1
|
Öksüz KE, Arslan S. Sustainable Synthesis of Multifunctionalized Amoxicillin-Loaded Biopolymer Foams. ACS OMEGA 2025; 10:15525-15539. [PMID: 40290945 PMCID: PMC12019502 DOI: 10.1021/acsomega.5c00442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 03/15/2025] [Accepted: 03/24/2025] [Indexed: 04/30/2025]
Abstract
The development of biocompatible biopolymer foams loaded with antibiotics is crucial to advancing drug delivery systems in biomedical engineering. These materials offer controlled drug release and specialized functionalities for improved therapeutic outcomes. This study presents the development and characterization of antimicrobial polymeric biofoam materials loaded with the drug amoxicillin (AMX). The sustainable synthesis of these biopolymer foams involves a cost-effective, eco-friendly method that incorporates natural starch within poly(vinyl alcohol) (PVA) through an aldehyde cross-linking/stabilizing process. The highly porous structure of the biofoams enabled effective impregnation of the AMX drug using an innovative process involving ultrasonication and vacuum pressure to maximize efficiency and minimize biomaterial loss. The findings demonstrate the potential of these PVA/starch-based biofoams as versatile drug delivery systems with desirable physicochemical and biological characteristics. Detailed investigations were conducted to evaluate morphological features, chemical properties, swelling behavior, in vitro biodegradability, drug release profiles, cell culture, and antimicrobial activity tests of the prepared biofoam samples. Investigating the effect of controlled loading of AMX under laboratory conditions on its release profile and studying its biodegradation in various environments over time represent a critical aspect of this research. The optimal release profile under physiological conditions and the potent inhibition of bacterial growth against Escherichia coli and Staphylococcus aureus microorganisms by AMX-loaded biofoam materials highlight their potential for biomedical applications. These materials show promise for the in vivo administration and local treatment of bacterial infections.
Collapse
Affiliation(s)
- Kerim Emre Öksüz
- Department
of Metallurgical and Materials Engineering, Sivas Cumhuriyet University, Sivas 58140, Türkiye
- Institute
of Science and Technology, Department of Bioengineering, Sivas Cumhuriyet University, Sivas 58140, Türkiye
| | - Saynur Arslan
- Department
of Metallurgical and Materials Engineering, Sivas Cumhuriyet University, Sivas 58140, Türkiye
| |
Collapse
|
2
|
ten Brink T, Damanik F, Rotmans JI, Moroni L. Unraveling and Harnessing the Immune Response at the Cell-Biomaterial Interface for Tissue Engineering Purposes. Adv Healthc Mater 2024; 13:e2301939. [PMID: 38217464 PMCID: PMC11468937 DOI: 10.1002/adhm.202301939] [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: 06/19/2023] [Revised: 12/14/2023] [Indexed: 01/15/2024]
Abstract
Biomaterials are defined as "engineered materials" and include a range of natural and synthetic products, designed for their introduction into and interaction with living tissues. Biomaterials are considered prominent tools in regenerative medicine that support the restoration of tissue defects and retain physiologic functionality. Although commonly used in the medical field, these constructs are inherently foreign toward the host and induce an immune response at the material-tissue interface, defined as the foreign body response (FBR). A strong connection between the foreign body response and tissue regeneration is suggested, in which an appropriate amount of immune response and macrophage polarization is necessary to trigger autologous tissue formation. Recent developments in this field have led to the characterization of immunomodulatory traits that optimizes bioactivity, the integration of biomaterials and determines the fate of tissue regeneration. This review addresses a variety of aspects that are involved in steering the inflammatory response, including immune cell interactions, physical characteristics, biochemical cues, and metabolomics. Harnessing the advancing knowledge of the FBR allows for the optimization of biomaterial-based implants, aiming to prevent damage of the implant, improve natural regeneration, and provide the tools for an efficient and successful in vivo implantation.
Collapse
Affiliation(s)
- Tim ten Brink
- Complex Tissue Regeneration DepartmentMERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityUniversiteitssingel 40Maastricht6229ERThe Netherlands
| | - Febriyani Damanik
- Complex Tissue Regeneration DepartmentMERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityUniversiteitssingel 40Maastricht6229ERThe Netherlands
| | - Joris I. Rotmans
- Department of Internal MedicineLeiden University Medical CenterAlbinusdreef 2Leiden2333ZAThe Netherlands
| | - Lorenzo Moroni
- Complex Tissue Regeneration DepartmentMERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityUniversiteitssingel 40Maastricht6229ERThe Netherlands
| |
Collapse
|
3
|
Yuan Y, Tyson C, Szyniec A, Agro S, Tavakol TN, Harmon A, Lampkins D, Pearson L, Dumas JE, Taite LJ. Bioactive Polyurethane-Poly(ethylene Glycol) Diacrylate Hydrogels for Applications in Tissue Engineering. Gels 2024; 10:108. [PMID: 38391438 PMCID: PMC10887679 DOI: 10.3390/gels10020108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 01/22/2024] [Accepted: 01/25/2024] [Indexed: 02/24/2024] Open
Abstract
Polyurethanes (PUs) are a highly adaptable class of biomaterials that are among some of the most researched materials for various biomedical applications. However, engineered tissue scaffolds composed of PU have not found their way into clinical application, mainly due to the difficulty of balancing the control of material properties with the desired cellular response. A simple method for the synthesis of tunable bioactive poly(ethylene glycol) diacrylate (PEGDA) hydrogels containing photocurable PU is described. These hydrogels may be modified with PEGylated peptides or proteins to impart variable biological functions, and the mechanical properties of the hydrogels can be tuned based on the ratios of PU and PEGDA. Studies with human cells revealed that PU-PEG blended hydrogels support cell adhesion and viability when cell adhesion peptides are crosslinked within the hydrogel matrix. These hydrogels represent a unique and highly tailorable system for synthesizing PU-based synthetic extracellular matrices for tissue engineering applications.
Collapse
Affiliation(s)
- Yixuan Yuan
- Department of Chemical Engineering, University of Virginia, Charlottesville, VA 22903, USA
| | - Caleb Tyson
- Department of Chemical Engineering, Hampton University, Hampton, VA 23668, USA
| | - Annika Szyniec
- Department of Chemical Engineering, University of Virginia, Charlottesville, VA 22903, USA
| | - Samuel Agro
- Department of Chemical Engineering, University of Virginia, Charlottesville, VA 22903, USA
| | - Tara N Tavakol
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
| | - Alexander Harmon
- Department of Chemical Engineering, Hampton University, Hampton, VA 23668, USA
| | - DessaRae Lampkins
- Department of Chemical Engineering, Hampton University, Hampton, VA 23668, USA
| | - Lauran Pearson
- Department of Chemical Engineering, Hampton University, Hampton, VA 23668, USA
| | - Jerald E Dumas
- Joint School of Nanoscience and Nanoengineering, North Carolina Agricultural & Technical State University, Greensboro, NC 27401, USA
| | - Lakeshia J Taite
- Department of Chemical Engineering, University of Virginia, Charlottesville, VA 22903, USA
| |
Collapse
|
4
|
Mu M, Leermakers FAM, Chen J, Holmes M, Ettelaie R. Effect of polymer architecture on the adsorption behaviour of amphiphilic copolymers: A theoretical study. J Colloid Interface Sci 2023; 644:333-345. [PMID: 37120882 DOI: 10.1016/j.jcis.2023.04.051] [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: 10/27/2022] [Revised: 04/05/2023] [Accepted: 04/12/2023] [Indexed: 05/02/2023]
Abstract
HYPOTHESIS Polymer architecture is known to have significant impact on its adsorption behaviour. Most studies have been concerned with the more concentrated, "close to surface saturation" regime of the isotherm, where complications such as lateral interactions and crowding also additionally affect the adsorption. We compare a variety of amphiphilic polymer architectures by determining their Henry's adsorption constant (kH), which, as with other surface active molecules, is the proportionality constant between surface coverage and bulk polymer concentration in a sufficiently dilute regime. It is speculated that not only the number of arms or branches, but also the position of adsorbing hydrophobes influence the adsorption, and that by controlling the latter the two can counteract each other. METHODOLOGY The Self-consistent field calculation of Scheutjens and Fleer was implemented to calculate the adsorbed amount of polymer for many different polymer architectures including linear, star and dendritic. Using the adsorption isotherms at very low bulk concentrations, we determined the value of kH for these. FINDINGS It is found that the branched structures (star polymers and dendrimers) can be viewed as analogues of linear block polymers based on the location of their adsorbing units. Polymers containing consecutive trains of adsorbing hydrophobes in all cases showed higher level of adsorption compared to their counterparts, where the hydrophobes were more uniformly distributed on the chains. While increasing the number of branches (or arms for star polymers) also confirmed the known result that the adsorption decreased with the number of arms, this trend can be partially offset by the appropriate choice of the location of anchoring groups.
Collapse
Affiliation(s)
- Mingduo Mu
- Food Colloids Group, School of Food Science and Nutrition, University of Leeds, Woodhouse Lane, Leeds LS2 9JT, UK.
| | - Frans A M Leermakers
- Wageningen Univ & Res, Phys Chem & Soft Matter, Stippeneng 4, 6708 WE Wageningen, Netherlands
| | - Jianshe Chen
- School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, Zhejiang 310018, China
| | - Melvin Holmes
- Food Colloids Group, School of Food Science and Nutrition, University of Leeds, Woodhouse Lane, Leeds LS2 9JT, UK
| | - Rammile Ettelaie
- Food Colloids Group, School of Food Science and Nutrition, University of Leeds, Woodhouse Lane, Leeds LS2 9JT, UK.
| |
Collapse
|
5
|
Trossmann VT, Scheibel T. Design of Recombinant Spider Silk Proteins for Cell Type Specific Binding. Adv Healthc Mater 2023; 12:e2202660. [PMID: 36565209 PMCID: PMC11468868 DOI: 10.1002/adhm.202202660] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 12/19/2022] [Indexed: 12/25/2022]
Abstract
Cytophilic (cell-adhesive) materials are very important for tissue engineering and regenerative medicine. However, for engineering hierarchically organized tissue structures comprising different cell types, cell-specific attachment and guidance are decisive. In this context, materials made of recombinant spider silk proteins are promising scaffolds, since they exhibit high biocompatibility, biodegradability, and the underlying proteins can be genetically functionalized. Here, previously established spider silk variants based on the engineered Araneus diadematus fibroin 4 (eADF4(C16)) are genetically modified with cell adhesive peptide sequences from extracellular matrix proteins, including IKVAV, YIGSR, QHREDGS, and KGD. Interestingly, eADF4(C16)-KGD as one of 18 tested variants is cell-selective for C2C12 mouse myoblasts, one out of 11 tested cell lines. Co-culturing with B50 rat neuronal cells confirms the cell-specificity of eADF4(C16)-KGD material surfaces for C2C12 mouse myoblast adhesion.
Collapse
Affiliation(s)
- Vanessa Tanja Trossmann
- Chair of BiomaterialsEngineering FacultyUniversity of BayreuthProf.‐Rüdiger‐Bormann‐Straße 195447BayreuthGermany
| | - Thomas Scheibel
- Chair of BiomaterialsEngineering FacultyUniversity of BayreuthProf.‐Rüdiger‐Bormann‐Straße 195447BayreuthGermany
- Bayreuth Center for Colloids and Interfaces (BZKG)Bavarian Polymer Institute (BPI)Bayreuth Center for Molecular Biosciences (BZMB)Bayreuth Center for Material Science (BayMAT)University of BayreuthUniversitätsstraße 3095447BayreuthGermany
| |
Collapse
|
6
|
López-Valverde N, Aragoneses J, López-Valverde A, Quispe-López N, Rodríguez C, Aragoneses JM. Effectiveness of biomolecule-based bioactive surfaces, on os-seointegration of titanium dental implants: A systematic review and meta-analysis of in vivo studies. Front Bioeng Biotechnol 2022; 10:986112. [PMID: 36225604 PMCID: PMC9548556 DOI: 10.3389/fbioe.2022.986112] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 09/12/2022] [Indexed: 12/09/2022] Open
Abstract
Titanium and alloy osseointegrated implants are used to replace missing teeth; however, some fail and are removed. Modifications of the implant surface with biologically active substances have been proposed. MEDLINE [via Pubmed], Embase and Web of Science were searched with the terms “titanium dental implants”, “surface properties”, “bioactive surface modifications”, “biomolecules”, “BMP”, “antibacterial agent”, “peptide”, “collagen”, “grown factor”, “osseointegration”, “bone apposition”, “osteogenic”, “osteogenesis”, “new bone formation”, “bone to implant contact”, “bone regeneration” and “in vivo studies”, until May 2022. A total of 10,697 references were iden-tified and 26 were included to analyze 1,109 implants, with follow-ups from 2 to 84 weeks. The ARRIVE guidelines and the SYRCLE tool were used to evaluate the methodology and scientific evidence. A meta-analysis was performed (RevMan 2020 software, Cochane Collaboration) with random effects that evaluated BIC at 4 weeks, with subgroups for the different coatings. The heterogeneity of the pooled studies was very high (95% CI, I2 = 99%). The subgroup of BMPs was the most favorable to coating. Surface modification of Ti implants by organic bioactive molecules seems to favor osseointegration in the early stages of healing, but long-term studies are necessary to corroborate the results of the experimental studies.
