1
|
Moghaddam AS, Dunne K, Breyer W, Wu Y, Pashuck ET. Hydrogels with multiple RGD presentations increase cell adhesion and spreading. Acta Biomater 2025:S1742-7061(25)00288-0. [PMID: 40254231 DOI: 10.1016/j.actbio.2025.04.037] [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: 12/11/2024] [Revised: 03/25/2025] [Accepted: 04/17/2025] [Indexed: 04/22/2025]
Abstract
A key challenge in designing hydrogels for cell culture is replicating the cell-matrix interactions found in tissues. Cells use integrins to bind their local matrix and form adhesions in which integrins dynamically move on the cell membrane while applying significant forces to the local matrix. Identifying the important biomaterial features for these interactions is challenging because it is difficult to independently adjust variables such as matrix stiffness, stress relaxation, the mobility of adhesion ligands, and the ability of these ligands to support cellular forces. In this work, we designed a hydrogel platform consisting of interpenetrating polymer networks of covalently crosslinked poly(ethylene glycol) (PEG) and self-assembled peptide amphiphiles (PA). We can tune the viscoelasticity of the hydrogel by modulating the composition of both networks. Ligand mobility can be adjusted independently of the matrix mechanical properties by attaching the arginine-glycine-aspartic acid (RGD) cell adhesion ligand to either the covalent PEG network, the dynamic PA network, or both networks at once. We find that endothelial cell adhesion formation and spreading is maximized in soft gels in which adhesion ligands are present on both the covalent and non-covalent networks. The dynamic nature of adhesion domains, coupled with their ability to exert substantial forces on the matrix, suggests that having different presentations of RGD ligands which are either mobile or capable of withstanding significant forces is needed to mimic different aspects of complex cell-matrix adhesions. These results will contribute to the design of hydrogels that better recapitulate physiological cell-matrix interactions. STATEMENT OF SIGNIFICANCE: Creating artificial environments that accurately mimic how cells interact with their surrounding matrix in natural tissues remains a fundamental challenge in biomaterials science. This study introduces a dual-network hydrogel platform that independently controls mechanical properties and adhesion ligand mobility by combining stable and dynamic polymer networks. A significant body of work has shown that matrix viscoelasticity and adhesion ligand mobility are important for cell adhesion and spreading. Our work builds on this by showing that endothelial cells function optimally when they can simultaneously engage with both mobile adhesion sites and force-resistant anchoring points, independent of matrix viscoelasticity. These insights will guide the design of more physiologically relevant hydrogels for tissue engineering applications and disease modeling.
Collapse
Affiliation(s)
| | | | - Wendy Breyer
- Department of Chemistry, Lehigh University, Bethlehem, PA 18015, USA
| | | | | |
Collapse
|
2
|
Kim TT, Malu D, He D, Hu Y, Kim J. Development of Bioorthogonally Degradable Tough Hydrogels Using Enamine N-Oxide Based Crosslinkers. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2414692. [PMID: 40018818 PMCID: PMC11962699 DOI: 10.1002/adma.202414692] [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: 09/27/2024] [Revised: 01/20/2025] [Indexed: 03/01/2025]
Abstract
Inducibly degradable polymers present new opportunities to integrate tough hydrogels into a wide range of biomaterials. Rapid and inducible degradation enables fast transition in material properties without sacrificing material integrity prior to removal. In pursuit of bioorthogonal chemical modalities that will enable inducible polymer degradation in biologically relevant environments, enamine N-oxide crosslinkers are developed for double network acrylamide-based polymer/alginate hydrogels. Bioorthogonal dissociation initiated by the application of aqueous diboron solution through several delivery mechanisms effectively lead to polymer degradation. Their degradation by aqueous B2(OH)4 solution results in a fracture energy half-life of <10 min. The biocompatibility of the degradable hydrogels and B2(OH)4 reagent is assessed, and the removability of strongly adhered tough hydrogels on mice skin is evaluated. Thermoresponsive PNiPAAm/Alg hydrogels are fabricated and application of the hydrogels as a chemically inducible degradable intraoral wound dressing is demonstrated. It is demonstrated through in vivo maximum tolerated dose studies that diboron solution administered to mice by oral gavage is well tolerated. Successful integration of enamine N-oxides within the tough double network hydrogels as chemically degradable motifs demonstrates the applicability of enamine N-oxides in the realm of polymer chemistry and highlights the importance of chemically induced bioorthogonal dissociation reactions for materials science.
Collapse
Affiliation(s)
- Thomas T. Kim
- School of Chemistry and BiochemistryGeorgia Institute of TechnologyAtlantaGA30332USA
- Department of Cancer BiologyDana‐Farber Cancer InstituteBostonMA02215USA
- Department of Biological Chemistry and Molecular PharmacologyHarvard Medical SchoolBostonMA02115USA
| | - Deep Malu
- George W. Woodruff School of Mechanical EngineeringGeorgia Institute of TechnologyAtlantaGA30332USA
- School of Chemical and Biomolecular EngineeringGeorgia Institute of TechnologyAtlantaGA30332USA
| | - Dongjing He
- George W. Woodruff School of Mechanical EngineeringGeorgia Institute of TechnologyAtlantaGA30332USA
- School of Chemical and Biomolecular EngineeringGeorgia Institute of TechnologyAtlantaGA30332USA
| | - Yuhang Hu
- George W. Woodruff School of Mechanical EngineeringGeorgia Institute of TechnologyAtlantaGA30332USA
- School of Chemical and Biomolecular EngineeringGeorgia Institute of TechnologyAtlantaGA30332USA
| | - Justin Kim
- School of Chemistry and BiochemistryGeorgia Institute of TechnologyAtlantaGA30332USA
- Department of Cancer BiologyDana‐Farber Cancer InstituteBostonMA02215USA
- Department of Biological Chemistry and Molecular PharmacologyHarvard Medical SchoolBostonMA02115USA
| |
Collapse
|
3
|
Friend NE, Zhang IW, Hu MM, McCoy AJ, Kent RN, DePalma SJ, Baker BM, Lesher-Pérez SC, Stegemann JP, Putnam AJ. Biofabrication and Characterization of Vascularizing PEG-Norbornene Microgels. J Biomed Mater Res A 2025; 113:e37900. [PMID: 40135515 PMCID: PMC11956422 DOI: 10.1002/jbm.a.37900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 03/04/2025] [Accepted: 03/08/2025] [Indexed: 03/27/2025]
Abstract
Establishing a robust, functional microvascular network remains a critical challenge for both the revascularization of damaged or diseased tissues and the development of engineered biological materials. Vascularizing microgels may aid in efforts to develop complex, multiphasic tissues by providing discrete, vascularized tissue modules that can be distributed throughout engineered constructs to vascularize large volumes. Here, we fabricated poly(ethylene glycol)-norbornene (PEGNB) microgels containing endothelial and stromal cells via flow-focusing microfluidic droplet generation. When embedded in bulk fibrin hydrogels, these cell-laden microgels initiated the formation and development of robust microvascular networks. Furthermore, extended preculture of cell-laden PEGNB microgels enabled the formation of vessel-like structures supported by basement membrane within the matrix without aggregation. Our findings highlight the suitability of PEG-based matrices for the development of vascularizing microgels capable of forming well-distributed, robust microvascular networks.
