College of Chemistry and Chemical Engineering, Anhui University, Hefei, China, 230601
| | - Gewen Xu
- College of Chemistry and Chemical Engineering, Anhui University, Hefei, China, 230601
| | - Junjie Bao
- College of Chemistry and Chemical Engineering, Anhui University, Hefei, China, 230601
| | - Qiansheng Xiong
- College of Chemistry and Chemical Engineering, Anhui University, Hefei, China, 230601
| | - Wen Yang
- Anhui Key Laboratory of advanced catalytic materials and reaction engineering, School of Chemistry and Chemical Engineering, Hefei University of Technology, Hefei, China, 230009
| |
Collapse
|
11
|
Visser D, Bakhshi H, Rogg K, Fuhrmann E, Wieland F, Schenke-Layland K, Meyer W, Hartmann H. Green Chemistry for Biomimetic Materials: Synthesis and Electrospinning of High-Molecular-Weight Polycarbonate-Based Nonisocyanate Polyurethanes. ACS OMEGA 2022; 7:39772-39781. [PMID: 36385898 PMCID: PMC9648058 DOI: 10.1021/acsomega.2c03731] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 10/14/2022] [Indexed: 06/16/2023]
Abstract
Conventional synthesis routes for thermoplastic polyurethanes (TPUs) still require the use of isocyanates and tin-based catalysts, which pose considerable safety and environmental hazards. To reduce both the ecological footprint and human health dangers for nonwoven TPU scaffolds, it is key to establish a green synthesis route, which eliminates the use of these toxic compounds and results in biocompatible TPUs with facile processability. In this study, we developed high-molecular-weight nonisocyanate polyurethanes (NIPUs) through transurethanization of 1,6-hexanedicarbamate with polycarbonate diols (PCDLs). Various molecular weights of PCDL were employed to maximize the molecular weight of NIPUs and consequently facilitate their electrospinnability. The synthesized NIPUs were characterized by nuclear magnetic resonance, Fourier-transform infrared spectroscopy, gel permeation chromatography, and differential scanning calorimetry. The highest achieved molecular weight (M w) was 58,600 g/mol. The NIPUs were consecutively electrospun into fibrous scaffolds with fiber diameters in the submicron range, as shown by scanning electron microscopy (SEM). To assess the suitability of electrospun NIPU mats as a possible biomimetic load-bearing pericardial substitute in cardiac tissue engineering, their cytotoxicity was investigated in vitro using primary human fibroblasts and a human epithelial cell line. The bare NIPU mats did not need further biofunctionalization to enhance cell adhesion, as it was not outperformed by collagen-functionalized NIPU mats and hence showed that the NIPU mats possess a great potential for use in biomimetic scaffolds.
Collapse
Affiliation(s)
- Dmitri Visser
- NMI
Natural and Medical Science Institute at the University of Tübingen, Markwiesenstrasse 55, 72770 Reutlingen, Germany
| | - Hadi Bakhshi
- Department
of Life Science and Bioprocesses, Fraunhofer
Institute for Applied Polymer Research IAP, Geiselbergstraße 69, 14476 Potsdam, Germany
- Department
of Functional Polymer Systems, Fraunhofer
Institute for Applied Polymer Research IAP, Geiselbergstraße 69, 14476 Potsdam, Germany
| | - Katharina Rogg
- NMI
Natural and Medical Science Institute at the University of Tübingen, Markwiesenstrasse 55, 72770 Reutlingen, Germany
| | - Ellena Fuhrmann
- NMI
Natural and Medical Science Institute at the University of Tübingen, Markwiesenstrasse 55, 72770 Reutlingen, Germany
| | - Franziska Wieland
- Department
of Functional Polymer Systems, Fraunhofer
Institute for Applied Polymer Research IAP, Geiselbergstraße 69, 14476 Potsdam, Germany
| | - Katja Schenke-Layland
- NMI
Natural and Medical Science Institute at the University of Tübingen, Markwiesenstrasse 55, 72770 Reutlingen, Germany
- Institute
of Biomedical Engineering, Dept. for Medical Technologies and Regenerative
Medicine, Eberhard Karls University Tübingen, 72076 Tübingen, Germany
- Cluster
of Excellence iFIT (EXC 2180) “Image-Guided and Functionally
Instructed Tumor Therapies”, Eberhard
Karls University Tübingen, 72076 Tübingen, Germany
| | - Wolfdietrich Meyer
- Department
of Life Science and Bioprocesses, Fraunhofer
Institute for Applied Polymer Research IAP, Geiselbergstraße 69, 14476 Potsdam, Germany
- Department
of Functional Polymer Systems, Fraunhofer
Institute for Applied Polymer Research IAP, Geiselbergstraße 69, 14476 Potsdam, Germany
| | - Hanna Hartmann
- NMI
Natural and Medical Science Institute at the University of Tübingen, Markwiesenstrasse 55, 72770 Reutlingen, Germany
| |
Collapse
|
12
|
Mehta SM, De Santos DR, Sridhar S, Aguayo VC, Meraz CA, Mikos M, Grande-Allen KJ. Fabricating a Low-Cost, Microscopy-Compatible Mechanical Testing Device. EXPERIMENTAL TECHNIQUES 2022; 46:731-743. [PMID: 39119455 PMCID: PMC11308025 DOI: 10.1007/s40799-021-00513-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 09/16/2021] [Indexed: 08/10/2024]
Abstract
Most commercially-available mechanical testing devices are bulky, expensive, and unable to evaluate changes in sample microstructure under load. This leaves a crucial gap in understanding between material structure and bulk mechanical properties. Our objective was to fabricate a mechanical testing device small enough to fit in most upright or inverted microscopy stages and able to position samples to allow for simultaneous mechanical and microstructural characterization. Parts were 3D printed using the hobbyist-friendly Fused Filament Fabrication technique, then assembled with commercial fasteners and translation components to create a mechanical testing device that utilized the deflection of plastic posts to determine sample reaction forces under applied strain. Video of sample deformation was analyzed using a custom processing script to calculate stress and strain behavior in an automated and high-throughput manner. This device was able to perform mechanical characterization with an accuracy comparable to commercial mechanical testing devices for a wide range of nonlinear and viscoelastic samples under dry and hydrated conditions. Additionally, the device showed compatibility with different upright and inverted microscopes and was able to demonstrate accurate mechanical testing results when used with these instruments. We successfully developed a device capable of accurately testing a majority of soft materials in the field of Biomedical Engineering with the ability to perform additional microstructural characterization using microscopy at a price point of $600.
Collapse
Affiliation(s)
- S M Mehta
- Department of Bioengineering, Rice University, Houston, TX, 77005
| | - D R De Santos
- Department of Mechanical Engineering, University of Texas at El Paso, El Paso, TX 79968
| | - S Sridhar
- Department of Bioengineering, Rice University, Houston, TX, 77005
| | - V C Aguayo
- Department of Biomedical Engineering, Texas A&M, College Station, TX 77843
| | - C A Meraz
- Department of Mechanical Engineering, University of Texas at El Paso, El Paso, TX 79968
| | - M Mikos
- Department of Bioengineering, Rice University, Houston, TX, 77005
| | - K J Grande-Allen
- Department of Bioengineering, Rice University, Houston, TX, 77005
| |
Collapse
|
13
|
Xu C, Hong Y. Rational design of biodegradable thermoplastic polyurethanes for tissue repair. Bioact Mater 2022; 15:250-271. [PMID: 35386346 PMCID: PMC8940769 DOI: 10.1016/j.bioactmat.2021.11.029] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 11/09/2021] [Accepted: 11/24/2021] [Indexed: 12/25/2022] Open
Abstract
As a type of elastomeric polymers, non-degradable polyurethanes (PUs) have a long history of being used in clinics, whereas biodegradable PUs have been developed in recent decades, primarily for tissue repair and regeneration. Biodegradable thermoplastic (linear) PUs are soft and elastic polymeric biomaterials with high mechanical strength, which mimics the mechanical properties of soft and elastic tissues. Therefore, biodegradable thermoplastic polyurethanes are promising scaffolding materials for soft and elastic tissue repair and regeneration. Generally, PUs are synthesized by linking three types of changeable blocks: diisocyanates, diols, and chain extenders. Alternating the combination of these three blocks can finely tailor the physio-chemical properties and generate new functional PUs. These PUs have excellent processing flexibilities and can be fabricated into three-dimensional (3D) constructs using conventional and/or advanced technologies, which is a great advantage compared with cross-linked thermoset elastomers. Additionally, they can be combined with biomolecules to incorporate desired bioactivities to broaden their biomedical applications. In this review, we comprehensively summarized the synthesis, structures, and properties of biodegradable thermoplastic PUs, and introduced their multiple applications in tissue repair and regeneration. A whole picture of their design and applications along with discussions and perspectives of future directions would provide theoretical and technical supports to inspire new PU development and novel applications.
Collapse
Affiliation(s)
- Cancan Xu
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX, 76019, USA
| | - Yi Hong
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX, 76019, USA
| |
Collapse
|
14
|
Design of a Mechanobioreactor to Apply Anisotropic, Biaxial Strain to Large Thin Biomaterials for Tissue Engineered Heart Valve Applications. Ann Biomed Eng 2022; 50:1073-1089. [PMID: 35622208 DOI: 10.1007/s10439-022-02984-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 05/16/2022] [Indexed: 01/05/2023]
Abstract
Repair and replacement solutions for congenitally diseased heart valves capable of post-surgery growth and adaptation have remained elusive. Tissue engineered heart valves (TEHVs) offer a potential biological solution that addresses the drawbacks of existing valve replacements. Typically, TEHVs are made from thin, fibrous biomaterials that either become cell populated in vitro or in situ. Often, TEHV designs poorly mimic the anisotropic mechanical properties of healthy native valves leading to inadequate biomechanical function. Mechanical conditioning of engineered tissues with anisotropic strain application can induce extracellular matrix remodelling to alter the anisotropic mechanical properties of a construct, but implementation has been limited to small-scale set-ups. To address this limitation for TEHV applications, we designed and built a mechanobioreactor capable of modulating biaxial strain anisotropy applied to large, thin, biomaterial sheets in vitro. The bioreactor can independently control two orthogonal stretch axes to modulate applied strain anisotropy on biomaterial sheets from 13 × 13 mm2 to 70 × 40 mm2. A design of experiments was performed using experimentally validated finite element (FE) models and demonstrated that biaxial strain was applied uniformly over a larger percentage of the cell seeded area for larger sheets (13 × 13 mm2: 58% of sheet area vs. 52 × 31 mm2: 86% of sheet area). Furthermore, bioreactor prototypes demonstrated that over 70% of the cell seeding area remained uniformly strained under different prescribed protocols: equibiaxial amplitudes between 5 to 40%, cyclic frequencies between 0.1 to 2.5 Hz and anisotropic strain ratios between 0:1 (constrained uniaxial) to 2:1. Lastly, proof-of-concept experiments were conducted where we applied equibiaxial (εx = εy = 8.75%) and anisotropic (εx = 12.5%, εy = 5%) strain protocols to cell-seeded, electrospun scaffolds. Cell nuclei and F-actin aligned to the vector-sum strain direction of each prescribed protocol (nuclei alignment: equibiaxial: 43.2° ± 1.8°, anisotropic: 17.5° ± 1.7°; p < 0.001). The abilities of this bioreactor to prescribe different strain amplitude, frequency and strain anisotropy protocols to cell-seeded scaffolds will enable future studies into the effects of anisotropic loading protocols on mechanically conditioned TEHVs and other engineered planar connective tissues.