Collapse
Affiliation(s)
- Nansi López-Valverde
- Department of Medicine and Medical Specialties, Faculty of Health Sciences, Universidad Alcalá de Henares, Madrid, Spain
| | - Javier Aragoneses
- Department of Medicine and Medical Specialties, Faculty of Health Sciences, Universidad Alcalá de Henares, Madrid, Spain
| | - Antonio López-Valverde
- Department of Surgery, Instituto de Investigación Biomédica de Salamanca (IBSAL), University of Salamanca, Salamanca, Spain
- Department of Dentistry, Universidad Federico Henríquez y Carvajal, Santo Domingo, Dominican Republic
- *Correspondence: Antonio López-Valverde,
| | - Norberto Quispe-López
- Department of Surgery, Instituto de Investigación Biomédica de Salamanca (IBSAL), University of Salamanca, Salamanca, Spain
| | - Cinthia Rodríguez
- Department of Dentistry, Universidad Federico Henríquez y Carvajal, Santo Domingo, Dominican Republic
| | | |
Collapse
|
7
|
Intracerebral Transplantation of Mesenchymal Stromal Cell Compounded with Recombinant Peptide Scaffold against Chronic Intracerebral Hemorrhage Model. Stem Cells Int 2022; 2022:8521922. [PMID: 35966129 PMCID: PMC9372516 DOI: 10.1155/2022/8521922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 06/17/2022] [Accepted: 06/28/2022] [Indexed: 11/18/2022] Open
Abstract
Background Due to the lack of effective therapies, stem cell transplantation is an anticipated treatment for chronic intracerebral hemorrhage (ICH), and higher cell survival and engraftment are considered to be the key for recovery. Mesenchymal stromal cells (MSCs) compounded with recombinant human collagen type I scaffolds (CellSaics) have a higher potential for cell survival and engraftment compared with solo-MSCs, and we investigated the validity of intracerebral transplantation of CellSaic in a chronic ICH model. Methods Rat CellSaics (rCellSaics) were produced by rat bone marrow-derived MSC (rBMSCs). The secretion potential of neurotrophic factors and the cell proliferation rate were compared under oxygen-glucose deprivation (OGD) conditions. rCellSaics, rBMSCs, or saline were transplanted into the hollow cavity of a rat chronic ICH model. Functional and histological analyses were evaluated, and single-photon emission computed tomography for benzodiazepine receptors was performed to monitor sequential changes in neuronal integrity. Furthermore, human CellSaics (hCellSaics) were transplanted into a chronic ICH model in immunodeficient rats. Antibodies neutralizing brain-derived neurotrophic factor (BDNF) were used to elucidate its mode of action. Results rCellSaics demonstrated a higher secretion potential of trophic factors and showed better cell proliferation in the OGD condition. Animals receiving rCellSaics displayed better neurological recovery, higher intracerebral BDNF, and better cell engraftment; they also showed a tendency for less brain atrophy and higher benzodiazepine receptor preservation. hCellSaics also promoted significant functional recovery, which was reversed by BDNF neutralization. Conclusion Intracerebral transplantation of CellSaics enabled neurological recovery in a chronic ICH model and may be a good option for clinical application.
Collapse
|
8
|
Schotman MJG, Fransen PP, Song J, Dankers PYW. Tuning the affinity of amphiphilic guest molecules in a supramolecular polymer transient network. RSC Adv 2022; 12:14052-14060. [PMID: 35558837 PMCID: PMC9088426 DOI: 10.1039/d2ra00346e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 04/11/2022] [Indexed: 11/25/2022] Open
Abstract
Dynamicity plays a central role in biological systems such as in the cellular microenvironment. Here, the affinity and dynamics of different guest molecules in a transient supramolecular polymer hydrogel system, i.e. the host network, are investigated. The hydrogel system consists of bifunctional ureido-pyrimidinone (UPy) poly(ethylene glycol) polymers. A monofunctional complementary UPy guest is introduced, designed to interact with the host network based on UPy–UPy interactions. Furthermore, two other guest molecules are synthesized, being cholesterol and dodecyl (c12) guests; both designed to interact with the host network via hydrophobic interactions. At the nanoscale in solution, differences in morphology of the guest molecules were observed. The UPy–guest molecule formed fibers, and the cholesterol and c12 guests formed aggregates. Furthermore, cellular internalization of fluorescent guest molecules was studied. No cellular uptake of the UPy–cy5 guest was observed, whereas the cholesterol–cy5 guest showed membrane binding and cellular uptake. Also the c12–cy5 guest showed cellular uptake. Formulation of the guest molecules into the UPy hydrogel system was done to study the guest–host affinity. No changes in mechanical properties as measured with rheology were found upon guest–hydrogel formulation. Fluorescence recovery after photobleaching showed the diffusive properties of the cy5-functionalized guests throughout the host network. The c12 guest displayed a relatively fast mobility, the UPy guest displayed a decrease in mobility, and the cholesterol–guest remained relatively stable in the host network with little mobility. This demonstrates the tunable dynamic differences of affinity-based interaction between guest molecules and the host network. Interestingly, the cholesterol guest is internalized in cells and is robustly incorporated in the hydrogel network, while the UPy guest is not taken up by cells but shows an affinity to the hydrogel network. These results show the importance of guest–hydrogel affinity for future drug release. However, if modified with cholesterol these guests, or future drugs, will be taken up by cells; if modified with a UPy unit this does not occur. In this way both the drug–hydrogel interaction and the cell internalization behavior can be tuned. Regulating the host–guest dynamics in transient hydrogels opens the door to various drug delivery purposes and tissue engineering. Dynamicity plays a central role in biological systems, which can be mimicked by tuning dynamicity in hydrogel networks.![]()
Collapse
Affiliation(s)
- Maaike J G Schotman
- Institute for Complex Molecular Systems, Eindhoven University of Technology P. O. Box 513 Eindhoven 5600 MB The Netherlands .,Department of Biomedical Engineering, Laboratory of Chemical Biology, Eindhoven University of Technology P. O. Box 513 Eindhoven 5600 MB The Netherlands
| | - Peter-Paul Fransen
- Institute for Complex Molecular Systems, Eindhoven University of Technology P. O. Box 513 Eindhoven 5600 MB The Netherlands .,Department of Biomedical Engineering, Laboratory of Chemical Biology, Eindhoven University of Technology P. O. Box 513 Eindhoven 5600 MB The Netherlands
| | - Jiankang Song
- Institute for Complex Molecular Systems, Eindhoven University of Technology P. O. Box 513 Eindhoven 5600 MB The Netherlands .,Department of Biomedical Engineering, Laboratory of Chemical Biology, Eindhoven University of Technology P. O. Box 513 Eindhoven 5600 MB The Netherlands
| | - Patricia Y W Dankers
- Institute for Complex Molecular Systems, Eindhoven University of Technology P. O. Box 513 Eindhoven 5600 MB The Netherlands .,Department of Biomedical Engineering, Laboratory of Chemical Biology, Eindhoven University of Technology P. O. Box 513 Eindhoven 5600 MB The Netherlands.,Department of Biomedical Engineering, Laboratory for Cell and Tissue Engineering, Eindhoven University of Technology P. W. Box 513 5600 MB The Netherlands
| |
Collapse
|
9
|
Ji YR, Hsu YH, Syue MH, Wang YC, Lin SY, Huang TW, Young TH. Controlled Decomposable Hydrogel Triggered with a Specific Enzyme. ACS OMEGA 2022; 7:3254-3261. [PMID: 35128237 PMCID: PMC8811883 DOI: 10.1021/acsomega.1c05178] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 01/12/2022] [Indexed: 06/14/2023]
Abstract
In this study, superabsorbent polyelectrolyte hydrogels were synthesized by cross-linking a nondegradable poly (allylamine hydrochloride) (PAH) and a recombinant protein with a specific enzymatic cleavage site. The recombinant protein was produced by E. coli with the pET-32b(+) plasmid, which is featured with the thioredoxin (Trx) gene containing a thrombin recognition site and a T7/lac hybrid promoter for high expression of recombinant protein. The swelling test shows that the composite hydrogel still maintained a high swelling ratio to 900% when 15% recombinant protein was cross-linked with PAH. The degradation test shows that such a PAH composite hydrogel could be decomposed by the addition of specific enzyme thrombin, which might lead to new biomedical applications of hydrogels needed to be decomposable by specific time not determined by the time period.
Collapse
Affiliation(s)
- You-Ren Ji
- Institute
of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, Taipei 100, Taiwan
| | - Ya-Hsiang Hsu
- Institute
of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, Taipei 100, Taiwan
| | - Ming-Hua Syue
- Institute
of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, Taipei 100, Taiwan
| | - Ying-Chu Wang
- Institute
of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, Taipei 100, Taiwan
| | - Shyr-Yi Lin
- Division
of Gastroenterology, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei 116, Taiwan
- Department
of General Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Tsung-Wei Huang
- Department
of Electrical Engineering, College of Electrical and Communication
Engineering, Yuan Ze University, Taoyuan 320, Taiwan
- Department
of Otolaryngology, Far Eastern Memorial
Hospital, New Taipei City 220, Taiwan
| | - Tai-Horng Young
- Institute
of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, Taipei 100, Taiwan
- Department
of Biomedical Engineering, National Taiwan
University Hospital, Taipei 100, Taiwan
| |
Collapse
|
10
|
Li J, Yu X, Martinez EE, Zhu J, Wang T, Shi S, Shin SR, Hassan S, Guo C. Emerging Biopolymer-Based Bioadhesives. Macromol Biosci 2021; 22:e2100340. [PMID: 34957668 DOI: 10.1002/mabi.202100340] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 11/23/2021] [Indexed: 12/13/2022]
Abstract
Bioadhesives have been widely used in healthcare and biomedical applications due to their ease-of-operation for wound closure and repair compared to conventional suturing and stapling. However, several challenges remain for developing ideal bioadhesives, such as unsatisfied mechanical properties, non-tunable biodegradability, and limited biological functions. Considering these concerns, naturally derived biopolymers have been considered good candidates for making bioadhesives owing to their ready availability, facile modification, tunable mechanical properties, and desired biocompatibility and biodegradability. Over the past several years, remarkable progress has been made on biopolymer-based adhesives, covering topics from novel materials designs and advanced processing to clinical translation. The developed bioadhesives have been applied for diverse applications, including tissue adhesion, hemostasis, antimicrobial, wound repair/tissue regeneration, and skin-interfaced bioelectronics. Here in this comprehensive review, recent progress on biopolymer-based bioadhesives is summarized with focuses on clinical translations and multifunctional bioadhesives. Furthermore, challenges and opportunities such as weak adhesion strength at the hydrated state, mechanical mismatch with tissues, and unfavorable immune responses are discussed with an aim to facilitate the future development of high-performance biopolymer-based bioadhesives.
Collapse
Affiliation(s)
- Jinghang Li
- School of Engineering, Westlake University, Hangzhou, Zhejiang Province, 310024, China.,School of Materials Science and Engineering, Wuhan Institute of Technology, Wuhan, Hubei Province, 430205, China
| | - Xin Yu
- School of Engineering, Westlake University, Hangzhou, Zhejiang Province, 310024, China
| | | | - Jiaqing Zhu
- School of Materials Science and Engineering, Wuhan Institute of Technology, Wuhan, Hubei Province, 430205, China
| | - Ting Wang
- Department of Laboratory Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, 210029, China
| | - Shengwei Shi
- School of Materials Science and Engineering, Wuhan Institute of Technology, Wuhan, Hubei Province, 430205, China
| | - Su Ryon Shin
- Division of Engineering in Medicine, Department of Medicine, Harvard Medical School, and Brigham and Women's Hospital, Cambridge, MA, 02139, USA
| | - Shabir Hassan
- Division of Engineering in Medicine, Department of Medicine, Harvard Medical School, and Brigham and Women's Hospital, Cambridge, MA, 02139, USA
| | - Chengchen Guo
- School of Engineering, Westlake University, Hangzhou, Zhejiang Province, 310024, China
| |
Collapse
|
11
|
Zhang Q, Li M, Hu W, Wang X, Hu J. Spidroin-Based Biomaterials in Tissue Engineering: General Approaches and Potential Stem Cell Therapies. Stem Cells Int 2021; 2021:7141550. [PMID: 34966432 PMCID: PMC8712125 DOI: 10.1155/2021/7141550] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 09/25/2021] [Accepted: 11/10/2021] [Indexed: 01/09/2023] Open
Abstract
Spider silks are increasingly gaining interest for potential use as biomaterials in tissue engineering and biomedical applications. Owing to their facile and versatile processability in native and regenerated forms, they can be easily tuned via chemical synthesis or recombinant technologies to address specific issues required for applications. In the past few decades, native spider silk and recombinant silk materials have been explored for a wide range of applications due to their superior strength, toughness, and elasticity as well as biocompatibility, biodegradation, and nonimmunogenicity. Herein, we present an overview of the recent advances in spider silk protein that fabricate biomaterials for tissue engineering and regenerative medicine. Beginning with a brief description of biological and mechanical properties of spidroin-based materials and the cellular regulatory mechanism, this review summarizes various types of spidroin-based biomaterials from genetically engineered spider silks and their prospects for specific biomedical applications (e.g., lung tissue engineering, vascularization, bone and cartilage regeneration, and peripheral nerve repair), and finally, we prospected the development direction and manufacturing technology of building more refined and customized spidroin-based protein scaffolds.