Collapse
Affiliation(s)
- Nicole E. Friend
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI
| | - Irene W. Zhang
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI
| | - Michael M. Hu
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI
| | - Atticus J. McCoy
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI
| | - Robert N. Kent
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI
| | - Samuel J. DePalma
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI
| | - Brendon M. Baker
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI
| | - Sasha Cai Lesher-Pérez
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI
| | - Jan P. Stegemann
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI
| | - Andrew J. Putnam
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI
| |
Collapse
|
4
|
Bokhari SMQ, Alioglu MA, Voronin GL, Catchmark JM. Mechanical, Thermal, and Rheological Properties of Fish-Porcine Gelatin Microparticle Composites for Advanced 3D Biofabrication. ACS APPLIED BIO MATERIALS 2025; 8:2614-2628. [PMID: 40030043 DOI: 10.1021/acsabm.4c01977] [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: 03/18/2025]
Abstract
Driven by the increasing need for the biofabrication of complex hydrogels, this work introduces a class of fish-porcine composite hydrogels that combine rapid, tunable photo-cross-linking with microparticle reinforcement for advanced 3D printing. Here, precross-linked porcine gelatin (methacrylated porcine gelatin, MPG) microparticles are incorporated into a methacrylated fish gelatin (MFG) matrix to produce robust yet easily processable hydrogels. Nuclear magnetic resonance (NMR) confirmed the degree of methacrylation, while scanning electron microscopy (SEM) revealed the hierarchical porosity vital for tissue integration. Detailed Mastersizer measurements characterized the size distributions of the MPG microparticles, and rheological tests demonstrated the composite hydrogels' strong shear-thinning behavior, an essential trait for extrusion-based and embedded 3D printing. Thermal (TGA, DSC) and mechanical (compression) analyses show that the microparticle-reinforced hydrogels achieve improved thermal stability, adjustable mass swelling ratio, and customizable compressive moduli. As a proof of concept, these composites are validated in digital light processing (DLP) printing of microfluidic constructs and as a support bath for embedded printing of complex geometries. This platform provides a unique synergy of easy UV cross-linkability, tunable mechanical features, and 3D printing versatility. This advancement underscores the potential of these materials as a foundational platform in tissue engineering, opening new avenues for creating complex, biocompatible structures with customizable properties.
Collapse
Affiliation(s)
- Syed M Q Bokhari
- Department of Agricultural and Biological Engineering, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
- Department of Electrical Engineering, The Technical University of Denmark, Kgs. Lyngby, Copenhagen 2800, Denmark
| | - Mecit A Alioglu
- Department of Engineering Science and Mechanics, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
- The Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Grace L Voronin
- Department of Food Science, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Jeffrey M Catchmark
- Department of Agricultural and Biological Engineering, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| |
Collapse
|
5
|
Anuța V, Nica MA, Prisada RM, Popa L, Velescu BȘ, Marinas IC, Gaboreanu DM, Ghica MV, Cocoș FI, Nicolae CA, Dinu-Pîrvu CE. Novel Buccal Xanthan Gum-Hyaluronic Acid Eutectogels with Dual Anti-Inflammatory and Antimicrobial Properties. Gels 2025; 11:208. [PMID: 40136913 PMCID: PMC11942315 DOI: 10.3390/gels11030208] [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: 08/22/2024] [Revised: 03/03/2025] [Accepted: 03/10/2025] [Indexed: 03/27/2025] Open
Abstract
Buccal drug delivery systems often struggle with poor drug solubility, limited adhesion, and rapid clearance, leading to suboptimal therapeutic outcomes. To address these limitations, we developed a novel hybrid eutectogel composed of xanthan gum (XTG), hyaluronic acid (HA), and a Natural Deep Eutectic Solvent (NADES) system (choline chloride, sorbitol, and glycerol in 2:1:1 mole ratio), incorporating 2.5% ibuprofen (IBU) as a model drug. The formulation was optimized using a face-centered central composite design to enhance the rheological, textural, and drug release properties. The optimized eutectogels exhibited shear-thinning behavior (flow behavior index, n = 0.26 ± 0.01), high mucoadhesion (adhesiveness: 2.297 ± 0.142 N·s), and sustained drug release over 24 h, governed by Higuchi kinetics (release rate: 237.34 ± 13.61 μg/cm2/min1/2). The ex vivo residence time increased substantially with NADES incorporation, reaching up to 176.7 ± 23.1 min. An in vivo anti-inflammatory evaluation showed that the eutectogel reduced λ-carrageenan-induced paw edema within 1 h and that its efficacy was sustained in the kaolin model up to 24 h (p < 0.05), achieving comparable efficacy to a commercial 5% IBU gel, despite a lower drug concentration. Additionally, the eutectogel presented a minimum inhibitory concentration for Gram-positive bacteria of 25 mg/mL, and through direct contact, it reduced microbial viability by up to 100%. Its efficacy against Bacillus cereus, Enterococcus faecium, and Klebsiella pneumoniae, combined with its significant anti-inflammatory properties, positions the NADES-based eutectogel as a promising multifunctional platform for buccal drug delivery, particularly for inflammatory conditions complicated by bacterial infections.
Collapse
Affiliation(s)
- Valentina Anuța
- Department of Physical and Colloidal Chemistry, Faculty of Pharmacy, “Carol Davila” University of Medicine and Pharmacy, 6 Traian Vuia Street, 020956 Bucharest, Romania; (V.A.); (M.-A.N.); (L.P.); (M.V.G.); (F.I.C.); (C.-E.D.-P.)
- Innovative Therapeutic Structures Research and Development Centre (InnoTher), “Carol Davila” University of Medicine and Pharmacy, 6 Traian Vuia Street, 020956 Bucharest, Romania;
| | - Mihaela-Alexandra Nica
- Department of Physical and Colloidal Chemistry, Faculty of Pharmacy, “Carol Davila” University of Medicine and Pharmacy, 6 Traian Vuia Street, 020956 Bucharest, Romania; (V.A.); (M.-A.N.); (L.P.); (M.V.G.); (F.I.C.); (C.-E.D.-P.)
- Innovative Therapeutic Structures Research and Development Centre (InnoTher), “Carol Davila” University of Medicine and Pharmacy, 6 Traian Vuia Street, 020956 Bucharest, Romania;
| | - Răzvan-Mihai Prisada
- Department of Physical and Colloidal Chemistry, Faculty of Pharmacy, “Carol Davila” University of Medicine and Pharmacy, 6 Traian Vuia Street, 020956 Bucharest, Romania; (V.A.); (M.-A.N.); (L.P.); (M.V.G.); (F.I.C.); (C.-E.D.-P.)
- Innovative Therapeutic Structures Research and Development Centre (InnoTher), “Carol Davila” University of Medicine and Pharmacy, 6 Traian Vuia Street, 020956 Bucharest, Romania;
| | - Lăcrămioara Popa
- Department of Physical and Colloidal Chemistry, Faculty of Pharmacy, “Carol Davila” University of Medicine and Pharmacy, 6 Traian Vuia Street, 020956 Bucharest, Romania; (V.A.); (M.-A.N.); (L.P.); (M.V.G.); (F.I.C.); (C.-E.D.-P.)
- Innovative Therapeutic Structures Research and Development Centre (InnoTher), “Carol Davila” University of Medicine and Pharmacy, 6 Traian Vuia Street, 020956 Bucharest, Romania;
| | - Bruno Ștefan Velescu
- Innovative Therapeutic Structures Research and Development Centre (InnoTher), “Carol Davila” University of Medicine and Pharmacy, 6 Traian Vuia Street, 020956 Bucharest, Romania;
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, “Carol Davila” University of Medicine and Pharmacy, 6 Traian Vuia Street, 020956 Bucharest, Romania
| | - Ioana Cristina Marinas
- Research Institute of the University of Bucharest—ICUB, University of Bucharest, 91–95 Spl. Independentei, 050095 Bucharest, Romania; (I.C.M.); (D.-M.G.)
| | - Diana-Madalina Gaboreanu
- Research Institute of the University of Bucharest—ICUB, University of Bucharest, 91–95 Spl. Independentei, 050095 Bucharest, Romania; (I.C.M.); (D.-M.G.)
- Departament of Botany and Microbiology, Faculty of Biology, University of Bucharest, Splaiul Independentei 91-95, 050095 Bucharest, Romania
| | - Mihaela Violeta Ghica
- Department of Physical and Colloidal Chemistry, Faculty of Pharmacy, “Carol Davila” University of Medicine and Pharmacy, 6 Traian Vuia Street, 020956 Bucharest, Romania; (V.A.); (M.-A.N.); (L.P.); (M.V.G.); (F.I.C.); (C.-E.D.-P.)
