1
|
Li N, Wang M, Luo H, Tse SD, Gao Y, Zhu Z, Guo H, He L, Zhu C, Yin K, Sun L, Guo J, Hong H. Processing and properties of graphene-reinforced polylactic acid nanocomposites for bioelectronic and tissue regenerative functions. BIOMATERIALS ADVANCES 2025; 167:214113. [PMID: 39549370 DOI: 10.1016/j.bioadv.2024.214113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 11/05/2024] [Accepted: 11/07/2024] [Indexed: 11/18/2024]
Abstract
An in-situ polymer-solution-processing approach enables the efficient production of uniform graphene-reinforced polylactic acid (G-PLA) nanocomposites with notable physical and biomedical properties. The approach effectively enhances the interfacial bonding between graphene and PLA by creating graphene dangling bonds and defects during exfoliation. As a result, an 182 % increase in Young's modulus and an 85 % increase in tensile strength can be achieved in G-PLA. Only 0.5 wt% graphene addition can reduce the contact angle of the composite from 75.3 to 70.4 and reduce its oxygen permeability by 23 %. The improved hydrophilicity, hermeticity, and mechanical properties make G-PLA an excellent encapsulation material for implantable bioelectronics. Moreover, the composite surface attributes and cell behaviors at the material-tissue interface are investigated histologically through the culture of stem cells on as-synthesized G-PLA. G-PLA composites can significantly boost cell proliferation and regulate cell differentiation towards vascular endothelium, offering tissue regeneration at the surface of implants to recover the injured tissues. The degradation rate of G-PLA nanocomposite can also be regulated since the graphene slows down the autocatalytic chain splitting induced by the terminal carboxylic acid groups of PLA. Therefore, such G-PLA nanocomposites with physical and biomedical properties regulated by graphene loading enable the development of next-generation implantable electronic systems providing both sensing and tissue engineering functions for complicated applications such as implanted sensors monitoring the healing of fractured bones or intracranial pressure.
Collapse
Affiliation(s)
- Nan Li
- SEU-FEI Nano-Pico Center, Key Lab of MEMS of Ministry of Education, Southeast University, Nanjing 210096, China
| | - Mengjia Wang
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Shandong Key Laboratory of Oral Tissue Regeneration, Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration, Shandong Provincial Clinical Research Center for Oral Diseases, China
| | - Haoyu Luo
- SEU-FEI Nano-Pico Center, Key Lab of MEMS of Ministry of Education, Southeast University, Nanjing 210096, China
| | - Stephen D Tse
- Department of Mechanical and Aerospace Engineering, Rutgers University-New Brunswick, Piscataway, NJ 08854, USA
| | - Yun Gao
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiaotong University, China
| | - Zhen Zhu
- SEU-FEI Nano-Pico Center, Key Lab of MEMS of Ministry of Education, Southeast University, Nanjing 210096, China
| | - Hongxuan Guo
- SEU-FEI Nano-Pico Center, Key Lab of MEMS of Ministry of Education, Southeast University, Nanjing 210096, China
| | - Longbing He
- SEU-FEI Nano-Pico Center, Key Lab of MEMS of Ministry of Education, Southeast University, Nanjing 210096, China
| | - Chao Zhu
- SEU-FEI Nano-Pico Center, Key Lab of MEMS of Ministry of Education, Southeast University, Nanjing 210096, China
| | - Kuibo Yin
- SEU-FEI Nano-Pico Center, Key Lab of MEMS of Ministry of Education, Southeast University, Nanjing 210096, China
| | - Litao Sun
- SEU-FEI Nano-Pico Center, Key Lab of MEMS of Ministry of Education, Southeast University, Nanjing 210096, China
| | - Jie Guo
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Shandong Key Laboratory of Oral Tissue Regeneration, Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration, Shandong Provincial Clinical Research Center for Oral Diseases, China
| | - Hua Hong
- SEU-FEI Nano-Pico Center, Key Lab of MEMS of Ministry of Education, Southeast University, Nanjing 210096, China.
| |
Collapse
|
2
|
Zhu R, Zheng Z, Zhu D, Wang X. Hydrogels with high sacrifice efficiency of sacrificial bonds and with high strength and toughness due to dense entanglements of polymer chains. J Colloid Interface Sci 2025; 677:687-696. [PMID: 39116566 DOI: 10.1016/j.jcis.2024.08.008] [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: 07/09/2024] [Revised: 08/01/2024] [Accepted: 08/02/2024] [Indexed: 08/10/2024]
Abstract
Introducing sacrificial bonds is a common method for increasing the toughness of hydrogels. Many sacrificial bonds have been extensively investigated, but the sacrifice efficiency has never been studied. In this study, polyacrylamide hydrogels with highly entangled polymer chains containing carboxyl-zirconium (-COO--Zr4+) sacrificial bonds are prepared to study the effect of polymer chain entanglement on the sacrificial bond efficiency. Unlike chemical crosslinking points, the dense physical entanglements do not affect the toughness (∼43 MJ/m3) of hydrogels but significantly improve the tensile strength (by two times) and Young's modulus (by six times). Physical entanglements enable the chains to slide and adjust the network structure under stress, which enables more polymer chains and sacrificial bonds to participate in the deformation process. Therefore, dense entanglements will greatly improve the sacrifice efficiency. However, a high density of chemical crosslinking points will limit the improvement in the sacrifice efficiency, which is attributed to the sliding limitations because of physical entanglement. The highly entangled polyacrylamide hydrogels toughened by -COO--Zr4+ have an excellent load-bearing capacity. This study provides a novel strategy for designing hydrogels with ultra-high strength and toughness, which paves the way for the development of many hydrogels used in engineering materials.
Collapse
Affiliation(s)
- Ruixin Zhu
- School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Zhen Zheng
- School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Dandan Zhu
- School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Xinling Wang
- School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China; State Key Laboratory of Metal Matrix Composites, Shanghai Jiao Tong University, Shanghai 200240, China.
| |
Collapse
|
3
|
Kim YH, Cidonio G, Kanczler JM, Oreffo ROC, Dawson JI. Human bone tissue-derived ECM hydrogels: Controlling physicochemical, biochemical, and biological properties through processing parameters. Bioact Mater 2025; 43:114-128. [PMID: 39376928 PMCID: PMC11456876 DOI: 10.1016/j.bioactmat.2024.09.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 08/21/2024] [Accepted: 09/04/2024] [Indexed: 10/09/2024] Open
Abstract
Decellularized tissues offer significant potential as biological materials for tissue regeneration given their ability to preserve the complex compositions and architecture of the native extracellular matrix (ECM). However, the evaluation and derivation of decellularized matrices from human bone tissue remains largely unexplored. We examined how the physiochemical and biological properties of ECM hydrogels derived from human bone ECM could be controlled by manipulating bone powder size (45-250 μm, 250-1000 μm, and 1000-2000 μm) and ECM composition through modulation of enzyme digestion time (3-5-7 days). A reduction in material bone powder size and an increase in ECM digestion time produced enhanced protein concentrations in the ECM hydrogels, accompanied by the presence of a diverse array of proteins and improved gelation strength. Human bone marrow-derived stromal cells (HBMSCs) cultured on ECM hydrogels from 45 to 250 μm bone powder, over 7 days, demonstrated enhanced osteogenic differentiation compared to hydrogels derived from larger bone powders and collagen gels confirming the potential of the hydrogels as biologically active materials for bone regeneration. Digestion time and bone powder size modulation enabled the generation of hydrogels with enhanced release of ECM proteins and appropriate gelation and rheological properties, offering new opportunities for application in bone repair.
Collapse
Affiliation(s)
- Yang-Hee Kim
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Institute of Developmental Sciences, University of Southampton, SO16 6YD, United Kingdom
| | - Gianluca Cidonio
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Institute of Developmental Sciences, University of Southampton, SO16 6YD, United Kingdom
- Department of Mechanical and Aerospace Engineering (DIMA), Sapienza University of Rome, Via Eudossiana 18, 00184, Rome, Italy
- Center for Life Nano- and Neuro-Science (CLN2S), Istituto Italiano di Tecnologia, Viale Regina Elena 291, 00161, Rome, Italy
| | - Janos M. Kanczler
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Institute of Developmental Sciences, University of Southampton, SO16 6YD, United Kingdom
| | - Richard OC. Oreffo
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Institute of Developmental Sciences, University of Southampton, SO16 6YD, United Kingdom
| | - Jonathan I. Dawson
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Institute of Developmental Sciences, University of Southampton, SO16 6YD, United Kingdom
| |
Collapse
|
4
|
Zakrzewska A, Kosik-Kozioł A, Zargarian SS, Zanoni M, Gualandi C, Lanzi M, Pierini F. Lemon Juice-Infused PVA Nanofibers for the Development of Sustainable Antioxidant and Antibacterial Electrospun Hydrogel Biomaterials. Biomacromolecules 2025. [PMID: 39743322 DOI: 10.1021/acs.biomac.4c01466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Abstract
Cross-linking bonds adjacent polymer chains into a three-dimensional network. Cross-linked poly(vinyl alcohol) (PVA) turns into a hydrogel, insoluble structure exhibiting outstanding sorption properties. As an electrospinnable polymer, PVA enables the creation of nanofibrous hydrogels resembling biological tissues, thus ideal for nature-inspired platforms. PVA properties are easily adjustable through additives and an appropriate cross-linking method. Drawing inspiration from environmentally safe approaches, this work developed a new "green" method of low-temperature PVA cross-linking. Nanofibers were electrospun from a precursor solution of PVA dissolved in fresh lemon juice, stabilized by heating at 60 °C for 7 days, and thoroughly characterized. The obtained nanoplatform demonstrated long-term stability and enhanced mechanical properties. Its biocompatibility was confirmed, and its antibacterial and health-promoting effects were attributed to lemon juice-rich in vitamin C, a potent antioxidant with anti-inflammatory properties. The developed system has future potential for use in the biomedical engineering field as a dressing accelerating wound healing.
Collapse
Affiliation(s)
- Anna Zakrzewska
- Department of Biosystems and Soft Matter, Institute of Fundamental Technological Research, Polish Academy of Sciences, Pawińskiego 5B, 02-106 Warsaw, Poland
| | - Alicja Kosik-Kozioł
- Department of Biosystems and Soft Matter, Institute of Fundamental Technological Research, Polish Academy of Sciences, Pawińskiego 5B, 02-106 Warsaw, Poland
| | - Seyed Shahrooz Zargarian
- Department of Biosystems and Soft Matter, Institute of Fundamental Technological Research, Polish Academy of Sciences, Pawińskiego 5B, 02-106 Warsaw, Poland
| | - Michele Zanoni
- Department of Chemistry "Giacomo Ciamician″, University of Bologna, Via Selmi 2, 40126 Bologna, Italy
| | - Chiara Gualandi
- Department of Chemistry "Giacomo Ciamician″, University of Bologna, Via Selmi 2, 40126 Bologna, Italy
- INSTM UdR of Bologna, University of Bologna, Via Selmi 2, 40126 Bologna, Italy
- Interdepartmental Center for Industrial Research on Advanced Applications in Mechanical Engineering and Materials Technology, CIRI-MAM, University of Bologna, Viale Risorgimento 2, 40136 Bologna, Italy
| | - Massimiliano Lanzi
- Department of Industrial Chemistry "Toso Montanari", University of Bologna, Viale del Risorgimento 4, 40136 Bologna, Italy
| | - Filippo Pierini
- Department of Biosystems and Soft Matter, Institute of Fundamental Technological Research, Polish Academy of Sciences, Pawińskiego 5B, 02-106 Warsaw, Poland
| |
Collapse
|
5
|
Jain N, Singh A, Bhatia D. DNA-amphiphilic nanostructures: synthesis, characterization and applications. NANOSCALE 2024; 17:18-52. [PMID: 39560070 DOI: 10.1039/d4nr03236e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2024]
Abstract
DNA's extraordinary potential reaches far beyond its role as a carrier of genetic information. It serves as a remarkably adaptable structural foundation for constructing intricate nanostructures with a diverse range of functionalities. This inherent programmability sets DNA apart from other biomolecules like peptides, proteins, and small molecules. By covalently attaching DNA to synthetic hydrophobic moieties, researchers create DNA amphiphiles capable of interacting with artificial lipid bilayers and cell membranes. These hybrid structures have rapidly gained prominence due to their promising potential in the medical field. This review provides a comprehensive overview of the latest advancements in the synthesis of DNA amphiphiles and their assembly into well-defined nanostructures. It explores the diverse applications of these nanostructures across various medical domains, including targeted drug delivery, innovative immunotherapies, and gene-silencing techniques. Moreover, the review delves into the current challenges and prospects of this rapidly evolving field, highlighting the potential of DNA hybrid materials to revolutionize medical treatments and diagnostics. By addressing the limitations and exploring new avenues of research, scientists aim to unlock the full potential of DNA nanotechnology for the benefit of human health.
Collapse
Affiliation(s)
- Nishkarsh Jain
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Prem Nagar, Patiala, Punjab 147004, India
| | - Ankur Singh
- Department of Biological Sciences & Engineering, Indian Institute of Technology Gandhinagar, Palaj, Gujarat 382355, India.
| | - Dhiraj Bhatia
- Department of Biological Sciences & Engineering, Indian Institute of Technology Gandhinagar, Palaj, Gujarat 382355, India.
| |
Collapse
|
6
|
Thai NLB, Beaman HT, Perlman M, Obeng EE, Du C, Monroe MBB. Chitosan Poly(vinyl alcohol) Methacrylate Hydrogels for Tissue Engineering Scaffolds. ACS APPLIED BIO MATERIALS 2024; 7:7818-7827. [PMID: 38380883 DOI: 10.1021/acsabm.3c01209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2024]
Abstract
A major challenge in tissue engineering scaffolds is controlling scaffold degradation rates during healing while maintaining mechanical properties to support tissue formation. Hydrogels are three-dimensional matrices that are widely applied as tissue scaffolds based on their unique properties that can mimic the extracellular matrix. In this study, we develop a hybrid natural/synthetic hydrogel platform to tune the properties for tissue engineering scaffold applications. We modified chitosan and poly(vinyl alcohol) (PVA) with photo-cross-linkable methacrylate functional groups and then synthesized a library of chitosan PVA methacrylate hydrogels (ChiPVAMA) with two different photoinitiators, Irgacure 2959 (I2959) and lithium phenyl-2,4,6-trimethylbenzoylphosphinate (LAP). ChiPVAMA hydrogels showed tunability in degradation rates and mechanical properties based on both the polymer content and photoinitiator type. This tunability could enable their application in a range of tissue scaffold applications. In a 2D scratch wound healing assay, all hydrogel samples induced faster wound closure compared to a gauze clinical wound dressing control. NIH/3T3 cells encapsulated in hydrogels showed a high viability (∼92%) over 14 days, demonstrating the capacity of this system as a supportive cell scaffold. In addition, hydrogels containing a higher chitosan content demonstrated a high antibacterial capacity. Overall, ChiPVAMA hydrogels provide a potential tissue engineering scaffold that is tunable, degradable, and suitable for cell growth.
Collapse
Affiliation(s)
- Nghia Le Ba Thai
- Department of Biomedical and Chemical Engineering, Syracuse Biomaterials Institute, and BioInspired Syracuse: Institute for Material and Living Systems, Syracuse University, Syracuse, New York 13244, United States
| | - Henry T Beaman
- Department of Biomedical and Chemical Engineering, Syracuse Biomaterials Institute, and BioInspired Syracuse: Institute for Material and Living Systems, Syracuse University, Syracuse, New York 13244, United States
| | - Megan Perlman
- Department of Biomedical and Chemical Engineering, Syracuse Biomaterials Institute, and BioInspired Syracuse: Institute for Material and Living Systems, Syracuse University, Syracuse, New York 13244, United States
| | - Ernest E Obeng
- Department of Biomedical and Chemical Engineering, Syracuse Biomaterials Institute, and BioInspired Syracuse: Institute for Material and Living Systems, Syracuse University, Syracuse, New York 13244, United States
| | - Changling Du
- Department of Biomedical and Chemical Engineering, Syracuse Biomaterials Institute, and BioInspired Syracuse: Institute for Material and Living Systems, Syracuse University, Syracuse, New York 13244, United States
| | - Mary Beth B Monroe
- Department of Biomedical and Chemical Engineering, Syracuse Biomaterials Institute, and BioInspired Syracuse: Institute for Material and Living Systems, Syracuse University, Syracuse, New York 13244, United States
| |
Collapse
|
7
|
Stern I, Barrera V, Randles M, Rooney P. Advances in preparation of acellular human dermis for tissue banking and transplantation. Cell Tissue Bank 2024; 26:3. [PMID: 39653869 PMCID: PMC11628444 DOI: 10.1007/s10561-024-10153-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 11/20/2024] [Indexed: 12/12/2024]
Abstract
Non-healing wounds cost the National Health Service over £5.6 billion annually in wound management. Skin allografts are used to treat non-healing wounds, ulcers and burns, offering the best protection against infection. In order to allow host cells to repopulate and to avoid immunogenicity, cell components are removed through decellularisation. Decellularisation of human dermis has so far been performed in NHS Blood and Transplant using a combination of two enzymes (RNase T1 and the recombinant human DNase Pulmozyme)®. This study aims at validating a new method to remove DNA from donated dermis via the use of a single enzyme, Benzonase, known for its effectiveness of DNA digestion. Skin samples were decellularised by removing the epidermis, lysing of dermal cells, removal of cellular fragments by a detergent wash and removal of nucleic acids by a nuclease incubation with either Benzonase or Pulmozyme + RNase T1. DNA quantification with PicoGreen, as well as histology on wax-embedded biopsies, stained with DAPI and haemotoxylin and eosin, were performed. In vitro toxicity test on human osteosarcoma immortalised cells and skin fibroblasts, and biomechanical (tensile) testing, were also performed. The effectiveness of DNA digestion with the new methodology was comparable to previous procedure. Mean DNA removal percentage following decellularisation with Pulmozyme + RNase was 99.9% (3.83 ng/mg). Mean DNA removal percentage with Benzonase was 99.8% (9.97 ng/mg). Histology staining showed complete decellularisation following either method. Benzonase was proven to be non-toxic to both cell lines used, and a one-way Anova test showed no significant difference in neither stress nor strain between acellular dermal matrix decellularised with either Benzonase or Pulmozyme + RNase T1. Benzonase was able to effectively decellularise dermis after prior removal of epidermis. It performed just as well as the combination of Pulmozyme + RNase T1, but represents significant advantages in terms of cost effectiveness, procurement and storage; Benzonase has been successfully used in the decellularisation of other tissues, thus would be better for Tissue Banking use. Switching to this combined DNase/RNase can have far-reaching consequences in the production of acellular human dermal matrix by NHSBT and in the treatment of patients requiring it.