Collapse
|
15
|
Natural Polymers in Heart Valve Tissue Engineering: Strategies, Advances and Challenges. Biomedicines 2022; 10:biomedicines10051095. [PMID: 35625830 PMCID: PMC9139175 DOI: 10.3390/biomedicines10051095] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 05/03/2022] [Accepted: 05/04/2022] [Indexed: 12/04/2022] Open
Abstract
In the history of biomedicine and biomedical devices, heart valve manufacturing techniques have undergone a spectacular evolution. However, important limitations in the development and use of these devices are known and heart valve tissue engineering has proven to be the solution to the problems faced by mechanical and prosthetic valves. The new generation of heart valves developed by tissue engineering has the ability to repair, reshape and regenerate cardiac tissue. Achieving a sustainable and functional tissue-engineered heart valve (TEHV) requires deep understanding of the complex interactions that occur among valve cells, the extracellular matrix (ECM) and the mechanical environment. Starting from this idea, the review presents a comprehensive overview related not only to the structural components of the heart valve, such as cells sources, potential materials and scaffolds fabrication, but also to the advances in the development of heart valve replacements. The focus of the review is on the recent achievements concerning the utilization of natural polymers (polysaccharides and proteins) in TEHV; thus, their extensive presentation is provided. In addition, the technological progresses in heart valve tissue engineering (HVTE) are shown, with several inherent challenges and limitations. The available strategies to design, validate and remodel heart valves are discussed in depth by a comparative analysis of in vitro, in vivo (pre-clinical models) and in situ (clinical translation) tissue engineering studies.
Collapse
|
16
|
Wu S, Li Y, Zhang C, Tao L, Kuss M, Lim JY, Butcher J, Duan B. Tri-Layered and Gel-Like Nanofibrous Scaffolds with Anisotropic Features for Engineering Heart Valve Leaflets. Adv Healthc Mater 2022; 11:e2200053. [PMID: 35289986 PMCID: PMC10976923 DOI: 10.1002/adhm.202200053] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 02/28/2022] [Indexed: 12/17/2022]
Abstract
3D heterogeneous and anisotropic scaffolds that approximate native heart valve tissues are indispensable for the successful construction of tissue engineered heart valves (TEHVs). In this study, novel tri-layered and gel-like nanofibrous scaffolds, consisting of poly(lactic-co-glycolic) acid (PLGA) and poly(aspartic acid) (PASP), are fabricated by a combination of positive/negative conjugate electrospinning and bioactive hydrogel post-processing. The nanofibrous PLGA-PASP scaffolds present tri-layered structures, resulting in anisotropic mechanical properties that are comparable with native heart valve leaflets. Biological tests show that nanofibrous PLGA-PASP scaffolds with high PASP ratios significantly promote the proliferation and collagen and glycosaminoglycans (GAGs) secretions of human aortic valvular interstitial cells (HAVICs), compared to PLGA scaffolds. Importantly, the nanofibrous PLGA-PASP scaffolds are found to effectively inhibit the osteogenic differentiation of HAVICs. Two types of porcine VICs, from young and adult age groups, are further seeded onto the PLGA-PASP scaffolds. The adult VICs secrete higher amounts of collagens and GAGs and undergo a significantly higher level of osteogenic differentiation than young VICs. RNA sequencing analysis indicates that age has a pivotal effect on the VIC behaviors. This study provides important guidance and a reference for the design and development of 3D tri-layered, gel-like nanofibrous PLGA-PASP scaffolds for TEHV applications.
Collapse
Affiliation(s)
- Shaohua Wu
- College of Textiles and Clothing, Qingdao University, Qingdao, 266071, China
- Mary & Dick Holland Regenerative Medicine Program and Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Yiran Li
- College of Textiles and Clothing, Qingdao University, Qingdao, 266071, China
| | - Caidan Zhang
- Key Laboratory of Yarn Materials Forming and Composite Processing Technology of Zhejiang Province, Jiaxing University, Jiaxing, 314001, China
| | - Litao Tao
- Department of Biomedical Science, Creighton University, Omaha, NE, 68178, USA
| | - Mitchell Kuss
- Mary & Dick Holland Regenerative Medicine Program and Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Jung Yul Lim
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA
| | - Jonathan Butcher
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, 14850, USA
| | - Bin Duan
- Mary & Dick Holland Regenerative Medicine Program and Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA
- Department of Surgery, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| |
Collapse
|
17
|
Pedersen DD, Kim S, Wagner WR. Biodegradable polyurethane scaffolds in regenerative medicine: Clinical translation review. J Biomed Mater Res A 2022; 110:1460-1487. [PMID: 35481723 DOI: 10.1002/jbm.a.37394] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 04/07/2022] [Accepted: 04/09/2022] [Indexed: 12/14/2022]
Abstract
Early explorations of tissue engineering and regenerative medicine concepts commonly utilized simple polyesters such as polyglycolide, polylactide, and their copolymers as scaffolds. These biomaterials were deemed clinically acceptable, readily accessible, and provided processability and a generally known biological response. With experience and refinement of approaches, greater control of material properties and integrated bioactivity has received emphasis and a broadened palette of synthetic biomaterials has been employed. Biodegradable polyurethanes (PUs) have emerged as an attractive option for synthetic scaffolds in a variety of tissue applications because of their flexibility in molecular design and ability to fulfill mechanical property objectives, particularly in soft tissue applications. Biodegradable PUs are highly customizable based on their composition and processability to impart tailored mechanical and degradation behavior. Additionally, bioactive agents can be readily incorporated into these scaffolds to drive a desired biological response. Enthusiasm for biodegradable PU scaffolds has soared in recent years, leading to rapid growth in the literature documenting novel PU chemistries, scaffold designs, mechanical properties, and aspects of biocompatibility. Despite the enthusiasm in the field, there are still few examples of biodegradable PU scaffolds that have achieved regulatory approval and routine clinical use. However, there is a growing literature where biodegradable PU scaffolds are being specifically developed for a wide range of pathologies and where relevant pre-clinical models are being employed. The purpose of this review is first to highlight examples of clinically used biodegradable PU scaffolds, and then to summarize the growing body of reports on pre-clinical applications of biodegradable PU scaffolds.
Collapse
Affiliation(s)
- Drake D Pedersen
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Seungil Kim
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - William R Wagner
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Chemical Engineering, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
18
|
Anisotropic elastic behavior of a hydrogel-coated electrospun polyurethane: Suitability for heart valve leaflets. J Mech Behav Biomed Mater 2022; 125:104877. [PMID: 34695661 PMCID: PMC8818123 DOI: 10.1016/j.jmbbm.2021.104877] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 09/20/2021] [Accepted: 09/30/2021] [Indexed: 01/03/2023]
Abstract
Although xenograft biomaterials have been used for decades in replacement heart valves, they continue to face multiple limitations, including limited durability, mineralization, and restricted design space due to their biological origins. These issues necessitate the need for novel replacement heart valve biomaterials that are durable, non-thrombogenic, and compatible with transcatheter aortic valve replacement devices. In this study, we explored the suitability of an electrospun poly(carbonate urethane) (ES-PCU) mesh coated with a poly(ethylene glycol) diacrylate (PEGDA) hydrogel as a synthetic biomaterial for replacement heart valve leaflets. In this material design, the mesh provides the mechanical support, while the hydrogel provides the required surface hemocompatibility. We conducted a comprehensive study to characterize the structural and mechanical properties of the uncoated mesh as well as the hydrogel-coated mesh (composite biomaterial) over the estimated operational range. We found that the composite biomaterial was functionally robust with reproducible stress-strain behavior within and beyond the functional ranges for replacement heart valves, and was able to withstand the rigors of mechanical evaluation without any observable damage. In addition, the composite biomaterial displayed a wide range of mechanical anisotropic responses, which were governed by fiber orientation of the mesh, which in turn, was controlled with the fabrication process. Finally, we developed a novel constitutive modeling approach to predict the mechanical behavior of the composite biomaterial under in-plane extension and shear deformation modes. This model identified the existence of fiber-fiber mechanical interactions in the mesh that have not previously been reported. Interestingly, there was no evidence of fiber-hydrogel mechanical interactions. This important finding suggests that the hydrogel coating can be optimized for hemocompatibility independent of the structural mechanical responses required by the leaflet. This initial study indicated that the composite biomaterial has mechanical properties well-suited for replacement heart valve applications and that the electrospun mesh microarchitecture and hydrogel biological properties can be optimized independently. It also reveals that the structural mechanisms contributing to the mechanical response are more complicated than what was previously established and paves the pathway for more detailed future studies.
Collapse
|
19
|
Li X, Yang Z, Fang L, Ma C, Zhao Y, Liu H, Che S, Zvyagin AV, Yang B, Lin Q. Hydrogel Composites with Different Dimensional Nanoparticles for Bone Regeneration. Macromol Rapid Commun 2021; 42:e2100362. [PMID: 34435714 DOI: 10.1002/marc.202100362] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 08/23/2021] [Indexed: 12/14/2022]
Abstract
The treatment of large segmental bone defects and complex types of fractures caused by trauma, inflammation, or tumor resection is still a challenge in the field of orthopedics. Various natural or synthetic biological materials used in clinical applications cannot fully replicate the structure and performance of raw bone. This highlights how to endow materials with multiple functions and biological properties, which is a problem that needs to be solved in practical applications. Hydrogels with outstanding biocompatibility, for their casting into any shape, size, or form, are suitable for different forms of bone defects. Therefore, they have been used in regenerative medicine more widely. In this review, versatile hydrogels are compounded with nanoparticles of different dimensions, and many desirable features of these materials in bone regeneration are introduced, including drug delivery, cell factor vehicle, cell scaffolds, which have potential in bone regeneration applications. The combination of hydrogels and nanoparticles of different dimensions encourages better filling of bone defect areas and has higher adaptability. This is due to the minimally invasive properties of the material and ability to match irregular defects. These biological characteristics make composite hydrogels with different dimensional nanoparticles become one of the most attractive options for bone regeneration materials.