- Innovative Therapeutic Structures Research and Development Centre (InnoTher), “Carol Davila” University of Medicine and Pharmacy, 6 Traian Vuia Street, 020956 Bucharest, Romania;
| | - Florentina Iuliana Cocoș
- Department of Physical and Colloidal Chemistry, Faculty of Pharmacy, “Carol Davila” University of Medicine and Pharmacy, 6 Traian Vuia Street, 020956 Bucharest, Romania; (V.A.); (M.-A.N.); (L.P.); (M.V.G.); (F.I.C.); (C.-E.D.-P.)
- Innovative Therapeutic Structures Research and Development Centre (InnoTher), “Carol Davila” University of Medicine and Pharmacy, 6 Traian Vuia Street, 020956 Bucharest, Romania;
| | - Cristian Andi Nicolae
- National Institute for Research and Development in Chemistry and Petrochemistry—ICECHIM Bucharest, 202 Spl. Independentei, 060021 Bucharest, Romania;
| | - Cristina-Elena Dinu-Pîrvu
- Department of Physical and Colloidal Chemistry, Faculty of Pharmacy, “Carol Davila” University of Medicine and Pharmacy, 6 Traian Vuia Street, 020956 Bucharest, Romania; (V.A.); (M.-A.N.); (L.P.); (M.V.G.); (F.I.C.); (C.-E.D.-P.)
- Innovative Therapeutic Structures Research and Development Centre (InnoTher), “Carol Davila” University of Medicine and Pharmacy, 6 Traian Vuia Street, 020956 Bucharest, Romania;
| |
Collapse
|
6
|
Serpe F, Nalin F, Tirelli MC, Posabella P, Celikkin N, Jaroszewicz J, Święszkowski W, Barbetta A, Şentürk E, Casciola CM, Ruocco G, Cidonio G, Scognamiglio C, Costantini M. Microfluidic 3D Bioprinting of Foamed Fibers with Controlled Micromorphology. ACS APPLIED MATERIALS & INTERFACES 2025; 17:13632-13645. [PMID: 39964244 DOI: 10.1021/acsami.4c22450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2025]
Abstract
The synergistic integration of microfluidic technologies with additive manufacturing systems is advancing the development of innovative platforms to 3D bioprint scaffolds for tissue engineering with unparalleled biological relevance. Significant interest is growing in realizing porous functionally graded materials (pFGMs) that can resemble the hierarchical organization of porosity found in bone tissue. This study introduces a method for fabricating porous scaffolds based on the real-time generation of a liquid foam, which is gelled, forming porous fibers that are organized into structured matrixes using a 3D bioprinting system. The primary advantage of this approach is the possibility to adjust bubble size during printing dynamically, modifying the characteristics of the deposited foamed filaments online and in one step. As a result, locally-defined and tailor-made pores can be distributed in 3D structures with high spatial accuracy. Besides the mechanical and morphological characterization of diverse microarchitectures, we also explored the biocompatibility of the proposed approach by directly embedding osteosarcoma cells within the biomaterial. Results demonstrated the biocompatibility of the proposed methodology and revealed the influence of the interior microporosity on cell proliferation, highlighting the potential for creating tailored tissue microenvironments. The findings underscore the versatility of the presented 3D bioprinting system and its potential in fabricating biomimetic scaffolds with tailored morphological gradients, representing a substantial advancement in pFGM synthesis, with direct implications in regenerative medicine and tissue engineering.
Collapse
Affiliation(s)
- Federico Serpe
- Department of Chemistry, University of Rome "La Sapienza", 00185 Rome, Italy
- Center for Life Nano- & Neuro-Science - CLN2S, Italian Institute of Technology (IIT), 00161 Rome, Italy
| | - Francesco Nalin
- Institute of Physical Chemistry, Polish Academy of Sciences, 01224 Warsaw, Poland
| | | | - Pasquale Posabella
- Warsaw University of Technology, Faculty of Materials Science and Engineering, 02507 Warsaw, Poland
| | - Nehar Celikkin
- Institute of Physical Chemistry, Polish Academy of Sciences, 01224 Warsaw, Poland
| | - Jakub Jaroszewicz
- Warsaw University of Technology, Faculty of Materials Science and Engineering, 02507 Warsaw, Poland
| | - Wojciech Święszkowski
- Warsaw University of Technology, Faculty of Materials Science and Engineering, 02507 Warsaw, Poland
| | - Andrea Barbetta
- Department of Chemistry, University of Rome "La Sapienza", 00185 Rome, Italy
| | - Efsun Şentürk
- Center for Life Nano- & Neuro-Science - CLN2S, Italian Institute of Technology (IIT), 00161 Rome, Italy
- Department of Mechanical and Aerospace Engineering (DIMA), University of Rome "La Sapienza", 00184 Rome, Italy
| | - Carlo Massimo Casciola
- Department of Mechanical and Aerospace Engineering (DIMA), University of Rome "La Sapienza", 00184 Rome, Italy
| | - Giancarlo Ruocco
- Center for Life Nano- & Neuro-Science - CLN2S, Italian Institute of Technology (IIT), 00161 Rome, Italy
| | - Gianluca Cidonio
- Center for Life Nano- & Neuro-Science - CLN2S, Italian Institute of Technology (IIT), 00161 Rome, Italy
- Department of Mechanical and Aerospace Engineering (DIMA), University of Rome "La Sapienza", 00184 Rome, Italy
| | - Chiara Scognamiglio
- Center for Life Nano- & Neuro-Science - CLN2S, Italian Institute of Technology (IIT), 00161 Rome, Italy
| | - Marco Costantini
- Institute of Physical Chemistry, Polish Academy of Sciences, 01224 Warsaw, Poland
| |
Collapse
|
7
|
Li R, Wang J, Lin Q, Yin Z, Zhou F, Chen X, Tan H, Su J. Mechano-Responsive Biomaterials for Bone Organoid Construction. Adv Healthc Mater 2025; 14:e2404345. [PMID: 39740101 DOI: 10.1002/adhm.202404345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 12/08/2024] [Indexed: 01/02/2025]
Abstract
Mechanical force is essential for bone development, bone homeostasis, and bone fracture healing. In the past few decades, various biomaterials have been developed to provide mechanical signals that mimic the natural bone microenvironment, thereby promoting bone regeneration. Bone organoids, emerging as a novel research approach, are 3D micro-bone tissues that possess the ability to self-renew and self-organize, exhibiting biomimetic spatial characteristics. Incorporating mechano-responsive biomaterials in the construction of bone organoids presents a promising avenue for simulating the mechanical bone microenvironment. Therefore, this review commences by elucidating the impact of mechanical force on bone health, encompassing both cellular interactions and alterations in bone structure. Furthermore, the most recent applications of mechano-responsive biomaterials within the realm of bone tissue engineering are highlighted. Three different types of mechano-responsive biomaterials are introduced with a focus on their responsive mechanisms, construction strategies, and efficacy in facilitating bone regeneration. Based on a comprehensive overview, the prospective utilization and future challenges of mechano-responsive biomaterials in the construction of bone organoids are discussed. As bone organoid technology advances, these biomaterials are poised to become powerful tools in bone regeneration.