Collapse
Affiliation(s)
- Irit Stern
- NHS Blood and Transplant, Tissue Services, 14 Estuary Banks, Speke, Liverpool, L24 8RB, UK.
| | - Valentina Barrera
- NHS Blood and Transplant, Tissue Services R&D, 14 Estuary Banks, Speke, Liverpool, L24 8RB, UK
| | - Michael Randles
- Faculty of Medicine and Life Sciences, Chester Medical School, University of Chester, Chester, UK
| | - Paul Rooney
- NHS Blood and Transplant, Tissue Services R&D, 14 Estuary Banks, Speke, Liverpool, L24 8RB, UK
| |
Collapse
|
8
|
García-Sobrino R, Martínez-Campos E, Marcos-Ríos D, Zepeda-Rodríguez Z, Valentín JL, Sanz-Horta R, León-Calero M, Reinecke H, Elvira C, Gallardo A, Rodríguez-Hernández J. Development of Thermosensitive Hydrogels with Tailor-Made Geometries to Modulate Cell Harvesting of Non-Flat Cell Cultures. Gels 2024; 10:802. [PMID: 39727560 DOI: 10.3390/gels10120802] [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: 10/13/2024] [Revised: 12/02/2024] [Accepted: 12/04/2024] [Indexed: 12/28/2024] Open
Abstract
Considering the complexity in terms of design that characterizes the different tissues of the human body, it is necessary to study and develop more precise therapies. In this sense, this article presents the possibility of fabricating photocurable thermosensitive hydrogels with free geometry and based on N-Vinyl Caprolactam (VCL) with the aim of modulating the adhesion of non-planar cell cultures. The fabrication process is based on the use as a mold of two-layer thick water-soluble polyvinyl alcohol (PVA) previously printed by Extrusion Material (MatEx). From this technology it has been possible to obtain hydrogels with different 3D geometries and different crosslinking percentages (2, 4 and 6 mol%). Studies have shown that networks reduce their thermosensitivity not only when the percentage of crosslinking in the formulation increases, but also when the thickness of the hydrogel obtained increases. Based on this reduction in thermosensitivity, the less crosslinked (2 mol%) hydrogels have been evaluated to carry out a novel direct application in which hydrogels with curved geometry have allowed cell adhesion and proliferation at 37 °C with the endothelial cell line C166-GFP; likewise, non-aggressive cell detachment was observed when the hydrogel temperature was reduced to values of 20 °C. Therefore, the present manuscript shows a novel application for the synthesis of free-form thermosensitive hydrogels that allows modulation of non-planar cell cultures.
Collapse
Affiliation(s)
- Rubén García-Sobrino
- Polymer Functionalization Group, Departamento de Química Macromolecular Aplicada, Instituto de Ciencia y Tecnología de Polímeros-Consejo Superior de Investigaciones Científicas (ICTP-CSIC), Calle Juan de la Cierva, n° 3, 28006 Madrid, Spain
- Group of Organic Synthesis and Bioevaluation, Instituto Pluridisciplinar, Universidad Complutense de Madrid (UCM), Associated Unit to the ICTP-IQM-CSIC, Paseo Juan XXIII, n° 1, 28040 Madrid, Spain
- Department of Applied Mathematics, Materials Science and Engineering and Electronic Technology, Universidad Rey Juan Carlos, Calle Tulipán s/n, 28933 Móstoles, Spain
| | - Enrique Martínez-Campos
- Polymer Functionalization Group, Departamento de Química Macromolecular Aplicada, Instituto de Ciencia y Tecnología de Polímeros-Consejo Superior de Investigaciones Científicas (ICTP-CSIC), Calle Juan de la Cierva, n° 3, 28006 Madrid, Spain
- Group of Organic Synthesis and Bioevaluation, Instituto Pluridisciplinar, Universidad Complutense de Madrid (UCM), Associated Unit to the ICTP-IQM-CSIC, Paseo Juan XXIII, n° 1, 28040 Madrid, Spain
| | - Daniel Marcos-Ríos
- Polymer Functionalization Group, Departamento de Química Macromolecular Aplicada, Instituto de Ciencia y Tecnología de Polímeros-Consejo Superior de Investigaciones Científicas (ICTP-CSIC), Calle Juan de la Cierva, n° 3, 28006 Madrid, Spain
- Group of Organic Synthesis and Bioevaluation, Instituto Pluridisciplinar, Universidad Complutense de Madrid (UCM), Associated Unit to the ICTP-IQM-CSIC, Paseo Juan XXIII, n° 1, 28040 Madrid, Spain
| | - Zenen Zepeda-Rodríguez
- Elastomers Group, Instituto de Ciencia y Tecnología de Polímeros-Consejo Superior de Investigaciones Científicas (ICTP-CSIC), Calle Juan de la Cierva, n° 3, 28006 Madrid, Spain
| | - Juan L Valentín
- Elastomers Group, Instituto de Ciencia y Tecnología de Polímeros-Consejo Superior de Investigaciones Científicas (ICTP-CSIC), Calle Juan de la Cierva, n° 3, 28006 Madrid, Spain
| | - Raúl Sanz-Horta
- Polymer Functionalization Group, Departamento de Química Macromolecular Aplicada, Instituto de Ciencia y Tecnología de Polímeros-Consejo Superior de Investigaciones Científicas (ICTP-CSIC), Calle Juan de la Cierva, n° 3, 28006 Madrid, Spain
| | - Marina León-Calero
- Polymer Functionalization Group, Departamento de Química Macromolecular Aplicada, Instituto de Ciencia y Tecnología de Polímeros-Consejo Superior de Investigaciones Científicas (ICTP-CSIC), Calle Juan de la Cierva, n° 3, 28006 Madrid, Spain
| | - Helmut Reinecke
- Polymer Functionalization Group, Departamento de Química Macromolecular Aplicada, Instituto de Ciencia y Tecnología de Polímeros-Consejo Superior de Investigaciones Científicas (ICTP-CSIC), Calle Juan de la Cierva, n° 3, 28006 Madrid, Spain
| | - Carlos Elvira
- Polymer Functionalization Group, Departamento de Química Macromolecular Aplicada, Instituto de Ciencia y Tecnología de Polímeros-Consejo Superior de Investigaciones Científicas (ICTP-CSIC), Calle Juan de la Cierva, n° 3, 28006 Madrid, Spain
| | - Alberto Gallardo
- Polymer Functionalization Group, Departamento de Química Macromolecular Aplicada, Instituto de Ciencia y Tecnología de Polímeros-Consejo Superior de Investigaciones Científicas (ICTP-CSIC), Calle Juan de la Cierva, n° 3, 28006 Madrid, Spain
| | - Juan Rodríguez-Hernández
- Polymer Functionalization Group, Departamento de Química Macromolecular Aplicada, Instituto de Ciencia y Tecnología de Polímeros-Consejo Superior de Investigaciones Científicas (ICTP-CSIC), Calle Juan de la Cierva, n° 3, 28006 Madrid, Spain
| |
Collapse
|
9
|
Banigo AT, Konings IBM, Nauta L, Zoetebier B, Karperien M. Synthesis and Engineering of Hyaluronic Acid-Gelatin Hydrogels with Improved Cellular Attachment and Growth. Polymers (Basel) 2024; 16:3410. [PMID: 39684154 DOI: 10.3390/polym16233410] [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: 10/17/2024] [Revised: 11/22/2024] [Accepted: 11/27/2024] [Indexed: 12/18/2024] Open
Abstract
Injectable hydrogels are promising materials for cartilage regeneration in tissue engineering due to their tunable crosslinking rates, mechanical properties, and biodegradation profiles. This study investigates the chondrogenic potential of hyaluronic acid (HA) hydrogels crosslinked via tyramine (TA) moieties, with and without gelatin modified with TA (Gel-TA). Incorporating Gel-TA improved cell viability, spreading, and cartilage matrix deposition, particularly in medium and high molecular weight (MMW and HMW) HA-TA/Gel-TA hydrogels. Although the hydrogels' molecular weight did not significantly alter stiffness, MMW and HMW HA-TA/Gel-TA formulations exhibited enhanced functional properties such as slower degradation and superior cartilage matrix deposition. These attributes, coupled with Gel-TA's effects, underscore the importance of both molecular weight and biofunctional components in hydrogel design for cartilage regeneration. While low molecular weight (LMW) HA-TA hydrogels offered excellent injectability and supported high cell viability, they degraded rapidly and exhibited reduced cartilage matrix formation. Gel-TA enhanced cell adhesion and spreading by providing integrin-binding sites and promoted collagen type II deposition, crucial for cartilage regeneration. Moreover, the increased stiffness of MMW and HMW HA-TA/Gel-TA hydrogels facilitated extracellular matrix production. These findings show the potential of Gel-TA-modified HA-TA hydrogels for cartilage tissue engineering, with the opportunity for further optimization through the incorporation of bioactive components.
Collapse
Affiliation(s)
- Alma Tamunonengiofori Banigo
- Department of Developmental BioEngineering, Faculty of Science and Technology and TechMed Centre, University of Twente, Drienerlolaan 5, 7522 NB Enschede, The Netherlands
| | - Irene B M Konings
- Department of Developmental BioEngineering, Faculty of Science and Technology and TechMed Centre, University of Twente, Drienerlolaan 5, 7522 NB Enschede, The Netherlands
| | - Laura Nauta
- Department of Developmental BioEngineering, Faculty of Science and Technology and TechMed Centre, University of Twente, Drienerlolaan 5, 7522 NB Enschede, The Netherlands
| | - Bram Zoetebier
- Department of Developmental BioEngineering, Faculty of Science and Technology and TechMed Centre, University of Twente, Drienerlolaan 5, 7522 NB Enschede, The Netherlands
| | - Marcel Karperien
- Department of Developmental BioEngineering, Faculty of Science and Technology and TechMed Centre, University of Twente, Drienerlolaan 5, 7522 NB Enschede, The Netherlands
| |
Collapse
|
10
|
Li S, Luo M, Li J, Huang Q, Lei B. Sprayable Nanocomposites Hydrogel for Wound Healing and Skin Regeneration. Adv Healthc Mater 2024; 13:e2402549. [PMID: 39400478 DOI: 10.1002/adhm.202402549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 09/23/2024] [Indexed: 10/15/2024]
Abstract
Wound management remains a critical challenge worldwide and imposes a huge financial burden on every nation. Hydrogels are promising for biomedical applications because of their extracellular matrix (ECM) like structure, good biocompatibility and multifunctional bioactivity. However, the poor mechanical properties and inconvenient operation of traditional hydrogels make it difficult to meet the complex and multifaceted needs of clinical practice. In recent years, the multifunctional nanocomposites hydrogel with especially sprayable feature have shown enhanced mechanical properties and facile operation, which enable their huge clinical applications value. A unique and powerful nanocomposite hydrogels (NCH) platform is developed by combining the many advantages of nanomaterials and hydrogels, which can achieve efficient trauma repair. This work reviews important advances on the preparation, functions and applications of sprayable NCH platforms. The challenges and future trends in the field with the aim of providing researchers with clarity on the past, present, and future of the emerging field of sprayable NCH are also proposed in detail.
Collapse
Affiliation(s)
- Sihua Li
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, 710000, China
| | - Meng Luo
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, 710000, China
- Academy of Advanced Interdisciplinary Research, Xidian University, Xi'an, 710126, China
| | - Juntang Li
- Research Centre of Immunity, Trauma and Environment Medicine, Collaborative Innovation Centre of Medical Equipment, PLA Key Laboratory of Biological Damage Effect and Protection, Luoyang, 471031, China
| | - Qian Huang
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, 710000, China
| | - Bo Lei
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, 710000, China
| |
Collapse
|
11
|
Pablos JL, Lozano D, Manzano M, Vallet-Regí M. Regenerative medicine: Hydrogels and mesoporous silica nanoparticles. Mater Today Bio 2024; 29:101342. [PMID: 39649249 PMCID: PMC11625165 DOI: 10.1016/j.mtbio.2024.101342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 11/06/2024] [Accepted: 11/13/2024] [Indexed: 12/10/2024] Open
Abstract
Hydrogels, that are crosslinked polymer networks, can absorb huge quantities of water and/or biological fluids. Their physical properties, such as elasticity and soft tissue, together with their biocompatibility and biodegradability, closely resemble living tissues. The versatility of hydrogels has fuelled their application in various fields, such as agriculture, biomaterials, the food industry, drug delivery, tissue engineering, and regenerative medicine. Their combination with nanoparticles, specifically with Mesoporous Silica Nanoparticles (MSNs), have elevated these composites to the next level, since MSNs could improve the hydrogel mechanical properties, their ability to encapsulate and controlled release great amounts of different therapeutic agents, and their responsiveness to a variety of external and internal stimuli. In this review, the main features of both MSNs and hydrogels are introduced, followed by the discussion of different hydrogels-MSNs structures and an overview of their use in different applications, such as drug delivery technologies and tissue engineering.
Collapse
Affiliation(s)
- Jesús L. Pablos
- Dpto. Química en Ciencias Farmacéuticas, U.D Química Inorgánica y Bioinorgánica. Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Hospital, 12 de Octubre i+12, Plaza Ramón y Cajal s/n, Madrid, 28040, Spain
| | - Daniel Lozano
- Dpto. Química en Ciencias Farmacéuticas, U.D Química Inorgánica y Bioinorgánica. Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Hospital, 12 de Octubre i+12, Plaza Ramón y Cajal s/n, Madrid, 28040, Spain
- Centro de Investigación Biomédica en Red, CIBER-BBN, Madrid, Spain
| | - Miguel Manzano
- Dpto. Química en Ciencias Farmacéuticas, U.D Química Inorgánica y Bioinorgánica. Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Hospital, 12 de Octubre i+12, Plaza Ramón y Cajal s/n, Madrid, 28040, Spain
- Centro de Investigación Biomédica en Red, CIBER-BBN, Madrid, Spain
| | - María Vallet-Regí
- Dpto. Química en Ciencias Farmacéuticas, U.D Química Inorgánica y Bioinorgánica. Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Hospital, 12 de Octubre i+12, Plaza Ramón y Cajal s/n, Madrid, 28040, Spain
- Centro de Investigación Biomédica en Red, CIBER-BBN, Madrid, Spain
| |
Collapse
|
12
|
Rovers MM, Rogkoti T, Bakker BK, Bakal KJ, van Genderen MH, Salmeron‐Sanchez M, Dankers PY. Using a Supramolecular Monomer Formulation Approach to Engineer Modular, Dynamic Microgels, and Composite Macrogels. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2405868. [PMID: 39463044 PMCID: PMC11636168 DOI: 10.1002/adma.202405868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 10/07/2024] [Indexed: 10/29/2024]
Abstract
Microgels show advantages over bulk hydrogels due to convenient control over microgel size and composition, and the ability to use microgels to modularly construct larger hierarchical scaffold hydrogel materials. Here, supramolecular chemistry is used to formulate supramolecular polymer, dynamic microgels solely held together by non-covalent interactions. Four-fold hydrogen bonding ureido-pyrimidinone (UPy) monomers with different functionalities are applied to precisely tune microgel properties in a modular way, via variations in monomer concentration, bifunctional crosslinker ratio, and the incorporation of supramolecular dyes and peptides. Functionalization with a bioactive supramolecular cell-adhesive peptide induced selectivity of cells toward the bioactive microgels over non-active, non-functionalized versions. Importantly, the supramolecular microgels can also be applied as microscale building blocks into supramolecular bulk macrogels with tunable dynamic behavior: a robust and weak macrogel, where the micro- and macrogels are composed of similar molecular building blocks. In a robust macrogel, microgels act as modular micro-building blocks, introducing multi-compartmentalization, while in a weak macrogel, microgels reinforce and enhance mechanical properties. This work demonstrates the potential to modularly engineer higher-length-scale structures using small molecule supramolecular monomers, wherein microgels serve as versatile and modular micro-building units.