Collapse
Affiliation(s)
- Qi Zhang
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong
| | - Min Li
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong
| | - Wenbo Hu
- Biological Science Research Center, Southwest University, Chongqing 400716, China
| | - Xin Wang
- Biological Science Research Center, Southwest University, Chongqing 400716, China
| | - Jinlian Hu
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong
| |
Collapse
|
12
|
Zhao G, Ge T, Yan Y, Shuai Q, Su WK. Highly Efficient Modular Construction of Functional Drug Delivery Platform Based on Amphiphilic Biodegradable Polymers via Click Chemistry. Int J Mol Sci 2021; 22:10407. [PMID: 34638747 PMCID: PMC8508947 DOI: 10.3390/ijms221910407] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 09/20/2021] [Accepted: 09/21/2021] [Indexed: 12/03/2022] Open
Abstract
Amphiphilic copolymers with pendant functional groups in polyester segments are widely used in nanomedicine. These enriched functionalities are designed to form covalent conjugates with payloads or provide additional stabilization effects for encapsulated drugs. A general method is successfully developed for the efficient preparation of functional biodegradable PEG-polyester copolymers via click chemistry. Firstly, in the presence of mPEG as initiator, Sn(Oct)2-catalyzed ring-opening polymerization of the α-alkynyl functionalized lactone with D,L-lactide or ε-caprolactone afforded linear mPEG-polyesters bearing multiple pendant alkynyl groups. Kinetic studies indicated the formation of random copolymers. Through copper-catalyzed azide-alkyne cycloaddition reaction, various small azido molecules with different functionalities to polyester segments are efficiently grafted. The molecular weights, polydispersities and grafting efficiencies of azido molecules of these copolymers were investigated by NMR and GPC. Secondly, it is demonstrated that the resulting amphiphilic functional copolymers with low CMC values could self-assemble to form nanoparticles in aqueous media. In addition, the in vitro degradation study and cytotoxicity assays indicated the excellent biodegradability and low cytotoxicity of these copolymers. This work provides a general approach toward the preparation of functional PEG-polyester copolymers in a quite efficient way, which may further facilitate the application of functional PEG-polyesters as drug delivery materials.
Collapse
Affiliation(s)
- Guangkuo Zhao
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014, China; (G.Z.); (T.G.)
| | - Tongtong Ge
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014, China; (G.Z.); (T.G.)
| | - Yunfeng Yan
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310014, China;
- State Key Laboratory of Molecular Engineering of Polymers, Fudan University, 2005 Songhu Road, Shanghai 200433, China
| | - Qi Shuai
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014, China; (G.Z.); (T.G.)
| | - Wei-Ke Su
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014, China; (G.Z.); (T.G.)
| |
Collapse
|
13
|
Chameettachal S, Puranik CJ, Veluthedathu MN, Chalil NB, John R, Pati F. Thickening of Ectatic Cornea through Regeneration Using Decellularized Corneal Matrix Injectable Hydrogel: A Strategic Advancement to Mitigate Corneal Ectasia. ACS APPLIED BIO MATERIALS 2021; 4:7300-7313. [PMID: 35006959 DOI: 10.1021/acsabm.1c00821] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Ectatic corneal diseases are a group of eye disorders characterized by progressive thinning and outward bulging of the cornea, resulting in vision impairment. A few attempts have been made to use cornea-derived extracellular matrix hydrogels for corneal tissue engineering; however, no studies have investigated its application in corneal ectasia. In this study, we have first developed an animal surgical model that mimics a few specific phenotypes of ectatic cornea. Later, we investigated the potential of decellularized cornea matrix hydrogels (dCMH) from both human and bovine sources in increasing the thickness of the cornea in the developed surgical model. Our data advocate that surgical stromal depletion can be followed to establish ectatic models and can also provide information on the biocompatibility of materials, its integration with native stroma, degradation over time, and tissue remodeling. We observed that dCMH from both sources could integrate with ectatic thin corneal stroma and helps in regaining the thickness by regenerating a reasonably functional and transparent stroma; however, no significant difference was spotted between the dCMH made from human and bovine corneal tissue sources. Hence, this study is a promising step toward developing a non-invasive technique for the treatment of corneal ectasia by using dCMH.
Collapse
Affiliation(s)
- Shibu Chameettachal
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi, Sangareddy Hyderabad, Telangana 502284, India
| | - Charuta J Puranik
- Oculus Regenerus Eye Care and Research Center, Nanalnagar, Hyderabad, Telangana 500008, India
| | - Mohamed Nijas Veluthedathu
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi, Sangareddy Hyderabad, Telangana 502284, India
| | - Najathulla Bhagavathi Chalil
- Department of Materials Science and Metallurgical Engineering, Indian Institute of Technology Hyderabad, Kandi, Sangareddy , Hyderabad, Telangana 502284, India
| | - Renu John
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi, Sangareddy Hyderabad, Telangana 502284, India
| | - Falguni Pati
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi, Sangareddy Hyderabad, Telangana 502284, India
| |
Collapse
|
14
|
Liu Z, Tamaddon M, Chen SM, Wang H, San Cheong V, Gang F, Sun X, Liu C. Determination of an Initial Stage of the Bone Tissue Ingrowth Into Titanium Matrix by Cell Adhesion Model. Front Bioeng Biotechnol 2021; 9:736063. [PMID: 34589474 PMCID: PMC8473621 DOI: 10.3389/fbioe.2021.736063] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Accepted: 07/21/2021] [Indexed: 11/30/2022] Open
Abstract
For achieving early intervention treatment to help patients delay or avoid joint replacement surgery, a personalized scaffold should be designed coupling the effects of mechanical, fluid mechanical, chemical, and biological factors on tissue regeneration, which results in time- and cost-consuming trial-and-error analyses to investigate the in vivo test and related experimental tests. To optimize the fluid mechanical and material properties to predict osteogenesis and cartilage regeneration for the in vivo and clinical trial, a simulation approach is developed for scaffold design, which is composed of a volume of a fluid model for simulating the bone marrow filling process of the bone marrow and air, as well as a discrete phase model and a cell impingement model for tracking cell movement during bone marrow fillings. The bone marrow is treated as a non-Newtonian fluid, rather than a Newtonian fluid, because of its viscoelastic property. The simulation results indicated that the biofunctional bionic scaffold with a dense layer to prevent the bone marrow flow to the cartilage layer and synovia to flow into the trabecular bone area guarantee good osteogenesis and cartilage regeneration, which leads to high-accuracy in vivo tests in sheep . This approach not only predicts the final bioperformance of the scaffold but also could optimize the scaffold structure and materials by their biochemical, biological, and biomechanical properties.
Collapse
Affiliation(s)
- Ziyu Liu
- Division of Surgery & Interventional Science, University College London, Royal National Orthopaedic Hospital, London, United Kingdom
- School of Engineering Medicine, Beihang University, Beijing, China
- Key Laboratory of Advanced Materials of Ministry of Education of China, School of Materials Science and Engineering, Tsinghua University, Beijing, China
| | - Maryam Tamaddon
- Division of Surgery & Interventional Science, University College London, Royal National Orthopaedic Hospital, London, United Kingdom
| | - Shen-Mao Chen
- Division of Surgery & Interventional Science, University College London, Royal National Orthopaedic Hospital, London, United Kingdom
| | - Haoyu Wang
- Division of Surgery & Interventional Science, University College London, Royal National Orthopaedic Hospital, London, United Kingdom
| | - Vee San Cheong
- Insigno Institute of in Silico Medicine and Department of Automatic Control and Systems Engineering, University of Sheffield, Sheffield, United Kingdom
| | - Fangli Gang
- School of Engineering Medicine, Beihang University, Beijing, China
- Key Laboratory of Advanced Materials of Ministry of Education of China, School of Materials Science and Engineering, Tsinghua University, Beijing, China
| | - Xiaodan Sun
- Key Laboratory of Advanced Materials of Ministry of Education of China, School of Materials Science and Engineering, Tsinghua University, Beijing, China
| | - Chaozong Liu
- Division of Surgery & Interventional Science, University College London, Royal National Orthopaedic Hospital, London, United Kingdom
| |
Collapse
|
15
|
Ledford B, Barron C, Van Dyke M, He JQ. Keratose hydrogel for tissue regeneration and drug delivery. Semin Cell Dev Biol 2021; 128:145-153. [PMID: 34219034 DOI: 10.1016/j.semcdb.2021.06.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 05/16/2021] [Accepted: 06/23/2021] [Indexed: 11/28/2022]
Abstract
Keratin (KRT), a natural fibrous structural protein, can be classified into two categories: "soft" cytosolic KRT that is primarily found in the epithelia tissues (e.g., skin, the inner lining of digestive tract) and "hard" KRT that is mainly found in the protective tissues (e.g., hair, horn). The latter is the predominant form of KRT widely used in biomedical research. The oxidized form of extracted KRT is exclusively denoted as keratose (KOS) while the reduced form of KRT is termed as kerateine (KRTN). KOS can be processed into various forms (e.g., hydrogel, films, fibers, and coatings) for different biomedical applications. KRT/KOS offers numerous advantages over other types of biomaterials, such as bioactivity, biocompatibility, degradability, immune/inflammatory privileges, mechanical resilience, chemical manipulability, and easy accessibility. As a result, KRT/KOS has attracted considerable attention and led to a large number of publications associated with this biomaterial over the past few decades; however, most (if not all) of the published review articles focus on KRT regarding its molecular structure, biochemical/biophysical properties, bioactivity, biocompatibility, drug/cell delivery, and in vivo transplantation, as well as its applications in biotechnical products and medical devices. Current progress that is directly associated with KOS applications in tissue regeneration and drug delivery appears an important topic that merits a commentary. To this end, the present review aims to summarize the current progress of KOS-associated biomedical applications, especially focusing on the in vitro and in vivo effects of KOS hydrogel on cultured cells and tissue regeneration following skin injury, skeletal muscle loss, peripheral nerve injury, and cardiac infarction.
Collapse
Affiliation(s)
- Benjamin Ledford
- Department of Biomedical Sciences and Pathobiology, College of Veterinary Medicine, Virginia Tech, Blacksburg, VA 24061, USA
| | - Catherine Barron
- Department of Biomedical Sciences and Pathobiology, College of Veterinary Medicine, Virginia Tech, Blacksburg, VA 24061, USA
| | - Mark Van Dyke
- Department of Biomedical Engineering, College of Engineering, University of Arizona, 1209 E. 2nd Street, Tucson, AZ 85721, USA
| | - Jia-Qiang He
- Department of Biomedical Sciences and Pathobiology, College of Veterinary Medicine, Virginia Tech, Blacksburg, VA 24061, USA.
| |
Collapse
|
16
|
Speer JE, Barcellona MN, Lu MY, Zha Z, Jing L, Gupta MC, Buchowski JM, Kelly MP, Setton LA. Development of a library of laminin-mimetic peptide hydrogels for control of nucleus pulposus cell behaviors. J Tissue Eng 2021; 12:20417314211021220. [PMID: 34188794 PMCID: PMC8211742 DOI: 10.1177/20417314211021220] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 05/12/2021] [Indexed: 12/15/2022] Open
Abstract
The nucleus pulposus (NP) of the intervertebral disc plays a critical role in
distributing mechanical loads to the axial skeleton. Alterations in NP cells and,
consequently, NP matrix are some of the earliest changes in the development of disc
degeneration. Previous studies demonstrated a role for laminin-presenting biomaterials in
promoting a healthy phenotype for human NP cells from degenerated tissue. Here we
investigate the use of laminin-mimetic peptides presented individually or in combination
on a poly(ethylene) glycol hydrogel as a platform to modulate the behaviors of
degenerative human NP cells. Data confirm that NP cells attach to select laminin-mimetic
peptides that results in cell signaling downstream of integrin and syndecan binding.
Furthermore, the peptide-functionalized hydrogels demonstrate an ability to promote cell
behaviors that mimic that of full-length laminins. These results identify a set of
peptides that can be used to regulate NP cell behaviors toward a regenerative engineering
strategy.
Collapse
Affiliation(s)
- Julie E Speer
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - Marcos N Barcellona
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - Michael Y Lu
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - Zizhen Zha
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - Liufang Jing
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - Munish C Gupta
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Jacob M Buchowski
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Michael P Kelly
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Lori A Setton
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA.,Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
17
|
Sharma P, Pal VK, Roy S. An overview of latest advances in exploring bioactive peptide hydrogels for neural tissue engineering. Biomater Sci 2021; 9:3911-3938. [PMID: 33973582 DOI: 10.1039/d0bm02049d] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Neural tissue engineering holds great potential in addressing current challenges faced by medical therapies employed for the functional recovery of the brain. In this context, self-assembling peptides have gained considerable interest owing to their diverse physicochemical properties, which enable them to closely mimic the biophysical characteristics of the native ECM. Additionally, in contrast to synthetic polymers, which lack inherent biological signaling, peptide-based nanomaterials could be easily designed to present essential biological cues to the cells to promote cellular adhesion. Moreover, injectability of these biomaterials further widens their scope in biomedicine. In this context, hydrogels obtained from short bioactive peptide sequences are of particular interest owing to their facile synthesis and highly tunable properties. In spite of their well-known advantages, the exploration of short peptides for neural tissue engineering is still in its infancy and thus detailed discussion is required to evoke interest in this direction. This review provides a general overview of various bioactive hydrogels derived from short peptide sequences explored for neural tissue engineering. The review also discusses the current challenges in translating the benefits of these hydrogels to clinical practices and presents future perspectives regarding the utilization of these hydrogels for advanced biomedical applications.