Collapse
Affiliation(s)
- Ruiyang Li
- Department of Orthopedics, Trauma Orthopedics Center, Institute of Musculoskeletal Injury and Translational Medicine of Organoids, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, P. R. China
- Institute of Translational Medicine, National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai, 200444, P. R. China
| | - Jian Wang
- Department of Orthopedics, Trauma Orthopedics Center, Institute of Musculoskeletal Injury and Translational Medicine of Organoids, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, P. R. China
- Institute of Translational Medicine, National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai, 200444, P. R. China
| | - Qiushui Lin
- Department of Spine Surgery, First Affiliated Hospital of Naval Medical University, Shanghai, 200433, P. R. China
| | - Zhifeng Yin
- Department of Orthopedics, Shanghai Zhongye Hospital, Shanghai, 200941, P. R. China
| | - Fengjin Zhou
- Department of Orthopedics, Honghui Hospital, Xi'an Jiao Tong University, Xi'an, 710000, P. R. China
| | - Xiao Chen
- Department of Orthopedics, Trauma Orthopedics Center, Institute of Musculoskeletal Injury and Translational Medicine of Organoids, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, P. R. China
| | - Hongbo Tan
- Department of Orthopedics, The 920th Hospital of Joint Logistics Support Force, Yunnan, 650020, P. R. China
| | - Jiacan Su
- Department of Orthopedics, Trauma Orthopedics Center, Institute of Musculoskeletal Injury and Translational Medicine of Organoids, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, P. R. China
- Institute of Translational Medicine, National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai, 200444, P. R. China
| |
Collapse
|
8
|
Aye SSS, Fang Z, Wu MCL, Lim KS, Ju LA. Integrating microfluidics, hydrogels, and 3D bioprinting for personalized vessel-on-a-chip platforms. Biomater Sci 2025; 13:1131-1160. [PMID: 39834160 DOI: 10.1039/d4bm01354a] [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: 01/22/2025]
Abstract
Thrombosis, a major cause of morbidity and mortality worldwide, presents a complex challenge in cardiovascular medicine due to the intricacy of clotting mechanisms in living organisms. Traditional research approaches, including clinical studies and animal models, often yield conflicting results due to the inability to control variables in these complex systems, highlighting the need for more precise investigative tools. This review explores the evolution of in vitro thrombosis models, from conventional polydimethylsiloxane (PDMS)-based microfluidic devices to advanced hydrogel-based systems and cutting-edge 3D bioprinted vascular constructs. We discuss how these emerging technologies, particularly vessel-on-a-chip platforms, are enabling researchers to control previously unmanageable factors, thereby offering unprecedented opportunities to pinpoint specific clotting mechanisms. While PDMS-based devices offer optical transparency and fabrication ease, their inherent limitations, including non-physiological rigidity and surface properties, have driven the development of hydrogel-based systems that better mimic the extracellular matrix of blood vessels. The integration of microfluidics with biomimetic materials and tissue engineering approaches has led to the development of sophisticated models capable of simulating patient-specific vascular geometries, flow dynamics, and cellular interactions under highly controlled conditions. The advent of 3D bioprinting further enables the creation of complex, multi-layered vascular structures with precise spatial control over geometry and cellular composition. Despite significant progress, challenges remain in achieving long-term stability, incorporating immune components, and translating these models to clinical applications. By providing a comprehensive overview of current advancements and future prospects, this review aims to stimulate further innovation in thrombosis research and accelerate the development of more effective, personalized approaches to thrombosis prevention and treatment.
Collapse
Affiliation(s)
- San Seint Seint Aye
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW 2008, Australia.
| | - Zhongqi Fang
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW 2008, Australia.
| | - Mike C L Wu
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW 2008, Australia.
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW 2006, Australia.
| | - Khoon S Lim
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW 2006, Australia.
- School of Medical Sciences, The University of Sydney, Camperdown, NSW 2006, Australia
| | - Lining Arnold Ju
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW 2008, Australia.
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW 2006, Australia.
- The University of Sydney Nano Institute (Sydney Nano), The University of Sydney, Camperdown, NSW 2006, Australia
- Heart Research Institute, Newtown, NSW 2042, Australia
| |
Collapse
|
9
|
Carvalho AF, Pereira T, Oliveira C, Figueiredo P, Carvalho A, Pereira DM, Hilliou L, Bañobre-López M, Xu B, Ferreira PMT, Martins JA. Tripeptides Featuring Dehydrophenylalanine and Homophenylalanine: Homo- Versus Hetero-Chirality and Sequence Effects on Self-Assembly and Gelation. Gels 2025; 11:164. [PMID: 40136869 PMCID: PMC11942182 DOI: 10.3390/gels11030164] [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: 01/12/2025] [Revised: 02/14/2025] [Accepted: 02/19/2025] [Indexed: 03/27/2025] Open
Abstract
Over the years, our research group developed dehydrodipeptides N-capped with aromatic moieties as protease-resistant efficacious hydrogelators, affording self-assembled hydrogels at low (critical) concentrations. Dehydrotripeptides, with different dipeptide sequences and (D,L) stereochemistry, open a wider chemical space for the development of self-assembled soft nanomaterials. In this work, a small library of N-succinylated dehydrotripeptides containing a C-terminal dehydrophenylalanine (∆Phe) residue and a scrambled dipeptide sequence with phenylalanine (Phe) and homophenylalanine (Hph) (L-Phe-L,D-Hph and L,D-Hph-L-Phe) was synthesized and characterized as a potential hydrogelator. Two pairs of diastereomeric tripeptides were synthesized, both as C-protected methyl esters and as deprotected dicarboxylic acids. Peptides with the sequence Hph-Phe-ΔPhe were obtained as a pair (D,L,Z)/(L,L,Z) of diastereomers. Their scrambled sequence analogues Phe-Hph-ΔPhe were obtained also as a diastereomeric (L,D,Z)/(L,L,Z) pair. The effect of stereochemistry (homo- vs. hetero-chirality) and sequence (Phe-∆Phe vs. Hph-∆Phe motif) on the self-assembly, biocompatibility, gelation and rheological properties of the hydrogels was studied in this work. Accessible, both as C-protected methyl esters and as dicarboxylic acids, N-succinylated dehydrotripeptides are interesting molecular architectures for the development of supramolecular nanomaterials. Interestingly, our results do not comply with the well-documented proposition that heterochiral peptides display much higher self-assembly propensity and gelation ability than their homochiral counterparts. Further studies will be necessary to fully understand the interplay between peptide sequence and homo- and hetero-chirality on peptide self-assembly and on the properties of their supramolecular materials.
Collapse
Affiliation(s)
- André F. Carvalho
- Center of Chemistry, University of Minho, 4710-057 Braga, Portugal (C.O.)
| | - Teresa Pereira
- Center of Chemistry, University of Minho, 4710-057 Braga, Portugal (C.O.)
| | - Carlos Oliveira
- Center of Chemistry, University of Minho, 4710-057 Braga, Portugal (C.O.)
| | - Pedro Figueiredo
- CNC—Center for Neuroscience and Cell Biology, Institute for Interdisciplinary Research (IIIUC), University of Coimbra, 3004-504 Coimbra, Portugal
- PhD Programme in Experimental Biology and Biomedicine, Institute for Interdisciplinary Research (IIIUC), University of Coimbra, Casa Costa Alemão, 3030-789 Coimbra, Portugal
| | - Alexandra Carvalho
- CNC—Center for Neuroscience and Cell Biology, Institute for Interdisciplinary Research (IIIUC), University of Coimbra, 3004-504 Coimbra, Portugal
- Almac Sciences, Department of Biocatalysis and Isotope Chemistry, Almac House, 20 Seagoe Industrial Estate, Craigavon BT63 5QD, UK
| | - David M. Pereira
- REQUIMTE/LAQV, Laboratório de Farmacognosia, Departamento de Química, Faculdade de Farmácia, Universidade do Porto, R. Jorge Viterbo Ferreira, n 228, 4050-313 Porto, Portugal
| | - Loic Hilliou
- Institute for Polymers and Composites, University of Minho, 4800-058 Guimarães, Portugal;
| | - Manuel Bañobre-López
- International Iberian Nanotechnology Laboratory (INL), Av. Mestre José Veiga s/n, 4715-330 Braga, Portugal;
| | - Bing Xu
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA 02453, USA
| | | | - José A. Martins
- Center of Chemistry, University of Minho, 4710-057 Braga, Portugal (C.O.)
| |
Collapse
|
10
|
Yang ZC, Lin H, Liu GJ, Pan H, Zhu JL, Zhang XH, Gao F, Wang Z, Wang ZH. CB-MNCs@ CS/HEC/GP promote wound healing in aged murine pressure ulcer model. Stem Cell Res Ther 2025; 16:52. [PMID: 39920794 PMCID: PMC11806723 DOI: 10.1186/s13287-025-04177-w] [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: 06/20/2024] [Accepted: 01/23/2025] [Indexed: 02/09/2025] Open
Abstract
BACKGROUND Non-healing pressure ulcers impose heavy burdens on patients and clinicians. Cord blood mononuclear cells (CB-MNCs) are a novel type of tissue repair seed cells. However, their clinical application is restricted by low retention and survival rates post-transplantation. This study aims to investigate the role of thermo-sensitive chitosan/hydroxyethyl cellulose/glycerophosphate (CS/HEC/GP) hydrogel encapsulated CB-MNCs in pressure ulcer wound healing. METHODS Pressure ulcers were induced on the backs of aged mice. After construction and validation of the characterization of thermo-sensitive CS/HEC/GP hydrogel, CB-MNCs are encapsulated in the hydrogel, called CB-MNCs@CS/HEC/GP which was locally applied to the mouse wounds. Mouse skin tissues were harvested for histological and molecular biology analyses. RESULTS CB-MNCs@CS/HEC/GP therapy accelerated pressure ulcer wound healing, attenuated inflammatory responses, promoted cell proliferation, angiogenesis, and collagen synthesis. Further investigation revealed that CB-MNCs@CS/HEC/GP exerted therapeutic effects by promoting changes in cell types, including fibroblasts, endothelial cells, keratinocytes, and smooth muscle cells. CONCLUSION CB-MNCs@CS/HEC/GP enhanced the delivery efficiency of CB-MNCs, preserved the cell viability, and contributed to pressure ulcer wound healing. Thus, CB-MNCs@CS/HEC/GP represents a novel therapeutic approach for skin regeneration of chronic wounds.