Collapse
Affiliation(s)
- Maritza M. Rovers
- Institute for Complex Molecular SystemsEindhoven University of TechnologyP.O. Box 513Eindhoven5600 MBThe Netherlands
- Department of Biomedical EngineeringLaboratory of Chemical BiologyEindhoven University of TechnologyP.O. Box 513Eindhoven5600 MBThe Netherlands
| | - Theodora Rogkoti
- Centre for the Cellular MicroenvironmentUniversity of Glasgow, Advanced Research Centre11 Chapel LaneGlasgowG11 6EWUK
| | - Bram K. Bakker
- Institute for Complex Molecular SystemsEindhoven University of TechnologyP.O. Box 513Eindhoven5600 MBThe Netherlands
- Department of Biomedical EngineeringLaboratory of Chemical BiologyEindhoven University of TechnologyP.O. Box 513Eindhoven5600 MBThe Netherlands
| | - Kalpit J. Bakal
- Institute for Complex Molecular SystemsEindhoven University of TechnologyP.O. Box 513Eindhoven5600 MBThe Netherlands
- Department of Mechanical EngineeringEindhoven University of TechnologyP.O. Box 513Eindhoven5600 MBThe Netherlands
| | - Marcel H.P. van Genderen
- Institute for Complex Molecular SystemsEindhoven University of TechnologyP.O. Box 513Eindhoven5600 MBThe Netherlands
- Department of Biomedical EngineeringLaboratory of Chemical BiologyEindhoven University of TechnologyP.O. Box 513Eindhoven5600 MBThe Netherlands
| | - Manuel Salmeron‐Sanchez
- Centre for the Cellular MicroenvironmentUniversity of Glasgow, Advanced Research Centre11 Chapel LaneGlasgowG11 6EWUK
- Institute for Bioengineering of Catalonia (IBEC)The Barcelona Institute for Science and Technology (BIST)Barcelona08028Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA)Barcelona08010Spain
| | - Patricia Y.W. Dankers
- Institute for Complex Molecular SystemsEindhoven University of TechnologyP.O. Box 513Eindhoven5600 MBThe Netherlands
- Department of Biomedical EngineeringLaboratory of Chemical BiologyEindhoven University of TechnologyP.O. Box 513Eindhoven5600 MBThe Netherlands
- Department of Chemical Engineering and ChemistryEindhoven University of TechnologyP.O. Box 513Eindhoven5600 MBThe Netherlands
| |
Collapse
|
13
|
Umarov AZ, Nikitina EA, Piryazev AA, Moutsios I, Rosenthal M, Kurbatov AO, Gordievskaya YD, Kramarenko EY, Dashtimoghadam E, Maw MR, Sheiko SS, Ivanov DA. Revealing Long-Range Order in Brush-like Graft Copolymers Through In Situ Measurements of X-Ray Scattering During Deformation. Polymers (Basel) 2024; 16:3309. [PMID: 39684055 DOI: 10.3390/polym16233309] [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: 10/28/2024] [Revised: 11/22/2024] [Accepted: 11/23/2024] [Indexed: 12/18/2024] Open
Abstract
Brush-like graft copolymers (A-g-B), in which linear A-blocks are randomly grafted onto the backbone of a brush-like B-block, exhibit intense strain-stiffening and high mechanical strength on par with load-bearing biological tissues such as skin and blood vessels. To elucidate molecular mechanisms underlying this tissue-mimetic behavior, in situ synchrotron X-ray scattering was measured during uniaxial stretching of bottlebrush- and comb-like graft copolymers with varying densities of poly(dimethyl siloxane) and poly(isobutylene) side chains. In an undeformed state, these copolymers revealed a single interference peak corresponding to the average spacing between the domains of linear A-blocks arranged in a disordered, liquid-like configuration. Under uniaxial stretching, the emergence of a distinct four-spot pattern in the small-angle region indicated the development of long-range order within the material. According to the affine deformation of a cubic lattice, the four-spot pattern's interference maxima correspond to 110 reflections upon stretching along the [111] axis of the body-centered unit cell. The experimental findings were corroborated by computer simulations of dissipative particle dynamics that confirmed the formation of a bcc domain structure.
Collapse
Affiliation(s)
- Akmal Z Umarov
- Department of Chemistry, Lomonosov Moscow State University (MSU), GSP-1, 1-3 Leninskiye Gory, 119991 Moscow, Russia
| | - Evgeniia A Nikitina
- Department of Chemistry, Lomonosov Moscow State University (MSU), GSP-1, 1-3 Leninskiye Gory, 119991 Moscow, Russia
| | - Alexey A Piryazev
- Department of Chemistry, Lomonosov Moscow State University (MSU), GSP-1, 1-3 Leninskiye Gory, 119991 Moscow, Russia
| | - Ioannis Moutsios
- Institut de Sciences des Matériaux de Mulhouse-IS2M, CNRS UMR 7361, F-68057 Mulhouse, France
| | - Martin Rosenthal
- Department of Chemistry, KU Leuven, Celestijnenlaan 200F, Box 2404, B-3001 Leuven, Belgium
| | - Andrey O Kurbatov
- Department of Chemistry, Lomonosov Moscow State University (MSU), GSP-1, 1-3 Leninskiye Gory, 119991 Moscow, Russia
| | - Yulia D Gordievskaya
- Department of Chemistry, Lomonosov Moscow State University (MSU), GSP-1, 1-3 Leninskiye Gory, 119991 Moscow, Russia
| | - Elena Yu Kramarenko
- Department of Chemistry, Lomonosov Moscow State University (MSU), GSP-1, 1-3 Leninskiye Gory, 119991 Moscow, Russia
| | - Erfan Dashtimoghadam
- Department of Chemistry, University of North Carolina, Chapel Hill, NC 27599-3290, USA
| | - Mitchell R Maw
- Department of Chemistry, University of North Carolina, Chapel Hill, NC 27599-3290, USA
| | - Sergei S Sheiko
- Department of Chemistry, University of North Carolina, Chapel Hill, NC 27599-3290, USA
| | - Dimitri A Ivanov
- Department of Chemistry, Lomonosov Moscow State University (MSU), GSP-1, 1-3 Leninskiye Gory, 119991 Moscow, Russia
- Institut de Sciences des Matériaux de Mulhouse-IS2M, CNRS UMR 7361, F-68057 Mulhouse, France
| |
Collapse
|
14
|
Duan Y, Li L, Hu J, Zheng B, He K. Engineering Gas-Releasing Nanomaterials for Efficient Wound Healing. Chembiochem 2024:e202400790. [PMID: 39592412 DOI: 10.1002/cbic.202400790] [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: 09/26/2024] [Revised: 11/25/2024] [Accepted: 11/26/2024] [Indexed: 11/28/2024]
Abstract
The escalating prevalence of tissue damage and its associated complications has elicited global apprehension. While nanomaterial-based wound healing exhibits significant potential in terms of curbing infections and surpassing conventional methods, unresolved concerns regarding nanomaterial controllability and precision remain unresolved, jeopardizing its practical applications. In recent years, a unique strategy for creating gas-releasing nanomaterials for wound repair has been proposed, involving the creation of gas-releasing nanomaterials to facilitate wound repair by generating gas donor moieties. The operational spatiotemporal responsiveness and broad-spectrum antibacterial properties of these gases, combined with their inability to generate bacterial resistance like traditional antibiotics, establish their efficacy in addressing chronic non-healing wounds, specifically diabetic foot ulcers (DFUs). In this review, we delve into the intricacies of wound healing process, emphasizing the chemical design, functionality, bactericidal activity, and potential of gas-release materials, encompassing NO, CO, H2S, O2, CO2, and H2, for effective wound healing. Furthermore, we explore the advancements in synergistic therapy utilizing these gases, aiming to enhance our overall comprehension of this field. The insights gleaned from this review will undoubtedly aid researchers and developers in the creation of promising gas-releasing nanomaterials, thus propelling efficient wound healing in the future.
Collapse
Affiliation(s)
- Yutian Duan
- SINOPEC Nanjing Research Institute of Chemical Industry Co., Ltd., Nanjing, 210048, China
| | - Lei Li
- China Petroleum & Chemical Corporation, Beijing, 100728, China
| | - Jinming Hu
- Hefei National Research Center for Physical Sciences at the Microscale, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, 230026, China
| | - Bin Zheng
- School of Chemistry and Pharmaceutical Engineering, Hefei Normal University, Hefei, Anhui, 230061, China
| | - Kewu He
- Imaging Center of the Third Affiliated Hospital of Anhui Medical University, Hefei, 230031, Anhui, China
| |
Collapse
|
15
|
K Jang L, T Ahlquist J, Ye C, Trujillo J, Triplett M, L Moya M, Robertson C, Hynes W, M Wasson E. Rapid curing dynamics of PEG-thiol-ene resins allow facile 3D bioprinting and in-air cell-laden microgel fabrication. Biomed Mater 2024; 20:015009. [PMID: 39584565 DOI: 10.1088/1748-605x/ad8540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 10/09/2024] [Indexed: 11/26/2024]
Abstract
Thiol-norbornene photoclick hydrogels are highly efficient in tissue engineering applications due to their fast gelation, cytocompatibility, and tunability. In this work, we utilized the advantageous features of polyethylene glycol (PEG)-thiol-ene resins to enable fabrication of complex and heterogeneous tissue scaffolds using 3D bioprinting and in-air drop encapsulation techniques. We demonstrated that photoclickable PEG-thiol-ene resins could be tuned by varying the ratio of PEG-dithiol to PEG norbornene to generate a wide range of mechanical stiffness (0.5-12 kPa) and swelling ratios. Importantly, all formulations maintained a constant, rapid gelation time (<0.5 s). We used this resin in biological projection microstereolithography (BioPµSL) to print complex structures with geometric fidelity and demonstrated biocompatibility by printing cell-laden microgrids. Moreover, the rapid gelling kinetics of this resin permitted high-throughput fabrication of tunable, cell-laden microgels in air using a biological in-air drop encapsulation apparatus (BioIDEA). We demonstrated that these microgels could support cell viability and be assembled into a gradient structure. This PEG-thiol-ene resin, along with BioPµSL and BioIDEA technology, will allow rapid fabrication of complex and heterogeneous tissues that mimic native tissues with cellular and mechanical gradients. The engineered tissue scaffolds with a controlled microscale porosity could be utilized in applications including gradient tissue engineering, biosensing, andin vitrotissue models.
Collapse
Affiliation(s)
- Lindy K Jang
- Materials Engineering Division, Lawrence Livermore National Laboratory, Livermore, CA, United States of America
| | - Jesse T Ahlquist
- Materials Engineering Division, Lawrence Livermore National Laboratory, Livermore, CA, United States of America
| | - Congwang Ye
- Materials Engineering Division, Lawrence Livermore National Laboratory, Livermore, CA, United States of America
| | - Juliana Trujillo
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, United States of America
| | - Michael Triplett
- Materials Engineering Division, Lawrence Livermore National Laboratory, Livermore, CA, United States of America
| | - Monica L Moya
- Materials Engineering Division, Lawrence Livermore National Laboratory, Livermore, CA, United States of America
| | - Claire Robertson
- Materials Engineering Division, Lawrence Livermore National Laboratory, Livermore, CA, United States of America
- UC Davis Comprehensive Cancer Center, Davis, CA, United States of America
| | - William Hynes
- Materials Engineering Division, Lawrence Livermore National Laboratory, Livermore, CA, United States of America
| | - Elisa M Wasson
- Materials Engineering Division, Lawrence Livermore National Laboratory, Livermore, CA, United States of America
- Bioscience Division, Los Alamos National Laboratory, Los Alamos, NM, United States of America
| |
Collapse
|
16
|
Hwang HS, Lee CS. Exosome-Integrated Hydrogels for Bone Tissue Engineering. Gels 2024; 10:762. [PMID: 39727520 DOI: 10.3390/gels10120762] [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: 10/31/2024] [Revised: 11/21/2024] [Accepted: 11/22/2024] [Indexed: 12/28/2024] Open
Abstract
Exosome-integrated hydrogels represent a promising frontier in bone tissue engineering, leveraging the unique biological properties of exosomes to enhance the regenerative capabilities of hydrogels. Exosomes, as naturally occurring extracellular vesicles, carry a diverse array of bioactive molecules that play critical roles in intercellular communication and tissue regeneration. When combined with hydrogels, these exosomes can be spatiotemporally delivered to target sites, offering a controlled and sustained release of therapeutic agents. This review aims to provide a comprehensive overview of the recent advancements in the development, engineering, and application of exosome-integrated hydrogels for bone tissue engineering, highlighting their potential to overcome current challenges in tissue regeneration. Furthermore, the review explores the mechanistic pathways by which exosomes embedded within hydrogels facilitate bone repair, encompassing the regulation of inflammatory pathways, enhancement of angiogenic processes, and induction of osteogenic differentiation. Finally, the review addresses the existing challenges, such as scalability, reproducibility, and regulatory considerations, while also suggesting future directions for research in this rapidly evolving field. Thus, we hope this review contributes to advancing the development of next-generation biomaterials that synergistically integrate exosome and hydrogel technologies, thereby enhancing the efficacy of bone tissue regeneration.
Collapse
Affiliation(s)
- Hee Sook Hwang
- Department of Pharmaceutical Engineering, Dankook University, Cheonan 31116, Republic of Korea
| | - Chung-Sung Lee
- Department of Pharmaceutical Engineering, Soonchunhyang University, Asan 31538, Republic of Korea
| |
Collapse
|
17
|
Yang L, Yuan QY, Lou CW, Lin JH, Li TT. Recent Advances of Cellulose-Based Hydrogels Combined with Natural Colorants in Smart Food Packaging. Gels 2024; 10:755. [PMID: 39727513 DOI: 10.3390/gels10120755] [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: 10/10/2024] [Revised: 11/02/2024] [Accepted: 11/13/2024] [Indexed: 12/28/2024] Open
Abstract
Due to the frequent occurrence of food safety problems in recent years, healthy diets are gradually receiving worldwide attention. Chemical pigments are used in smart food packaging because of their bright colors and high visibility. However, due to shortcomings such as carcinogenicity, people are gradually looking for natural pigments to be applied in the field of smart food packaging. In traditional smart food packaging, the indicator and the packaging bag substrate have different degrees of toxicity. Smart food packaging that combines natural colorants and cellulose-based hydrogels is becoming more and more popular with consumers for being natural, non-toxic, environmentally friendly, and renewable. This paper reviews the synthesis methods and characteristics of cellulose-based hydrogels, as well as the common types and characteristics of natural pigments, and discusses the application of natural colorants and cellulose-based hydrogels in food packaging, demonstrating their great potential in smart food packaging.
Collapse
Affiliation(s)
- Lan Yang
- School of Textile Science and Engineering, Tiangong University, Tianjin 300387, China
| | - Qian-Yu Yuan
- School of Textile Science and Engineering, Tiangong University, Tianjin 300387, China
| | - Ching-Wen Lou
- School of Textile Science and Engineering, Tiangong University, Tianjin 300387, China
- Fujian Key Laboratory of Novel Functional Textile Fibers and Materials, Minjiang University, Fuzhou 350108, China
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung City 413305, Taiwan
- Advanced Medical Care and Protection Technology Research Center, College of Textile and Clothing, Qingdao University, Qingdao 266071, China
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung City 404333, Taiwan
| | - Jia-Horng Lin
- School of Textile Science and Engineering, Tiangong University, Tianjin 300387, China
- College of Material and Chemical Engineering, Minjiang University, Fuzhou 350108, China
- Advanced Medical Care and Protection Technology Research Center, Department of Fiber and Composite Materials, Feng Chia University, Taichung City 407102, Taiwan
- School of Chinese Medicine, China Medical University, Taichung City 404333, Taiwan
| | - Ting-Ting Li
- School of Textile Science and Engineering, Tiangong University, Tianjin 300387, China
- Tianjin and Ministry of Education Key Laboratory for Advanced Textile Composite Materials, Tiangong University, Tianjin 300387, China
| |
Collapse
|
18
|
Rector Iv JA, McBride L, Weber CM, Grossman K, Sorets A, Ventura-Antunes L, Holtz I, Young K, Schrag M, Lippmann ES, Bellan LM. Fabrication of endothelialized capillary-like microchannel networks using sacrificial thermoresponsive microfibers. Biofabrication 2024; 17:015023. [PMID: 39401530 PMCID: PMC11575475 DOI: 10.1088/1758-5090/ad867d] [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: 02/22/2024] [Revised: 07/28/2024] [Accepted: 10/14/2024] [Indexed: 11/20/2024]
Abstract
In the body, capillary beds fulfill the metabolic needs of cells by acting as the sites of diffusive transport for vital gasses and nutrients. In artificial tissues, replicating the scale and complexity of capillaries has proved challenging, especially in a three-dimensional context. In order to better develop thick artificial tissues, it will be necessary to recreate both the form and function of capillaries. Here we demonstrate a top-down method of patterning hydrogels using sacrificial templates formed from thermoresponsive microfibers whose size and architecture approach those of natural capillaries. Within the resulting microchannels, we cultured endothelial monolayers that remain viable for over three weeks and exhibited functional barrier properties. Additionally, we cultured endothelialized microchannels within hydrogels containing fibroblasts and characterized the viability of the co-cultures to demonstrate this approach's potential when applied to cell-laden hydrogels. This method represents a step forward in the evolution of artificial tissues and a path towards producing viable capillary-scale microvasculature for engineered organs.