Collapse
Affiliation(s)
- Xingchen Li
- State Key Laboratory of Supramolecular Structure and Material, College of Chemistry, Jilin University, Changchun, 130012, China
| | - Zhe Yang
- State Key Laboratory of Supramolecular Structure and Material, College of Chemistry, Jilin University, Changchun, 130012, China
| | - Linan Fang
- Department of Thoracic Surgery, the First Hospital of Jilin University, Changchun, 130000, China
| | - Chengyuan Ma
- Department of Neurosurgery, the First Hospital of Jilin University, Changchun, 130021, China
| | - Yue Zhao
- State Key Laboratory of Supramolecular Structure and Material, College of Chemistry, Jilin University, Changchun, 130012, China
| | - Hou Liu
- State Key Laboratory of Supramolecular Structure and Material, College of Chemistry, Jilin University, Changchun, 130012, China
| | - Songtian Che
- Department of Ocular Fundus Disease, the Second Hospital of Jilin University, Changchun, 130022, China
| | - Andrei V Zvyagin
- Australian Research Council Centre of Excellence for Nanoscale Biophotonics, Macquarie University, Sydney, NSW, 2109, Australia
| | - Bai Yang
- State Key Laboratory of Supramolecular Structure and Material, College of Chemistry, Jilin University, Changchun, 130012, China
| | - Quan Lin
- State Key Laboratory of Supramolecular Structure and Material, College of Chemistry, Jilin University, Changchun, 130012, China
| |
Collapse
|
20
|
Electrospun Nanofibrous Membranes for Tissue Engineering and Cell Growth. APPLIED SCIENCES-BASEL 2021. [DOI: 10.3390/app11156929] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
In biotechnology, the field of cell cultivation is highly relevant. Cultivated cells can be used, for example, for the development of biopharmaceuticals and in tissue engineering. Commonly, mammalian cells are grown in bioreactors, T-flasks, well plates, etc., without a specific substrate. Nanofibrous mats, however, have been reported to promote cell growth, adhesion, and proliferation. Here, we give an overview of the different attempts at cultivating mammalian cells on electrospun nanofiber mats for biotechnological and biomedical purposes. Starting with a brief overview of the different electrospinning methods, resulting in random or defined fiber orientations in the nanofiber mats, we describe the typical materials used in cell growth applications in biotechnology and tissue engineering. The influence of using different surface morphologies and polymers or polymer blends on the possible application of such nanofiber mats for tissue engineering and other biotechnological applications is discussed. Polymer blends, in particular, can often be used to reach the required combination of mechanical and biological properties, making such nanofiber mats highly suitable for tissue engineering and other biotechnological or biomedical cell growth applications.
Collapse
|
21
|
Zang L, Finnerty C, Zheng S, Conway K, Sun L, Ma J, Mi B. Interfacial solar vapor generation for desalination and brine treatment: Evaluating current strategies of solving scaling. WATER RESEARCH 2021; 198:117135. [PMID: 33895587 DOI: 10.1016/j.watres.2021.117135] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 04/06/2021] [Accepted: 04/07/2021] [Indexed: 06/12/2023]
Abstract
Interfacial solar vapor generation, an efficient, sustainable, and low-cost method for producing clean water, has attracted great interest for application in solar desalination and wastewater treatment. Although recent studies indicated significant enhancement of overall performance by developing photothermal materials and constructing different dimensional systems, stable evaporation performance and long-term operation of the evaporator are hindered by severe scaling issues. In this critical review, we present the latest strategies in reducing salt accumulation on the evaporator for solar desalination and brine treatment. We first demonstrate the consequences of salt accumulation, and then discuss various self-cleaning methods based on bio-inspired concepts and other strategies such as physical cleaning, ion rejection and exchange, fast ion diffusion, and controlled crystallization, etc. Importantly, we discuss and address the rational design of the evaporator via establishing a relationship model between its porosity, thickness, and thermal conductivity. Lastly, we evaluate salt-resistance strategies, evaporation performance, and possibilities of real application in different evaporation systems with scaling-resistant abilities.
Collapse
Affiliation(s)
- Linlin Zang
- State Key Laboratory of Urban Water Resource and Environment, School of Environment, Harbin Institute of Technology, Harbin 150090, China; Department of Civil and Environmental Engineering, University of California, Berkeley, California 94720, United States
| | - Casey Finnerty
- Department of Civil and Environmental Engineering, University of California, Berkeley, California 94720, United States
| | - Sunxiang Zheng
- Department of Civil and Environmental Engineering, University of California, Berkeley, California 94720, United States
| | - Kelly Conway
- Department of Civil and Environmental Engineering, University of California, Berkeley, California 94720, United States
| | - Liguo Sun
- School of Chemical Engineering and Materials, Heilongjiang University, Harbin 150080, China
| | - Jun Ma
- State Key Laboratory of Urban Water Resource and Environment, School of Environment, Harbin Institute of Technology, Harbin 150090, China.
| | - Baoxia Mi
- Department of Civil and Environmental Engineering, University of California, Berkeley, California 94720, United States.
| |
Collapse
|
22
|
Zhu AS, Mustafa T, Connell JP, Grande-Allen KJ. Tumor necrosis factor alpha and interleukin 1 beta suppress myofibroblast activation via nuclear factor kappa B signaling in 3D-cultured mitral valve interstitial cells. Acta Biomater 2021; 127:159-168. [PMID: 33831572 DOI: 10.1016/j.actbio.2021.03.075] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 03/25/2021] [Accepted: 03/30/2021] [Indexed: 12/12/2022]
Abstract
Mitral valve disease is a major cause of cardiovascular morbidity throughout the world. Many different mitral valve pathologies feature fibrotic remodeling, often accompanied by an inflammatory state. Mitral valve fibrosis is mediated by valvular interstitial cells (VICs), which reside in the valve leaflets and often differentiate into myofibroblast-like cells during disease conditions. In this study, we investigated the effects of tumor necrosis factor alpha (TNF-α) and interleukin 1 beta (IL-1β) on mitral VICs, since these pro-inflammatory cytokines have been shown to exert pleiotropic effects on various cell types in other fibrotic disorders. Using biomimetic three-dimensional culture systems, we demonstrated that TNF-α and IL-1β suppress myofibroblast differentiation in mitral VICs, as evidenced by gene and protein expression of alpha smooth muscle actin and smooth muscle 22 alpha. Addition of TNF-α and IL-1β also inhibited mitral VIC-mediated contraction of collagen gels. Furthermore, inhibition of NF-κB, which is downstream of TNF-α and IL-1β, reversed these effects. These results reveal targetable pathways for potential development of pharmaceutical treatments for alleviating fibrosis during mitral valve disease. STATEMENT OF SIGNIFICANCE: Mitral valve disease is a common cardiovascular condition that is often accompanied by fibrotic tissue remodeling. Valvular interstitial cells (VICs), the fibroblast-like cells that reside in heart valve leaflets, are thought to drive fibrosis during valve disease by differentiating into activated myofibroblasts. However, the signaling pathways that regulate this process in the mitral valve are not fully understood. In the present study, we cultured mitral VICs in collagen and poly(ethylene glycol) scaffolds designed to mimic the heart valve microenvironment and treated the cell-seeded scaffolds with cytokines. Using these 3D culture models, we found that the pro-inflammatory cytokines TNF-α and IL-1β downregulate myofibroblast and fibrosis markers in mitral VICs via the canonical NF-κB signaling pathway.
Collapse
|
23
|
Bronzeri LB, Gauche C, Gudimard L, Courtial EJ, Marquette C, Felisberti MI. Amphiphilic and segmented polyurethanes based on poly(ε-caprolactone)diol and poly(2-ethyl-2-oxazoline)diol: Synthesis, properties, and a preliminary performance study of the 3D printing. Eur Polym J 2021. [DOI: 10.1016/j.eurpolymj.2021.110449] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
|
24
|
Mirani B, Parvin Nejad S, Simmons CA. Recent Progress Toward Clinical Translation of Tissue-Engineered Heart Valves. Can J Cardiol 2021; 37:1064-1077. [PMID: 33839245 DOI: 10.1016/j.cjca.2021.03.022] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 03/04/2021] [Accepted: 03/14/2021] [Indexed: 01/02/2023] Open
Abstract
Surgical replacement remains the primary option to treat the rapidly growing number of patients with severe valvular heart disease. Although current valve replacements-mechanical, bioprosthetic, and cryopreserved homograft valves-enhance survival and quality of life for many patients, the ideal prosthetic heart valve that is abundantly available, immunocompatible, and capable of growth, self-repair, and life-long performance has yet to be developed. These features are essential for pediatric patients with congenital defects, children and young adult patients with rheumatic fever, and active adult patients with valve disease. Heart valve tissue engineering promises to address these needs by providing living valve replacements that function similarly to their native counterparts. This is best evidenced by the long-term clinical success of decellularised pulmonary and aortic homografts, but the supply of homografts cannot meet the demand for replacement valves. A more abundant and consistent source of replacement valves may come from cellularised valves grown in vitro or acellular off-the-shelf biomaterial/tissue constructs that recellularise in situ, but neither tissue engineering approach has yet achieved long-term success in preclinical testing. Beyond the technical challenges, heart valve tissue engineering faces logistical, economic, and regulatory challenges. In this review, we summarise recent progress in heart valve tissue engineering, highlight important outcomes from preclinical and clinical testing, and discuss challenges and future directions toward clinical translation.
Collapse
Affiliation(s)
- Bahram Mirani
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, Ontario, Canada; Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada; Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Shouka Parvin Nejad
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, Ontario, Canada; Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Craig A Simmons
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, Ontario, Canada; Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada; Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
25
|
Iop L. Toward the Effective Bioengineering of a Pathological Tissue for Cardiovascular Disease Modeling: Old Strategies and New Frontiers for Prevention, Diagnosis, and Therapy. Front Cardiovasc Med 2021; 7:591583. [PMID: 33748193 PMCID: PMC7969521 DOI: 10.3389/fcvm.2020.591583] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 12/08/2020] [Indexed: 12/18/2022] Open
Abstract
Cardiovascular diseases (CVDs) still represent the primary cause of mortality worldwide. Preclinical modeling by recapitulating human pathophysiology is fundamental to advance the comprehension of these diseases and propose effective strategies for their prevention, diagnosis, and treatment. In silico, in vivo, and in vitro models have been applied to dissect many cardiovascular pathologies. Computational and bioinformatic simulations allow developing algorithmic disease models considering all known variables and severity degrees of disease. In vivo studies based on small or large animals have a long tradition and largely contribute to the current treatment and management of CVDs. In vitro investigation with two-dimensional cell culture demonstrates its suitability to analyze the behavior of single, diseased cellular types. The introduction of induced pluripotent stem cell technology and the application of bioengineering principles raised the bar toward in vitro three-dimensional modeling by enabling the development of pathological tissue equivalents. This review article intends to describe the advantages and disadvantages of past and present modeling approaches applied to provide insights on some of the most relevant congenital and acquired CVDs, such as rhythm disturbances, bicuspid aortic valve, cardiac infections and autoimmunity, cardiovascular fibrosis, atherosclerosis, and calcific aortic valve stenosis.