Collapse
Affiliation(s)
- Zhi-Cheng Yang
- Department of Geriatric Medicine & Laboratory of Gerontology and Anti-Aging Research, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
- School of Nursing and Rehabilitation, Shandong University, Jinan, 250012, Shandong, China
| | - He Lin
- Department of Geriatric Medicine & Laboratory of Gerontology and Anti-Aging Research, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Guo-Jun Liu
- Shandong Qilu Stem Cell Engineering Co., Ltd, Jinan, 250012, Shandong, China
| | - Hui Pan
- Department of Geriatric Medicine & Laboratory of Gerontology and Anti-Aging Research, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Jun-Lu Zhu
- School of Nursing and Rehabilitation, Shandong University, Jinan, 250012, Shandong, China
| | - Xiao-Hong Zhang
- School of Nursing and Rehabilitation, Shandong University, Jinan, 250012, Shandong, China
| | - Feng Gao
- Shandong Qilu Stem Cell Engineering Co., Ltd, Jinan, 250012, Shandong, China
| | - Zhong Wang
- Shandong Qilu Stem Cell Engineering Co., Ltd, Jinan, 250012, Shandong, China
| | - Zhi-Hao Wang
- Department of Geriatric Medicine & Laboratory of Gerontology and Anti-Aging Research, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China.
| |
Collapse
|
11
|
Zhang H, Li X, Jia Z, Jiao K, Liu C, Deng Z, Bai Y, Wei X, Zhou X. Bioprinted hydrogels in bone regeneration: a bibliometric analysis. Front Pharmacol 2025; 16:1532629. [PMID: 39963238 PMCID: PMC11830744 DOI: 10.3389/fphar.2025.1532629] [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: 11/22/2024] [Accepted: 01/13/2025] [Indexed: 02/20/2025] Open
Abstract
Background The application of bioprinted hydrogels in the field of bone regeneration is garnering increasing attention. The objective of this study is to provide a comprehensive overview of the current research status, hotspots and research directions in this field through bibliometric methods, and to predict the development trend of this field. Methods A search was conducted on 27 December 2024, for papers published on the Web of Science from 2010 to 2025. We used the bibliometrix package in the software program R to analyze the retrieved data and VOSviewer and CiteSpace to visualize hotspots and research trends in bioprinted hydrogels for bone regeneration. Results We identified and reviewed 684 articles published in this field between 2010 and 2025. A total of 811 institutions and 1,166 researchers from 41 countries/regions contributed to these publications. Among them, China led in terms of the number of articles published, single-country publications (SCP), and multi-country publications (MCP). Our bibliometric-based visualization analysis revealed that the mechanical properties and osteogenic differentiation capacity of biomaterials have been a focal research topic over the past decade, while emerging research has also concentrated on the in vitro fabrication of stem cells for bone regeneration and osteogenic differentiation, particularly the precise application of in situ stem cell-loaded bioprinted organoids. Conclusion This study provides an in-depth analysis of the research trajectory in the application of bioprinted hydrogels for bone regeneration. The number of research papers in this field is increasing annually, and the main research hotspots include bone regeneration, 3D printing, scaffolds, and hydrogels. Future research directions may focus on gelatin, additive manufacturing, and growth factors. Additionally, international collaboration is essential to enhance the effectiveness of bioprinted hydrogels in bone regeneration applications.
Collapse
Affiliation(s)
- Huijie Zhang
- Department of Orthopedics, Shanghai Changhai Hospital, Shanghai, China
| | - Xiaoyu Li
- Department of Orthopedics, Shanghai Changhai Hospital, Shanghai, China
| | - Zhenyu Jia
- Department of Orthopedics, General Hospital of Southern Theater Command, Guangzhou, China
| | - Kun Jiao
- Department of Orthopedics, Shanghai Changhai Hospital, Shanghai, China
- Department of Orthopedics, Shanghai Changzheng Hospital, Shanghai, China
| | - Chen Liu
- Department of Orthopedics, Shanghai Changhai Hospital, Shanghai, China
- Department of Outpatient Service, Military District Shenyang No. 1 Retreat Center for Separated Cadres, Liaoning, China
| | - Zixiang Deng
- Department of Orthopedics, Shanghai Changhai Hospital, Shanghai, China
| | - Yushu Bai
- Department of Orthopedics, Shanghai Changhai Hospital, Shanghai, China
| | - Xianzhao Wei
- Department of Orthopedics, Shanghai Changhai Hospital, Shanghai, China
| | - Xiaoyi Zhou
- Department of Orthopedics, Shanghai Changhai Hospital, Shanghai, China
| |
Collapse
|
12
|
Sim B, Chang JJ, Lin Q, Wong JHM, Ow V, Leow Y, Wong YJ, Boo YJ, Goh R, Loh XJ. Hydrogels Based on Polyelectrolyte Complexes: Underlying Principles and Biomedical Applications. Biomacromolecules 2024; 25:7563-7580. [PMID: 39557662 DOI: 10.1021/acs.biomac.4c01240] [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: 11/20/2024]
Abstract
Ionic complexes of electrostatically charged biomacromolecules are key players in various biological processes like nucleotide transportation, organelle formation, and protein folding. These complexes, abundant in biological systems, contribute to the function, responsiveness, and mechanical properties of organisms. Coherent with these natural phenomena, hydrogels formed through the complexation of oppositely charged polymers exhibit unique attributes, such as rapid self-assembly, hierarchical microstructures, tunable properties, and protective encapsulation. Consequently, polyelectrolyte complex (PEC) hydrogels have garnered considerable interest, emerging as an up-and-coming platform for various biomedical applications. This review outlines the underlying principles governing PEC hydrogels. The classification of polyelectrolytes and the self-assembly of PEC hydrogels are discussed, including the factors influencing their self-assembly process. Recent developments of PEC hydrogels for biomedical applications, including drug delivery, tissue engineering, wound healing and management, and wearable sensors, are summarized. This review concludes with the prospective directions for the next generation of PEC hydrogel research.