Collapse
Affiliation(s)
- John A Rector Iv
- Department of Mechanical Engineering, Vanderbilt University, Nashville, TN, United States of America
| | - Lucas McBride
- Department of Mechanical Engineering, Vanderbilt University, Nashville, TN, United States of America
| | - Callie M Weber
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, United States of America
| | - Kira Grossman
- Department of Mechanical Engineering, Vanderbilt University, Nashville, TN, United States of America
| | - Alexander Sorets
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, United States of America
| | - Lissa Ventura-Antunes
- School of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States of America
| | - Isabella Holtz
- Department of Cognitive Studies, Vanderbilt University, Nashville, TN, United States of America
- Department of Medicine, Health, and Society, Vanderbilt University, Nashville, TN, United States of America
| | - Katherine Young
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, United States of America
| | - Matthew Schrag
- School of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States of America
| | - Ethan S Lippmann
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, United States of America
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, United States of America
| | - Leon M Bellan
- Department of Mechanical Engineering, Vanderbilt University, Nashville, TN, United States of America
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, United States of America
| |
Collapse
|
19
|
Zheng Y, Celik U, Vorwald C, Leach JK, Liu GY. High-Resolution Atomic Force Microscopy Investigation of Alginate Hydrogel Materials in Aqueous Media. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2024. [PMID: 39558643 DOI: 10.1021/acs.langmuir.4c03554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2024]
Abstract
Alginate hydrogels are frequently used in 3D bioprinting and tissue repair and regeneration. Establishing the structure-property-performance correlation of these materials would benefit significantly from high-resolution structural characterization in aqueous environments from the molecular level to continuum. This study overcomes technical challenges and enables high-resolution atomic force microscopy (AFM) imaging of hydrated alginate hydrogels in aqueous media. By combining a new sample preparation protocol with extremely gentle tapping mode AFM imaging, we characterized the morphology and regional mechanical properties of the hydrated alginate. Upon cross-linking, basic units of these hydrogel materials consist of egg-box dimers, which assemble into long fibrils. These fibrils congregate and pile up, forming a sponge-like structure, whose pore size and distribution depend on the cross-linking conditions. At the exterior, surface tension impacts the piling of fibrils, leading to stripe-like features. These structural features contribute to local, regional, and macroscopic mechanics. The outcome provides new insights into its structural characteristics from nanometers to tens of micrometers, i.e., at the dimensions pertaining to biomaterial and hydrogel-cell interactions. Collectively, the results advance our knowledge of the structure and mechanics from the nanometer to continuum, facilitating advanced applications in hydrogel biomaterials.
Collapse
Affiliation(s)
- Yunbo Zheng
- Department of Chemistry, University of California, Davis, California 95616, United States
| | - Umit Celik
- Department of Chemistry, University of California, Davis, California 95616, United States
| | - Charlotte Vorwald
- Department of Biomedical Engineering, University of California, Davis, California 95616, United States
| | - J Kent Leach
- Department of Biomedical Engineering, University of California, Davis, California 95616, United States
- Department of Orthopaedic Surgery, UC Davis Health, Sacramento, California 95817, United States
| | - Gang-Yu Liu
- Department of Chemistry, University of California, Davis, California 95616, United States
| |
Collapse
|
20
|
Hughes SM, Aykanat A, Pierini NG, Paiva WA, Weeks AA, Edwards AS, Durant OC, Oldenhuis NJ. DNA-Intercalating Supramolecular Hydrogels for Tunable Thermal and Viscoelastic Properties. Angew Chem Int Ed Engl 2024; 63:e202411115. [PMID: 39102520 DOI: 10.1002/anie.202411115] [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: 06/12/2024] [Revised: 07/23/2024] [Accepted: 08/05/2024] [Indexed: 08/07/2024]
Abstract
Polymeric supramolecular hydrogels (PSHs) leverage the thermodynamic and kinetic properties of non-covalent interactions between polymer chains to govern their structural characteristics. As these materials are formed via endothermic or exothermic equilibria, their thermal response is challenging to control without drastically changing the nature of the chemistry used to join them. In this study, we introduce a novel class of PSHs utilizing the intercalation of double-stranded DNA (dsDNA) as the primary dynamic non-covalent interaction. The resulting dsDNA intercalating supramolecular hydrogels (DISHs) can be tuned to exhibit both endothermically or exothermically driven binding through strategic selection of intercalators. Bifunctional polyethylene glycol (MW~2000 Da) capped with intercalators of varying hydrophobicity, charge, and size (acridine, psoralen, thiazole orange, and phenanthridine) produced DISHs with comparable moduli (500-1000 Pa), but unique thermal viscoelastic responses. Notably, acridine-based cross-linkers displayed invariant and even increasing relaxation times with temperature, suggesting an endothermic binding mechanism. This methodology expands the set of structure-properties available to biomass-derived DNA biomaterials and promises a new material system where a broad set of thermal and viscoelastic responses can be obtained due to the sheer number and variety of intercalating molecules.
Collapse
Affiliation(s)
- Shaina M Hughes
- Department of Chemistry, College of Engineering and Physical Science, University of New Hampshire, 23 Academic Way, Parsons Hall, Durham, NH 03824, United States of America
| | - Aylin Aykanat
- Department of Chemistry, College of Engineering and Physical Science, University of New Hampshire, 23 Academic Way, Parsons Hall, Durham, NH 03824, United States of America
| | - Nicholas G Pierini
- Department of Chemistry, College of Engineering and Physical Science, University of New Hampshire, 23 Academic Way, Parsons Hall, Durham, NH 03824, United States of America
| | - Wynter A Paiva
- Department of Chemistry, College of Engineering and Physical Science, University of New Hampshire, 23 Academic Way, Parsons Hall, Durham, NH 03824, United States of America
| | - April A Weeks
- Department of Chemistry, College of Engineering and Physical Science, University of New Hampshire, 23 Academic Way, Parsons Hall, Durham, NH 03824, United States of America
| | - Austin S Edwards
- Department of Chemistry, College of Engineering and Physical Science, University of New Hampshire, 23 Academic Way, Parsons Hall, Durham, NH 03824, United States of America
| | - Owen C Durant
- Department of Chemistry, College of Engineering and Physical Science, University of New Hampshire, 23 Academic Way, Parsons Hall, Durham, NH 03824, United States of America
| | - Nathan J Oldenhuis
- Department of Chemistry, College of Engineering and Physical Science, University of New Hampshire, 23 Academic Way, Parsons Hall, Durham, NH 03824, United States of America
| |
Collapse
|
21
|
Liu Y, Gilchrist AE, Heilshorn SC. Engineered Protein Hydrogels as Biomimetic Cellular Scaffolds. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2407794. [PMID: 39233559 PMCID: PMC11573243 DOI: 10.1002/adma.202407794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 08/01/2024] [Indexed: 09/06/2024]
Abstract
The biochemical and biophysical properties of the extracellular matrix (ECM) play a pivotal role in regulating cellular behaviors such as proliferation, migration, and differentiation. Engineered protein-based hydrogels, with highly tunable multifunctional properties, have the potential to replicate key features of the native ECM. Formed by self-assembly or crosslinking, engineered protein-based hydrogels can induce a range of cell behaviors through bioactive and functional domains incorporated into the polymer backbone. Using recombinant techniques, the amino acid sequence of the protein backbone can be designed with precise control over the chain-length, folded structure, and cell-interaction sites. In this review, the modular design of engineered protein-based hydrogels from both a molecular- and network-level perspective are discussed, and summarize recent progress and case studies to highlight the diverse strategies used to construct biomimetic scaffolds. This review focuses on amino acid sequences that form structural blocks, bioactive blocks, and stimuli-responsive blocks designed into the protein backbone for highly precise and tunable control of scaffold properties. Both physical and chemical methods to stabilize dynamic protein networks with defined structure and bioactivity for cell culture applications are discussed. Finally, a discussion of future directions of engineered protein-based hydrogels as biomimetic cellular scaffolds is concluded.
Collapse
Affiliation(s)
- Yueming Liu
- Department of Materials Science & Engineering, Stanford University, Stanford, CA, 94305, USA
| | - Aidan E Gilchrist
- Department of Biomedical Engineering, University of California, Davis 451 Health Sciences Dr, GBSF 3315, Davis, CA, 95616, USA
| | - Sarah C Heilshorn
- Department of Materials Science & Engineering, 476 Lomita Mall, McCullough Room 246, Stanford, CA, 94305, USA
| |
Collapse
|
22
|
Lee YJ, Lee E, Kim SE, Shin H, Huh KM. Synthesis and characterization of methacryl glycol chitosan as a novel functionally advanced thermogel for biomedical applications. Int J Biol Macromol 2024; 280:135858. [PMID: 39307499 DOI: 10.1016/j.ijbiomac.2024.135858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 08/31/2024] [Accepted: 09/19/2024] [Indexed: 11/20/2024]
Abstract
Thermo-responsive hydrogels (thermogels), known for their sol-gel transition capabilities, have garnered significant interest for biomedical applications over recent decades. However, conventional thermogels are hindered by intrinsic physicochemical and functional limitations that impede their broader utility. This study introduces methacryl glycol chitosan (MGC) as a novel thermogel, offering enhanced functionality and addressing these limitations. MGCs, synthesized through N-methacrylation of glycol chitosan, exhibit tunable thermogelling and photo-crosslinking behaviors. The thermo-reversible sol-gel transition of MGCs occurs within a 21-54 °C range, adjustable by polymer concentration and methacryl substitution degree. Photo-crosslinking using UV light further enhances the mechanical properties of MGC thermogels, creating thermo-irreversible, chemically crosslinked hydrogels. MGCs show no cytotoxic effects and effectively support cell encapsulation. In vivo studies demonstrate stable crosslinking with minimal UV-induced skin damage. Due to their unique thermo-sensitivity, multi-functionality, and customizable properties, MGC thermogels are promising novel biomaterials for various biomedical applications, particularly injectable tissue engineering and cell encapsulation, thus overcoming the limitations of conventional thermogels.
Collapse
Affiliation(s)
- Young Ju Lee
- Department of Polymer Science and Engineering, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Eunjin Lee
- Department of Bioengineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul 04763, Republic of Korea; BK21 FOUR, Education and Research Group for Biopharmaceutical Innovation Leader, Hanyang University, Seoul 04763, Republic of Korea
| | - Seong Eun Kim
- Department of Materials Science and Engineering, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Heungsoo Shin
- Department of Bioengineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul 04763, Republic of Korea; BK21 FOUR, Education and Research Group for Biopharmaceutical Innovation Leader, Hanyang University, Seoul 04763, Republic of Korea; Institute of Nano Science and Technology, Hanyang University, Seoul 04763, Republic of Korea.
| | - Kang Moo Huh
- Department of Polymer Science and Engineering, Chungnam National University, Daejeon 34134, Republic of Korea; Department of Materials Science and Engineering, Chungnam National University, Daejeon 34134, Republic of Korea.
| |
Collapse
|
23
|
Maiti S, Maji B, Badwaik H, Pandey MM, Lakra P, Yadav H. Oxidized ionic polysaccharide hydrogels: Review on derived scaffolds characteristics and tissue engineering applications. Int J Biol Macromol 2024; 280:136089. [PMID: 39357721 DOI: 10.1016/j.ijbiomac.2024.136089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 09/11/2024] [Accepted: 09/25/2024] [Indexed: 10/04/2024]
Abstract
Polysaccharide-based hydrogels have gained prominence due to their non-toxicity, biocompatibility, and structural adaptability for constructing tissue engineering scaffolds. Polysaccharide crosslinking is necessary for hydrogel stability in vivo. The periodate oxidation enables the modification of native polysaccharide characteristics for wound healing and tissue engineering applications. It produces dialdehydes, which are used to crosslink biocompatible amine-containing macromolecules such as chitosan, gelatin, adipic acid dihydrazide, silk fibroin, and peptides via imine/hydrazone linkages. Crosslinked oxidized ionic polysaccharide hydrogels have been studied for wound healing, cardiac and liver tissue engineering, bone, cartilage, corneal tissue regeneration, abdominal wall repair, nucleus pulposus regeneration, and osteoarthritis. Several modified hydrogel systems have been synthesized using antibiotics and inorganic substances to improve porosity, mechanical and viscoelastic properties, desired swelling propensity, and antibacterial efficacy. Thus, the injectable hydrogels provide a host-tissue-mimetic environment with high cell adhesion and viability, making them appropriate for scarless wound healing and tissue engineering applications. This review describes the oxidation procedure for alginate, hyaluronic acid, gellan gum, pectin, xanthan gum and chitosan, as well as the characteristics of the resulting materials. Furthermore, a critical review of scientific advances in wound healing and tissue engineering applications has been provided.
Collapse
Affiliation(s)
- Sabyasachi Maiti
- Department of Pharmacy, Indira Gandhi National Tribal University, Amarkantak, Madhya Pradesh, India.
| | - Biswajit Maji
- Department of Chemistry, Indira Gandhi National Tribal University, Amarkantak, Madhya Pradesh, India
| | - Hemant Badwaik
- Department of Pharmaceutical Chemistry, Shri Shankaracharya Institute of Pharmaceutical Sciences and Research, Junwani, Bhilai, Chhattisgarh, India
| | - Murali Monohar Pandey
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani, Rajasthan, India
| | - Preeti Lakra
- Department of Pharmacy, Indira Gandhi National Tribal University, Amarkantak, Madhya Pradesh, India
| | - Harsh Yadav
- Department of Pharmacy, Indira Gandhi National Tribal University, Amarkantak, Madhya Pradesh, India
| |
Collapse
|
24
|
Zhang W, Rao Y, Wong SH, Wu Y, Zhang Y, Yang R, Tsui SKW, Ker DFE, Mao C, Frith JE, Cao Q, Tuan RS, Wang DM. Transcriptome-Optimized Hydrogel Design of a Stem Cell Niche for Enhanced Tendon Regeneration. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024:e2313722. [PMID: 39417770 DOI: 10.1002/adma.202313722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 09/04/2024] [Indexed: 10/19/2024]
Abstract
Bioactive hydrogels have emerged as promising artificial niches for enhancing stem cell-mediated tendon repair. However, a substantial knowledge gap remains regarding the optimal combination of niche features for targeted cellular responses, which often leads to lengthy development cycles and uncontrolled healing outcomes. To address this critical gap, an innovative, data-driven materiomics strategy is developed. This approach is based on in-house RNA-seq data that integrates bioinformatics and mathematical modeling, which is a significant departure from traditional trial-and-error methods. It aims to provide both mechanistic insights and quantitative assessments and predictions of the tenogenic effects of adipose-derived stem cells induced by systematically modulated features of a tendon-mimetic hydrogel (TenoGel). The knowledge generated has enabled a rational approach for TenoGel design, addressing key considerations, such as tendon extracellular matrix concentration, uniaxial tensile loading, and in vitro pre-conditioning duration. Remarkably, our optimized TenoGel demonstrated robust tenogenesis in vitro and facilitated tendon regeneration while preventing undesired ectopic ossification in a rat tendon injury model. These findings shed light on the importance of tailoring hydrogel features for efficient tendon repair. They also highlight the tremendous potential of the innovative materiomics strategy as a powerful predictive and assessment tool in biomaterial development for regenerative medicine.