Collapse
Affiliation(s)
- Laura Iop
- Department of Cardiac Thoracic Vascular Sciences, and Public Health, University of Padua Medical School, Padua, Italy
| |
Collapse
|
26
|
Almanza M, Clavica F, Chavanne J, Moser D, Obrist D, Carrel T, Civet Y, Perriard Y. Feasibility of a Dielectric Elastomer Augmented Aorta. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:2001974. [PMID: 33747718 PMCID: PMC7967089 DOI: 10.1002/advs.202001974] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 10/09/2020] [Indexed: 06/12/2023]
Abstract
Although heart transplantation is a gold standard for severe heart failure, there is a need for alternative effective therapies. A dielectric-elastomer aorta is used to augment the physiological role of the aorta in the human circulatory system. To this end, the authors developed a tubular dielectric elastomer actuator (DEA) able to assist the heart by easing the deformation of the aorta in the systole and by increasing its recoil force in the diastole. In vitro experiments using a pulsatile flow-loop, replicating human physiological flow and pressure conditions, show a reduction of 5.5% (47 mJ per cycle) of the heart energy with this device. Here, the controlled stiffness of the DEA graft, which is usually difficult to exploit for actuators, is perfectly matching the assistance principle. At the same time, the physiological aortic pressure is exploited to offer a prestretch to the DEA which otherwise would require an additional bulky pre-stretching system to reach high performances.
Collapse
Affiliation(s)
- Morgan Almanza
- Integrated Actuators LaboratoryÉcole Polytechnique fédérale de Lausanne (EPFL)Neuchâtel2000Switzerland
| | - Francesco Clavica
- Integrated Actuators LaboratoryÉcole Polytechnique fédérale de Lausanne (EPFL)Neuchâtel2000Switzerland
- ARTORG Center for Biomedical Engineering ResearchUniversity of BernBern3012Switzerland
| | - Jonathan Chavanne
- Integrated Actuators LaboratoryÉcole Polytechnique fédérale de Lausanne (EPFL)Neuchâtel2000Switzerland
| | - David Moser
- Integrated Actuators LaboratoryÉcole Polytechnique fédérale de Lausanne (EPFL)Neuchâtel2000Switzerland
| | - Dominik Obrist
- ARTORG Center for Biomedical Engineering ResearchUniversity of BernBern3012Switzerland
| | - Thierry Carrel
- Department of Cardiovascular SurgeryUniversity Hospital and University of BernBern3012Switzerland
| | - Yoan Civet
- Integrated Actuators LaboratoryÉcole Polytechnique fédérale de Lausanne (EPFL)Neuchâtel2000Switzerland
| | - Yves Perriard
- Integrated Actuators LaboratoryÉcole Polytechnique fédérale de Lausanne (EPFL)Neuchâtel2000Switzerland
| |
Collapse
|
27
|
Howsmon DP, Sacks MS. On Valve Interstitial Cell Signaling: The Link Between Multiscale Mechanics and Mechanobiology. Cardiovasc Eng Technol 2021; 12:15-27. [PMID: 33527256 PMCID: PMC11046423 DOI: 10.1007/s13239-020-00509-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 12/05/2020] [Indexed: 01/02/2023]
Abstract
Heart valves function in one of the most mechanically demanding environments in the body to ensure unidirectional blood flow. The resident valve interstitial cells respond to this mechanical environment and maintain the structure and integrity of the heart valve tissues to preserve homeostasis. While the mechanics of organ-tissue-cell heart valve function has progressed, the intracellular signaling network downstream of mechanical stimuli has not been fully elucidated. This has hindered efforts to both understand heart valve mechanobiology and rationally identify drug targets for treating valve disease. In the present work, we review the current literature on VIC mechanobiology and then propose mechanistic mathematical modeling of the mechanically-stimulated VIC signaling response to comprehend the coupling between VIC mechanobiology and valve mechanics.
Collapse
Affiliation(s)
- Daniel P Howsmon
- James T. Willerson Center for Cardiovascular Modeling and Simulation, The Oden Institute for Computational Engineering and Sciences and the Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Michael S Sacks
- James T. Willerson Center for Cardiovascular Modeling and Simulation, The Oden Institute for Computational Engineering and Sciences and the Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA.
| |
Collapse
|
28
|
Wancura MM, Talanker M, Toubbeh S, Bryan A, Cosgriff-Hernandez E. Bioactive hydrogel coatings of complex substrates using diffusion-mediated redox initiation. J Mater Chem B 2020; 8:4289-4298. [PMID: 32322860 PMCID: PMC9207961 DOI: 10.1039/d0tb00055h] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Hydrogels have long been established as materials with tunable stiffness and chemistry that enable controlled cellular interactions. When applied as coatings, hydrogels can be used to introduce biofunctionality to medical devices with minimal effect on bulk properties. However, it remains challenging to uniformly apply hydrogel coatings to three dimensional geometries without substantially changing the manufacturing process and potentially affecting device function. Herein, we report a new redox-based crosslinking method for applying conformable hydrogel coatings with tunable thickness and chemistry. This new diffusion-mediated strategy of redox initiation and hydrogel crosslinking enabled coating of a variety of three dimensional substrates without changing the primary fabrication process. Following adsorption of the reducing agent to the construct, hydrogel coating thickness was readily controlled by immersion time with desorption and diffusion of the reducing agent initiating hydrogel crosslinking from the surface. The process was used to generate a range of hydrogel properties by varying the macromer molecular weight and concentration. In addition, we demonstrated that these coatings can be applied sequentially to generate multilayered constructs with distinct features. Finally, incorporation of proteins into the bulk of the hydrogel coating or as a final surface layer permitted the controlled introduction of bioactivity that supported cell attachment. This work provides a versatile method for assembling bioactive coatings with a simple post-fabrication process that is amenable to diverse geometric substrates and chemistries.
Collapse
Affiliation(s)
- Megan M. Wancura
- Department of Chemistry, The University of Texas at Austin, Austin TX 78712, USA
| | - Michael Talanker
- Department of Biomedical Engineering, The University of Texas at Austin, Austin TX 78712, USA
| | - Shireen Toubbeh
- Department of Biomedical Engineering, The University of Texas at Austin, Austin TX 78712, USA
| | - Alex Bryan
- Department of Biomedical Engineering, The University of Texas at Austin, Austin TX 78712, USA
| | | |
Collapse
|
29
|
In Vivo Stability of Polyurethane-Based Electrospun Vascular Grafts in Terms of Chemistry and Mechanics. Polymers (Basel) 2020; 12:polym12040845. [PMID: 32272564 PMCID: PMC7240619 DOI: 10.3390/polym12040845] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 03/25/2020] [Accepted: 03/27/2020] [Indexed: 12/31/2022] Open
Abstract
The biostability of the polyurethanes Tecoflex EG-80A and Pellethane 2363-80A, used as basic polymers of the vascular grafts (VGs) produced by electrospinning, as well as the tensile strength of Tecoflex VGs, are studied. Solutions of Tecoflex or Pellethane with gelatin and bivalirudin in 1,1,1,3,3,3-hexafluoroisopropanol are used for VG production. After 1, 12, and 24 weeks of VG implantation in the infrarenal position of the abdominal aorta of Wistar rats, VGs are explanted, fixed in formalin, freed from outer tissues, dialyzed, and dried. The polyurethanes are extracted from VGs by dispersion/extraction in tetrahydrofuran (THF) and freed from the excess of THF-insoluble biopolymers. The stability of polyurethanes is assessed by IR spectroscopy and gel permeation chromatography. Pellethane has emerged to be stable at all experimental points. Tecoflex loses approximately 10% of its molecular weight (both Mn and Mw) after 3 months and restored its initial value within 6 months of its functioning as a graft. Mechanical testing demonstrates a 30% reduction in the tensile strength after 3 months in VG and a 10% increase after 6 months. The stability and mechanical properties of polyurethane-based VGs demonstrate their utility for the reconstitution of damaged arteries.
Collapse
|
30
|
Lin J, Wang W, Cheng J, Cui Z, Si J, Wang Q, Chen W. Modification of thermoplastic polyurethane nanofiber membranes by in situ polydopamine coating for tissue engineering. J Appl Polym Sci 2020. [DOI: 10.1002/app.49252] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Affiliation(s)
- Jixin Lin
- School of Materials Science and Engineering Fujian University of Technology Fujian China
- Fujian Provincial Engineering Research Center of Die and Mold Fujian University of Technology Fujian China
- Mould Technology Development Base of Fujian Province Fujian University of Technology Fujian China
| | - Weiwen Wang
- School of Materials Science and Engineering Fujian University of Technology Fujian China
- Fujian Provincial Engineering Research Center of Die and Mold Fujian University of Technology Fujian China
- Mould Technology Development Base of Fujian Province Fujian University of Technology Fujian China
| | - Jiaqi Cheng
- School of Materials Science and Engineering Fujian University of Technology Fujian China
- Fujian Provincial Engineering Research Center of Die and Mold Fujian University of Technology Fujian China
- Mould Technology Development Base of Fujian Province Fujian University of Technology Fujian China
| | - Zhixiang Cui
- School of Materials Science and Engineering Fujian University of Technology Fujian China
- Fujian Provincial Engineering Research Center of Die and Mold Fujian University of Technology Fujian China
- Mould Technology Development Base of Fujian Province Fujian University of Technology Fujian China
| | - Junhui Si
- School of Materials Science and Engineering Fujian University of Technology Fujian China
- Fujian Provincial Engineering Research Center of Die and Mold Fujian University of Technology Fujian China
- Mould Technology Development Base of Fujian Province Fujian University of Technology Fujian China
| | - Qianting Wang
- School of Materials Science and Engineering Fujian University of Technology Fujian China
- Fujian Provincial Engineering Research Center of Die and Mold Fujian University of Technology Fujian China
- Mould Technology Development Base of Fujian Province Fujian University of Technology Fujian China
| | - Wenzhe Chen
- School of Materials Science and Engineering Fujian University of Technology Fujian China
- Fujian Provincial Engineering Research Center of Die and Mold Fujian University of Technology Fujian China
- Mould Technology Development Base of Fujian Province Fujian University of Technology Fujian China
| |
Collapse
|
31
|
Oveissi F, Naficy S, Lee A, Winlaw D, Dehghani F. Materials and manufacturing perspectives in engineering heart valves: a review. Mater Today Bio 2020; 5:100038. [PMID: 32211604 PMCID: PMC7083765 DOI: 10.1016/j.mtbio.2019.100038] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 11/26/2019] [Accepted: 11/27/2019] [Indexed: 12/27/2022] Open
Abstract
Valvular heart diseases (VHD) are a major health burden, affecting millions of people worldwide. The treatments for such diseases rely on medicine, valve repair, and artificial heart valves including mechanical and bioprosthetic valves. Yet, there are countless reports on possible alternatives noting long-term stability and biocompatibility issues and highlighting the need for fabrication of more durable and effective replacements. This review discusses the current and potential materials that can be used for developing such valves along with existing and developing fabrication methods. With this perspective, we quantitatively compare mechanical properties of various materials that are currently used or proposed for heart valves along with their fabrication processes to identify challenges we face in creating new materials and manufacturing techniques to better mimick the performance of native heart valves.