Collapse
Affiliation(s)
- Pooja Sharma
- Institute of Nano Science and Technology, Sector 81, Knowledge city, Mohali, 140306, Punjab, India.
| | - Vijay Kumar Pal
- Institute of Nano Science and Technology, Sector 81, Knowledge city, Mohali, 140306, Punjab, India.
| | - Sangita Roy
- Institute of Nano Science and Technology, Sector 81, Knowledge city, Mohali, 140306, Punjab, India.
| |
Collapse
|
18
|
Effect of Biomedical Materials in the Implementation of a Long and Healthy Life Policy. Processes (Basel) 2021. [DOI: 10.3390/pr9050865] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
This paper is divided into seven main parts. Its purpose is to review the literature to demonstrate the importance of developing bioengineering and global production of biomaterials to care for the level of healthcare in the world. First, the general description of health as a universal human value and assumptions of a long and healthy life policy is presented. The ethical aspects of the mission of medical doctors and dentists were emphasized. The coronavirus, COVID-19, pandemic has had a significant impact on health issues, determining the world’s health situation. The scope of the diseases is given, and specific methods of their prevention are discussed. The next part focuses on bioengineering issues, mainly medical engineering and dental engineering, and the need for doctors to use technical solutions supporting medicine and dentistry, taking into account the current stage Industry 4.0 of the industrial revolution. The concept of Dentistry 4.0 was generally presented, and a general Bioengineering 4.0 approach was suggested. The basics of production management and the quality loop of the product life cycle were analyzed. The general classification of medical devices and biomedical materials necessary for their production was presented. The paper contains an analysis of the synthesis and characterization of biomedical materials supporting medicine and dentistry, emphasizing additive manufacturing methods. Numerous examples of clinical applications supported considerations regarding biomedical materials. The economic conditions for implementing various biomedical materials groups were supported by forecasts for developing global markets for biomaterials, regenerative medicine, and tissue engineering. In the seventh part, recapitulation and final remarks against the background of historical retrospection, it was emphasized that the technological processes of production and processing of biomedical materials and the systematic increase in their global production are a determinant of the implementation of a long and healthy policy.
Collapse
|
19
|
Maru V, Dixit U, Patil RSB, Parekh R. Cytotoxicity and Bioactivity of Mineral Trioxide Aggregate and Bioactive Endodontic Type Cements: A Systematic Review. Int J Clin Pediatr Dent 2021; 14:30-39. [PMID: 34326580 PMCID: PMC8311779 DOI: 10.5005/jp-journals-10005-1880] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Background Knowledge of the cytotoxicity and bioactivity of endodontic materials may assist in understanding their ability to promote dental pulp stem cell activity and pulp healing in primary teeth. Materials and methods This systematic review was carried out by searching the electronic databases such as PubMed, Google Scholar, and Cochrane reviews for the articles published between January 2000 and December 2018 using the appropriate MeSH keywords. An independent investigator evaluated the abstracts and titles for possible inclusion, as per the stipulated inclusion and exclusion criteria. The topics considered for extracting data from each study were: cell lineage, cytotoxicity assay used, and type of material tested. Results Seven eligible studies were selected for assessing the quality of evidence on the bioactivity of bioactive endodontic cements (BECs) (1 human cell line, 2 animal cell lines, and 4 in vitro, animal, and human studies) and 13 studies were selected for reviewing the quality of evidence on cytotoxicity (7 human cell lines, 4 animal cell lines, and 2 animal model studies). Very limited studies had been conducted on the bioactivity of materials other than mineral trioxide aggregate (MTA). With regards to cytotoxicity, the studies were diverse and most of the studies were based on MTT assay. Mineral trioxide aggregate is the most frequently used as well as studied root-end filling cement, and the literature evidence corroborated its reduced cytotoxicity and enhanced bioavailability. Conclusion There was a lack of sufficient evidence to arrive at a consensus on the ideal material with minimal cytotoxicity and optimal bioactivity. More focused human/cell line-based studies are needed on the available root filling materials. Clinical significance The present systematic review provides an update on the available literature evidence on the cytotoxicity and bioactivity of various BECs including MTAs and their influence on the different cells with respect to their composition and strength. How to cite this article Maru V, Dixit U, Patil RSB, et al. Cytotoxicity and Bioactivity of Mineral Trioxide Aggregate and Bioactive Endodontic Type Cements: A Systematic Review. Int J Clin Pediatr Dent 2021;14(1):30–39.
Collapse
Affiliation(s)
- Viral Maru
- Department of Pediatric and Preventive Dentistry, DY Patil School of Dentistry, Nerul, Navi Mumbai, Maharashtra, India
| | - Uma Dixit
- Department of Pediatric and Preventive Dentistry, DY Patil School of Dentistry, Nerul, Navi Mumbai, Maharashtra, India
| | - Rucha Shivajirao Bhise Patil
- Department of Pediatric and Preventive Dentistry, DY Patil School of Dentistry, Nerul, Navi Mumbai, Maharashtra, India
| | - Rupanshi Parekh
- Department of Pediatric and Preventive Dentistry, DY Patil School of Dentistry, Nerul, Navi Mumbai, Maharashtra, India
| |
Collapse
|
20
|
Buie T, McCune J, Cosgriff-Hernandez E. Gelatin Matrices for Growth Factor Sequestration. Trends Biotechnol 2020; 38:546-557. [PMID: 31954527 PMCID: PMC11577444 DOI: 10.1016/j.tibtech.2019.12.005] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 11/15/2019] [Accepted: 12/06/2019] [Indexed: 01/07/2023]
Abstract
Gelatin is used in a broad range of tissue engineering applications because of its bioactivity, mild processing conditions, and ease of modification, which have increased interest in its use as a growth factor delivery vehicle. Traditional methods to control growth factor sequestration and delivery have relied on controlling hydrogel mesh size via chemical crosslinking with corollary changes to the physical properties of the hydrogel. To decouple growth factor release from scaffold properties, affinity sequestration modalities have been developed to preserve the bioactivity of the growth factor through interactions with the modified gelatin. This review provides a summary of these mechanisms, highlights current gelatin growth factor delivery systems, and addresses the future perspective of gelatin matrices for growth factor delivery in tissue engineering.
Collapse
Affiliation(s)
- Taneidra Buie
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX 78712, USA
| | - Joshua McCune
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX 78712, USA
| | | |
Collapse
|
21
|
Sun W, Liu W, Wu Z, Chen H. Chemical Surface Modification of Polymeric Biomaterials for Biomedical Applications. Macromol Rapid Commun 2020; 41:e1900430. [DOI: 10.1002/marc.201900430] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 02/08/2020] [Accepted: 02/16/2020] [Indexed: 12/12/2022]
Affiliation(s)
- Wei Sun
- College of ChemistryChemical Engineering and Materials ScienceCollaborative Innovation Center for New Type Urbanization and Social Governance of Jiangsu ProvinceSoochow University Suzhou 215123 P. R. China
| | - Wenying Liu
- College of ChemistryChemical Engineering and Materials ScienceCollaborative Innovation Center for New Type Urbanization and Social Governance of Jiangsu ProvinceSoochow University Suzhou 215123 P. R. China
| | - Zhaoqiang Wu
- College of ChemistryChemical Engineering and Materials ScienceCollaborative Innovation Center for New Type Urbanization and Social Governance of Jiangsu ProvinceSoochow University Suzhou 215123 P. R. China
| | - Hong Chen
- College of ChemistryChemical Engineering and Materials ScienceCollaborative Innovation Center for New Type Urbanization and Social Governance of Jiangsu ProvinceSoochow University Suzhou 215123 P. R. China
| |
Collapse
|
22
|
Bar A, Cohen S. Inducing Endogenous Cardiac Regeneration: Can Biomaterials Connect the Dots? Front Bioeng Biotechnol 2020; 8:126. [PMID: 32175315 PMCID: PMC7056668 DOI: 10.3389/fbioe.2020.00126] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Accepted: 02/10/2020] [Indexed: 12/19/2022] Open
Abstract
Heart failure (HF) after myocardial infarction (MI) due to blockage of coronary arteries is a major public health issue. MI results in massive loss of cardiac muscle due to ischemia. Unfortunately, the adult mammalian myocardium presents a low regenerative potential, leading to two main responses to injury: fibrotic scar formation and hypertrophic remodeling. To date, complete heart transplantation remains the only clinical option to restore heart function. In the last two decades, tissue engineering has emerged as a promising approach to promote cardiac regeneration. Tissue engineering aims to target processes associated with MI, including cardiomyogenesis, modulation of extracellular matrix (ECM) remodeling, and fibrosis. Tissue engineering dogmas suggest the utilization and combination of two key components: bioactive molecules and biomaterials. This chapter will present current therapeutic applications of biomaterials in cardiac regeneration and the challenges still faced ahead. The following biomaterial-based approaches will be discussed: Nano-carriers for cardiac regeneration-inducing biomolecules; corresponding matrices for their controlled release; injectable hydrogels for cell delivery and cardiac patches. The concept of combining cardiac patches with controlled release matrices will be introduced, presenting a promising strategy to promote endogenous cardiac regeneration.
Collapse
Affiliation(s)
- Assaf Bar
- The Avram and Stella Goldstein-Goren Department of Biotechnology Engineering, Faculty of Engineering Sciences, Ben-Gurion University of the Negev, Beersheba, Israel
| | - Smadar Cohen
- The Avram and Stella Goldstein-Goren Department of Biotechnology Engineering, Faculty of Engineering Sciences, Ben-Gurion University of the Negev, Beersheba, Israel
- Regenerative Medicine and Stem Cell Research Center, Ben-Gurion University of the Negev, Beersheba, Israel
- Ilse Katz Institute for Nanoscale Science and Technology, Ben-Gurion University of the Negev, Beersheba, Israel
| |
Collapse
|
23
|
Ippel BD, Arts B, Keizer HM, Dankers PYW. Combinatorial functionalization with bisurea-peptides and antifouling bisurea additives of a supramolecular elastomeric biomaterial. JOURNAL OF POLYMER SCIENCE. PART B, POLYMER PHYSICS 2019; 57:1725-1735. [PMID: 32025088 PMCID: PMC6988465 DOI: 10.1002/polb.24907] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 11/08/2019] [Accepted: 11/08/2019] [Indexed: 12/13/2022]
Abstract
The bioactive additive toolbox to functionalize supramolecular elastomeric materials expands rapidly. Here we have set an explorative step toward screening of complex combinatorial functionalization with antifouling and three peptide-containing additives in a bisurea-based supramolecular system. Thorough investigation of surface properties of thin films with contact angle measurements, X-ray photoelectron spectroscopy and atomic force microscopy, was correlated to cell-adhesion of endothelial and smooth muscle cells to apprehend their respective predictive values for functional biomaterial development. Peptides were presented at the surface alone, and in combinatorial functionalization with the oligo(ethylene glycol)-based non-cell adhesive additive. The bisurea-RGD additive was cell-adhesive in all conditions, whereas the endothelial cell-specific bisurea-REDV showed limited bioactive properties in all chemical nano-environments. Also, aspecific functionality was observed for a bisurea-SDF1α peptide. These results emphasize that special care should be taken in changing the chemical nano-environment with peptide functionalization. © 2019 The Authors. Journal of Polymer Science Part B: Polymer Physics published by Wiley Periodicals, Inc. J. Polym. Sci., Part B: Polym. Phys. 2019, 57, 1725-1735.
Collapse
Affiliation(s)
- Bastiaan D. Ippel
- Institute for Complex Molecular SystemsEindhoven University of TechnologyPO Box 513 5600EindhovenManitobaThe Netherlands
- Department of Biomedical Engineering, Laboratory for Cell and Tissue EngineeringEindhoven University of TechnologyPO Box 513 5600EindhovenManitobaThe Netherlands
| | - Boris Arts
- Institute for Complex Molecular SystemsEindhoven University of TechnologyPO Box 513 5600EindhovenManitobaThe Netherlands
- Department of Biomedical Engineering, Laboratory of Chemical BiologyEindhoven University of TechnologyPO Box 513, 5600EindhovenManitobaThe Netherlands
| | - Henk M. Keizer
- SyMO‐Chem B.VDen Dolech 2, 5612EindhovenArizonaThe Netherlands
| | - Patricia Y. W. Dankers
- Institute for Complex Molecular SystemsEindhoven University of TechnologyPO Box 513 5600EindhovenManitobaThe Netherlands
- Department of Biomedical Engineering, Laboratory for Cell and Tissue EngineeringEindhoven University of TechnologyPO Box 513 5600EindhovenManitobaThe Netherlands
- Department of Biomedical Engineering, Laboratory of Chemical BiologyEindhoven University of TechnologyPO Box 513, 5600EindhovenManitobaThe Netherlands
| |
Collapse
|
24
|
Clegg JR, Wagner AM, Shin SR, Hassan S, Khademhosseini A, Peppas NA. Modular Fabrication of Intelligent Material-Tissue Interfaces for Bioinspired and Biomimetic Devices. PROGRESS IN MATERIALS SCIENCE 2019; 106:100589. [PMID: 32189815 PMCID: PMC7079701 DOI: 10.1016/j.pmatsci.2019.100589] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
One of the goals of biomaterials science is to reverse engineer aspects of human and nonhuman physiology. Similar to the body's regulatory mechanisms, such devices must transduce changes in the physiological environment or the presence of an external stimulus into a detectable or therapeutic response. This review is a comprehensive evaluation and critical analysis of the design and fabrication of environmentally responsive cell-material constructs for bioinspired machinery and biomimetic devices. In a bottom-up analysis, we begin by reviewing fundamental principles that explain materials' responses to chemical gradients, biomarkers, electromagnetic fields, light, and temperature. Strategies for fabricating highly ordered assemblies of material components at the nano to macro-scales via directed assembly, lithography, 3D printing and 4D printing are also presented. We conclude with an account of contemporary material-tissue interfaces within bioinspired and biomimetic devices for peptide delivery, cancer theranostics, biomonitoring, neuroprosthetics, soft robotics, and biological machines.