Collapse
Affiliation(s)
- Wanqi Zhang
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Ying Rao
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Shing Hei Wong
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- Hong Kong Bioinformatics Centre, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Yeung Wu
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Yuanhao Zhang
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Rui Yang
- Department of Sports Medicine, Orthopedics, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Stephen Kwok-Wing Tsui
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- Hong Kong Bioinformatics Centre, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Dai Fei Elmer Ker
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong SAR, China
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Hong Kong SAR, China
- Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, 999077, China
| | - Chuanbin Mao
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Jessica E Frith
- Materials Science and Engineering, Monash University, Clayton, 3800, VIC, Australia
- Australian Regenerative Medicine Institute, Monash University, Clayton, 3800, VIC, Australia
- Australian Research Council Training Centre for Cell and Tissue Engineering Technologies, Monash University, Clayton, 3800, VIC, Australia
| | - Qin Cao
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- Hong Kong Bioinformatics Centre, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Rocky S Tuan
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Hong Kong SAR, China
- Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, 999077, China
| | - Dan Michelle Wang
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Hong Kong SAR, China
- Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, 999077, China
| |
Collapse
|
25
|
Wu KY, Qian SY, Faucher A, Tran SD. Advancements in Hydrogels for Corneal Healing and Tissue Engineering. Gels 2024; 10:662. [PMID: 39451315 PMCID: PMC11507397 DOI: 10.3390/gels10100662] [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: 08/31/2024] [Revised: 10/08/2024] [Accepted: 10/14/2024] [Indexed: 10/26/2024] Open
Abstract
Hydrogels have garnered significant attention for their versatile applications across various fields, including biomedical engineering. This review delves into the fundamentals of hydrogels, exploring their definition, properties, and classification. Hydrogels, as three-dimensional networks of crosslinked polymers, possess tunable properties such as biocompatibility, mechanical strength, and hydrophilicity, making them ideal for medical applications. Uniquely, this article offers original insights into the application of hydrogels specifically for corneal tissue engineering, bridging a gap in current research. The review further examines the anatomical and functional complexities of the cornea, highlighting the challenges associated with corneal pathologies and the current reliance on donor corneas for transplantation. Considering the global shortage of donor corneas, this review discusses the potential of hydrogel-based materials in corneal tissue engineering. Emphasis is placed on the synthesis processes, including physical and chemical crosslinking, and the integration of bioactive molecules. Stimuli-responsive hydrogels, which react to environmental triggers, are identified as promising tools for drug delivery and tissue repair. Additionally, clinical applications of hydrogels in corneal pathologies are explored, showcasing their efficacy in various trials. Finally, the review addresses the challenges of regulatory approval and the need for further research to fully realize the potential of hydrogels in corneal tissue engineering, offering a promising outlook for future developments in this field.
Collapse
Affiliation(s)
- Kevin Y. Wu
- Department of Surgery, Division of Ophthalmology, University of Sherbrooke, Sherbrooke, QC J1G 2E8, Canada; (K.Y.W.)
| | - Shu Yu Qian
- Faculty of Medicine, University of Sherbrooke, Sherbrooke, QC J1G 2E8, Canada
| | - Anne Faucher
- Department of Surgery, Division of Ophthalmology, University of Sherbrooke, Sherbrooke, QC J1G 2E8, Canada; (K.Y.W.)
| | - Simon D. Tran
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC H3A 1G1, Canada
| |
Collapse
|
26
|
Zimmermann J, Farooqi AR, van Rienen U. Electrical stimulation for cartilage tissue engineering - A critical review from an engineer's perspective. Heliyon 2024; 10:e38112. [PMID: 39416819 PMCID: PMC11481755 DOI: 10.1016/j.heliyon.2024.e38112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 08/31/2024] [Accepted: 09/18/2024] [Indexed: 10/19/2024] Open
Abstract
Cartilage has a limited intrinsic healing capacity. Hence, cartilage degradation and lesions pose a huge clinical challenge, particularly in an ageing society. Osteoarthritis impacts a significant number of the population and requires the development of repair and tissue engineering methods for hyaline articular cartilage. In this context, electrical stimulation has been investigated for more than 50 years already. Yet, no well-established clinical therapy to treat osteoarthritis by means of electrical stimulation exists. We argue that one reason is the lack of replicability of electrical stimulation devices from a technical perspective together with lacking hypotheses of the biophysical mechanism. Hence, first, the electrical stimulation studies reported in the context of cartilage tissue engineering with a special focus on technical details are summarized. Then, an experimental and numerical approach is discussed to make the electrical stimulation experiments replicable. Finally, biophysical hypotheses have been reviewed on the interaction of electric fields and cells that are relevant for cartilage tissue engineering. With that, the aim is to inspire future research to enable clinical electrical stimulation therapies to fight osteoarthritis.
Collapse
Affiliation(s)
- Julius Zimmermann
- Institute of General Electrical Engineering, University of Rostock, 18051 Rostock, Germany
| | - Abdul Razzaq Farooqi
- Institute of General Electrical Engineering, University of Rostock, 18051 Rostock, Germany
- Department of Electronic Engineering, Faculty of Engineering, The Islamia University of Bahawalpur, 63100 Bahawalpur, Pakistan
| | - Ursula van Rienen
- Institute of General Electrical Engineering, University of Rostock, 18051 Rostock, Germany
- Department of Ageing of Individuals and Society, Interdisciplinary Faculty, University of Rostock, 18051 Rostock, Germany
- Department Life, Light & Matter, University of Rostock, 18051 Rostock, Germany
| |
Collapse
|
27
|
Gao S, Liu Z, Zeng W, Zhang Y, Zhang F, Wu D, Wang Y. Biocompatible Hydrogel Coating on Silicone Rubber with Improved Antifouling and Durable Lubricious Properties. Gels 2024; 10:647. [PMID: 39451300 PMCID: PMC11507538 DOI: 10.3390/gels10100647] [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: 09/11/2024] [Revised: 10/03/2024] [Accepted: 10/08/2024] [Indexed: 10/26/2024] Open
Abstract
Silicone rubber is widely used in various medical applications. However, silicone rubber is prone to biofouling due to their affinity for lipids and has a high friction coefficient, which can significantly impact their efficacy and performance used as medical devices. Thus, the development of hydrogels with antifouling and lubricious abilities for the modification of silicone rubber is in high demand. We herein prepared a variety of hydrogel coatings mainly based on polyvinylpyrrolidone (PVP) and poly (ethylene glycol) diacrylate (PEGDA). We modified the silicone rubber using the prepared hydrogel coatings and cured it using a heating method. Then, we characterized its surface and evaluated the antifouling property, lubricious property, cytotoxicity, sensitization, and vaginal irritation. The results of water contact angle (WCA), protein adsorption, and friction coefficient indicated the success of the modification of the silicone rubber, leading to a significant decrease in the corresponding test values. Meanwhile, the results of cytotoxicity, sensitization, and vaginal irritation tests showed that the hydrogel coating-modified silicone rubbers have an excellent biocompatibility. This study describes how the silicone rubber could be modified with a biocompatible hydrogel coating. The hydrogel coating-modified silicone rubbers have improved antifouling and durable lubricious properties.
Collapse
Affiliation(s)
- Shuai Gao
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610065, China; (S.G.); (F.Z.)
- Chengdu Daxan Innovative Medical Tech. Co., Ltd., Chengdu 611137, China; (Z.L.); (W.Z.); (Y.Z.)
| | - Zheng Liu
- Chengdu Daxan Innovative Medical Tech. Co., Ltd., Chengdu 611137, China; (Z.L.); (W.Z.); (Y.Z.)
| | - Wei Zeng
- Chengdu Daxan Innovative Medical Tech. Co., Ltd., Chengdu 611137, China; (Z.L.); (W.Z.); (Y.Z.)
| | - Yunfeng Zhang
- Chengdu Daxan Innovative Medical Tech. Co., Ltd., Chengdu 611137, China; (Z.L.); (W.Z.); (Y.Z.)
| | - Fanjun Zhang
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610065, China; (S.G.); (F.Z.)
| | - Dimeng Wu
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610065, China; (S.G.); (F.Z.)
| | - Yunbing Wang
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610065, China; (S.G.); (F.Z.)
| |
Collapse
|
28
|
Begum B, Koduru TS, Madni SN, Fathima Anjum N, Seetharaman S, Veeranna B, Gupta VK. Dual-Self-Crosslinking Effect of Alginate-Di-Aldehyde with Natural and Synthetic Co-Polymers as Injectable In Situ-Forming Biodegradable Hydrogel. Gels 2024; 10:649. [PMID: 39451302 PMCID: PMC11507163 DOI: 10.3390/gels10100649] [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: 08/05/2024] [Revised: 09/29/2024] [Accepted: 10/02/2024] [Indexed: 10/26/2024] Open
Abstract
Injectable, in situ-forming hydrogels, both biocompatible and biodegradable, have garnered significant attention in tissue engineering due to their potential for creating adaptable scaffolds. The adaptability of these hydrogels, made from natural proteins and polysaccharides, opens up a world of possibilities. In this study, sodium alginate was used to synthesize alginate di-aldehyde (ADA) through periodate oxidation, resulting in a lower molecular weight and reduced viscosity, with different degrees of oxidation (54% and 70%). The dual-crosslinking mechanism produced an injectable in situ hydrogel. Initially, physical crosslinking occurred between ADA and borax via borax complexation, followed by chemical crosslinking with gelatin through a Schiff's base reaction, which takes place between the amino groups of gelatin and the aldehyde groups of ADA, without requiring an external crosslinking agent. The formation of Schiff's base was confirmed by Fourier-transform infrared (FT-IR) spectroscopy. At the same time, the aldehyde groups in ADA were characterized using FT-IR, proton nuclear magnetic resonance (¹H NMR), and gel permeation chromatography (GPC), which determined its molecular weight. Furthermore, borax complexation was validated through boron-11 nuclear magnetic resonance (¹¹B NMR). The hydrogel formulation containing 70% ADA, polyethylene glycol (PEG), and 9% gelatin exhibited a decreased gelation time at physiological temperature, attributed to the increased gelatin content and higher degree of oxidation. Rheological analysis mirrored these findings, showing a correlation with gelation time. The swelling capacity was also enhanced due to the increased oxidation degree of PEG and the system's elevated gelatin content and hydrophilicity. The hydrogel demonstrated an average pore size of 40-60 µm and a compressive strength of 376.80 kPa. The lower molecular weight and varied pH conditions influenced its degradation behavior. Notably, the hydrogel's syringeability was deemed sufficient for practical applications, further enhancing its potential in tissue engineering. Given these properties, the 70% ADA/gelatin/PEG hydrogel is a promising candidate and a potential game-changer for injectable, self-crosslinking applications in tissue engineering. Its potential to revolutionize the field is inspiring and should motivate further exploration.
Collapse
Affiliation(s)
- Bushra Begum
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Sri Shivarathreeshwara Nagar, Mysuru 570015, India; (B.B.); (T.S.K.); (B.V.)
- Department of Pharmaceutics, Farooqia College of Pharmacy, Mysuru 570019, India;
| | - Trideva Sastri Koduru
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Sri Shivarathreeshwara Nagar, Mysuru 570015, India; (B.B.); (T.S.K.); (B.V.)
| | - Syeda Noor Madni
- Department of Pharmaceutics, Farooqia College of Pharmacy, Mysuru 570019, India;
| | - Noor Fathima Anjum
- Department of Pharmaceutical Chemistry, Farooqia College of Pharmacy, Mysuru 570015, India;
| | | | - Balamuralidhara Veeranna
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Sri Shivarathreeshwara Nagar, Mysuru 570015, India; (B.B.); (T.S.K.); (B.V.)
| | - Vishal Kumar Gupta
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Sri Shivarathreeshwara Nagar, Mysuru 570015, India; (B.B.); (T.S.K.); (B.V.)
| |
Collapse
|
29
|
Faheem S, Hameed H, Paiva-Santos AC, Khan MA, Ghumman SA, Hameed A. The role of chondroitin sulphate as a potential biomaterial for hepatic tissue regeneration: A comprehensive review. Int J Biol Macromol 2024; 280:136332. [PMID: 39482129 DOI: 10.1016/j.ijbiomac.2024.136332] [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: 04/05/2024] [Revised: 08/26/2024] [Accepted: 10/03/2024] [Indexed: 11/03/2024]
Abstract
Chondroitin sulphate is an anionic hetero-polysaccharide, having numerous structural affinities for building the bio-active components. In addition to biodegradable/biocompatible activities, chondroitin sulphate also possesses anti-coagulant/anti-thrombogenic, anti-inflammatory, anti-oxidant as well as anti-tumor activities. Chondroitin sulphate has an inherited affinity for glycosylation enzymes and receptors, which are overexpressed over degenerated cells and organelles. Because of this affinity, chondroitin sulphate is nominated as an active cellular/subcellular targeted biological macromolecule to assist in site-specific delivery. Chondroitin sulphate is mainly considered a promising biomaterial for drug targeting and tissue engineering due to its specific physicochemical, mechanical, bio-degradation, and biological characteristics. In this review, the fundamental applications of chondroitin sulphate in hepatic tissue engineering are discussed. Chondroitin sulphate along with mesenchymal stem cells (MSCs) based scaffold and hydrogels for biopharmaceuticals' delivery in hepatic tissue engineering are critically discussed. In addition, the manuscript also describes leading features and markers involved in hepatic damage, and the potential role of chondroitin sulphate-based delivery systems in hepatic tissue engineering.
Collapse
Affiliation(s)
- Saleha Faheem
- Faculty of Pharmaceutical Sciences, University of Central Punjab, Lahore 54000, Pakistan.
| | - Huma Hameed
- Faculty of Pharmaceutical Sciences, University of Central Punjab, Lahore 54000, Pakistan.
| | - Ana Cláudia Paiva-Santos
- Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, 3000-548 Coimbra, Portugal; REQUIMTE/LAQV, Group of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, 3000-548 Coimbra, Portugal.
| | - Mahtab Ahmad Khan
- Faculty of Pharmaceutical Sciences, University of Central Punjab, Lahore 54000, Pakistan.
| | | | - Anam Hameed
- Department of Human Nutrition and Dietetics, Faculty of Rehabilitation and Allied Health Sciences, Riphah International University, Gulberg III, Lahore 54000, Pakistan.
| |
Collapse
|
30
|
Kim J, Choi YJ, Gal CW, Sung A, Utami SS, Park H, Yun HS. Enhanced Osteogenesis in 2D and 3D Culture Systems Using RGD Peptide and α-TCP Phase Transition within Alginate-Based Hydrogel. Macromol Biosci 2024; 24:e2400190. [PMID: 39116430 DOI: 10.1002/mabi.202400190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 07/21/2024] [Indexed: 08/10/2024]
Abstract
Cell-laden hydrogels have been extensively investigated in various tissue engineering fields by their potential capacity to deposit numerous types of cells in a specific area. They are largely used in soft-tissue engineering applications because of their low mechanical strength. In addition, sodium alginate is well-known for its encapsulation, loading capacity and for being easily controllable; however, it lacks cell-binding ligands and hence the ability to adhere cells. In this study, it is aimed to enhance osteogenesis in cells encapsulated in alginate and improve its mechanical properties by introducing a synthetic peptide and calcium phosphate phase transition. To increase cell-hydrogel interactions and increasing cell viability, an RGD peptide is added to a photocrosslinkable methacrylate-modified alginate, and alpha-tricalcium phosphate (α-TCP) is added to the hydrogel to increase its mechanical strength via phase transition. Cell proliferation, growth, and differentiation are assessed in both 2D and 3D cell cultures. The addition of α-TCP significantly improved the mechanical properties of the hydrogel. Moreover, the RGD peptide and α-TCP showed a synergistic effect with significantly improved cell adhesion and osteogenesis in both 2D and 3D cell cultures. Therefore, the functional hydrogel developed in this study can potentially be used for bone tissue regeneration.
Collapse
Affiliation(s)
- Jueun Kim
- Advanced Bio and Healthcare Materials Research Division, Korea Institute of Materials Science, 797 Changwon-daero, Seongasna-gu, Changwon, South Korea
| | - Yeong-Jin Choi
- Advanced Bio and Healthcare Materials Research Division, Korea Institute of Materials Science, 797 Changwon-daero, Seongasna-gu, Changwon, South Korea
| | - Chang-Woo Gal
- Advanced Bio and Healthcare Materials Research Division, Korea Institute of Materials Science, 797 Changwon-daero, Seongasna-gu, Changwon, South Korea
| | - Aram Sung
- Advanced Bio and Healthcare Materials Research Division, Korea Institute of Materials Science, 797 Changwon-daero, Seongasna-gu, Changwon, South Korea
| | - Siwi Setya Utami
- Advanced Bio and Healthcare Materials Research Division, Korea Institute of Materials Science, 797 Changwon-daero, Seongasna-gu, Changwon, South Korea
- Department of Advanced Materials Engineering, University of Science and Technology, 217 Gajeon-ro, Yeseong-gu, Daejeon, Republic of Korea
| | - Honghyun Park
- Advanced Bio and Healthcare Materials Research Division, Korea Institute of Materials Science, 797 Changwon-daero, Seongasna-gu, Changwon, South Korea
| | - Hui-Suk Yun
- Advanced Bio and Healthcare Materials Research Division, Korea Institute of Materials Science, 797 Changwon-daero, Seongasna-gu, Changwon, South Korea
- Department of Advanced Materials Engineering, University of Science and Technology, 217 Gajeon-ro, Yeseong-gu, Daejeon, Republic of Korea
| |
Collapse
|
31
|
Li N, He J. Hydrogel-based therapeutic strategies for spinal cord injury repair: Recent advances and future prospects. Int J Biol Macromol 2024; 277:134591. [PMID: 39127289 DOI: 10.1016/j.ijbiomac.2024.134591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 08/06/2024] [Accepted: 08/06/2024] [Indexed: 08/12/2024]
Abstract
Spinal cord injury (SCI) is a debilitating condition that can result in significant functional impairment and loss of quality of life. There is a growing interest in developing new therapies for SCI, and hydrogel-based multimodal therapeutic strategies have emerged as a promising approach. They offer several advantages for SCI repair, including biocompatibility, tunable mechanical properties, low immunogenicity, and the ability to deliver therapeutic agents. This article provides an overview of the recent advances in hydrogel-based therapy strategies for SCI repair, particularly within the past three years. We summarize the SCI hydrogels with varied characteristics such as phase-change hydrogels, self-healing hydrogel, oriented fibers hydrogel, and self-assembled microspheres hydrogel, as well as different functional hydrogels such as conductive hydrogels, stimuli-responsive hydrogels, adhesive hydrogel, antioxidant hydrogel, sustained-release hydrogel, etc. The composition, preparation, and therapeutic effect of these hydrogels are briefly discussed and comprehensively evaluated. In the end, the future development of hydrogels in SCI repair is prospected to inspire more researchers to invest in this promising field.