Collapse
Affiliation(s)
- Belynn Sim
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, #08-03 Innovis, Singapore 138634, Republic of Singapore
- School of Materials Science and Engineering, Nanyang Technological University (NTU), Singapore 639798, Republic of Singapore
| | - Jun Jie Chang
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, #08-03 Innovis, Singapore 138634, Republic of Singapore
| | - Qianyu Lin
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, #08-03 Innovis, Singapore 138634, Republic of Singapore
| | - Joey Hui Min Wong
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, #08-03 Innovis, Singapore 138634, Republic of Singapore
| | - Valerie Ow
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, #08-03 Innovis, Singapore 138634, Republic of Singapore
- Department of Biomedical Engineering, National University of Singapore (NUS), 4 Engineering Drive 3, Singapore 117583, Singapore
| | - Yihao Leow
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, #08-03 Innovis, Singapore 138634, Republic of Singapore
- School of Materials Science and Engineering, Nanyang Technological University (NTU), Singapore 639798, Republic of Singapore
| | - Yi Jing Wong
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, #08-03 Innovis, Singapore 138634, Republic of Singapore
- School of Materials Science and Engineering, Nanyang Technological University (NTU), Singapore 639798, Republic of Singapore
| | - Yi Jian Boo
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, #08-03 Innovis, Singapore 138634, Republic of Singapore
| | - Rubayn Goh
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, #08-03 Innovis, Singapore 138634, Republic of Singapore
| | - Xian Jun Loh
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, #08-03 Innovis, Singapore 138634, Republic of Singapore
- School of Materials Science and Engineering, Nanyang Technological University (NTU), Singapore 639798, Republic of Singapore
- Department of Materials Science and Engineering, National University of Singapore (NUS), 9 Engineering Drive, Singapore 117576, Singapore
| |
Collapse
|
13
|
Wu KY, Osman R, Kearn N, Kalevar A. Three-Dimensional Bioprinting for Retinal Tissue Engineering. Biomimetics (Basel) 2024; 9:733. [PMID: 39727737 DOI: 10.3390/biomimetics9120733] [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: 11/01/2024] [Revised: 11/20/2024] [Accepted: 11/25/2024] [Indexed: 12/28/2024] Open
Abstract
Three-dimensional bioprinting (3DP) is transforming the field of regenerative medicine by enabling the precise fabrication of complex tissues, including the retina, a highly specialized and anatomically complex tissue. This review provides an overview of 3DP's principles, its multi-step process, and various bioprinting techniques, such as extrusion-, droplet-, and laser-based methods. Within the scope of biomimicry and biomimetics, emphasis is placed on how 3DP potentially enables the recreation of the retina's natural cellular environment, structural complexity, and biomechanical properties. Focusing on retinal tissue engineering, we discuss the unique challenges posed by the retina's layered structure, vascularization needs, and the complex interplay between its numerous cell types. Emphasis is placed on recent advancements in bioink formulations, designed to emulate retinal characteristics and improve cell viability, printability, and mechanical stability. In-depth analyses of bioinks, scaffold materials, and emerging technologies, such as microfluidics and organ-on-a-chip, highlight the potential of bioprinted models to replicate retinal disease states, facilitating drug development and testing. While challenges remain in achieving clinical translation-particularly in immune compatibility and long-term integration-continued innovations in bioinks and scaffolding are paving the way toward functional retinal constructs. We conclude with insights into future research directions, aiming to refine 3DP for personalized therapies and transformative applications in vision restoration.
Collapse
Affiliation(s)
- Kevin Y Wu
- Department of Surgery, Division of Ophthalmology, University of Sherbrooke, Sherbrooke, QC J1G 2E8, Canada
| | - Rahma Osman
- Department of Medicine, School of Medicine, Queen's University, Kingston, ON K7L 3N6, Canada
| | - Natalie Kearn
- Department of Medicine, School of Medicine, Queen's University, Kingston, ON K7L 3N6, Canada
| | - Ananda Kalevar
- Department of Surgery, Division of Ophthalmology, University of Sherbrooke, Sherbrooke, QC J1G 2E8, Canada
| |
Collapse
|
14
|
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
|
15
|
Schrope JH, Horn A, Lazorchak K, Tinnen CW, Stevens JJ, Farooqui M, Li J, Bennin D, Robertson T, Juang TD, Ahmed A, Li C, Huttenlocher A, Beebe D. Confinement by liquid-liquid interface replicates in vivo neutrophil deformations and elicits bleb based migration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.06.14.544898. [PMID: 38106211 PMCID: PMC10723256 DOI: 10.1101/2023.06.14.544898] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Leukocytes navigate through interstitial spaces resulting in deformation of both the motile leukocytes and surrounding cells. Creating an in vitro system that models the deformable cellular environment encountered in vivo has been challenging. Here, we engineer microchannels with a liquid-liquid interface that exerts confining pressures (200-3000 Pa) similar to cells in tissues, and, thus, is deformable by cell generated forces. Consequently, the balance between migratory cell-generated and interfacial pressures determines the degree of confinement. Pioneer cells that first contact the interfacial barrier require greater deformation forces to forge a path for migration, and as a result migrate slower than trailing cells. Critically, resistive pressures are tunable by controlling the curvature of the liquid interface, which regulates motility. By granting cells autonomy in determining their confinement, and tuning environmental resistance, interfacial deformations are made to match those of surrounding cells in vivo during interstitial neutrophil migration in a larval zebrafish model. We discover that, in this context, neutrophils employ a bleb-based mechanism of force generation to deform a barrier exerting cell-scale confining pressures.
Collapse
|
16
|
Anggelia MR, Cheng HY, Lin CH. Thermosensitive Hydrogels as Targeted and Controlled Drug Delivery Systems: Potential Applications in Transplantation. Macromol Biosci 2024; 24:e2400064. [PMID: 38991045 DOI: 10.1002/mabi.202400064] [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: 02/14/2024] [Revised: 06/27/2024] [Indexed: 07/13/2024]
Abstract
Drug delivery in transplantation plays a vital role in promoting graft survival, preventing rejection, managing complications, and contributing to positive patient outcomes. Targeted and controlled drug delivery can minimize systemic effects. Thermosensitive hydrogels, due to their unique sol-gel transition properties triggered by thermo-stimuli, have attracted significant research interest as a potential drug delivery system in transplantation. This review describes the current status, characteristics, and recent applications of thermosensitive hydrogels for drug delivery. Studies aimed at improving allotransplantation outcomes using thermosensitive hydrogels are then elaborated on. Finally, the challenges and opportunities associated with their use are discussed. Understanding the progress of research will serve as a guide for future improvements in their application as a means of targeted and controlled drug delivery in translational therapeutic applications for transplantation.
Collapse
Affiliation(s)
- Madonna Rica Anggelia
- Center for Vascularized Composite Allotransplantation, Department of Plastic and Reconstructive Surgery, Chang Gung Memorial Hospital, Chang Gung Medical College and Chang Gung University, Taoyuan, 333, Taiwan
| | - Hui-Yun Cheng
- Center for Vascularized Composite Allotransplantation, Department of Plastic and Reconstructive Surgery, Chang Gung Memorial Hospital, Chang Gung Medical College and Chang Gung University, Taoyuan, 333, Taiwan
| | - Cheng-Hung Lin
- Center for Vascularized Composite Allotransplantation, Department of Plastic and Reconstructive Surgery, Chang Gung Memorial Hospital, Chang Gung Medical College and Chang Gung University, Taoyuan, 333, Taiwan
| |
Collapse
|
17
|
Mierke CT. Bioprinting of Cells, Organoids and Organs-on-a-Chip Together with Hydrogels Improves Structural and Mechanical Cues. Cells 2024; 13:1638. [PMID: 39404401 PMCID: PMC11476109 DOI: 10.3390/cells13191638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 09/25/2024] [Accepted: 10/01/2024] [Indexed: 10/19/2024] Open
Abstract
The 3D bioprinting technique has made enormous progress in tissue engineering, regenerative medicine and research into diseases such as cancer. Apart from individual cells, a collection of cells, such as organoids, can be printed in combination with various hydrogels. It can be hypothesized that 3D bioprinting will even become a promising tool for mechanobiological analyses of cells, organoids and their matrix environments in highly defined and precisely structured 3D environments, in which the mechanical properties of the cell environment can be individually adjusted. Mechanical obstacles or bead markers can be integrated into bioprinted samples to analyze mechanical deformations and forces within these bioprinted constructs, such as 3D organoids, and to perform biophysical analysis in complex 3D systems, which are still not standard techniques. The review highlights the advances of 3D and 4D printing technologies in integrating mechanobiological cues so that the next step will be a detailed analysis of key future biophysical research directions in organoid generation for the development of disease model systems, tissue regeneration and drug testing from a biophysical perspective. Finally, the review highlights the combination of bioprinted hydrogels, such as pure natural or synthetic hydrogels and mixtures, with organoids, organoid-cell co-cultures, organ-on-a-chip systems and organoid-organ-on-a chip combinations and introduces the use of assembloids to determine the mutual interactions of different cell types and cell-matrix interferences in specific biological and mechanical environments.