Collapse
Key Words
- 3D printing
- Biofabrication
- Biomaterials
- E, Young's modulus
- Electrospinning
- Gal, galactose-α1,3-galactose
- GelMa, gelatin methacrylate
- HA, hyaluronic acid
- HAVIC, human aortic valvular interstitial cells
- MA-HA, methacrylated hyaluronic acid
- NeuGc, N-glycolylneuraminic acid
- P4HB, poly(4-hydroxybutyrate)
- PAAm, polyacrylamide
- PCE, polycitrate-(ε-polypeptide)
- PCL, polycaprolactone
- PE, polyethylene
- PEG, polyethylene glycol
- PEGDA, polyethylene glycol diacrylate
- PGA, poly(glycolic acid)
- PHA, poly(hydroxyalkanoate)
- PLA, polylactide
- PMMA, poly(methyl methacrylate)
- PPG, polypropylene glycol
- PTFE, polytetrafluoroethylene
- PU, polyurethane
- SIBS, poly(styrene-b-isobutylene-b-styrene)
- SMC, smooth muscle cells
- VHD, valvular heart disease
- VIC, aortic valve leaflet interstitial cells
- Valvular heart diseases
- dECM, decellularized extracellular matrix
- ePTFE, expanded PTFE
- xSIBS, crosslinked version of SIBS
- α-SMA, alpha-smooth muscle actin
Collapse
Affiliation(s)
- F. Oveissi
- School of Chemical and Biomolecular Engineering, The University of Sydney, Sydney, New South Wales, 2006, Australia
| | - S. Naficy
- School of Chemical and Biomolecular Engineering, The University of Sydney, Sydney, New South Wales, 2006, Australia
| | - A. Lee
- School of Chemical and Biomolecular Engineering, The University of Sydney, Sydney, New South Wales, 2006, Australia
- Discipline of Child and Adolescent Health, Sydney Medical School, Faculty of Health and Medicine, The University of Sydney, New South Wales, 2006, Australia
- Heart Centre for Children, The Children's Hospital at Westmead, New South Wales, 2145, Australia
| | - D.S. Winlaw
- Discipline of Child and Adolescent Health, Sydney Medical School, Faculty of Health and Medicine, The University of Sydney, New South Wales, 2006, Australia
- Heart Centre for Children, The Children's Hospital at Westmead, New South Wales, 2145, Australia
| | - F. Dehghani
- School of Chemical and Biomolecular Engineering, The University of Sydney, Sydney, New South Wales, 2006, Australia
| |
Collapse
|
32
|
Tao ZW, Wu S, Cosgriff-Hernandez EM, Jacot JG. Evaluation of a polyurethane-reinforced hydrogel patch in a rat right ventricle wall replacement model. Acta Biomater 2020; 101:206-218. [PMID: 31654774 PMCID: PMC6960327 DOI: 10.1016/j.actbio.2019.10.026] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 09/30/2019] [Accepted: 10/18/2019] [Indexed: 12/16/2022]
Abstract
Congenital heart defects affect about 1% births in the United States. Many of the defects are treated with surgically implanted patches made from inactive materials or fixed pericardium that do not grow with the patients, leading to an increased risk of arrhythmia, sudden cardiac death, and heart failure. This study investigated an angiogenic poly(ethylene glycol) fibrin-based hydrogel reinforced with an electrospun biodegradable poly(ether ester urethane) urea (BPUR) mesh layer that was designed to encourage cell invasion, angiogenesis, and regenerative remodeling in the repair of an artificial defect created onto the rat right ventricle wall. Electrocardiogram signals were analyzed, heart function was measured, and fibrosis, macrophage infiltration, muscularization, vascularization, and defect size were evaluated at 4- and 8-weeks post-surgery. Compared with rats with fixed pericardium patches, rats with BPUR-reinforced hydrogel patches had fewer arrhythmias and greater right ventricular ejection fraction and cardiac output, as well as greater left ventricular ejection fraction, fractional shorting, stroke work and cardiac output. Histology and immunofluorescence staining showed less fibrosis and less patch material remaining in rats with BPUR-reinforced hydrogel patches at 4- and 8-weeks. Rats with BPUR-reinforced hydrogel patches also had a greater volume of granular tissue, a greater volume of muscularized tissue, more blood vessels, and a greater number of leukocytes, pan-macrophages, and M2 macrophages at 8 weeks. Overall, this study demonstrated that the engineered BPUR-reinforced hydrogel patch initiated greater regenerative vascular and muscular remodeling with a limited fibrotic response, resulting in fewer incidences of arrhythmia and improved heart function compared with fixed pericardium patches when applied to heal the defects created on the rat right ventricle wall. STATEMENT OF SIGNIFICANCE: The study tested a polyurethane-reinforced hydrogel patch in a rat right ventricle wall replacement model. Compared with fixed pericardium patches, these reinforced hydrogel patches initiated greater regenerative vascular and muscular remodeling with a reduced fibrotic response, resulting in fewer incidences of arrhythmia and improved heart function at 4- and 8-weeks post surgery. Overall, the new BPUR-reinforced hydrogel patches resulted in better heart function when replacing contractile myocardium than fixed pericardium patches.
Collapse
Affiliation(s)
- Ze-Wei Tao
- Department of Bioengineering, University of Colorado Anschutz Medical Campus, 12705 E Montview Blvd, Suite 100, Aurora 80045, CO, USA
| | - Siliang Wu
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, USA
| | | | - Jeffrey G Jacot
- Department of Bioengineering, University of Colorado Anschutz Medical Campus, 12705 E Montview Blvd, Suite 100, Aurora 80045, CO, USA; Department of Pediatrics, Children's Hospital Colorado, Aurora, CO, USA.
| |
Collapse
|
33
|
Velu R, Calais T, Jayakumar A, Raspall F. A Comprehensive Review on Bio-Nanomaterials for Medical Implants and Feasibility Studies on Fabrication of Such Implants by Additive Manufacturing Technique. MATERIALS (BASEL, SWITZERLAND) 2019; 13:E92. [PMID: 31878040 PMCID: PMC6981457 DOI: 10.3390/ma13010092] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 12/17/2019] [Accepted: 12/18/2019] [Indexed: 02/08/2023]
Abstract
Nanomaterials have allowed significant breakthroughs in bio-engineering and medical fields. In the present paper a holistic assessment on diverse biocompatible nanocomposites are studied. Their compatibility with advanced fabrication methods such as additive manufacturing for the design of functional medical implants is also critically reviewed. The significance of nanocomposites and processing techniques is also envisaged comprehensively in regard with the needs and futures of implantable medical device industries.
Collapse
Affiliation(s)
- Rajkumar Velu
- Digital Manufacturing and Design Centre (DManD), Singapore University of Technology and Design, Singapore 486842, Singapore; (T.C.); (F.R.)
| | - Theo Calais
- Digital Manufacturing and Design Centre (DManD), Singapore University of Technology and Design, Singapore 486842, Singapore; (T.C.); (F.R.)
| | | | - Felix Raspall
- Digital Manufacturing and Design Centre (DManD), Singapore University of Technology and Design, Singapore 486842, Singapore; (T.C.); (F.R.)
| |
Collapse
|
34
|
W. King M, Chen J, Deshpande M, He T, Ramakrishna H, Xie Y, Zhang F, Zhao F. Structural Design, Fabrication and Evaluation of Resorbable Fiber-Based Tissue Engineering Scaffolds. Biotechnol Bioeng 2019. [DOI: 10.5772/intechopen.84643] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
35
|
Cardiac cell differentiation of muscle satellite cells on aligned composite electrospun polyurethane with reduced graphene oxide. JOURNAL OF POLYMER RESEARCH 2019. [DOI: 10.1007/s10965-019-1936-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
36
|
Mathew E, Domínguez-Robles J, Stewart SA, Mancuso E, O'Donnell K, Larrañeta E, Lamprou DA. Fused Deposition Modeling as an Effective Tool for Anti-Infective Dialysis Catheter Fabrication. ACS Biomater Sci Eng 2019; 5:6300-6310. [PMID: 33405537 DOI: 10.1021/acsbiomaterials.9b01185] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Catheter-associated infections are a common complication that occurs in dialysis patients. Current strategies to prevent infection include catheter coatings containing heparin, pyrogallol, or silver nanoparticles, which all have an increased risk of causing resistance in bacteria. Therefore, a novel approach for manufacture, such as the use of additive manufacturing (AM), also known as three-dimensional (3D) printing, is required. Filaments were produced by extrusion using thermoplastic polyurethane (TPU) and tetracycline hydrochloride (TC) in various concentrations (e.g., 0, 0.25, 0.5, and 1%). The extruded filaments were used in a fused deposition modeling (FDM) 3D printer to print catheter constructs at varying concentrations. Release studies in phosphate-buffered saline, microbiology studies, thermal analysis, contact angle, attenuated total reflection-Fourier transform infrared, scanning electron microscopy, and X-ray microcomputer tomography (μCT) analysis were conducted on the printed catheters. The results suggested that TC was uniformly distributed within the TPU matrix. The microbiology testing of the catheters showed that devices containing TC had an inhibitory effect on the growth of Staphylococcus aureus NCTC 10788 bacteria. Catheters containing 1% TC maintained inhibitory effect after 10 day release studies. After an initial burst release in the first 24 h, there was a steady release of TC in all concentrations of catheters. 3D-printed antibiotic catheters were successfully printed with inhibitory effect on S. aureus bacteria. Finally, TC containing catheters showed resistance to S. aureus adherence to their surfaces when compared with catheters containing no TC. Catheters containing 1% of TC showed a bacterial adherence reduction of up to 99.97%. Accordingly, the incorporation of TC to TPU materials can be effectively used to prepare anti-infective catheters using FDM. This study highlights the potential for drug-impregnated medical devices to be created through AM.