Collapse
Affiliation(s)
- John R Clegg
- Department of Biomedical Engineering, the University of Texas at Austin, Austin, Texas, USA
| | - Angela M Wagner
- McKetta Department of Chemical Engineering, the University of Texas at Austin, Austin, Texas, USA
| | - Su Ryon Shin
- Division of Engineering in Medicine, Department of Medicine, Brigham Women's Hospital, Harvard Medical School, Cambridge, Massachusetts, USA
| | - Shabir Hassan
- Division of Engineering in Medicine, Department of Medicine, Brigham Women's Hospital, Harvard Medical School, Cambridge, Massachusetts, USA
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Ali Khademhosseini
- Center for Minimally Invasive Therapeutics (C-MIT), University of California - Los Angeles, Los Angeles, California, USA
- California NanoSystems Institute (CNSI), University of California - Los Angeles, Los Angeles, California, USA
- Department of Bioengineering, University of California - Los Angeles, Los Angeles, California, USA
- Department of Bioindustrial Technologies, College of Animal Bioscience and Technology, Konkuk University, Seoul, Republic of Korea
| | - Nicholas A Peppas
- Department of Biomedical Engineering, the University of Texas at Austin, Austin, Texas, USA
- McKetta Department of Chemical Engineering, the University of Texas at Austin, Austin, Texas, USA
- Institute for Biomaterials, Drug Delivery, and Regenerative Medicine, the University of Texas at Austin, Austin, Texas, USA
- Department of Surgery and Perioperative Care, Dell Medical School, the University of Texas at Austin, Austin, Texas, USA
- Department of Pediatrics, Dell Medical School, the University of Texas at Austin, Austin, Texas, USA
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, the University of Texas at Austin, Austin, Texas, USA
| |
Collapse
|
25
|
Padiolleau L, Chanseau C, Durrieu S, Ayela C, Laroche G, Durrieu M. Directing hMSCs fate through geometrical cues and mimetics peptides. J Biomed Mater Res A 2019; 108:201-211. [DOI: 10.1002/jbm.a.36804] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 06/11/2019] [Accepted: 09/16/2019] [Indexed: 12/23/2022]
Affiliation(s)
- Laurence Padiolleau
- Chimie et Biologie des Membranes et Nano‐Objets (UMR5248 CBMN) University Bordeaux Pessac France
- CNRS, CBMN UMR5248 Pessac France
- Bordeaux INP, CBMN UMR5248 Pessac France
- Laboratoire d'Ingénierie de Surface (LIS), Département de Génie des Mines, de la Métallurgie et des Matériaux Centre de Recherche sur les Matériaux Avancés (CERMA), Université Laval Québec Canada
- Centre de Recherche du Centre Hospitalier Universitaire de Québec (CRCHUQ), Hôpital St‐François d'Assise Québec Canada
| | - Christel Chanseau
- Chimie et Biologie des Membranes et Nano‐Objets (UMR5248 CBMN) University Bordeaux Pessac France
- CNRS, CBMN UMR5248 Pessac France
- Bordeaux INP, CBMN UMR5248 Pessac France
| | - Stéphanie Durrieu
- ARNA Laboratory Université de Bordeaux Bordeaux France
- ARNA Laboratory INSERM, U1212 – CNRS UMR 5320 Bordeaux France
| | - Cédric Ayela
- Université de Bordeaux, IMS, UMR CNRS 5218 Talence France
| | - Gaétan Laroche
- Laboratoire d'Ingénierie de Surface (LIS), Département de Génie des Mines, de la Métallurgie et des Matériaux Centre de Recherche sur les Matériaux Avancés (CERMA), Université Laval Québec Canada
- Centre de Recherche du Centre Hospitalier Universitaire de Québec (CRCHUQ), Hôpital St‐François d'Assise Québec Canada
| | - Marie‐Christine Durrieu
- Chimie et Biologie des Membranes et Nano‐Objets (UMR5248 CBMN) University Bordeaux Pessac France
- CNRS, CBMN UMR5248 Pessac France
- Bordeaux INP, CBMN UMR5248 Pessac France
| |
Collapse
|
26
|
Shadish JA, Strange AC, DeForest CA. Genetically Encoded Photocleavable Linkers for Patterned Protein Release from Biomaterials. J Am Chem Soc 2019; 141:15619-15625. [PMID: 31525979 DOI: 10.1021/jacs.9b07239] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Given the critical role that proteins play in almost all biological processes, there is great interest in controlling their presentation within and release from biomaterials. Despite such outstanding enthusiasm, previously developed strategies in this regard result in ill-defined and heterogeneous populations with substantially decreased activity, precluding their successful application to fragile species including growth factors. Here, we introduce a modular and scalable method for creating monodisperse, genetically encoded chimeras that enable bioactive proteins to be immobilized within and subsequently photoreleased from polymeric hydrogels. Building upon recent developments in chemoenzymatic reactions, bioorthogonal chemistry, and optogenetics, we tether fluorescent proteins, model enzymes, and growth factors site-specifically to gel biomaterials through a photocleavable protein (PhoCl) that undergoes irreversible backbone photoscission upon exposure to cytocompatible visible light (λ ≈ 400 nm) in a dose-dependent manner. Mask-based and laser-scanning lithographic strategies using commonly available light sources are employed to spatiotemporally pattern protein release from hydrogels while retaining their full activity. The photopatterned epidermal growth factor presentation is exploited to promote anisotropic cellular proliferation in 3D. We expect these methods to be broadly useful for applications in diagnostics, drug delivery, and regenerative medicine.
Collapse
Affiliation(s)
| | | | - Cole A DeForest
- Department of Bioengineering , University of Washington , Seattle , Washington 98105 , United States.,Institute for Stem Cell & Regenerative Medicine , University of Washington , Seattle , Washington 98109 , United States
| |
Collapse
|
27
|
Cereceres S, Lan Z, Bryan L, Whitely M, Wilems T, Fabela N, Whitfield-Cargile C, Cosgriff-Hernandez E. In Vivo Characterization of Poly(ethylene glycol) Hydrogels with Thio-β Esters. Ann Biomed Eng 2019; 48:953-967. [PMID: 31139974 DOI: 10.1007/s10439-019-02271-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 04/11/2019] [Indexed: 01/14/2023]
Abstract
Resorbable hydrogels have numerous potential applications in tissue engineering and drug delivery due to their highly tunable properties and soft tissue-like mechanical properties. The incorporation of esters into the backbone of poly(ethylene glycol) hydrogels has been used to develop libraries of hydrogels with tunable degradation rates. However, these synthetic strategies used to increase degradation rate often result in undesired changes in the hydrogel physical properties such as matrix modulus or swelling. In an effort to decouple degradation rate from other hydrogel properties, we inserted thio-β esters into the poly(ethylene glycol)-diacrylate backbone to introduce labile bonds without changing macromer molecular weight. This allowed the number of hydrolytically labile thio-β esters to be controlled through changing the ratios of this modified macromer to the original macromer without affecting network properties. The retention of hydrogel properties at different macromer ratios was confirmed by measuring gel fraction, swelling ratio, and compressive modulus. The tunable degradation profiles were characterized both in vitro and in vivo. Following confirmation of cytocompatibility after exposure to the hydrogel degradation products, the in vivo host response was evaluated in comparison to medical grade silicone. Collectively, this work demonstrates the utility and tunability of these hydrolytically degradable hydrogels for a wide variety of tissue engineering applications.
Collapse
Affiliation(s)
- Stacy Cereceres
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, 77843, USA
| | - Ziyang Lan
- Department of Biomedical Engineering, The University of Texas at Austin, 107 W. Dean Keeton, BME 3.503D, 1 University Station, C0800, Austin, TX, 78712, USA
| | - Laura Bryan
- Department of Veterinary Pathobiology, Texas A&M University, College Station, TX, 77843, USA
| | - Michael Whitely
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, 77843, USA
| | - Thomas Wilems
- Department of Biomedical Engineering, The University of Texas at Austin, 107 W. Dean Keeton, BME 3.503D, 1 University Station, C0800, Austin, TX, 78712, USA
| | - Natalia Fabela
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, 77843, USA
| | - Canaan Whitfield-Cargile
- Department of Large Animal Clinical Sciences, Texas A&M University, College Station, TX, 77843, USA
| | - Elizabeth Cosgriff-Hernandez
- Department of Biomedical Engineering, The University of Texas at Austin, 107 W. Dean Keeton, BME 3.503D, 1 University Station, C0800, Austin, TX, 78712, USA.
| |
Collapse
|
28
|
Wu Y, Yu C, Xing M, Wang L, Guan G. Surface modification of polyvinyl alcohol (PVA)/polyacrylamide (PAAm) hydrogels with polydopamine and REDV for improved applicability. J Biomed Mater Res B Appl Biomater 2019; 108:117-127. [PMID: 30912304 DOI: 10.1002/jbm.b.34371] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2018] [Revised: 03/01/2019] [Accepted: 03/05/2019] [Indexed: 02/03/2023]
Abstract
Developing a small-diameter vascular graft with a satisfactory performance in terms of mechanical and biological properties remains a challenging issue because of comprehensive requirements from clinical applications. Polyvinyl alcohol (PVA)/polyacrylamide (PAAm) hydrogels exhibit many desirable characteristics for small-diameter vascular grafts because of their tunable mechanical properties, especially high compliance. However, poor cells adhesion hinders their application for endothelialization in situ. Therefore, in the present work, polydopamine (PDA) and tetrapeptide Arg-Glu-Asp-Val (REDV) were used to functionalize the hydrogels surface and improve cells adhesion. A series of characterizations were systematically conducted to examine the applicability of coated hydrogels to small-diameter vascular grafts. Results showed that bare and coated hydrogels have appropriate structural stability, and no significant differences in tensile properties could be found after being coated with PDA or PDA-REDV. The hydrophilicity of the hydrogels decreased with the coatings of PDA and especially PDA-REDV to improve protein adsorption, porcine iliac artery endothelial cells (PIECs) adhesion, viability, proliferation, and spreading on the hydrogels. Lower hemolysis percentages and higher blood clotting index values were attained for the hydrogels, suggesting their satisfactory hemocompatibility. Overall, the present work provided insights into the development of a novel hydrogel-based small-diameter vascular graft. © 2019 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater 108B:117-127, 2020.
Collapse
Affiliation(s)
- Yufen Wu
- Key Laboratory of Textile Science & Technology, Ministry of Education, College of Textiles, Donghua University, Shanghai, 201620, China
| | - Chenglong Yu
- Key Laboratory of Textile Science & Technology, Ministry of Education, College of Textiles, Donghua University, Shanghai, 201620, China
| | - Meiyi Xing
- Key Laboratory of Textile Science & Technology, Ministry of Education, College of Textiles, Donghua University, Shanghai, 201620, China
| | - Lu Wang
- Key Laboratory of Textile Science & Technology, Ministry of Education, College of Textiles, Donghua University, Shanghai, 201620, China
| | - Guoping Guan
- Key Laboratory of Textile Science & Technology, Ministry of Education, College of Textiles, Donghua University, Shanghai, 201620, China
| |
Collapse
|
29
|
Onwuka E, King N, Heuer E, Breuer C. The Heart and Great Vessels. Cold Spring Harb Perspect Med 2018; 8:cshperspect.a031922. [PMID: 28289246 DOI: 10.1101/cshperspect.a031922] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Cardiovascular disease is the leading cause of mortality worldwide. We have made large strides over the past few decades in management, but definitive therapeutic options to address this health-care burden are still limited. Given the ever-increasing need, much effort has been spent creating engineered tissue to replaced diseased tissue. This article gives a general overview of this work as it pertains to the development of great vessels, myocardium, and heart valves. In each area, we focus on currently studied methods, limitations, and areas for future study.