Collapse
Affiliation(s)
- Claudia Tanja Mierke
- Faculty of Physics and Earth System Science, Peter Debye Institute of Soft Matter Physics, Biological Physics Division, Leipzig University, 04103 Leipzig, Germany
| |
Collapse
|
18
|
Lambrecht S, Gazizova A, Kara S, Meyer J, Jopp S. Antimicrobial properties and biocompatibility of semi-synthetic carbohydrate-based ionic hydrogels. RSC Adv 2024; 14:30719-30731. [PMID: 39328876 PMCID: PMC11425042 DOI: 10.1039/d4ra05695g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 09/20/2024] [Indexed: 09/28/2024] Open
Abstract
Hydrogels have gained significant interest in the last decades, especially in the medical sector, due to their versatile properties. While hydrogels from naturally occurring polysaccharides (e.g. cellulose) are well-known, those produced from polymerizable carbohydrate-based monomers remain underexplored. However, these semi-synthetic hydrogels offer the great advantage of having adjustable properties for customization depending on their application. The objective of this study was to characterize semi-synthetic carbohydrate-based ionic hydrogels produced from GVIM-I (glucosyl vinyl imidazolium iodide). The antimicrobial activity was evaluated using the disk diffusion method, which demonstrated that all samples exhibit inhibitory effects on the growth of Candida auris. In vitro biocompatibility was determined by cell viability studies with L929 mouse fibroblasts, and a correlation was observed between eluate concentration and cell viability. In particular, the type of initiator system employed for polymerization was found to affect cell viability. The direct contact assessments showed that specific pre-treatments of the hydrogels resulted in higher cell viability than non-treated hydrogels. The results also revealed the impact of crosslinker concentration and type and identified poly(ethylene glycol)diacrylate (PEGDA) 575 as a promising crosslinker for future medical applications. LC-MS analysis of the wash medium identified unreacted GVIM-I as the leached material, which is presumed to be the cause of the observed cytotoxicity. Overall, the study provides valuable insights into the characteristics of GVIM-I based hydrogels and sheds light on the factors that influence their cytotoxicity and potential for medical application.
Collapse
Affiliation(s)
- Sina Lambrecht
- Department Life, Light & Matter, University of Rostock Albert-Einstein-Str. 25 18059 Rostock Germany
| | - Alina Gazizova
- Institute of Chemistry, University of Rostock Albert-Einstein-Str. 3a 18059 Rostock Germany
| | - Selin Kara
- Institute of Technical Chemistry, Leibniz University Hannover Callinstraße 5 30167 Hannover Germany
- Biocatalysis and Bioprocessing Group, Department of Biological and Chemical Engineering, Aarhus University Gustav Wieds Vej 10 8000 Aarhus Denmark
| | - Johanna Meyer
- Institute of Technical Chemistry, Leibniz University Hannover Callinstraße 5 30167 Hannover Germany
| | - Stefan Jopp
- Department Life, Light & Matter, University of Rostock Albert-Einstein-Str. 25 18059 Rostock Germany
| |
Collapse
|
19
|
Moghaddam AS, Dunne K, Breyer W, Wu Y, Pashuck ET. Hydrogels with Independently Controlled Adhesion Ligand Mobility and Viscoelasticity Increase Cell Adhesion and Spreading. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.23.614501. [PMID: 39386463 PMCID: PMC11463488 DOI: 10.1101/2024.09.23.614501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
A primary objective in designing hydrogels for cell culture is recreating the cell-matrix interactions found within human tissues. Identifying the most important biomaterial features for these interactions is challenging because it is difficult to independently adjust variables such as matrix stiffness, stress relaxation, the mobility of adhesion ligands and the ability of these ligands to support cellular forces. In this work we designed a hydrogel platform consisting of interpenetrating polymer networks of covalently crosslinked poly(ethylene glycol) (PEG) and self-assembled peptide amphiphiles (PA). We can tailor the storage modulus of the hydrogel by altering the concentration and composition of each network, and we can tune the stress relaxation half-life through the non-covalent bonding in the PA network. Ligand mobility can be adjusted independently of the matrix mechanical properties by attaching the RGD cell adhesion ligand to either the covalent PEG network, the dynamic PA network, or both networks at once. Interestingly, our findings show that endothelial cell adhesion formation and spreading is maximized in soft, viscoelastic gels in which RGD adhesion ligands are present on both the covalent PEG and non-covalent PA networks. The dynamic nature of cell adhesion domains, coupled with their ability to exert substantial forces on the matrix, suggests that having different presentations of RGD ligands which are either mobile or are capable of withstanding significant forces are needed mimic different aspects of complex cell-matrix adhesions. By demonstrating how different presentations of RGD ligands affect cell behavior independently of viscoelastic properties, these results contribute to the rational design of hydrogels that facilitate desired cell-matrix interactions, with the potential of improving in vitro models and regenerative therapies.
Collapse
Affiliation(s)
| | - Katelyn Dunne
- Department of Bioengineering, Lehigh University, Bethlehem PA, USA, 18015
| | - Wendy Breyer
- Department of Chemistry, Lehigh University, Bethlehem PA, USA, 18015
| | - Yingjie Wu
- Department of Bioengineering, Lehigh University, Bethlehem PA, USA, 18015
| | - E Thomas Pashuck
- Department of Bioengineering, Lehigh University, Bethlehem PA, USA, 18015
| |
Collapse
|
20
|
Cui H, Li P, Su Z, Guan S, Dong H, Dong X. Preparation and Stability Study of an Injectable Hydrogel for Artificial Intraocular Lenses. Polymers (Basel) 2024; 16:2562. [PMID: 39339025 PMCID: PMC11434676 DOI: 10.3390/polym16182562] [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: 07/26/2024] [Revised: 08/31/2024] [Accepted: 09/06/2024] [Indexed: 09/30/2024] Open
Abstract
Currently available intraocular lenses (IOLs) on the market often differ significantly in elastic modulus compared to the natural human lens, which impairs their ability to respond effectively to the tension of the ciliary muscles for focal adjustment after implantation. In this study, we synthesized a polyacrylamide-sodium acrylate hydrogel (PAH) through the cross-linking polymerization of acrylamide and sodium acrylate. This hydrogel possesses excellent biocompatibility and exhibits several favorable properties. Notably, the hydrogel demonstrates high transparency (94%) and a refractive index (1.41 ± 0.07) that closely matches that of the human lens (1.42). Additionally, it shows strong compressive strength (14.00 kPa), good extensibility (1400%), and an appropriate swelling ratio (50 ± 2.5%). Crucially, the tensile modulus of the hydrogel is 2.07 kPa, which closely aligns with the elastic modulus of the human lens (1.70-2.10 kPa), enabling continuous focal adjustment under the tension exerted by the ciliary muscles.
Collapse
Affiliation(s)
- Haifeng Cui
- School of Materials Science and Engineering, Dalian University of Technology, Dalian 116024, China; (H.C.); (P.L.); (Z.S.); (S.G.)
| | - Pengfei Li
- School of Materials Science and Engineering, Dalian University of Technology, Dalian 116024, China; (H.C.); (P.L.); (Z.S.); (S.G.)
| | - Zekun Su
- School of Materials Science and Engineering, Dalian University of Technology, Dalian 116024, China; (H.C.); (P.L.); (Z.S.); (S.G.)
| | - Shiqiang Guan
- School of Materials Science and Engineering, Dalian University of Technology, Dalian 116024, China; (H.C.); (P.L.); (Z.S.); (S.G.)
| | - He Dong
- Department Ophthalmology, The Third People’s Hospital of Dalian, Dalian 116033, China
| | - Xufeng Dong
- School of Materials Science and Engineering, Dalian University of Technology, Dalian 116024, China; (H.C.); (P.L.); (Z.S.); (S.G.)
| |
Collapse
|
21
|
Kang S, Woo Y, Seo Y, Yoo D, Kwon D, Park H, Lee SD, Yoo HY, Lee T. A Descriptive Review on the Potential Use of Diatom Biosilica as a Powerful Functional Biomaterial: A Natural Drug Delivery System. Pharmaceutics 2024; 16:1171. [PMID: 39339207 PMCID: PMC11434644 DOI: 10.3390/pharmaceutics16091171] [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: 07/24/2024] [Revised: 08/29/2024] [Accepted: 09/02/2024] [Indexed: 09/30/2024] Open
Abstract
Although various chemically synthesized materials are essential in medicine, food, and agriculture, they can exert unexpected side effects on the environment and human health by releasing certain toxic chemicals. Therefore, eco-friendly and biocompatible biomaterials based on natural resources are being actively explored. Recently, biosilica derived from diatoms has attracted attention in various biomedical fields, including drug delivery systems (DDS), due to its uniform porous nano-pattern, hierarchical structure, and abundant silanol functional groups. Importantly, the structural characteristics of diatom biosilica improve the solubility of poorly soluble substances and enable sustained release of loaded drugs. Additionally, diatom biosilica predominantly comprises SiO2, has high biocompatibility, and can easily hybridize with other DDS platforms, including hydrogels and cationic DDS, owing to its strong negative charge and abundant silanol groups. This review explores the potential applications of various diatom biosilica-based DDS in various biomedical fields, with a particular focus on hybrid DDS utilizing them.