Collapse
Affiliation(s)
- Essyrose Mathew
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, U.K
| | - Juan Domínguez-Robles
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, U.K
| | - Sarah A Stewart
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, U.K
| | - Elena Mancuso
- Nanotechnology and Integrated Bio-Engineering Centre (NIBEC), Ulster University, Jordanstown Campus BT37 0QB, U.K
| | - Kieran O'Donnell
- Nanotechnology and Integrated Bio-Engineering Centre (NIBEC), Ulster University, Jordanstown Campus BT37 0QB, U.K
| | - Eneko Larrañeta
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, U.K
| | - Dimitrios A Lamprou
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, U.K
| |
Collapse
|
37
|
Khang A, Gonzalez Rodriguez A, Schroeder ME, Sansom J, Lejeune E, Anseth KS, Sacks MS. Quantifying heart valve interstitial cell contractile state using highly tunable poly(ethylene glycol) hydrogels. Acta Biomater 2019; 96:354-367. [PMID: 31323351 PMCID: PMC6717677 DOI: 10.1016/j.actbio.2019.07.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 07/04/2019] [Accepted: 07/06/2019] [Indexed: 01/19/2023]
Abstract
Valve interstitial cells (VIC) are the primary cell type residing within heart valve tissues. In many valve pathologies, VICs become activated and will subsequently profoundly remodel the valve tissue extracellular matrix (ECM). A primary indicator of VIC activation is the upregulation of α-smooth muscle actin (αSMA) stress fibers, which in turn increase VIC contractility. Thus, contractile state reflects VIC activation and ECM biosynthesis levels. In general, cell contraction studies have largely utilized two-dimensional substrates, which are a vastly different micro mechanical environment than 3D native leaflet tissue. To address this limitation, hydrogels have been a popular choice for studying cells in a three-dimensional environment due to their tunable properties and optical transparency, which allows for direct cell visualization. In the present study, we extended the use of hydrogels to study the active contractile behavior of VICs. Aortic VICs (AVIC) were encapsulated within poly(ethylene glycol) (PEG) hydrogels and were subjected to flexural-deformation tests to assess the state of AVIC contraction. Using a finite element model of the experimental setup, we determined the effective shear modulus μ of the constructs. An increase in μ resulting from AVIC active contraction was observed. Results further indicated that AVIC contraction had a more pronounced effect on μ in softer gels (72 ± 21% increase in μ within 2.5 kPa gels) and was dependent upon the availability of adhesion sites (0.5-1 mM CRGDS). The transparency of the gel allowed us to image AVICs directly within the hydrogel, where we observed a time-dependent decrease in volume (time constant τ=3.04 min) when the AVICs were induced into a hypertensive state. Our results indicated that AVIC contraction was regulated by both the intrinsic (unseeded) gel stiffness and the CRGDS peptide concentrations. This finding suggests that AVIC contractile state can be profoundly modulated through their local micro environment using modifiable PEG gels in a 3D micromechanical-emulating environment. Moving forward, this approach has the potential to be used towards delineating normal and diseased VIC biomechanical properties using highly tunable PEG biomaterials. STATEMENT OF SIGNIFICANCE.
Collapse
Affiliation(s)
- Alex Khang
- James T. Willerson Center for Cardiovascular Modeling and Simulation, The Oden Institute for Computational Engineering and Sciences and the Department of Biomedical Engineering, The University of Texas at Austin, 240 East 24th Street, Austin, TX 78712, United States
| | - Andrea Gonzalez Rodriguez
- Department of Chemical and Biological Engineering, University of Colorado at Boulder, 3415 Colorado Avenue, Boulder, CO 80309, United States
| | - Megan E Schroeder
- Department of Materials Science and Engineering, University of Colorado at Boulder, 3415 Colorado Avenue, Boulder, CO 80309, United States
| | - Jacob Sansom
- James T. Willerson Center for Cardiovascular Modeling and Simulation, The Oden Institute for Computational Engineering and Sciences and the Department of Biomedical Engineering, The University of Texas at Austin, 240 East 24th Street, Austin, TX 78712, United States
| | - Emma Lejeune
- James T. Willerson Center for Cardiovascular Modeling and Simulation, The Oden Institute for Computational Engineering and Sciences and the Department of Biomedical Engineering, The University of Texas at Austin, 240 East 24th Street, Austin, TX 78712, United States
| | - Kristi S Anseth
- Department of Chemical and Biological Engineering, University of Colorado at Boulder, 3415 Colorado Avenue, Boulder, CO 80309, United States; Department of Materials Science and Engineering, University of Colorado at Boulder, 3415 Colorado Avenue, Boulder, CO 80309, United States; Biofrontiers Institute, University of Colorado at Boulder, 3415 Colorado Avenue, Boulder, CO80309, United States
| | - Michael S Sacks
- James T. Willerson Center for Cardiovascular Modeling and Simulation, The Oden Institute for Computational Engineering and Sciences and the Department of Biomedical Engineering, The University of Texas at Austin, 240 East 24th Street, Austin, TX 78712, United States.
| |
Collapse
|
38
|
Zhalmuratova D, La TG, Yu KTT, Szojka ARA, Andrews SHJ, Adesida AB, Kim CI, Nobes DS, Freed DH, Chung HJ. Mimicking "J-Shaped" and Anisotropic Stress-Strain Behavior of Human and Porcine Aorta by Fabric-Reinforced Elastomer Composites. ACS APPLIED MATERIALS & INTERFACES 2019; 11:33323-33335. [PMID: 31464413 DOI: 10.1021/acsami.9b10524] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
An ex vivo heart perfusion device preserves the donor heart in a warm beating state during transfer between extraction and implantation surgeries. One of the current challenges includes the use of rigid and noncompliant plastic tubes, which causes injuries to the heart at the junction between the tissue and the tube. The compliant and rapidly strain-stiffening mechanical property that generates a "J-shaped" stress-strain behavior is necessary for producing the Windkessel effect, which ensures continuous flow of blood through the aorta. In this study, we mimic the J-shaped and anisotropic stress-strain behavior of human aorta in synthetic elastomers to replace the problematic noncompliant plastic tube. First, we assess the mechanical properties of human (n = 1) and porcine aorta (n = 14) to quantify the nonlinear and anisotropic behavior under uniaxial tensile stress from five different regions of the aorta. Second, fabric-reinforced elastomer composites were prepared by reinforcing silicone elastomers with embedded fabrics in a trilayer geometry. The knitted structures of the fabric provide strain-stiffening as well as anisotropic mechanical properties of the resulting composite in a deterministic manner. By optimizing the combination between different elastomers and fabrics, the resulting composites matched the J-shaped and anisotropic stress-strain behavior of natural human and porcine aorta. Finally, improved analytical constitutive models based on Gent's and Mooney-Rivlin's constitutive model (to describe the elastomer matrix) combined with Holzapfel-Gasser-Ogden's model (to represent the stiffer fabrics) were developed to describe the J-shaped behavior of the natural aortas and the fabric-reinforced composites. We anticipate that the suggested fabric-reinforced silicone elastomer composite design concept can be used to develop complex soft biomaterials, as well as in emerging engineering fields such as soft robotics and microfluidics, where the Windkessel effect can be useful in regulating the flow of fluids.
Collapse
Affiliation(s)
| | | | | | - Alexander R A Szojka
- Department of Surgery , University of Alberta , Edmonton , Alberta T6G 2E1 , Canada
| | - Stephen H J Andrews
- Department of Surgery , University of Alberta , Edmonton , Alberta T6G 2E1 , Canada
| | - Adetola B Adesida
- Department of Surgery , University of Alberta , Edmonton , Alberta T6G 2E1 , Canada
| | | | | | - Darren H Freed
- Department of Surgery , University of Alberta , Edmonton , Alberta T6G 2E1 , Canada
| | | |
Collapse
|
39
|
Zhu H, Fu L, He L, Zhang J, Zhang L, Yang L, Li Y, Zhao Y, Wang Y, Mo H, Shen J. Polyurethane–Cardiolipin Nanoparticle-Modified Decellularized Scaffold-Based Vascular Patches for Tissue Engineering Applications. ACS APPLIED BIO MATERIALS 2019; 2:1696-1702. [DOI: 10.1021/acsabm.9b00079] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Haomiao Zhu
- Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, National and Local Joint Engineering Research Center of Biomedical Functional Materials, Jiangsu Engineering Research Center for Biomedical Function Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Lei Fu
- Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, National and Local Joint Engineering Research Center of Biomedical Functional Materials, Jiangsu Engineering Research Center for Biomedical Function Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Lei He
- Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, National and Local Joint Engineering Research Center of Biomedical Functional Materials, Jiangsu Engineering Research Center for Biomedical Function Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Jun Zhang
- Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, National and Local Joint Engineering Research Center of Biomedical Functional Materials, Jiangsu Engineering Research Center for Biomedical Function Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Luxia Zhang
- Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, National and Local Joint Engineering Research Center of Biomedical Functional Materials, Jiangsu Engineering Research Center for Biomedical Function Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Lutao Yang
- Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, National and Local Joint Engineering Research Center of Biomedical Functional Materials, Jiangsu Engineering Research Center for Biomedical Function Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Yajuan Li
- Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, National and Local Joint Engineering Research Center of Biomedical Functional Materials, Jiangsu Engineering Research Center for Biomedical Function Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Yue Zhao
- Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, National and Local Joint Engineering Research Center of Biomedical Functional Materials, Jiangsu Engineering Research Center for Biomedical Function Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Yutong Wang
- College of Materials Science and Engineering, Nanjing Forestry University, Nanjing 210037, China
| | - Hong Mo
- Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, National and Local Joint Engineering Research Center of Biomedical Functional Materials, Jiangsu Engineering Research Center for Biomedical Function Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Jian Shen
- Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, National and Local Joint Engineering Research Center of Biomedical Functional Materials, Jiangsu Engineering Research Center for Biomedical Function Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| |
Collapse
|
40
|
Zang L, Lin R, Dou T, Ma J, Sun L. Electrospun superhydrophilic membranes for effective removal of Pb(ii) from water. NANOSCALE ADVANCES 2019; 1:389-394. [PMID: 36132483 PMCID: PMC9473238 DOI: 10.1039/c8na00044a] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2018] [Accepted: 09/26/2018] [Indexed: 06/10/2023]
Abstract
Nanofibrous membranes have a high specific surface area and large porosity, which are beneficial for being used as adsorbents to remove heavy metal ions from water. In this work, electrospun nanofibers were wrapped with a hydrogel layer with a tunable thickness, which endowed the membrane with excellent superhydrophilic performance. Because of good water-retention properties and abundant functional groups originating from the hydrogel layer, as a static adsorbent, the maximum adsorption capacity of Pb(ii) was up to 146.21 mg g-1 according to the Langmuir model. Meanwhile, the electrospun membrane also possessed water permeability as a flow-through membrane for dynamic adsorption, which was obviously different from traditional hydrogel adsorbents. As a result, the rejection ratio of Pb(ii) can remain over 55% after running for 72 h under high pH conditions and at low initial ion concentrations. Apart from these, cycle operations confirmed the regeneration of the membrane, and competitive adsorption experiments illustrated the selective removal of Pb(ii) in a mixed ion solution.