Collapse
Affiliation(s)
- Ekene Onwuka
- Tissue Engineering and Surgical Research, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio 43205.,College of Medicine, The Ohio State University, Columbus, Ohio 43210
| | - Nakesha King
- Tissue Engineering and Surgical Research, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio 43205.,College of Medicine, The Ohio State University, Columbus, Ohio 43210
| | - Eric Heuer
- Tissue Engineering and Surgical Research, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio 43205
| | - Christopher Breuer
- Tissue Engineering and Surgical Research, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio 43205.,College of Medicine, The Ohio State University, Columbus, Ohio 43210.,Department of Pediatric Surgery, Nationwide Children's Hospital, Columbus, Ohio 43205
| |
Collapse
|
30
|
Sever M, Tansik G, Arslan E, Yergoz F, Ozkan AD, Tekinay AB, Guler MO. Self-assembled peptide nanostructures and their gels for regenerative medicine applications. SELF-ASSEMBLING BIOMATERIALS 2018:455-473. [DOI: 10.1016/b978-0-08-102015-9.00022-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
31
|
Kim MS, Lee MH, Kwon BJ, Koo MA, Seon GM, Kim D, Hong SH, Park JC. Influence of Biomimetic Materials on Cell Migration. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1064:93-107. [DOI: 10.1007/978-981-13-0445-3_6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
32
|
Ahadian S, Civitarese R, Bannerman D, Mohammadi MH, Lu R, Wang E, Davenport-Huyer L, Lai B, Zhang B, Zhao Y, Mandla S, Korolj A, Radisic M. Organ-On-A-Chip Platforms: A Convergence of Advanced Materials, Cells, and Microscale Technologies. Adv Healthc Mater 2018; 7. [PMID: 29034591 DOI: 10.1002/adhm.201700506] [Citation(s) in RCA: 170] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 06/15/2017] [Indexed: 12/11/2022]
Abstract
Significant advances in biomaterials, stem cell biology, and microscale technologies have enabled the fabrication of biologically relevant tissues and organs. Such tissues and organs, referred to as organ-on-a-chip (OOC) platforms, have emerged as a powerful tool in tissue analysis and disease modeling for biological and pharmacological applications. A variety of biomaterials are used in tissue fabrication providing multiple biological, structural, and mechanical cues in the regulation of cell behavior and tissue morphogenesis. Cells derived from humans enable the fabrication of personalized OOC platforms. Microscale technologies are specifically helpful in providing physiological microenvironments for tissues and organs. In this review, biomaterials, cells, and microscale technologies are described as essential components to construct OOC platforms. The latest developments in OOC platforms (e.g., liver, skeletal muscle, cardiac, cancer, lung, skin, bone, and brain) are then discussed as functional tools in simulating human physiology and metabolism. Future perspectives and major challenges in the development of OOC platforms toward accelerating clinical studies of drug discovery are finally highlighted.
Collapse
Affiliation(s)
- Samad Ahadian
- Institute of Biomaterials and Biomedical Engineering; University of Toronto; Toronto M5S 3G9 Ontario Canada
| | - Robert Civitarese
- Institute of Biomaterials and Biomedical Engineering; University of Toronto; Toronto M5S 3G9 Ontario Canada
| | - Dawn Bannerman
- Institute of Biomaterials and Biomedical Engineering; University of Toronto; Toronto M5S 3G9 Ontario Canada
- Department of Chemical Engineering and Applied Chemistry; University of Toronto; Toronto M5S 3G9 Ontario Canada
| | - Mohammad Hossein Mohammadi
- Institute of Biomaterials and Biomedical Engineering; University of Toronto; Toronto M5S 3G9 Ontario Canada
- Department of Chemical Engineering and Applied Chemistry; University of Toronto; Toronto M5S 3G9 Ontario Canada
| | - Rick Lu
- Institute of Biomaterials and Biomedical Engineering; University of Toronto; Toronto M5S 3G9 Ontario Canada
| | - Erika Wang
- Institute of Biomaterials and Biomedical Engineering; University of Toronto; Toronto M5S 3G9 Ontario Canada
| | - Locke Davenport-Huyer
- Institute of Biomaterials and Biomedical Engineering; University of Toronto; Toronto M5S 3G9 Ontario Canada
- Department of Chemical Engineering and Applied Chemistry; University of Toronto; Toronto M5S 3G9 Ontario Canada
| | - Ben Lai
- Institute of Biomaterials and Biomedical Engineering; University of Toronto; Toronto M5S 3G9 Ontario Canada
| | - Boyang Zhang
- Institute of Biomaterials and Biomedical Engineering; University of Toronto; Toronto M5S 3G9 Ontario Canada
- Department of Chemical Engineering and Applied Chemistry; University of Toronto; Toronto M5S 3G9 Ontario Canada
| | - Yimu Zhao
- Department of Chemical Engineering and Applied Chemistry; University of Toronto; Toronto M5S 3G9 Ontario Canada
| | - Serena Mandla
- Department of Chemical Engineering and Applied Chemistry; University of Toronto; Toronto M5S 3G9 Ontario Canada
| | - Anastasia Korolj
- Institute of Biomaterials and Biomedical Engineering; University of Toronto; Toronto M5S 3G9 Ontario Canada
- Department of Chemical Engineering and Applied Chemistry; University of Toronto; Toronto M5S 3G9 Ontario Canada
| | - Milica Radisic
- Institute of Biomaterials and Biomedical Engineering; University of Toronto; Toronto M5S 3G9 Ontario Canada
- Department of Chemical Engineering and Applied Chemistry; University of Toronto; Toronto M5S 3G9 Ontario Canada
| |
Collapse
|
33
|
Abstract
This review is focused on the use of membranes for the specific application of bone regeneration. The first section focuses on the relevance of membranes in this context and what are the specifications that they should possess to improve the regeneration of bone. Afterward, several techniques to engineer bone membranes by using "bulk"-like methods are discussed, where different parameters to induce bone formation are disclosed in a way to have desirable structural and functional properties. Subsequently, the production of nanostructured membranes using a bottom-up approach is discussed by highlighting the main advances in the field of bone regeneration. Primordial importance is given to the promotion of osteoconductive and osteoinductive capability during the membrane design. Whenever possible, the films prepared using different techniques are compared in terms of handability, bone guiding ability, osteoinductivity, adequate mechanical properties, or biodegradability. A last chapter contemplates membranes only composed by cells, disclosing their potential to regenerate bone.
Collapse
Affiliation(s)
- Sofia G Caridade
- Department of Chemistry CICECO, Aveiro Institute of Materials, University of Aveiro , Aveiro, Portugal
| | - João F Mano
- Department of Chemistry CICECO, Aveiro Institute of Materials, University of Aveiro , Aveiro, Portugal
| |
Collapse
|
34
|
Bilem I, Chevallier P, Plawinski L, Sone ED, Durrieu MC, Laroche G. Interplay of Geometric Cues and RGD/BMP-2 Crosstalk in Directing Stem Cell Fate. ACS Biomater Sci Eng 2017; 3:2514-2523. [PMID: 33465907 DOI: 10.1021/acsbiomaterials.7b00279] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Within the native microenvironment, extracellular matrix (ECM) components are thought to display a complex and heterogeneous distribution, spanning several length scales. Herein, the objective is to mimic, in vitro, the hierarchical organization of proteins and growth factors as well as their crosstalk. Photolithography technique was used to adjacently pattern geometrically defined regions of RGD and BMP-2 mimetic peptides onto glass substrates. These ECM-derived ligands are known to jointly regulate mesenchymal stem cells (MSCs) osteogenic differentiation. By manipulating the spatial distribution of dually grafted peptides, the extent of human MSCs osteogenic differentiation was significantly affected, depending on the shape of peptide micropatterns. Our data highlight the existence of a strong interplay between geometric cues and biochemical signals. Such in vitro systems provide a valuable tool to investigate mechanisms by which multiple ECM cues overlap to regulate stem cell fate, thereby contributing to the design of bioinspired biomaterials for bone tissue engineering applications.
Collapse
Affiliation(s)
- Ibrahim Bilem
- Laboratoire d'Ingénierie de Surface, Centre de Recherche sur les Matériaux Avancés, Département de Génie des Mines, de la Métallurgie et des Matériaux, Université Laval, 1065 Avenue de la médecine, Québec G1V 0A6, Canada.,Centre de Recherche du Centre Hospitalier Universitaire de Québec, Hôpital St-François d'Assise, 10 rue de l'Espinay, Québec G1L 3L5, Canada.,CBMN, UMR 5248, Université de Bordeaux, Pessac F-33600, France.,Institute of Chemistry & Biology of Membranes & Nanoobjects (CBMN 5248), Centre National de la Recherche Scientifique (CNRS), Pessac F-33600, France.,CBMN, UMR 5248, Bordeaux INP, F-33600, Pessac, France
| | - Pascale Chevallier
- Laboratoire d'Ingénierie de Surface, Centre de Recherche sur les Matériaux Avancés, Département de Génie des Mines, de la Métallurgie et des Matériaux, Université Laval, 1065 Avenue de la médecine, Québec G1V 0A6, Canada.,Centre de Recherche du Centre Hospitalier Universitaire de Québec, Hôpital St-François d'Assise, 10 rue de l'Espinay, Québec G1L 3L5, Canada
| | - Laurent Plawinski
- CBMN, UMR 5248, Université de Bordeaux, Pessac F-33600, France.,Institute of Chemistry & Biology of Membranes & Nanoobjects (CBMN 5248), Centre National de la Recherche Scientifique (CNRS), Pessac F-33600, France.,CBMN, UMR 5248, Bordeaux INP, F-33600, Pessac, France
| | - Eli D Sone
- Institute of Biomaterials and Biomedical Engineering, Department of Materials Science and Engineering, and Faculty of Dentistry, University of Toronto, Toronto, Ontario M5S 3G9, Canada
| | - Marie-Christine Durrieu
- CBMN, UMR 5248, Université de Bordeaux, Pessac F-33600, France.,Institute of Chemistry & Biology of Membranes & Nanoobjects (CBMN 5248), Centre National de la Recherche Scientifique (CNRS), Pessac F-33600, France.,CBMN, UMR 5248, Bordeaux INP, F-33600, Pessac, France
| | - Gaétan Laroche
- Laboratoire d'Ingénierie de Surface, Centre de Recherche sur les Matériaux Avancés, Département de Génie des Mines, de la Métallurgie et des Matériaux, Université Laval, 1065 Avenue de la médecine, Québec G1V 0A6, Canada.,Centre de Recherche du Centre Hospitalier Universitaire de Québec, Hôpital St-François d'Assise, 10 rue de l'Espinay, Québec G1L 3L5, Canada
| |
Collapse
|
35
|
Biocomposite nanofiber matrices to support ECM remodeling by human dermal progenitors and enhanced wound closure. Sci Rep 2017; 7:10291. [PMID: 28860484 PMCID: PMC5579010 DOI: 10.1038/s41598-017-10735-x] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 08/14/2017] [Indexed: 01/14/2023] Open
Abstract
Cell-based therapies have recently been the focus of much research to enhance skin wound healing. An important challenge will be to develop vehicles for cell delivery that promote survival and uniform distribution of cells across the wound bed. These systems should be stiff enough to facilitate handling, whilst soft enough to limit damage to newly synthesized wound tissue and minimize patient discomfort. Herein, we developed several novel modifiable nanofibre scaffolds comprised of Poly (ε-caprolactone) (PCL) and gelatin (GE). We asked whether they could be used as a functional receptacle for adult human Skin-derived Precursor Cells (hSKPs) and how naked scaffolds impact endogenous skin wound healing. PCL and GE were electrospun in a single facile solvent to create composite scaffolds and displayed unique morphological and mechanical properties. After seeding with adult hSKPs, deposition of extracellular matrix proteins and sulphated glycosaminoglycans was found to be enhanced in composite grafts. Moreover, composite scaffolds exhibited significantly higher cell proliferation, greater cell spreading and integration within the nanofiber mats. Transplantation of acellular scaffolds into wounds revealed scaffolds exhibited improvement in dermal-epidermal thickness, axonal density and collagen deposition. These results demonstrate that PCL-based nanofiber scaffolds show promise as a cell delivery system for wound healing.
Collapse
|
36
|
Chekh BOC, Ferens MV, Ostapiv DD, Samaryk VY, Varvarenko SM, Vlizlo VV. Characteristics of novel polymer based on pseudo-polyamino acids GluLa-DPG-PEG600: binding of albumin, biocompatibility, biodistribution and potential crossing the blood-brain barrier in rats. UKRAINIAN BIOCHEMICAL JOURNAL 2017. [DOI: 10.15407/ubj89.04.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
37
|
Mansour A, Mezour MA, Badran Z, Tamimi F. * Extracellular Matrices for Bone Regeneration: A Literature Review. Tissue Eng Part A 2017; 23:1436-1451. [PMID: 28562183 DOI: 10.1089/ten.tea.2017.0026] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The gold standard material for bone regeneration is still autologous bone, a mesenchymal tissue that consists mainly of extracellular matrix (ECM) (90% v/v) and little cellular content (10% v/v). However, the fact that decellularized allogenic bone grafts often present a clinical performance comparable to autologous bone grafts demonstrates the crucial role of ECM in bone regeneration. For long, the mechanism by which bone allografts function was not clear, but recent research has unveiled many unique characteristics of ECM that seem to play a key role in tissue regeneration. This is further confirmed by the fact that synthetic biomaterials with composition and properties resembling bone ECM present excellent bone regeneration properties. In this context, ECM molecules such as glycosaminoglycans (GAGs) and self-assembly peptides (SAPs) can improve the performance of bone regeneration biomaterials. Moreover, decellularized ECM derived either from native tissues such as bone, cartilage, skin, and tooth germs or from cells such as osteoblasts, chondrocytes, and stem cells has shown promising results in bone regeneration applications. Understanding the role of ECM in bone regeneration is crucial for the development of the next generation of biomaterials for bone tissue engineering. In this sense, this review addresses the state-of-the-art on this subject matter.