Collapse
Affiliation(s)
- Na Li
- School of Rehabilitation Sciences and Engineering, University of Health and Rehabilitation Sciences, Qingdao 266113, China
| | - Jintao He
- School of Rehabilitation Sciences and Engineering, University of Health and Rehabilitation Sciences, Qingdao 266113, China.
| |
Collapse
|
32
|
Quigley RM, Kearney M, Kennedy OD, Duncan HF. Tissue engineering approaches for dental pulp regeneration: The development of novel bioactive materials using pharmacological epigenetic inhibitors. Bioact Mater 2024; 40:182-211. [PMID: 38966600 PMCID: PMC11223092 DOI: 10.1016/j.bioactmat.2024.06.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 06/05/2024] [Accepted: 06/06/2024] [Indexed: 07/06/2024] Open
Abstract
The drive for minimally invasive endodontic treatment strategies has shifted focus from technically complex and destructive root canal treatments towards more conservative vital pulp treatment. However, novel approaches to maintaining dental pulp vitality after disease or trauma will require the development of innovative, biologically-driven regenerative medicine strategies. For example, cell-homing and cell-based therapies have recently been developed in vitro and trialled in preclinical models to study dental pulp regeneration. These approaches utilise natural and synthetic scaffolds that can deliver a range of bioactive pharmacological epigenetic modulators (HDACis, DNMTis, and ncRNAs), which are cost-effective and easily applied to stimulate pulp tissue regrowth. Unfortunately, many biological factors hinder the clinical development of regenerative therapies, including a lack of blood supply and poor infection control in the necrotic root canal system. Additional challenges include a need for clinically relevant models and manufacturing challenges such as scalability, cost concerns, and regulatory issues. This review will describe the current state of bioactive-biomaterial/scaffold-based engineering strategies to stimulate dentine-pulp regeneration, explicitly focusing on epigenetic modulators and therapeutic pharmacological inhibition. It will highlight the components of dental pulp regenerative approaches, describe their current limitations, and offer suggestions for the effective translation of novel epigenetic-laden bioactive materials for innovative therapeutics.
Collapse
Affiliation(s)
- Ross M. Quigley
- Division of Restorative Dentistry & Periodontology, Dublin Dental University Hospital, Trinity College Dublin (TCD), University of Dublin, Lincoln Place, Dublin, Ireland
- Department of Anatomy and Regenerative Medicine, and Tissue Engineering Research Group, Royal College of Surgeons in Ireland (RCSI) University of Medicine and Health Sciences, Dublin, Ireland
| | - Michaela Kearney
- Division of Restorative Dentistry & Periodontology, Dublin Dental University Hospital, Trinity College Dublin (TCD), University of Dublin, Lincoln Place, Dublin, Ireland
| | - Oran D. Kennedy
- Department of Anatomy and Regenerative Medicine, and Tissue Engineering Research Group, Royal College of Surgeons in Ireland (RCSI) University of Medicine and Health Sciences, Dublin, Ireland
- The Trinity Centre for Biomedical Engineering (TCBE) and the Advanced Materials and Bioengineering Research Centre (AMBER), Royal College of Surgeons in Ireland (RCSI) and Trinity College Dublin (TCD), Dublin, Ireland
| | - Henry F. Duncan
- Division of Restorative Dentistry & Periodontology, Dublin Dental University Hospital, Trinity College Dublin (TCD), University of Dublin, Lincoln Place, Dublin, Ireland
- The Trinity Centre for Biomedical Engineering (TCBE) and the Advanced Materials and Bioengineering Research Centre (AMBER), Royal College of Surgeons in Ireland (RCSI) and Trinity College Dublin (TCD), Dublin, Ireland
| |
Collapse
|
33
|
Feng PY, Jing X. Novel shish-kebab structured nanofibrous decorating chitosan unidirectional scaffolds to mimic extracellular matrix for tissue engineering. J Mech Behav Biomed Mater 2024; 158:106677. [PMID: 39068847 DOI: 10.1016/j.jmbbm.2024.106677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 07/11/2024] [Accepted: 07/24/2024] [Indexed: 07/30/2024]
Abstract
Electrospun nanofibrous scaffolds are renowned for their ability to mimic the microstructure of the extracellular matrix (ECM). However, they often fail to replicate the geometry of target tissues, and the biocompatibility of these scaffolds those made from synthetic polymers is always limited due to the lack of cell binding sites. To address these issues, we proposed an innovative approach that combined unidirectional freeze-drying and electrospinning. During this process, electrospun polycaprolactone (PCL) nanofibers were chopped into nanofibrils, which range in size up to several hundred micrometers, and were incorporated into the chitosan scaffolds via unidirectional freeze-drying. In these scaffolds, the chitosan phase was responsible for maintaining the structural integrity at the macroscale, while the embedded nanofibers enhanced the surface topography at the microscale. The resulting scaffolds exhibited a high porosity of 90% and an impressive water uptake capacity of 2500%. Furthermore, 3T3 fibroblast cells showed strong interactions with the scaffolds, characterized by high rates of cell proliferation and viability. The cells also displayed significant orientation along the direction of the pores, suggesting that the scaffolds effectively guided cellular growth.
Collapse
Affiliation(s)
- Pei-Yong Feng
- Key Laboratory of Advanced Packaging Materials and Technology of Hunan Province, Hunan University of Technology, Zhuzhou, Hunan, 412007, China
| | - Xin Jing
- Key Laboratory of Advanced Packaging Materials and Technology of Hunan Province, Hunan University of Technology, Zhuzhou, Hunan, 412007, China.
| |
Collapse
|
34
|
Poyraz Y, Baltacı N, Hassan G, Alayoubi O, Uysal BÖ, Pekcan Ö. Composite Hydrogel of Polyacrylamide/Starch/Gelatin as a Novel Amoxicillin Delivery System. Gels 2024; 10:625. [PMID: 39451278 PMCID: PMC11507288 DOI: 10.3390/gels10100625] [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: 08/20/2024] [Revised: 09/16/2024] [Accepted: 09/24/2024] [Indexed: 10/26/2024] Open
Abstract
This study investigates the development and characterization of a novel composite hydrogel composed of polyacrylamide (PAAm), starch, and gelatin for use as an amoxicillin delivery system. The optical properties, swelling behavior, and drug release profile of the composite hydrogel's were studied to evaluate its efficacy and potential applications. UV-visible spectroscopy was employed to determine the optical properties, revealing significant transparency in the visible range, which is essential for biomedical applications. The incorporation of starch and gelatin into the polyacrylamide matrix significantly enhanced the hydrogel's swelling capacity and biocompatibility. Studies on drug delivery demonstrated a sustained release profile of amoxicillin in simulated gastrointestinal fluids, which is essential for maintaining therapeutic levels for a prolonged amount of time. The results indicate that the composite hydrogel of PAAm/starch/gelatin has good swelling behavior, appealing optical characteristics, and a promising controlled drug release mechanism. These results point to this hydrogel's considerable potential as a drug delivery method, providing a viable path toward enhancing the medicinal effectiveness of amoxicillin and maybe other medications.
Collapse
Affiliation(s)
- Yağmur Poyraz
- Computational Sciences and Engineering, School of Graduate Studies, Kadir Has University, Cibali, Fatih, Istanbul 34083, Turkey
| | - Nisa Baltacı
- Materials Science and Nanotechnology, School of Graduate Studies, Kadir Has University, Cibali, Fatih, Istanbul 34083, Turkey
| | - Gana Hassan
- Materials Science and Nanotechnology, School of Graduate Studies, Kadir Has University, Cibali, Fatih, Istanbul 34083, Turkey
| | - Oubadah Alayoubi
- Materials Science and Nanotechnology, School of Graduate Studies, Kadir Has University, Cibali, Fatih, Istanbul 34083, Turkey
| | - Bengü Özuğur Uysal
- Faculty of Engineering and Natural Sciences, Kadir Has University, Cibali, Fatih, Istanbul 34083, Turkey;
| | - Önder Pekcan
- Faculty of Engineering and Natural Sciences, Kadir Has University, Cibali, Fatih, Istanbul 34083, Turkey;
| |
Collapse
|
35
|
Bocheng X, França R. Innovative 3D bioprinting approaches for advancing brain science and medicine: a literature review. Biomed Phys Eng Express 2024; 10:062002. [PMID: 39260389 DOI: 10.1088/2057-1976/ad795c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 09/11/2024] [Indexed: 09/13/2024]
Abstract
The rapid advancements in 3D printing technology have revolutionized the field of tissue engineering, particularly in the development of neural tissues for the treatment of nervous system diseases. Brain neural tissue, composed of neurons and glial cells, plays a crucial role in the functioning of the brain, spinal cord, and peripheral nervous system by transmitting nerve impulses and processing information. By leveraging 3D bioprinting and bioinks, researchers can create intricate neural scaffolds that facilitate the proliferation and differentiation of nerve cells, thereby promoting the repair and regeneration of damaged neural tissues. This technology allows for the precise spatial arrangement of various cell types and scaffold materials, enabling the construction of complex neural tissue models that closely mimic the natural architecture of the brain. Human-induced pluripotent stem cells (hiPSCs) have emerged as a groundbreaking tool in neuroscience research and the potential treatment of neurological diseases. These cells can differentiate into diverse cell types within the nervous system, including neurons, astrocytes, microglia, oligodendrocytes, and Schwann cells, providing a versatile platform for studying neural networks, neurodevelopment, and neurodegenerative disorders. The use of hiPSCs also opens new avenues for personalized medicine, allowing researchers to model diseases and develop targeted therapies based on individual patient profiles. Despite the promise of direct hiPSC injections for therapeutic purposes, challenges such as poor localization and limited integration have led to the exploration of biomaterial scaffolds as supportive platforms for cell delivery and tissue regeneration. This paper reviews the integration of 3D bioprinting technologies and bioink materials in neuroscience applications, offering a unique platform to create complex brain and tissue architectures that mimic the mechanical, architectural, and biochemical properties of native tissues. These advancements provide robust tools for modelling, repair, and drug screening applications. The review highlights current research, identifies research gaps, and offers recommendations for future studies on 3D bioprinting in neuroscience. The investigation demonstrates the significant potential of 3D bioprinting to fabricate brain-like tissue constructs, which holds great promise for regenerative medicine and drug testing models. This approach offers new avenues for studying brain diseases and potential treatments.
Collapse
Affiliation(s)
- Xu Bocheng
- Department of Biomedical Engineering, Price Faculty of Engineering, University of Manitoba, Winnipeg, R3E 0W2, Canada
| | - Rodrigo França
- Department of Biomedical Engineering, Price Faculty of Engineering, University of Manitoba, Winnipeg, R3E 0W2, Canada
- Rady Faculty of Health Science, Dental Biomaterials Research Lab, University of Manitoba, Winnipeg, R3E 0W2, Canada
| |
Collapse
|
36
|
Rijns L, Rutten MGTA, Vrehen AF, Aldana AA, Baker MB, Dankers PYW. Mimicking the extracellular world: from natural to fully synthetic matrices utilizing supramolecular biomaterials. NANOSCALE 2024; 16:16290-16312. [PMID: 39161293 DOI: 10.1039/d4nr02088j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/21/2024]
Abstract
The extracellular matrix (ECM) has evolved around complex covalent and non-covalent interactions to create impressive function-from cellular signaling to constant remodeling. A major challenge in the biomedical field is the de novo design and control of synthetic ECMs for applications ranging from tissue engineering to neuromodulation to bioelectronics. As we move towards recreating the ECM's complexity in hydrogels, the field has taken several approaches to recapitulate the main important features of the native ECM (i.e. mechanical, bioactive and dynamic properties). In this review, we first describe the wide variety of hydrogel systems that are currently used, ranging from fully natural to completely synthetic to hybrid versions, highlighting the advantages and limitations of each class. Then, we shift towards supramolecular hydrogels that show great potential for their use as ECM mimics due to their biomimetic hierarchical structure, inherent (controllable) dynamic properties and their modular design, allowing for precise control over their mechanical and biochemical properties. In order to make the next step in the complexity of synthetic ECM-mimetic hydrogels, we must leverage the supramolecular self-assembly seen in the native ECM; we therefore propose to use supramolecular monomers to create larger, hierarchical, co-assembled hydrogels with complex and synergistic mechanical, bioactive and dynamic features.
Collapse
Affiliation(s)
- Laura Rijns
- Institute for Complex Molecular Systems, Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands.
- Department of Biomedical Engineering, Laboratory of Chemical Biology, Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands
| | - Martin G T A Rutten
- Institute for Complex Molecular Systems, Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands.
- Department of Biomedical Engineering, Laboratory of Chemical Biology, Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands
| | - Annika F Vrehen
- Institute for Complex Molecular Systems, Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands.
- Department of Biomedical Engineering, Laboratory of Chemical Biology, Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands
| | - Ana A Aldana
- Department of Complex Tissue Regeneration, MERLN Institute for Technology Inspired Regenerative Medicine, Maastricht University, 6200 MD Maastricht, The Netherlands
| | - Matthew B Baker
- Department of Complex Tissue Regeneration, MERLN Institute for Technology Inspired Regenerative Medicine, Maastricht University, 6200 MD Maastricht, The Netherlands
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology Inspired Regenerative Medicine, Maastricht University, 6200 MD Maastricht, The Netherlands
| | - Patricia Y W Dankers
- Institute for Complex Molecular Systems, Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands.
- Department of Biomedical Engineering, Laboratory of Chemical Biology, Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands
- Department of Chemical Engineering and Chemistry, Eindhoven University of Technology, 5600 MB, Eindhoven, The Netherlands
| |
Collapse
|
37
|
Zhao Y, Dai Z, Huang H, Tian J, Cai H. Injectable Silver Nanoparticle-Based Hydrogel Dressings with Rapid Shape Adaptability and Antimicrobial Activity. Appl Biochem Biotechnol 2024:10.1007/s12010-024-05048-5. [PMID: 39254796 DOI: 10.1007/s12010-024-05048-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/19/2024] [Indexed: 09/11/2024]
Abstract
Burns and scalds often result in deep wounds that challenge adequate debridement and inflammation control using traditional sheet-like hydrogel dressings. Herein, we developed an antibacterial, injectable, and self-healing hydrogel (ADCM@Ag) by employing carboxymethyl chitosan (CMCS) for in situ green reduction of silver ions and utilizing a spontaneous Schiff base reaction with aldehyde-functionalized dextran (AD). SEM analysis revealed a porous structure within the hydrogel. Swelling and enzymatic degradation assays demonstrated that ADCM@Ag hydrogel possesses excellent fluid absorption capacity and biodegradability. Mechanical tests indicated good mechanical properties, allowing the hydrogel to withstand external forces when applied to animal wounds. The hydrogel exhibited good injectability, shape adaptability, and self-healing capability. Cell experiments showed that the ADCM@Ag hydrogel avoided the cytotoxicity caused by high concentrations of silver ions and had good cell compatibility. Antimicrobial assays showed that ADCM@Ag exhibited potent bactericidal effects against Gram-negative and Gram-positive bacteria, achieving at least 85% killing efficacy. Collectively, ADCM@Ag hydrogel has good potential for wound dressing applications.