Collapse
Affiliation(s)
- Linlin Zang
- State Key Laboratory of Urban Water Resource and Environment, School of Environment, Harbin Institute of Technology Harbin 150090 PR China
| | - Ru Lin
- State Key Laboratory of Urban Water Resource and Environment, School of Environment, Harbin Institute of Technology Harbin 150090 PR China
| | - Tianwei Dou
- Key Laboratory of Chemical Engineering Process & Technology for High-efficiency Conversion, College of Heilongjiang Province, School of Chemical Engineering and Materials, Heilongjiang University Harbin 150080 PR China
| | - Jun Ma
- State Key Laboratory of Urban Water Resource and Environment, School of Environment, Harbin Institute of Technology Harbin 150090 PR China
| | - Liguo Sun
- Key Laboratory of Chemical Engineering Process & Technology for High-efficiency Conversion, College of Heilongjiang Province, School of Chemical Engineering and Materials, Heilongjiang University Harbin 150080 PR China
| |
Collapse
|
41
|
Gao S, Tang G, Hua D, Xiong R, Han J, Jiang S, Zhang Q, Huang C. Stimuli-responsive bio-based polymeric systems and their applications. J Mater Chem B 2019; 7:709-729. [DOI: 10.1039/c8tb02491j] [Citation(s) in RCA: 401] [Impact Index Per Article: 80.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
This article highlights the properties of stimuli-responsive bio-based polymeric systems and their main intelligent applications.
Collapse
Affiliation(s)
- Shuting Gao
- College of Chemical Engineering, Jiangsu Provincial Key Lab for the Chemistry and Utilization of Agro-forest Biomass, Jiangsu Key Lab of Biomass-based Green Fuels and Chemicals, Nanjing Forestry University (NFU)
- Nanjing 210037
- P. R. China
| | - Guosheng Tang
- College of Chemical Engineering, Jiangsu Provincial Key Lab for the Chemistry and Utilization of Agro-forest Biomass, Jiangsu Key Lab of Biomass-based Green Fuels and Chemicals, Nanjing Forestry University (NFU)
- Nanjing 210037
- P. R. China
| | - Dawei Hua
- College of Chemical Engineering, Jiangsu Provincial Key Lab for the Chemistry and Utilization of Agro-forest Biomass, Jiangsu Key Lab of Biomass-based Green Fuels and Chemicals, Nanjing Forestry University (NFU)
- Nanjing 210037
- P. R. China
| | - Ranhua Xiong
- Lab General Biochemistry & Physical Pharmacy, Department of Pharmaceutics, Ghent University
- Belgium
| | - Jingquan Han
- College of Materials Science and Engineering, Nanjing Forestry University (NFU)
- Nanjing 210037
- P. R. China
| | - Shaohua Jiang
- College of Materials Science and Engineering, Nanjing Forestry University (NFU)
- Nanjing 210037
- P. R. China
| | - Qilu Zhang
- School of Material Science and Engineering, Xi’an Jiaotong University
- Xi’an 710049
- P. R. China
| | - Chaobo Huang
- College of Chemical Engineering, Jiangsu Provincial Key Lab for the Chemistry and Utilization of Agro-forest Biomass, Jiangsu Key Lab of Biomass-based Green Fuels and Chemicals, Nanjing Forestry University (NFU)
- Nanjing 210037
- P. R. China
- Laboratory of Biopolymer based Functional Materials, Nanjing Forestry University
- Nanjing
| |
Collapse
|
42
|
Dekker S, van Geemen D, van den Bogaerdt AJ, Driessen-Mol A, Aikawa E, Smits AIPM. Sheep-Specific Immunohistochemical Panel for the Evaluation of Regenerative and Inflammatory Processes in Tissue-Engineered Heart Valves. Front Cardiovasc Med 2018; 5:105. [PMID: 30159315 PMCID: PMC6104173 DOI: 10.3389/fcvm.2018.00105] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2018] [Accepted: 07/13/2018] [Indexed: 12/27/2022] Open
Abstract
The creation of living heart valve replacements via tissue engineering is actively being pursued by many research groups. Numerous strategies have been described, aimed either at culturing autologous living valves in a bioreactor (in vitro) or inducing endogenous regeneration by the host via resorbable scaffolds (in situ). Whereas a lot of effort is being invested in the optimization of heart valve scaffold parameters and culturing conditions, the pathophysiological in vivo remodeling processes to which tissue-engineered heart valves are subjected upon implantation have been largely under-investigated. This is partly due to the unavailability of suitable immunohistochemical tools specific to sheep, which serves as the gold standard animal model in translational research on heart valve replacements. Therefore, the goal of this study was to comprise and validate a comprehensive sheep-specific panel of antibodies for the immunohistochemical analysis of tissue-engineered heart valve explants. For the selection of our panel we took inspiration from previous histopathological studies describing the morphology, extracellular matrix composition and cellular composition of native human heart valves throughout development and adult stages. Moreover, we included a range of immunological markers, which are particularly relevant to assess the host inflammatory response evoked by the implanted heart valve. The markers specifically identifying extracellular matrix components and cell phenotypes were tested on formalin-fixed paraffin-embedded sections of native sheep aortic valves. Markers for inflammation and apoptosis were tested on ovine spleen and kidney tissues. Taken together, this panel of antibodies could serve as a tool to study the spatiotemporal expression of proteins in remodeling tissue-engineered heart valves after implantation in a sheep model, thereby contributing to our understanding of the in vivo processes which ultimately determine long-term success or failure of tissue-engineered heart valves.
Collapse
Affiliation(s)
- Sylvia Dekker
- Soft Tissue Engineering & Mechanobiology Division, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
| | - Daphne van Geemen
- Soft Tissue Engineering & Mechanobiology Division, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
| | | | - Anita Driessen-Mol
- Soft Tissue Engineering & Mechanobiology Division, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
- Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, Netherlands
| | - Elena Aikawa
- Division of Cardiovascular Medicine, Department of Medicine, Center for Excellence in Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA, United States
| | - Anthal I. P. M. Smits
- Soft Tissue Engineering & Mechanobiology Division, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
- Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, Netherlands
| |
Collapse
|
43
|
Bas O, Catelas I, De-Juan-Pardo EM, Hutmacher DW. The quest for mechanically and biologically functional soft biomaterials via soft network composites. Adv Drug Deliv Rev 2018; 132:214-234. [PMID: 30048654 DOI: 10.1016/j.addr.2018.07.015] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 07/18/2018] [Accepted: 07/20/2018] [Indexed: 12/15/2022]
Abstract
Developing multifunctional soft biomaterials capable of addressing all the requirements of the complex tissue regeneration process is a multifaceted problem. In order to tackle the current challenges, recent research efforts are increasingly being directed towards biomimetic design concepts that can be translated into soft biomaterials via advanced manufacturing technologies. Among those, soft network composites consisting of a continuous hydrogel matrix and a reinforcing fibrous network closely resemble native soft biological materials in terms of design and composition as well as physicochemical properties. This article reviews soft network composite systems with a particular emphasis on the design, biomaterial and fabrication aspects within the context of soft tissue engineering and drug delivery applications.
Collapse
Affiliation(s)
- Onur Bas
- ARC Industrial Transformation Training Centre in Additive Biomanufacturing, Queensland University of Technology (QUT), Kelvin Grove, Brisbane, QLD 4059, Australia; Centre in Regenerative Medicine, Institute of Health and Biomedical Innovation, Queensland University of Technology (QUT), Brisbane, QLD 4059, Australia; School of Chemistry, Physics and Mechanical Engineering, Science and Engineering Faculty (SEF), Queensland University of Technology (QUT), Brisbane, QLD 4001, Australia
| | - Isabelle Catelas
- School of Chemistry, Physics and Mechanical Engineering, Science and Engineering Faculty (SEF), Queensland University of Technology (QUT), Brisbane, QLD 4001, Australia; Department of Mechanical Engineering, University of Ottawa, Ottawa, ON K1N 6N5, Canada
| | - Elena M De-Juan-Pardo
- ARC Industrial Transformation Training Centre in Additive Biomanufacturing, Queensland University of Technology (QUT), Kelvin Grove, Brisbane, QLD 4059, Australia; School of Chemistry, Physics and Mechanical Engineering, Science and Engineering Faculty (SEF), Queensland University of Technology (QUT), Brisbane, QLD 4001, Australia
| | - Dietmar W Hutmacher
- ARC Industrial Transformation Training Centre in Additive Biomanufacturing, Queensland University of Technology (QUT), Kelvin Grove, Brisbane, QLD 4059, Australia; Centre in Regenerative Medicine, Institute of Health and Biomedical Innovation, Queensland University of Technology (QUT), Brisbane, QLD 4059, Australia; School of Chemistry, Physics and Mechanical Engineering, Science and Engineering Faculty (SEF), Queensland University of Technology (QUT), Brisbane, QLD 4001, Australia; Institute for Advanced Study, Technische Universität München, 85748 Garching, Germany.
| |
Collapse
|
44
|
Bouten CVC, Smits AIPM, Baaijens FPT. Can We Grow Valves Inside the Heart? Perspective on Material-based In Situ Heart Valve Tissue Engineering. Front Cardiovasc Med 2018; 5:54. [PMID: 29896481 PMCID: PMC5987128 DOI: 10.3389/fcvm.2018.00054] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 05/09/2018] [Indexed: 12/14/2022] Open
Abstract
In situ heart valve tissue engineering using cell-free synthetic, biodegradable scaffolds is under development as a clinically attractive approach to create living valves right inside the heart of a patient. In this approach, a valve-shaped porous scaffold "implant" is rapidly populated by endogenous cells that initiate neo-tissue formation in pace with scaffold degradation. While this may constitute a cost-effective procedure, compatible with regulatory and clinical standards worldwide, the new technology heavily relies on the development of advanced biomaterials, the processing thereof into (minimally invasive deliverable) scaffolds, and the interaction of such materials with endogenous cells and neo-tissue under hemodynamic conditions. Despite the first positive preclinical results and the initiation of a small-scale clinical trial by commercial parties, in situ tissue formation is not well understood. In addition, it remains to be determined whether the resulting neo-tissue can grow with the body and preserves functional homeostasis throughout life. More important yet, it is still unknown if and how in situ tissue formation can be controlled under conditions of genetic or acquired disease. Here, we discuss the recent advances of material-based in situ heart valve tissue engineering and highlight the most critical issues that remain before clinical application can be expected. We argue that a combination of basic science - unveiling the mechanisms of the human body to respond to the implanted biomaterial under (patho)physiological conditions - and technological advancements - relating to the development of next generation materials and the prediction of in situ tissue growth and adaptation - is essential to take the next step towards a realistic and rewarding translation of in situ heart valve tissue engineering.