Collapse
Affiliation(s)
- Alaa Mansour
- 1 Faculty of Dentistry, McGill University , Montreal, Canada
| | | | - Zahi Badran
- 1 Faculty of Dentistry, McGill University , Montreal, Canada .,2 Department of Periodontology (CHU/UIC 11, INSERM UMR 1229-RMeS), Faculty of Dental Surgery, University of Nantes , Nantes, France
| | - Faleh Tamimi
- 1 Faculty of Dentistry, McGill University , Montreal, Canada
| |
Collapse
|
38
|
Abstract
This review highlights the synthesis, properties, and advanced applications of synthetic and natural polymers 3D printed using stereolithography for soft tissue engineering applications. Soft tissue scaffolds are of great interest due to the number of musculoskeletal, cardiovascular, and connective tissue injuries and replacements humans face each year. Accurately replacing or repairing these tissues is challenging due to the variation in size, shape, and strength of different types of soft tissue. With advancing processing techniques such as stereolithography, control of scaffold resolution down to the μm scale is achievable along with the ability to customize each fabricated scaffold to match the targeted replacement tissue. Matching the advanced manufacturing technique to polymer properties as well as maintaining the proper chemical, biological, and mechanical properties for tissue replacement is extremely challenging. This review discusses the design of polymers with tailored structure, architecture, and functionality for stereolithography, while maintaining chemical, biological, and mechanical properties to mimic a broad range of soft tissue types.
Collapse
|
39
|
Seki A, Ishizone T, Oyane A, Yokoyama H. A Segregation and Deprotection Approach for Hydrophilic Surfaces Using Amphiphilic Block Copolymers Possessing Polystyrene and Poly[(tri(ethylene glycol) methacrylate)] Segments. MACROMOL CHEM PHYS 2017. [DOI: 10.1002/macp.201700048] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Akiko Seki
- Department of Chemical Science and Engineering; Tokyo Institute of Technology; 2-12-1-S1-13 Ohokayama Meguro-ku Tokyo 152-8552 Japan
| | - Takashi Ishizone
- Department of Chemical Science and Engineering; Tokyo Institute of Technology; 2-12-1-S1-13 Ohokayama Meguro-ku Tokyo 152-8552 Japan
| | - Ayako Oyane
- Nanomaterials Research Institute; National Institute of Advanced Industrial Science and Technology; Central 5, 1-1-1 Higashi Tsukuba Ibaraki 305-8565 Japan
| | - Hideaki Yokoyama
- Department of Advanced Materials Science; Graduate School of Frontier Sciences; The University of Tokyo; 603 Transdisciplinary Sciences Bldg, 5-1-5 Kashiwa-no-ha Kashiwa Chiba 277-8561 Japan
| |
Collapse
|
40
|
Huang W, Ebrahimi D, Dinjaski N, Tarakanova A, Buehler MJ, Wong JY, Kaplan DL. Synergistic Integration of Experimental and Simulation Approaches for the de Novo Design of Silk-Based Materials. Acc Chem Res 2017; 50:866-876. [PMID: 28191922 DOI: 10.1021/acs.accounts.6b00616] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Tailored biomaterials with tunable functional properties are crucial for a variety of task-specific applications ranging from healthcare to sustainable, novel bio-nanodevices. To generate polymeric materials with predictive functional outcomes, exploiting designs from nature while morphing them toward non-natural systems offers an important strategy. Silks are Nature's building blocks and are produced by arthropods for a variety of uses that are essential for their survival. Due to the genetic control of encoded protein sequence, mechanical properties, biocompatibility, and biodegradability, silk proteins have been selected as prototype models to emulate for the tunable designs of biomaterial systems. The bottom up strategy of material design opens important opportunities to create predictive functional outcomes, following the exquisite polymeric templates inspired by silks. Recombinant DNA technology provides a systematic approach to recapitulate, vary, and evaluate the core structure peptide motifs in silks and then biosynthesize silk-based polymers by design. Post-biosynthesis processing allows for another dimension of material design by controlled or assisted assembly. Multiscale modeling, from the theoretical prospective, provides strategies to explore interactions at different length scales, leading to selective material properties. Synergy among experimental and modeling approaches can provide new and more rapid insights into the most appropriate structure-function relationships to pursue while also furthering our understanding in terms of the range of silk-based systems that can be generated. This approach utilizes nature as a blueprint for initial polymer designs with useful functions (e.g., silk fibers) but also employs modeling-guided experiments to expand the initial polymer designs into new domains of functional materials that do not exist in nature. The overall path to these new functional outcomes is greatly accelerated via the integration of modeling with experiment. In this Account, we summarize recent advances in understanding and functionalization of silk-based protein systems, with a focus on the integration of simulation and experiment for biopolymer design. Spider silk was selected as an exemplary protein to address the fundamental challenges in polymer designs, including specific insights into the role of molecular weight, hydrophobic/hydrophilic partitioning, and shear stress for silk fiber formation. To expand current silk designs toward biointerfaces and stimuli responsive materials, peptide modules from other natural proteins were added to silk designs to introduce new functions, exploiting the modular nature of silk proteins and fibrous proteins in general. The integrated approaches explored suggest that protein folding, silk volume fraction, and protein amino acid sequence changes (e.g., mutations) are critical factors for functional biomaterial designs. In summary, the integrated modeling-experimental approach described in this Account suggests a more rationally directed and more rapid method for the design of polymeric materials. It is expected that this combined use of experimental and computational approaches has a broad applicability not only for silk-based systems, but also for other polymer and composite materials.
Collapse
Affiliation(s)
- Wenwen Huang
- Department
of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, Massachusetts 02155, United States
| | - Davoud Ebrahimi
- Laboratory
for Atomistic and Molecular Mechanics (LAMM), Department of Civil
and Environmental Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Nina Dinjaski
- Department
of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, Massachusetts 02155, United States
| | - Anna Tarakanova
- Laboratory
for Atomistic and Molecular Mechanics (LAMM), Department of Civil
and Environmental Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Markus J. Buehler
- Laboratory
for Atomistic and Molecular Mechanics (LAMM), Department of Civil
and Environmental Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Joyce Y. Wong
- Department
of Biomedical Engineering, Boston University, 44 Cummington Street, Boston, Massachusetts 02215, United States
| | - David L. Kaplan
- Department
of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, Massachusetts 02155, United States
| |
Collapse
|
41
|
Yang J, Zhang YS, Lei P, Hu X, Wang M, Liu H, Shen X, Li K, Huang Z, Huang J, Ju J, Hu Y, Khademhosseini A. "Steel-Concrete" Inspired Biofunctional Layered Hybrid Cage for Spine Fusion and Segmental Bone Reconstruction. ACS Biomater Sci Eng 2017; 3:637-647. [PMID: 33429631 DOI: 10.1021/acsbiomaterials.6b00666] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
In this paper we report a "steel-concrete" inspired layered hybrid spine cage combining a titanium mesh and a bioceramic scaffold, which were welded together through a bioglass bonding layer using a novel multistep manufacturing methodology including three-dimensional slip deposition, gel casting, freeze-drying, and cosintering. The interfacial welding strength achieved 27 ± 0.7 MPa, indicating an excellent structural integrity of the hybrid cage construct. The biocramic scaffold layer consisting of wollastonite and hydroxyapatite had an interconnected, highly porous structure with a pore size of 100-500 μm and a porosity of >85%, well fufilling the structural requirements of bone regeneration. Simulated body fluid immersion assay showed that the hybrid cage exhibited excellent biodegradability to facilitate rapid bone-like apatite formation. In vitro studies demonstrated that the bioceramic scaffold on the hybrid cage supported attachment, spreading, growth, and migration of bone/vessel-forming cells and triggered osteogenic differentiation of human mesenchymal stem cells. In vivo studies further suggested that the bioceramic scaffold on the hybrid cage could actively promote fast generation of new bone tissues within 12 weeks of implantation in a rabbit femoral condyle model. This study has provided a new design and fabrication methodology of hybrid cages by integrating strong mechanical properties with excellent biological activities including osteoinductivity and bone regeneration ability, for spine fusion and segmental bone reconstruction.
Collapse
Affiliation(s)
- Jingzhou Yang
- School of Mechanical and Chemical Engineering, University of Western Australia, 35 Stirling Highway, Perth, Western Australia 6009, Australia.,Biomaterials Innovation Research Center, Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 65 Landsdowne Street, Cambridge, Massachusetts 02139, United States.,Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Yu Shrike Zhang
- Biomaterials Innovation Research Center, Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 65 Landsdowne Street, Cambridge, Massachusetts 02139, United States.,Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Pengfei Lei
- Biomaterials Innovation Research Center, Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 65 Landsdowne Street, Cambridge, Massachusetts 02139, United States.,Orthopedics Department, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan 410008, People's Republic of China.,Department of Orthopedic Surgery, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, Massachusetts 02115, United States
| | - Xiaozhi Hu
- School of Mechanical and Chemical Engineering, University of Western Australia, 35 Stirling Highway, Perth, Western Australia 6009, Australia
| | - Mian Wang
- Biomaterials Innovation Research Center, Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 65 Landsdowne Street, Cambridge, Massachusetts 02139, United States.,Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States.,School of Chemistry and Chemical Engineering, Guangxi University, 100 University East Road, Nanning, Guangxi 530004, People's Republic of China
| | - Haitao Liu
- School of Materials Sciences and Technology, China University of Geosciences, 29 Xueyuan Road, Beijing 100086, People's Republic of China
| | - Xiulin Shen
- School of Materials Sciences and Technology, China University of Geosciences, 29 Xueyuan Road, Beijing 100086, People's Republic of China
| | - Kun Li
- Orthopedics Department, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan 410008, People's Republic of China
| | - Zhaohui Huang
- School of Materials Sciences and Technology, China University of Geosciences, 29 Xueyuan Road, Beijing 100086, People's Republic of China
| | - Juntong Huang
- School of Materials Science and Engineering, Nanchang Hangkong University, 696 Fenghe Nan Street, Nanchang, Jiangxi 330063, People's Republic of China
| | - Jie Ju
- Biomaterials Innovation Research Center, Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 65 Landsdowne Street, Cambridge, Massachusetts 02139, United States.,Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Yihe Hu
- Orthopedics Department, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan 410008, People's Republic of China
| | - Ali Khademhosseini
- Biomaterials Innovation Research Center, Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 65 Landsdowne Street, Cambridge, Massachusetts 02139, United States.,Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States.,Department of Physics, King Abdulaziz University, Abdullah Sulayman Street, Jeddah 21569, Saudi Arabia
| |
Collapse
|
42
|
Dinjaski N, Plowright R, Zhou S, Belton DJ, Perry CC, Kaplan DL. Osteoinductive recombinant silk fusion proteins for bone regeneration. Acta Biomater 2017; 49:127-139. [PMID: 27940162 DOI: 10.1016/j.actbio.2016.12.002] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2016] [Revised: 11/03/2016] [Accepted: 12/02/2016] [Indexed: 01/12/2023]
Abstract
Protein polymers provide a unique opportunity for tunable designs of material systems due to the genetic basis of sequence control. To address the challenge of biomineralization interfaces with protein based materials, we genetically engineered spider silks to design organic-inorganic hybrid systems. The spider silk inspired domain (SGRGGLGGQG AGAAAAAGGA GQGGYGGLGSQGT)15 served as an organic scaffold to control material stability and to allow multiple modes of processing, whereas the hydroxyapatite binding domain VTKHLNQISQSY (VTK), provided control over osteogenesis. The VTK domain was fused either to the N-, C- or both terminals of the spider silk domain to understand the effect of position on material properties and mineralization. The addition of the VTK domain to silk did not affect the physical properties of the silk recombinant constructs, but it had a critical role in the induction of biomineralization. When the VTK domain was placed on both the C- and N-termini the formation of crystalline hydroxyapatite was significantly increased. In addition, all of the recombinant proteins in film format supported the growth and proliferation of human mesenchymal stem cells (hMSCs). Importantly, the presence of the VTK domain enhanced osteoinductive properties up to 3-fold compared to the control (silk alone without VTK). Therefore, silk-VTK fusion proteins have been shown suitable for mineralization and functionalization for specific biomedical applications. STATEMENT OF SIGNIFICANCE Organic-inorganic interfaces are integral to biomaterial functions in many areas of repair and regeneration. Several protein polymers have been investigated for this purpose. Despite their success the limited options to fine-tune their material properties, degradation patterns and functionalize them for each specific biomedical application limits their application. Various studies have shown that the biological performance of such proteins can be improved by genetic engineering. The present study provides data relating protein design parameters and functional outcome quantified by biomineralization and human mesenchymal stem cell differentiation. As such, it helps the design of osteoinductive recombinant biomaterials for bone regeneration.