Collapse
Affiliation(s)
- Yuanyuan Zhao
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, 130 Meilong Road, P. O. Box 309#, Shanghai, 200237, People's Republic of China
| | - Zhaobo Dai
- Shanghai Key Laboratory of Functional Materials Chemistry, East China University of Science and Technology, Meilong Road No. 130, Shanghai, 200237, People's Republic of China
| | - Huimin Huang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, 130 Meilong Road, P. O. Box 309#, Shanghai, 200237, People's Republic of China
| | - Jia Tian
- Shanghai Key Laboratory of Functional Materials Chemistry, East China University of Science and Technology, Meilong Road No. 130, Shanghai, 200237, People's Republic of China
| | - Haibo Cai
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, 130 Meilong Road, P. O. Box 309#, Shanghai, 200237, People's Republic of China.
| |
Collapse
|
38
|
Wiita EG, Toprakcioglu Z, Jayaram AK, Knowles TPJ. Formation of Nanofibrillar Self-Healing Hydrogels Using Antimicrobial Peptides. ACS APPLIED MATERIALS & INTERFACES 2024; 16:46167-46176. [PMID: 39171944 PMCID: PMC11378157 DOI: 10.1021/acsami.4c11542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/23/2024]
Abstract
The rise of drug-resistant microorganisms has prompted the development of innovative strategies with the aim of addressing this challenge. Among the alternative approaches gaining increased attention are antimicrobial peptides (AMPs), a group of peptides with the ability to combat microbial pathogens. Here, we investigated a small peptide, KLVFF, derived from the Alzheimer's amyloid-β (Aβ) protein. While Aβ has been associated with the development of neurodegenerative diseases, the core part of the Aβ protein, namely the Aβ 16-20 fragment, has also been exploited to obtain highly functional biomaterials. In this study we found that KLVFF is capable of self-assembling into a fibrillar network to form a self-healing hydrogel. Moreover, this small peptide can undergo a transition from a gel to a liquid state following application of shear stress, in a reversible manner. As an AMP, this material exhibited both antibacterial and antifungal properties while remaining highly biocompatible and noncytotoxic toward mammalian cells. The propensity of the KLVFF hydrogel to rapidly assemble into highly ordered macroscopic structures makes it an ideal candidate for biomedical applications necessitating antimicrobial activity, such as wound healing.
Collapse
Affiliation(s)
- Elizabeth G Wiita
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, U.K
| | - Zenon Toprakcioglu
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, U.K
| | - Akhila K Jayaram
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, U.K
- Cavendish Laboratory, Department of Physics, University of Cambridge, J J Thomson Avenue, Cambridge CB3 0HE, U.K
| | - Tuomas P J Knowles
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, U.K
- Cavendish Laboratory, Department of Physics, University of Cambridge, J J Thomson Avenue, Cambridge CB3 0HE, U.K
| |
Collapse
|
39
|
Ahmad N, Kiriako G, Saliba J, Abla K, El-Sabban M, Mhanna R. Engineering a 3D Biomimetic Peptides Functionalized-Polyethylene Glycol Hydrogel Model Cocultured with Endothelial Cells and Astrocytes: Enhancing In Vitro Blood-Brain Barrier Biomimicry. Mol Pharm 2024; 21:4664-4672. [PMID: 39133897 PMCID: PMC11372828 DOI: 10.1021/acs.molpharmaceut.4c00599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/03/2024]
Abstract
The blood-brain barrier (BBB) poses a significant challenge for drug delivery and is linked to various neurovascular disorders. In vitro BBB models provide a tool to investigate drug permeation across the BBB and the barrier's response to external injury events. Yet, existing models lack fidelity in replicating the BBB's complexity, hindering a comprehensive understanding of its functions. This study introduces a three-dimensional (3D) model using polyethylene glycol (PEG) hydrogels modified with biomimetic peptides that represent recognition sequences of key proteins in the brain. Hydrogels were functionalized with recognition sequences for laminin (IKVAV) and fibronectin peptides (RGD) and chemically cross-linked with matrix metalloprotease-sensitive peptides (MMPs) to mimic the extracellular matrix of the BBB. Astrocytes and endothelial cells were seeded within and on the surface of the hydrogels, respectively. The barrier integrity was assessed through different tests including transendothelial electrical resistance (TEER), the permeability of sodium fluorescence (Na-F), the permeability of Evan's blue bound to albumin (EBA), and the expression of zonula occluden-1 (ZO-1) in seeded endothelial cells. Hydrogels with a combination of RGD and IKVAV peptides displayed superior performance, exhibiting significantly higher TEER values (55.33 ± 1.47 Ω·cm2) at day 5 compared to other 2D controls including HAECs-monoculture and HAECs-cocultured with NHAs seeded on well inserts and 3D controls including RGD hydrogel and RGD-IKVAV monoculture with HAECs and RGD hydrogel cocultured with HAECs and NHAs. The designed 3D system resulted in the lowest Evan's blue permeability at 120 min (0.215 ± 0.055 μg/mL) compared to controls. ZO-1 expression was significantly higher and formed a relatively larger network in the functionalized hydrogel cocultured with astrocytes and endothelial cells compared to the controls. Thus, the designed 3D model effectively recapitulates the main BBB structure and function in vitro and is expected to contribute to a deeper understanding of pathological CNS angiogenesis and the development of effective CNS medications.
Collapse
Affiliation(s)
- Nesrine Ahmad
- Biomedical Engineering Program, Maroun Semaan Faculty of Engineering and Architecture, American University of Beirut, Beirut 1107-2020, Lebanon
| | - Georges Kiriako
- Biomedical Engineering Program, Maroun Semaan Faculty of Engineering and Architecture, American University of Beirut, Beirut 1107-2020, Lebanon
| | - John Saliba
- Biomedical Engineering Program, Maroun Semaan Faculty of Engineering and Architecture, American University of Beirut, Beirut 1107-2020, Lebanon
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 1107-2020, Lebanon
| | - Kawthar Abla
- Biomedical Engineering Program, Maroun Semaan Faculty of Engineering and Architecture, American University of Beirut, Beirut 1107-2020, Lebanon
| | - Marwan El-Sabban
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 1107-2020, Lebanon
| | - Rami Mhanna
- Biomedical Engineering Program, Maroun Semaan Faculty of Engineering and Architecture, American University of Beirut, Beirut 1107-2020, Lebanon
| |
Collapse
|
40
|
Su W, Yang Q, Li T, Xu J, Yin P, Han M, Lin Z, Deng Y, Wu Y, Huang W, Wang L. Electrospun Aligned Nanofiber Yarns Constructed Biomimetic M-Type Interface Integrated into Precise Co-Culture System as Muscle-Tendon Junction-on-a-Chip for Drug Development. SMALL METHODS 2024; 8:e2301754. [PMID: 38593371 DOI: 10.1002/smtd.202301754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 03/21/2024] [Indexed: 04/11/2024]
Abstract
The incorporation of engineered muscle-tendon junction (MTJ) with organ-on-a-chip technology provides promising in vitro models for the understanding of cell-cell interaction at the interface between muscle and tendon tissues. However, developing engineered MTJ tissue with biomimetic anatomical interface structure remains challenging, and the precise co-culture of engineered interface tissue is further regarded as a remarkable obstacle. Herein, an interwoven waving approach is presented to develop engineered MTJ tissue with a biomimetic "M-type" interface structure, and further integrated into a precise co-culture microfluidic device for functional MTJ-on-a-chip fabrication. These multiscale MTJ scaffolds based on electrospun nanofiber yarns enabled 3D cellular alignment and differentiation, and the "M-type" structure led to cellular organization and interaction at the interface zone. Crucially, a compartmentalized co-culture system is integrated into an MTJ-on-a-chip device for the precise co-culture of muscle and tendon zones using their medium at the same time. Such an MTJ-on-a-chip device is further served for drug-associated MTJ toxic or protective efficacy investigations. These results highlight that these interwoven nanofibrous scaffolds with biomimetic "M-type" interface are beneficial for engineered MTJ tissue development, and MTJ-on-a-chip with precise co-culture system indicated their promising potential as in vitro musculoskeletal models for drug development and biological mechanism studies.
Collapse
Affiliation(s)
- Weiwei Su
- Guangdong Engineering Research Center for Translation of Medical 3D Printing Application, Guangdong Provincial Key Laboratory of Digital Medicine and Biomechanics, National Key Discipline of Human Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Qiao Yang
- School of Biomedical Engineering, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Ting Li
- Guangdong Engineering Research Center for Translation of Medical 3D Printing Application, Guangdong Provincial Key Laboratory of Digital Medicine and Biomechanics, National Key Discipline of Human Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Jie Xu
- School of Biomedical Engineering, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Panjing Yin
- Guangdong Engineering Research Center for Translation of Medical 3D Printing Application, Guangdong Provincial Key Laboratory of Digital Medicine and Biomechanics, National Key Discipline of Human Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Mingying Han
- School of Biomedical Engineering, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Zhuosheng Lin
- Guangdong Engineering Research Center for Translation of Medical 3D Printing Application, Guangdong Provincial Key Laboratory of Digital Medicine and Biomechanics, National Key Discipline of Human Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Yuping Deng
- Guangdong Engineering Research Center for Translation of Medical 3D Printing Application, Guangdong Provincial Key Laboratory of Digital Medicine and Biomechanics, National Key Discipline of Human Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Yaobin Wu
- Guangdong Engineering Research Center for Translation of Medical 3D Printing Application, Guangdong Provincial Key Laboratory of Digital Medicine and Biomechanics, National Key Discipline of Human Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Wenhua Huang
- Guangdong Engineering Research Center for Translation of Medical 3D Printing Application, Guangdong Provincial Key Laboratory of Digital Medicine and Biomechanics, National Key Discipline of Human Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
- Department of Orthopedics, Affiliated Hospital of Putian University, Putian, 351100, China
| | - Ling Wang
- School of Biomedical Engineering, Southern Medical University, Guangzhou, Guangdong, 510515, China
| |
Collapse
|
41
|
Chen H, Zhang B, Huang J. Recent advances and applications of artificial intelligence in 3D bioprinting. BIOPHYSICS REVIEWS 2024; 5:031301. [PMID: 39036708 PMCID: PMC11260195 DOI: 10.1063/5.0190208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 06/11/2024] [Indexed: 07/23/2024]
Abstract
3D bioprinting techniques enable the precise deposition of living cells, biomaterials, and biomolecules, emerging as a promising approach for engineering functional tissues and organs. Meanwhile, recent advances in 3D bioprinting enable researchers to build in vitro models with finely controlled and complex micro-architecture for drug screening and disease modeling. Recently, artificial intelligence (AI) has been applied to different stages of 3D bioprinting, including medical image reconstruction, bioink selection, and printing process, with both classical AI and machine learning approaches. The ability of AI to handle complex datasets, make complex computations, learn from past experiences, and optimize processes dynamically makes it an invaluable tool in advancing 3D bioprinting. The review highlights the current integration of AI in 3D bioprinting and discusses future approaches to harness the synergistic capabilities of 3D bioprinting and AI for developing personalized tissues and organs.
Collapse
Affiliation(s)
| | - Bin Zhang
- Department of Mechanical and Aerospace Engineering, Brunel University London, London, United Kingdom
| | - Jie Huang
- Department of Mechanical Engineering, University College London, London, United Kingdom
| |
Collapse
|
42
|
Mishra A, Omoyeni T, Singh PK, Anandakumar S, Tiwari A. Trends in sustainable chitosan-based hydrogel technology for circular biomedical engineering: A review. Int J Biol Macromol 2024; 276:133823. [PMID: 39002912 DOI: 10.1016/j.ijbiomac.2024.133823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 07/08/2024] [Accepted: 07/10/2024] [Indexed: 07/15/2024]
Abstract
Eco-friendly materials have emerged in biomedical engineering, driving major advances in chitosan-based hydrogels. These hydrogels offer a promising green alternative to conventional polymers due to their non-toxicity, biodegradability, biocompatibility, environmental friendliness, affordability, and easy accessibility. Known for their remarkable properties such as drug encapsulation, delivery capabilities, biosensing, functional scaffolding, and antimicrobial behavior, chitosan hydrogels are at the forefront of biomedical research. This paper explores the fabrication and modification methods of chitosan hydrogels for diverse applications, highlighting their role in advancing climate-neutral healthcare technologies. It reviews significant scientific advancements and trends chitosan hydrogels focusing on cancer diagnosis, drug delivery, and wound care. Additionally, it addresses current challenges and green synthesis practices that support a circular economy, enhancing biomedical sustainability. By providing an in-depth analysis of the latest evidence on climate-neutral management, this review aims to facilitate informed decision-making and foster the development of sustainable strategies leveraging chitosan hydrogel technology. The insights from this comprehensive examination are pivotal for steering future research and applications in sustainable biomedical solutions.
Collapse
Affiliation(s)
- Anshuman Mishra
- Institute of Advanced Materials, IAAM, Gammalkilsvägen 18, Ulrika 59053, Sweden
| | - Temitayo Omoyeni
- Institute of Advanced Materials, IAAM, Gammalkilsvägen 18, Ulrika 59053, Sweden; Cyprus International University Faculty of Engineering, Nicosia 99258, TRNC, Cyprus
| | - Pravin Kumar Singh
- Institute of Advanced Materials, IAAM, Gammalkilsvägen 18, Ulrika 59053, Sweden
| | - S Anandakumar
- Department of Chemistry, Anna University, Chennai 600025, India
| | - Ashutosh Tiwari
- Institute of Advanced Materials, IAAM, Gammalkilsvägen 18, Ulrika 59053, Sweden.
| |
Collapse
|
43
|
Han H, Li H, Wang L, Zhu Y, Guan H, Yao J, Xiao W, Li B, Liao X. Preparation of Autoclavable and Injectable Silk Fibroin Cryogels for Tissue Engineering Applications. Macromol Biosci 2024; 24:e2400038. [PMID: 38843388 DOI: 10.1002/mabi.202400038] [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: 01/31/2024] [Revised: 05/19/2024] [Indexed: 06/19/2024]
Abstract
A cryogel is a supermacroporous gel network that is generated at subzero temperatures by polymerizing monomers or gelating polymeric precursors. Since cryogels possess inherent characteristics such as interconnected macroporous structures, excellent mechanical properties, and high resistance to autoclave sterilization, they are highly desirable for tissue engineering and regenerative medicine. Silk fibroin, a natural protein obtained from Bombyx mori silkworms, is an excellent raw material for cryogel preparation. The aim of this study is to establish a controlled method for preparing silk fibroin cryogels with suitable properties for application as tissue engineering scaffolds. Using a dual crosslinking strategy consisting of low-temperature radical polymerization coupled with methanol-induced conformational transformation, porous cryogels are prepared. The cryogels display many unique characteristics, such as an interconnected macroporous structure, a high water absorption capacity, water-triggered shape memory, syringe injectability, and strong resilience to autoclave sterilization. Furthermore, the cryogels demonstrate excellent biocompatibility and cell affinity, facilitating cell adhesion, migration, and proliferation. The interconnected supermacroporous architecture resembling the native extracellular matrix, together with their unique physical properties and autoclaving stability, suggests that cryogels are promising candidate scaffolds for tissue engineering and cell therapy.
Collapse
Affiliation(s)
- Hongjuan Han
- Chongqing Key Laboratory of Nano/Micro Composite Materials and Devices, School of Metallurgy and Materials Engineering, Chongqing University of Science and Technology, Chongqing, 401331, P. R. China
| | - Haiyan Li
- Chongqing Engineering Laboratory of Nano/Micro Biomedical Detection Technology, School of Metallurgy and Materials Engineering, Chongqing University of Science and Technology, Chongqing, 401331, P. R. China
| | - Lu Wang
- Chongqing Key Laboratory of Nano/Micro Composite Materials and Devices, School of Metallurgy and Materials Engineering, Chongqing University of Science and Technology, Chongqing, 401331, P. R. China
| | - Yong Zhu
- Chongqing Key Laboratory of Nano/Micro Composite Materials and Devices, School of Metallurgy and Materials Engineering, Chongqing University of Science and Technology, Chongqing, 401331, P. R. China
| | - Haoqing Guan
- Chongqing Engineering Laboratory of Nano/Micro Biomedical Detection Technology, School of Metallurgy and Materials Engineering, Chongqing University of Science and Technology, Chongqing, 401331, P. R. China
| | - Jingzhi Yao
- Chongqing Engineering Laboratory of Nano/Micro Biomedical Detection Technology, School of Metallurgy and Materials Engineering, Chongqing University of Science and Technology, Chongqing, 401331, P. R. China
| | - Wenqian Xiao
- Chongqing Key Laboratory of Nano/Micro Composite Materials and Devices, School of Metallurgy and Materials Engineering, Chongqing University of Science and Technology, Chongqing, 401331, P. R. China
| | - Bo Li
- Chongqing Key Laboratory of Nano/Micro Composite Materials and Devices, School of Metallurgy and Materials Engineering, Chongqing University of Science and Technology, Chongqing, 401331, P. R. China
| | - Xiaoling Liao
- Chongqing Engineering Laboratory of Nano/Micro Biomedical Detection Technology, School of Metallurgy and Materials Engineering, Chongqing University of Science and Technology, Chongqing, 401331, P. R. China
| |
Collapse
|
44
|
Dong L, Li L, Chen H, Cao Y, Lei H. Mechanochemistry: Fundamental Principles and Applications. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024:e2403949. [PMID: 39206931 DOI: 10.1002/advs.202403949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 07/30/2024] [Indexed: 09/04/2024]
Abstract
Mechanochemistry is an emerging research field at the interface of physics, mechanics, materials science, and chemistry. Complementary to traditional activation methods in chemistry, such as heat, electricity, and light, mechanochemistry focuses on the activation of chemical reactions by directly or indirectly applying mechanical forces. It has evolved as a powerful tool for controlling chemical reactions in solid state systems, sensing and responding to stresses in polymer materials, regulating interfacial adhesions, and stimulating biological processes. By combining theoretical approaches, simulations and experimental techniques, researchers have gained intricate insights into the mechanisms underlying mechanochemistry. In this review, the physical chemistry principles underpinning mechanochemistry are elucidated and a comprehensive overview of recent significant achievements in the discovery of mechanically responsive chemical processes is provided, with a particular emphasis on their applications in materials science. Additionally, The perspectives and insights into potential future directions for this exciting research field are offered.