Collapse
Affiliation(s)
- Carlijn V. C. Bouten
- Soft Tissue Engineering and Mechanobiology, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
- Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, Netherlands
| | - Anthal I. P. M. Smits
- Soft Tissue Engineering and Mechanobiology, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
- Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, Netherlands
| | - Frank P. T. Baaijens
- Soft Tissue Engineering and Mechanobiology, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
- Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, Netherlands
| |
Collapse
|
45
|
Multi-length scale bioprinting towards simulating microenvironmental cues. Biodes Manuf 2018; 1:77-88. [PMID: 30546920 PMCID: PMC6267274 DOI: 10.1007/s42242-018-0014-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 05/02/2018] [Indexed: 02/08/2023]
Abstract
It is envisaged that the creation of cellular environments at multiple length scales, that recapitulate in vivo bioactive and structural roles, may hold the key to creating functional, complex tissues in the laboratory. This review considers recent advances in biofabrication and bioprinting techniques across different length scales. Particular focus is placed on 3D printing of hydrogels and fabrication of biomaterial fibres that could extend the feature resolution and material functionality of soft tissue constructs. The outlook from this review discusses how one might create and simulate microenvironmental cues in vitro. A fabrication platform that integrates the competencies of different biofabrication technologies is proposed. Such a multi-process, multiscale fabrication strategy may ultimately translate engineering capability into an accessible life sciences toolkit, fulfilling its potential to deliver in vitro disease models and engineered tissue implants.
Collapse
|
46
|
Zhang J, Wang Y, Liu C, Feng F, Wang D, Mo H, Si L, Wei G, Shen J. Polyurethane/polyurethane nanoparticle-modified expanded poly(tetrafluoroethylene) vascular patches promote endothelialization. J Biomed Mater Res A 2018; 106:2131-2140. [DOI: 10.1002/jbm.a.36419] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Revised: 03/23/2018] [Accepted: 03/27/2018] [Indexed: 02/04/2023]
Affiliation(s)
- Jun Zhang
- Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, College of Chemistry and Materials Science, Nanjing Normal University; Nanjing 210023 People's Republic of China
| | - Yutong Wang
- Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, College of Chemistry and Materials Science, Nanjing Normal University; Nanjing 210023 People's Republic of China
- College of Materials Science and Engineering; Nanjing Forestry University; Nanjing 210037 People's Republic of China
| | - Cheng Liu
- Medical School, Nanjing University; Nanjing 210093 People's Republic of China
| | - Fuling Feng
- Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, College of Chemistry and Materials Science, Nanjing Normal University; Nanjing 210023 People's Republic of China
| | - Dawei Wang
- Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, College of Chemistry and Materials Science, Nanjing Normal University; Nanjing 210023 People's Republic of China
| | - Hong Mo
- Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, College of Chemistry and Materials Science, Nanjing Normal University; Nanjing 210023 People's Republic of China
| | - Ling Si
- Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, College of Chemistry and Materials Science, Nanjing Normal University; Nanjing 210023 People's Republic of China
| | - Guo Wei
- Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, College of Chemistry and Materials Science, Nanjing Normal University; Nanjing 210023 People's Republic of China
| | - Jian Shen
- Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, College of Chemistry and Materials Science, Nanjing Normal University; Nanjing 210023 People's Republic of China
| |
Collapse
|
47
|
Nachlas ALY, Li S, Jha R, Singh M, Xu C, Davis ME. Human iPSC-derived mesenchymal stem cells encapsulated in PEGDA hydrogels mature into valve interstitial-like cells. Acta Biomater 2018; 71:235-246. [PMID: 29505894 PMCID: PMC5907941 DOI: 10.1016/j.actbio.2018.02.025] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Revised: 02/06/2018] [Accepted: 02/22/2018] [Indexed: 02/07/2023]
Abstract
Despite recent advances in tissue engineered heart valves (TEHV), a major challenge is identifying a cell source for seeding TEHV scaffolds. Native heart valves are durable because valve interstitial cells (VICs) maintain tissue homeostasis by synthesizing and remodeling the extracellular matrix. This study demonstrates that induced pluripotent stem cells (iPSC)-derived mesenchymal stem cells (iMSCs) can be derived from iPSCs using a feeder-free protocol and then further matured into VICs by encapsulation within 3D hydrogels. The differentiation efficiency was characterized using flow cytometry, immunohistochemistry staining, and trilineage differentiation. Using our feeder-free differentiation protocol, iMSCs were differentiated from iPSCs and had CD90+, CD44+, CD71+, αSMA+, and CD45- expression. Furthermore, iMSCs underwent trilineage differentiation when cultured in induction media for 21 days. iMSCs were then encapsulated in poly(ethylene glycol)diacrylate (PEGDA) hydrogels grafted with adhesion peptide (RGDS) to promote remodeling and further maturation into VIC-like cells. VIC phenotype was assessed by the expression of alpha-smooth muscle actin (αSMA), vimentin, and collagen production after 28 days. When MSC-derived cells were encapsulated in PEGDA hydrogels that mimic the leaflet modulus, a decrease in αSMA expression and increase in vimentin was observed. In addition, iMSCs synthesized collagen type I after 28 days in 3D hydrogel culture. Thus, the results from this study suggest that iMSCs may be a promising cell source for TEHV. STATEMENT OF SIGNIFICANCE Developing a suitable cell source is a critical component for the success and durability of tissue engineered heart valves. The significance of this study is the generation of iPSCs-derived mesenchymal stem cells (iMSCs) that have the capacity to mature into valve interstitial-like cells when introduced into a 3D cell culture designed to mimic the layers of the valve leaflet. iMSCs were generated using a feeder-free protocol, which is one major advantage over other methods, as it is more clinically relevant. In addition to generating a potential new cell source for heart valve tissue engineering, this study also highlights the importance of a 3D culture environment to influence cell phenotype and function.
Collapse
Affiliation(s)
- Aline L Y Nachlas
- Wallace H Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA, USA
| | - Siyi Li
- Wallace H Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA, USA
| | - Rajneesh Jha
- Children's Heart Research & Outcomes (HeRO) Center, Children's Healthcare of Atlanta & Emory University, Atlanta, GA, USA; Division of Pediatric Cardiology, Department of Pediatrics, Emory University School of Medicine, USA
| | - Monalisa Singh
- Children's Heart Research & Outcomes (HeRO) Center, Children's Healthcare of Atlanta & Emory University, Atlanta, GA, USA; Division of Pediatric Cardiology, Department of Pediatrics, Emory University School of Medicine, USA
| | - Chunhui Xu
- Wallace H Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA, USA; Children's Heart Research & Outcomes (HeRO) Center, Children's Healthcare of Atlanta & Emory University, Atlanta, GA, USA; Division of Pediatric Cardiology, Department of Pediatrics, Emory University School of Medicine, USA
| | - Michael E Davis
- Wallace H Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA, USA; Children's Heart Research & Outcomes (HeRO) Center, Children's Healthcare of Atlanta & Emory University, Atlanta, GA, USA.
| |
Collapse
|
48
|
Zhu AS, Grande-Allen KJ. Heart valve tissue engineering for valve replacement and disease modeling. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2018. [DOI: 10.1016/j.cobme.2017.12.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
|
49
|
D'Amato AR, Bramson MTK, Puhl DL, Johnson J, Corr DT, Gilbert RJ. Solvent retention in electrospun fibers affects scaffold mechanical properties. ELECTROSPINNING 2018; 2:15-28. [PMID: 31032427 PMCID: PMC6482971 DOI: 10.1515/esp-2018-0002] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Electrospinning is a robust material fabrication method allowing for fine control of mechanical, chemical, and functional properties in scaffold manufacturing. Electrospun fiber scaffolds have gained prominence for their potential in a variety of applications such as tissue engineering and textile manufacturing, yet none have assessed the impact of solvent retention in fibers on the scaffold's mechanical properties. In this study, we hypothesized that retained electrospinning solvent acts as a plasticizer, and gradual solvent evaporation, by storing fibers in ambient air, will cause significant increases in electrospun fiber scaffold brittleness and stiffness, and a significant decrease in scaffold toughness. Thermogravimetric analysis indicated solvent retention in PGA, PLCL, and PET fibers, and not in PU and PCL fibers. Differential scanning calorimetry revealed that polymers that were electrospun below their glass transition temperature (T g ) retained solvent and polymers electrospun above T g did not. Young's moduli increased and yield strain decreased for solventretaining PGA, PLCL, and PET fiber scaffolds as solvent evaporated from the scaffolds over a period of 14 days. Toughness and failure strain decreased for PGA and PET scaffolds as solvent evaporated. No significant differences were observed in the mechanical properties of PU and PCL scaffolds that did not retain solvent. These observations highlight the need to consider solvent retention following electrospinning and its potential effects on scaffold mechanical properties.
Collapse
Affiliation(s)
- Anthony R D'Amato
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, 110 8th St., Troy, New York 12180, United States of America; Center for Biotechnology and Interdisciplinary Sciences, Rensselaer Polytechnic Institute, 1623 15th Street, Troy, New York 12180, United States of America
| | - Michael T K Bramson
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, 110 8th St., Troy, New York 12180, United States pf America
| | - Devan L Puhl
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, 110 8th St., Troy, New York 12180, United States of America; Center for Biotechnology and Interdisciplinary Sciences, Rensselaer Polytechnic Institute, 1623 15th Street, Troy, New York 12180, United States of America
| | - Jed Johnson
- Nanofiber Solutions, 4389 Weaver Court North, Hilliard, OH 43026, United States of America
| | - David T Corr
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, 110 8th St., Troy, New York 12180, United States pf America
| | - Ryan J Gilbert
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, 110 8th St., Troy, New York 12180, United States of America; Center for Biotechnology and Interdisciplinary Sciences, Rensselaer Polytechnic Institute, 1623 15th Street, Troy, New York 12180, United States of America
| |
Collapse
|
50
|
Khorshidi S, Karkhaneh A. A review on gradient hydrogel/fiber scaffolds for osteochondral regeneration. J Tissue Eng Regen Med 2018; 12:e1974-e1990. [PMID: 29243352 DOI: 10.1002/term.2628] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2016] [Revised: 07/17/2017] [Accepted: 11/27/2017] [Indexed: 12/31/2022]
Abstract
Osteochondral tissue regeneration is a complicated field due to the distinct properties and healing potential of osseous and chondral phases. In a natural osteochondral region, the composition, mechanics, and structure vary smoothly from bony to cartilaginous phase. Therefore, a homogeneous scaffold cannot satisfy the complexity of the osteochondral matrix. In essence, a natural extracellular matrix is composed of fibrous proteins elongated into a gelatinous background. A hydrogel/fiber scaffold possessing gradient in both phases would be of the utmost interest to imitate tissue arrangement of a native osteochondral interface. However, there are limited research works that exploit hydrogel/fiber scaffolds for osteochondral restoration. In the present review, currently used fibrous or gelatinous scaffolds for osteochondral damages are discussed. Moreover, superiority of using gradient hydrogel/fiber composites for osteochondral regeneration and practical approaches to develop those scaffolds is debated.
Collapse
Affiliation(s)
- Sajedeh Khorshidi
- Biomedical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran
| | - Akbar Karkhaneh
- Biomedical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran
| |
Collapse
|