Collapse
Affiliation(s)
- Nina Dinjaski
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, MA 02155, United States
| | - Robyn Plowright
- Biomolecular and Materials Interface Research Group, Interdisciplinary Biomedical Research Centre, School of Science and Technology, Nottingham Trent University, Clifton Lane, Nottingham NG11 8NS, UK
| | - Shun Zhou
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, MA 02155, United States
| | - David J Belton
- Biomolecular and Materials Interface Research Group, Interdisciplinary Biomedical Research Centre, School of Science and Technology, Nottingham Trent University, Clifton Lane, Nottingham NG11 8NS, UK
| | - Carole C Perry
- Biomolecular and Materials Interface Research Group, Interdisciplinary Biomedical Research Centre, School of Science and Technology, Nottingham Trent University, Clifton Lane, Nottingham NG11 8NS, UK.
| | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, MA 02155, United States.
| |
Collapse
|
43
|
|
44
|
Manfrini M, Mazzoni E, Barbanti-Brodano G, Nocini P, D'agostino A, Trombelli L, Tognon M. Osteoconductivity of Complex Biomaterials Assayed by Fluorescent-Engineered Osteoblast-like Cells. Cell Biochem Biophys 2016; 71:1509-15. [PMID: 25388843 DOI: 10.1007/s12013-014-0374-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Biomaterials employed for the bone regeneration can be assayed for specific features such as osteoconductivity and gene expression. In this study, the composite HA/collagen/chondroitin-sulfate biomaterial was investigated using an engineered human cell line, named Saos-eGFP. This cell line, a green fluorescent engineered human osteoblast-like cell, was employed as a cellular model for the in vitro study of biomaterial characteristics. The cytotoxicity was indirectly evaluated by fluorescence detection, osteoconductivity was assayed both by fluorescence and electron microscope analysis as well as cell morphology, whereas the RT-PCR technique was employed to assay gene expression. Saos-eGFP cells viability detection after 24 and 96 h of incubation showed that biomaterial enables the adhesion and proliferation of seeded cells as well as that of the plastic surface, the control. Fluorescence and scanning electron microscopy (SEM) analyses indicated that Saos-eGFP cells were homogeneously distributed on the HA granule surfaces, exhibiting cytoplasmic bridges, and were localized on the collagen-chondroitin sulfate extra-cellular matrix. An expression analysis of specific genes encoding for differentiation markers, showed that biomaterial assayed did not alter the osteogenic pathway of the Saos-eGFP cell line. Our assays confirm the cytocompatibility of this biomaterial, suggesting an osteoconductive capacity mediated by its chemical contents. We showed that the Saos-eGFP cellular model is suitable for in vitro biomaterial assays, and more specifically for assessing osteoconductivity. This result suggests that the cytocompatibility and osteoconductive features of the biomaterial assayed as bone substitute, could have a positive downstream effect on implant osteo-integration.
Collapse
Affiliation(s)
- Marco Manfrini
- Department of Morphology, Surgery and Experimental Medicine, School of Medicine, University of Ferrara, Ferrara, Italy
| | - Elisa Mazzoni
- Department of Morphology, Surgery and Experimental Medicine, School of Medicine, University of Ferrara, Ferrara, Italy
| | | | | | - Antonio D'agostino
- Department of Surgery, School of Medicine, University of Verona, Verona, Italy
| | - Leonardo Trombelli
- Research Centre for the Study of Periodontal and Peri-Implant Diseases, University of Ferrara, Ferrara, Italy
| | - Mauro Tognon
- Department of Morphology, Surgery and Experimental Medicine, School of Medicine, University of Ferrara, Ferrara, Italy.
| |
Collapse
|
45
|
Chaudhari AA, Vig K, Baganizi DR, Sahu R, Dixit S, Dennis V, Singh SR, Pillai SR. Future Prospects for Scaffolding Methods and Biomaterials in Skin Tissue Engineering: A Review. Int J Mol Sci 2016; 17:E1974. [PMID: 27898014 PMCID: PMC5187774 DOI: 10.3390/ijms17121974] [Citation(s) in RCA: 323] [Impact Index Per Article: 35.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Revised: 11/21/2016] [Accepted: 11/22/2016] [Indexed: 01/17/2023] Open
Abstract
Over centuries, the field of regenerative skin tissue engineering has had several advancements to facilitate faster wound healing and thereby restoration of skin. Skin tissue regeneration is mainly based on the use of suitable scaffold matrices. There are several scaffold types, such as porous, fibrous, microsphere, hydrogel, composite and acellular, etc., with discrete advantages and disadvantages. These scaffolds are either made up of highly biocompatible natural biomaterials, such as collagen, chitosan, etc., or synthetic materials, such as polycaprolactone (PCL), and poly-ethylene-glycol (PEG), etc. Composite scaffolds, which are a combination of natural or synthetic biomaterials, are highly biocompatible with improved tensile strength for effective skin tissue regeneration. Appropriate knowledge of the properties, advantages and disadvantages of various biomaterials and scaffolds will accelerate the production of suitable scaffolds for skin tissue regeneration applications. At the same time, emphasis on some of the leading challenges in the field of skin tissue engineering, such as cell interaction with scaffolds, faster cellular proliferation/differentiation, and vascularization of engineered tissues, is inevitable. In this review, we discuss various types of scaffolding approaches and biomaterials used in the field of skin tissue engineering and more importantly their future prospects in skin tissue regeneration efforts.
Collapse
Affiliation(s)
- Atul A Chaudhari
- Center for Nanobiotechnology Research, Alabama State University, Montgomery, AL 36104, USA.
| | - Komal Vig
- Center for Nanobiotechnology Research, Alabama State University, Montgomery, AL 36104, USA.
| | | | - Rajnish Sahu
- Center for Nanobiotechnology Research, Alabama State University, Montgomery, AL 36104, USA.
| | - Saurabh Dixit
- Center for Nanobiotechnology Research, Alabama State University, Montgomery, AL 36104, USA.
| | - Vida Dennis
- Center for Nanobiotechnology Research, Alabama State University, Montgomery, AL 36104, USA.
| | - Shree Ram Singh
- Center for Nanobiotechnology Research, Alabama State University, Montgomery, AL 36104, USA.
| | - Shreekumar R Pillai
- Center for Nanobiotechnology Research, Alabama State University, Montgomery, AL 36104, USA.
| |
Collapse
|
46
|
Synthesis and characterization of well-defined ligand-terminated block copolymer brushes for multifunctional biointerfaces. POLYMER 2016. [DOI: 10.1016/j.polymer.2016.03.031] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
47
|
Lee H, Chung HJ, Park TG. Perspectives On: Local and Sustained Delivery of Angiogenic Growth Factors. J BIOACT COMPAT POL 2016. [DOI: 10.1177/0883911506073363] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
This review emphasizes the role of angiogenesis in tissue engineering, introduces various angiogenic growth factors, and highlights current status of delivery systems for angiogenic growth factors using natural and synthetic biomaterials. A short overview of angiogenic growth factors is presented, followed by the introduction of emerging strategies for designing smart delivery carriers.
Collapse
Affiliation(s)
- Hyukjin Lee
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 305-701, Korea
| | - Hyun Jung Chung
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 305-701, Korea
| | - Tae Gwan Park
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 305-701, Korea,
| |
Collapse
|
48
|
Zhao W, Xu Z, Cui Q, Sahai N. Predicting the Structure-Activity Relationship of Hydroxyapatite-Binding Peptides by Enhanced-Sampling Molecular Simulation. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2016; 32:7009-7022. [PMID: 27329793 DOI: 10.1021/acs.langmuir.6b01582] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Understanding the molecular structural and energetic basis of the interactions between peptides and inorganic surfaces is critical to their applications in tissue engineering and biomimetic material synthesis. Despite recent experimental progresses in the identification and functionalization of hydroxyapatite (HAP)-binding peptides, the molecular mechanisms of their interactions with HAP surfaces are yet to be explored. In particular, the traditional method of molecular dynamics (MD) simulation suffers from insufficient sampling at the peptide-inorganic interface that renders the molecular-level observation dubious. Here we demonstrate that an integrated approach combining bioinformatics, MD, and metadynamics provides a powerful tool for investigating the structure-activity relationship of HAP-binding peptides. Four low charge density peptides, previously identified by phage display, have been considered. As revealed by bioinformatics and MD, the binding conformation of the peptides is controlled by both the sequence and the amino acid composition. It was found that formation of hydrogen bonds between lysine residue and phosphate ions on the surface dictates the binding of positively charged peptide to HAP. The binding affinities of the peptides to the surface are estimated by free energy calculation using parallel-tempering metadynamics, and the results compare favorably to measurements reported in previous experimental studies. The calculation suggests that the charge density of the peptide primarily controls the binding affinity to the surface, while the backbone secondary structure that may restrain side chain orientation toward the surface plays a minor role. We also report that the application of enhanced-sampling metadynamics effects a major advantage over the steered MD method by significantly improving the reliability of binding free energy calculation. In general, our novel integration of diverse sampling techniques should contribute to the rational design of surface-recognition peptides in biomedical applications.
Collapse
Affiliation(s)
| | | | - Qiang Cui
- Department of Chemistry and Theoretical Chemistry Institute, University of Wisconsin-Madison , Madison, Wisconsin 53706-1322, United States
| | | |
Collapse
|
49
|
A tenascin-C mimetic peptide amphiphile nanofiber gel promotes neurite outgrowth and cell migration of neurosphere-derived cells. Acta Biomater 2016; 37:50-8. [PMID: 27063496 DOI: 10.1016/j.actbio.2016.04.010] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Revised: 03/18/2016] [Accepted: 04/07/2016] [Indexed: 12/30/2022]
Abstract
UNLABELLED Biomimetic materials that display natural bioactive signals derived from extracellular matrix molecules like laminin and fibronectin hold promise for promoting regeneration of the nervous system. In this work, we investigated a biomimetic peptide amphiphile (PA) presenting a peptide derived from the extracellular glycoprotein tenascin-C, known to promote neurite outgrowth through interaction with β1 integrin. The tenascin-C mimetic PA (TN-C PA) was found to self-assemble into supramolecular nanofibers and was incorporated through co-assembly into PA gels formed by highly aligned nanofibers. TN-C PA content in these gels increased the length and number of neurites produced from neurons differentiated from encapsulated P19 cells. Furthermore, gels containing TN-C PA were found to increase migration of cells out of neurospheres cultured on gel coatings. These bioactive gels could serve as artificial matrix therapies in regions of neuronal loss to guide neural stem cells and promote through biochemical cues neurite extension after differentiation. One example of an important target would be their use as biomaterial therapies in spinal cord injury. STATEMENT OF SIGNIFICANCE Tenascin-C is an important extracellular matrix molecule in the nervous system and has been shown to play a role in regenerating the spinal cord after injury and guiding neural progenitor cells during brain development, however, minimal research has been reported exploring the use of biomimetic biomaterials of tenascin-C. In this work, we describe a selfassembling biomaterial system in which peptide amphiphiles present a peptide derived from tenascin-C that promotes neurite outgrowth. Encapsulation of neurons in hydrogels of aligned nanofibers formed by tenascin-C-mimetic peptide amphiphiles resulted in enhanced neurite outgrowth. Additionally, these peptide amphiphiles promoted migration of neural progenitor cells cultured on nanofiber coatings. Tenascin-C biomimetic biomaterials such as the one described here have significant potential in neuroregenerative medicine.
Collapse
|
50
|
C K Rajendran SR, Mason B, Udenigwe CC. Peptidomics of Peptic Digest of Selected Potato Tuber Proteins: Post-Translational Modifications and Limited Cleavage Specificity. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2016; 64:2432-2437. [PMID: 26947758 DOI: 10.1021/acs.jafc.6b00418] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Bioinformatic tools are useful in predicting bioactive peptides from food proteins. This study was focused on using bioinformatics and peptidomics to evaluate the specificity of peptide release and post-translational modifications (PTMs) in a peptic digest of potato protein isolate. Peptides in the protein hydrolysate were identified by LC-MS/MS and subsequently aligned to their parent potato tuber proteins. Five major proteins were selected for further analysis, namely, lipoxygenase, α-1,4-glucan phosphorylase, annexin, patatin, and polyubiquitin, based on protein coverage, abundance, confidence levels, and function. Comparison of the in silico peptide profile generated with ExPASy PeptideCutter and experimental peptidomics data revealed several differences. The experimental peptic cleavage sites were found to vary in number and specificity from PeptideCutter predictions. Average peptide chain length was also found to be higher than predicted with hexapeptides as the smallest detected peptides. Moreover, PTMs, particularly Met oxidation and Glu/Asp deamidation, were observed in some peptides, and these were unaccounted for during in silico analysis. PTMs can be formed during aging of potato tubers, or as a result of processing conditions during protein isolation and hydrolysis. The findings provide insights on the limitations of current bioinformatics tools for predicting bioactive peptide release from proteins, and on the existence of structural modifications that can alter the peptide bioactivity and functionality.
Collapse
Affiliation(s)
- Subin R C K Rajendran
- Department of Environmental Sciences, Dalhousie University , Truro, Nova Scotia B2N 5E3, Canada
| | - Beth Mason
- Verschuren Centre for Sustainability in Energy and the Environment, Cape Breton University , Sydney, Nova Scotia B1P 6L2, Canada
| | - Chibuike C Udenigwe
- Department of Environmental Sciences, Dalhousie University , Truro, Nova Scotia B2N 5E3, Canada
| |
Collapse
|