Collapse
Affiliation(s)
- Liang Dong
- Collaborative Innovation Center of Advanced Microstructures, National Laboratory of Solid State Microstructure, Department of Physics, Nanjing University, Nanjing, Jiangsu, 210093, P. R. China
| | - Luofei Li
- Collaborative Innovation Center of Advanced Microstructures, National Laboratory of Solid State Microstructure, Department of Physics, Nanjing University, Nanjing, Jiangsu, 210093, P. R. China
| | - Huiyan Chen
- Collaborative Innovation Center of Advanced Microstructures, National Laboratory of Solid State Microstructure, Department of Physics, Nanjing University, Nanjing, Jiangsu, 210093, P. R. China
| | - Yi Cao
- Collaborative Innovation Center of Advanced Microstructures, National Laboratory of Solid State Microstructure, Department of Physics, Nanjing University, Nanjing, Jiangsu, 210093, P. R. China
| | - Hai Lei
- School of Physics, Zhejiang University, Hangzhou, Zhejiang, 310027, P. R. China
- Institute of Advanced Physics, Zhejiang University, Hangzhou, Zhejiang, 310027, P. R. China
| |
Collapse
|
45
|
Choi W, Kohane DS. Hybrid Nanoparticle-Hydrogel Systems for Drug Delivery Depots and Other Biomedical Applications. ACS NANO 2024; 18:22780-22792. [PMID: 39140388 DOI: 10.1021/acsnano.4c06888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/15/2024]
Abstract
Hydrogel-based depots typically tend to remain where injected and have excellent biocompatibility but are relatively poor at controlling drug release. Nanoparticles (NPs) typically have the opposite properties. The smaller the NPs are, the more likely they are to leave the site of injection. Their biocompatibility is variable depending on the material but can be poor. However, NPs can be good at controlling drug release. In these and other properties, combining NPs and hydrogels can leverage their advantages and negate their disadvantages. This review highlights the rationale for hybrid NP-hydrogel systems in drug delivery, the basic methods of producing them, and examples where combining the two systems addressed specific problems.
Collapse
Affiliation(s)
- Wonmin Choi
- Laboratory for Biomaterials and Drug Delivery, Department of Anesthesiology, Division of Critical Care Medicine, Boston Children's Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, Massachusetts 02115, United States
| | - Daniel S Kohane
- Laboratory for Biomaterials and Drug Delivery, Department of Anesthesiology, Division of Critical Care Medicine, Boston Children's Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, Massachusetts 02115, United States
| |
Collapse
|
46
|
Hasan MM, Ahmad A, Akter MZ, Choi YJ, Yi HG. Bioinks for bioprinting using plant-derived biomaterials. Biofabrication 2024; 16:042004. [PMID: 39079554 DOI: 10.1088/1758-5090/ad6932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 07/30/2024] [Indexed: 08/23/2024]
Abstract
Three-dimensional (3D) bioprinting has revolutionized tissue engineering by enabling the fabrication of complex and functional human tissues and organs. An essential component of successful 3D bioprinting is the selection of an appropriate bioink capable of supporting cell proliferation and viability. Plant-derived biomaterials, because of their abundance, biocompatibility, and tunable properties, hold promise as bioink sources, thus offering advantages over animal-derived biomaterials, which carry immunogenic concerns. This comprehensive review explores and analyzes the potential of plant-derived biomaterials as bioinks for 3D bioprinting of human tissues. Modification and optimization of these materials to enhance printability and biological functionality are discussed. Furthermore, cancer research and drug testing applications of the use of plant-based biomaterials in bioprinting various human tissues such as bone, cartilage, skin, and vascular tissues are described. Challenges and limitations, including mechanical integrity, cell viability, resolution, and regulatory concerns, along with potential strategies to overcome them, are discussed. Additionally, this review provides insights into the potential use of plant-based decellularized ECM (dECM) as bioinks, future prospects, and emerging trends in the use of plant-derived biomaterials for 3D bioprinting applications. The potential of plant-derived biomaterials as bioinks for 3D bioprinting of human tissues is highlighted herein. However, further research is necessary to optimize their processing, standardize their properties, and evaluate their long-termin vivoperformance. Continued advancements in plant-derived biomaterials have the potential to revolutionize tissue engineering and facilitate the development of functional and regenerative therapies for diverse clinical applications.
Collapse
Affiliation(s)
- Md Mehedee Hasan
- Department of Convergence Biosystems Engineering, College of Agriculture and Life Sciences (CALS), Chonnam National University, Gwangju 61186, Republic of Korea
| | - Ashfaq Ahmad
- Department of Convergence Biosystems Engineering, College of Agriculture and Life Sciences (CALS), Chonnam National University, Gwangju 61186, Republic of Korea
- Interdisciplinary Program in IT-Bio Convergence System, Chonnam National University, Gwangju, Republic of Korea
| | - Mst Zobaida Akter
- Department of Convergence Biosystems Engineering, College of Agriculture and Life Sciences (CALS), Chonnam National University, Gwangju 61186, Republic of Korea
- Interdisciplinary Program in IT-Bio Convergence System, Chonnam National University, Gwangju, Republic of Korea
| | - Yeong-Jin Choi
- Advanced Bio and Healthcare Materials Research Division, Korea Institute of Materials Science (KIMS), Changwon 51508, Republic of Korea
| | - Hee-Gyeong Yi
- Department of Convergence Biosystems Engineering, College of Agriculture and Life Sciences (CALS), Chonnam National University, Gwangju 61186, Republic of Korea
- Interdisciplinary Program in IT-Bio Convergence System, Chonnam National University, Gwangju, Republic of Korea
| |
Collapse
|
47
|
Shen L, Cao Y, Wang L, Zhang X, Zhang A, Li W. Compressible Hydrogels with Stabilized Chirality from Thermoresponsive Helical Dendronized Poly(phenylacetylene)s. Angew Chem Int Ed Engl 2024; 63:e202407552. [PMID: 38770786 DOI: 10.1002/anie.202407552] [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: 04/21/2024] [Revised: 05/17/2024] [Accepted: 05/21/2024] [Indexed: 05/22/2024]
Abstract
Fabrication of chiral hydrogels from thermoresponsive helical dendronized phenylacetylene copolymers (PPAs) carrying three-fold dendritic oligoethylene glycols (OEGs) is reported. Three different temperatures, i.e. below or above cloud point temperatures (Tcps) of the copolymers, and under freezing condition, were utilized, affording thermoresponsive hydrogels with different morphologies and mechanical properties. At room temperature, transparent hydrogels were obtained through crosslinking among different copolymer chains. Differently, opaque hydrogels with much improved mechanical properties were formed at elevated temperatures through crosslinking from the thermally dehydrated and collapsed copolymer aggregates, leading to heterogeneity for the hydrogels with highly porous morphology. While crosslinking at freezing temperature synergistically through ice templating, these amphiphilic dendronized copolymers formed hydrogels with highly porous lamellar structures, which exhibited remarkable compressible properties as human articular cartilage with excellent fatigue resistance. Amphiphilicity of the dendronized copolymers played a pivotal role in modulating the network formation during the gelation, as well as morphology and mechanical performance of the resulting hydrogels. Through crosslinking, these dendronized copolymers featured with typical dynamic helical conformations were transformed into hydrogels with unprecedently stabilized helicities due to the restrained chain mobilities in the three-dimensional networks.
Collapse
Affiliation(s)
- Lefei Shen
- International Joint Laboratory of Biomimetic and Smart Polymers, School of Materials Science & Engineering, Shanghai University, 333 Nanchen Road, Shanghai, 200444, China
| | - Yuexin Cao
- International Joint Laboratory of Biomimetic and Smart Polymers, School of Materials Science & Engineering, Shanghai University, 333 Nanchen Road, Shanghai, 200444, China
| | - Lei Wang
- International Joint Laboratory of Biomimetic and Smart Polymers, School of Materials Science & Engineering, Shanghai University, 333 Nanchen Road, Shanghai, 200444, China
| | - Xiacong Zhang
- International Joint Laboratory of Biomimetic and Smart Polymers, School of Materials Science & Engineering, Shanghai University, 333 Nanchen Road, Shanghai, 200444, China
| | - Afang Zhang
- International Joint Laboratory of Biomimetic and Smart Polymers, School of Materials Science & Engineering, Shanghai University, 333 Nanchen Road, Shanghai, 200444, China
| | - Wen Li
- International Joint Laboratory of Biomimetic and Smart Polymers, School of Materials Science & Engineering, Shanghai University, 333 Nanchen Road, Shanghai, 200444, China
| |
Collapse
|
48
|
Zhai Z, Zhou Y, Sarkar I, Liu Y, Yao Y, Zhang J, Bortner MJ, Matson JB, Johnson BN, Edgar KJ. Synthesis and real-time characterization of self-healing, injectable, fast-gelling hydrogels based on alginate multi-reducing end polysaccharides (MREPs). Carbohydr Polym 2024; 338:122172. [PMID: 38763719 DOI: 10.1016/j.carbpol.2024.122172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 04/13/2024] [Accepted: 04/15/2024] [Indexed: 05/21/2024]
Abstract
Polysaccharide-based hydrogels are promising for many biomedical applications including drug delivery, wound healing, and tissue engineering. We illustrate herein self-healing, injectable, fast-gelling hydrogels prepared from multi-reducing end polysaccharides, recently introduced by the Edgar group. Simple condensation of reducing ends from multi-reducing end alginate (M-Alg) with amines from polyethylene imine (PEI) in water affords a dynamic, hydrophilic polysaccharide network. Trace amounts of acetic acid can accelerate the gelation time from hours to seconds. The fast-gelation behavior is driven by rapid Schiff base formation and strong ionic interactions induced by acetic acid. A cantilever rheometer enables real-time monitoring of changes in viscoelastic properties during hydrogel formation. The reversible nature of these crosslinks (imine bonds, ionic interactions) provides a hydrogel with low toxicity in cell studies as well as self-healing and injectable properties. Therefore, the self-healing, injectable, and fast-gelling M-Alg/PEI hydrogel holds substantial promise for biomedical, agricultural, controlled release, and other applications.
Collapse
Affiliation(s)
- Zhenghao Zhai
- Macromolecules Innovation Institute, Virginia Tech, Blacksburg, VA 24061, United States
| | - Yang Zhou
- Department of Sustainable Biomaterials, Virginia Tech, Blacksburg, VA 24061, United States
| | - Ishani Sarkar
- Department of Chemistry, Virginia Tech, Blacksburg, VA 24061, United States
| | - Yang Liu
- Macromolecules Innovation Institute, Virginia Tech, Blacksburg, VA 24061, United States
| | - Yimin Yao
- Macromolecules Innovation Institute, Virginia Tech, Blacksburg, VA 24061, United States; Department of Chemical Engineering, Virginia Tech, Blacksburg, VA 24061, United States
| | - Junru Zhang
- Department of Industrial & Systems Engineering, Virginia Tech, Blacksburg, VA 24061, United States
| | - Michael J Bortner
- Macromolecules Innovation Institute, Virginia Tech, Blacksburg, VA 24061, United States; Department of Chemical Engineering, Virginia Tech, Blacksburg, VA 24061, United States
| | - John B Matson
- Macromolecules Innovation Institute, Virginia Tech, Blacksburg, VA 24061, United States; Department of Chemistry, Virginia Tech, Blacksburg, VA 24061, United States
| | - Blake N Johnson
- Macromolecules Innovation Institute, Virginia Tech, Blacksburg, VA 24061, United States; Department of Chemical Engineering, Virginia Tech, Blacksburg, VA 24061, United States; Department of Industrial & Systems Engineering, Virginia Tech, Blacksburg, VA 24061, United States
| | - Kevin J Edgar
- Macromolecules Innovation Institute, Virginia Tech, Blacksburg, VA 24061, United States; Department of Sustainable Biomaterials, Virginia Tech, Blacksburg, VA 24061, United States.
| |
Collapse
|
49
|
Mondal P, Chatterjee K. Multibiofunctional Self-healing Adhesive Injectable Nanocomposite Polysaccharide Hydrogel. Biomacromolecules 2024; 25:4762-4779. [PMID: 38989826 DOI: 10.1021/acs.biomac.4c00016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/12/2024]
Abstract
Injectable hydrogels with good antimicrobial and antioxidant properties, self-healing characteristics, suitable mechanical properties, and therapeutic effects have great practical significance for developing treatments for pressing healthcare challenges. Herein, we have designed a novel, self-healing injectable hydrogel composite incorporating cross-linked biofunctional nanomaterials by mixing alginate aldehyde (Ox-Alg), quaternized chitosan (QCS), adipic acid dihydrazide (ADH), and copper oxide nanosheets surface functionalized with folic acid as the bioligand (F-CuO). Gelation was achieved under physiological conditions via the dynamic Schiff base cross-linking mechanism. The developed nanocomposite injectable hydrogel demonstrated the fast self-healing ability essential to bear deformation and outstanding antibacterial properties along with ROS scavenging ability. Furthermore, the optimized formulation of our F-CuO-embedded injectable hydrogel exhibited excellent cytocompatibility, blood compatibility, and in vitro wound healing performance. Taken together, the F-CuO nanosheet cross-linked injectable hydrogel composite presented herein offers a promising candidate biomaterial with multifunctional properties to develop solutions for addressing clinical challenges.
Collapse
Affiliation(s)
- Pritiranjan Mondal
- Department of Materials Engineering, Indian Institute of Science, C.V. Raman Avenue, Bangalore 560012, India
| | - Kaushik Chatterjee
- Department of Materials Engineering, Indian Institute of Science, C.V. Raman Avenue, Bangalore 560012, India
| |
Collapse
|
50
|
Mišković V, Greco I, Minetti C, Cialdai F, Monici M, Gazzi A, Marcellino J, Samad YA, Delogu LG, Ferrari AC, Iorio CS. Hydrogel mechanical properties in altered gravity. NPJ Microgravity 2024; 10:83. [PMID: 39117674 PMCID: PMC11310329 DOI: 10.1038/s41526-024-00388-2] [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: 02/19/2022] [Accepted: 03/21/2024] [Indexed: 08/10/2024] Open
Abstract
Exposure to altered gravity influences cellular behaviour in cell cultures. Hydrogels are amongst the most common materials used to produce tissue-engineering scaffolds, and their mechanical properties play a crucial role in cell-matrix interaction. However, little is known about the influence of altered gravity on hydrogel properties. Here we study the mechanical properties of Poly (ethylene glycol) diacrylate (PEGDA) and PEGDA incorporated with graphene oxide (GO) by performing tensile tests in micro and hypergravity during a Parabolic flight campaign, and by comparing them to the same tests performed in Earth gravity. We show that gravity levels do not result in a statistically significant difference in Young's modulus.
Collapse
Affiliation(s)
- Vanja Mišković
- Centre for Research and Engineering in Space Technologies, École Polytechnique de Bruxelles, Université libre de Bruxelles, Brussels, Belgium
| | - Immacolata Greco
- Centre for Research and Engineering in Space Technologies, École Polytechnique de Bruxelles, Université libre de Bruxelles, Brussels, Belgium
| | - Christophe Minetti
- Centre for Research and Engineering in Space Technologies, École Polytechnique de Bruxelles, Université libre de Bruxelles, Brussels, Belgium
| | - Francesca Cialdai
- ASAcampus Joint Laboratory, ASA Research Division, Department of Experimental and Clinical Biomedical Sciences « Mario Serio », University of Florence, Florence, Italy
| | - Monica Monici
- ASAcampus Joint Laboratory, ASA Research Division, Department of Experimental and Clinical Biomedical Sciences « Mario Serio », University of Florence, Florence, Italy
| | - Arianna Gazzi
- Department of Chemical and Pharmaceutical Sciences, University of Trieste, Trieste, Italy
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | | | - Yarjan Abdul Samad
- Cambridge Graphene Centre, University of Cambridge, Cambridge, CB3 0FA, UK
- Department of Aerospace Engineering, Khalifa university of Science and Technology, Abu Dhabi, 127788, UAE
| | - Lucia Gemma Delogu
- Department of Biomedical Sciences, University of Padua, Padua, Italy
- Department of Biological Science, Khalifa university of Science and Technology, Abu Dhabi, UAE
| | - Andrea C Ferrari
- Cambridge Graphene Centre, University of Cambridge, Cambridge, CB3 0FA, UK
| | - Carlo Saverio Iorio
- Centre for Research and Engineering in Space Technologies, École Polytechnique de Bruxelles, Université libre de Bruxelles, Brussels, Belgium.
| |
Collapse
|