Collapse
Affiliation(s)
- Sunggu Kang
- Department of Chemical Engineering, Kwangwoon University, 20 Kwangwoon-ro, Nowon-gu, Seoul 01897, Republic of Korea
| | - Yeeun Woo
- Department of Chemical Engineering, Kwangwoon University, 20 Kwangwoon-ro, Nowon-gu, Seoul 01897, Republic of Korea
| | - Yoseph Seo
- Department of Chemical Engineering, Kwangwoon University, 20 Kwangwoon-ro, Nowon-gu, Seoul 01897, Republic of Korea
| | - Daehyeon Yoo
- Department of Chemical Engineering, Kwangwoon University, 20 Kwangwoon-ro, Nowon-gu, Seoul 01897, Republic of Korea
| | - Daeryul Kwon
- Protist Research Division, Biological Resources Research Department, Nakdonggang National Institute of Biological Resources (NNIBR), 137, Donam 2-gil, Sangju-si 37242, Gyeongsangbuk-do, Republic of Korea
| | - Hyunjun Park
- Department of Chemical Engineering, Kwangwoon University, 20 Kwangwoon-ro, Nowon-gu, Seoul 01897, Republic of Korea
| | - Sang Deuk Lee
- Protist Research Division, Biological Resources Research Department, Nakdonggang National Institute of Biological Resources (NNIBR), 137, Donam 2-gil, Sangju-si 37242, Gyeongsangbuk-do, Republic of Korea
| | - Hah Young Yoo
- Department of Biotechnology, Sangmyung University, 20, Hongjimun 2-gil, Jongno-gu, Seoul 03016, Republic of Korea
| | - Taek Lee
- Department of Chemical Engineering, Kwangwoon University, 20 Kwangwoon-ro, Nowon-gu, Seoul 01897, Republic of Korea
| |
Collapse
|
22
|
Prieto-López L, Pereiro X, Vecino E. The mechanics of the retina: Müller glia role on retinal extracellular matrix and modelling. Front Med (Lausanne) 2024; 11:1393057. [PMID: 39296899 PMCID: PMC11410058 DOI: 10.3389/fmed.2024.1393057] [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: 03/04/2024] [Accepted: 08/13/2024] [Indexed: 09/21/2024] Open
Abstract
The retina is a highly heterogeneous tissue, both cell-wise but also regarding its extracellular matrix (ECM). The stiffness of the ECM is pivotal in retinal development and maturation and has also been associated with the onset and/or progression of numerous retinal pathologies, such as glaucoma, proliferative vitreoretinopathy (PVR), age-related macular degeneration (AMD), epiretinal membrane (ERM) formation or uveitis. Nonetheless, much remains unknown about the biomechanical milieu of the retina, and specifically the role that Müller glia play as principal mechanosensors and major producers of ECM constituents. So far, new approaches need to be developed to further the knowledge in the field of retinal mechanobiology for ECM-target applications to arise. In this review, we focus on the involvement of Müller glia in shaping and altering the retinal ECM under both physiological and pathological conditions and look into various biomaterial options to more accurately replicate the impact of matrix stiffness in vitro.
Collapse
Affiliation(s)
- Laura Prieto-López
- Experimental Ophthalmo-Biology Group, Department of Cell Biology and Histology, University of Basque Country UPV/EHU, Leioa, Spain
| | - Xandra Pereiro
- Experimental Ophthalmo-Biology Group, Department of Cell Biology and Histology, University of Basque Country UPV/EHU, Leioa, Spain
- Begiker-Ophthalmology Research Group, BioCruces Health Research Institute, Cruces Hospital, Barakaldo, Spain
| | - Elena Vecino
- Experimental Ophthalmo-Biology Group, Department of Cell Biology and Histology, University of Basque Country UPV/EHU, Leioa, Spain
- Begiker-Ophthalmology Research Group, BioCruces Health Research Institute, Cruces Hospital, Barakaldo, Spain
| |
Collapse
|
23
|
Wu H, Wang J, Fan W, Zhong Q, Xue R, Li S, Song Z, Tao Y. Eye of the future: Unlocking the potential utilization of hydrogels in intraocular lenses. Bioeng Transl Med 2024; 9:e10664. [PMID: 39553434 PMCID: PMC11561835 DOI: 10.1002/btm2.10664] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 03/13/2024] [Accepted: 03/27/2024] [Indexed: 11/19/2024] Open
Abstract
Hydrogels are distinguished by their exceptional ability to absorb and retain large volumes of water within their complex three-dimensional polymer networks, which is advantageous for the development of intraocular lenses (IOLs). Their innate hydrophilicity offers an optimal substrate for the fabrication of IOLs that simulate the natural lens' accommodation, thereby reducing irritation and facilitating healing after surgery. The swelling and water retention characteristics of hydrogels contribute to their notable biocompatibility and versatile mechanical properties. However, the clinical application of hydrogels faces challenges, including managing potential adverse postimplantation effects. Rigorous research is essential to ascertain the safety and effectiveness of hydrogels. This review systematically examines the prospects and constraints of hydrogels as innovative materials for IOLs. Our comprehensive analysis examines their inherent properties, various classification strategies, cross-linking processes, and sensitivity to external stimuli. Additionally, we thoroughly evaluate their interactions with ocular tissues, underscoring the potential for hydrogels to be refined into seamless and biologically integrated visual aids. We also discuss the anticipated technological progress and clinical uses of hydrogels in IOL manufacturing. With ongoing technological advancements, the promise of hydrogels is poised to evolve from concept to clinical reality, marking a significant leap forward in ophthalmology characterized by improved patient comfort, enhanced functionality, and reliable safety.
Collapse
Affiliation(s)
- Hao Wu
- Henan Eye Institute, Henan Eye Hospital, People's Hospital of Zhengzhou UniversityZhengzhouChina
- Zhengzhou University School of MedicineZhengzhouChina
| | - Jiale Wang
- Henan Eye Institute, Henan Eye Hospital, People's Hospital of Zhengzhou UniversityZhengzhouChina
- Zhengzhou University School of MedicineZhengzhouChina
| | - Wenhui Fan
- Henan Eye Institute, Henan Eye Hospital, People's Hospital of Zhengzhou UniversityZhengzhouChina
- Zhengzhou University School of MedicineZhengzhouChina
| | - Qi Zhong
- Henan Eye Institute, Henan Eye Hospital, People's Hospital of Zhengzhou UniversityZhengzhouChina
- Zhengzhou University School of MedicineZhengzhouChina
| | - Rongyue Xue
- Henan Eye Institute, Henan Eye Hospital, People's Hospital of Zhengzhou UniversityZhengzhouChina
- Zhengzhou University School of MedicineZhengzhouChina
| | - Siyu Li
- Henan Eye Institute, Henan Eye Hospital, People's Hospital of Zhengzhou UniversityZhengzhouChina
- Zhengzhou University School of MedicineZhengzhouChina
| | - Zongming Song
- Henan Eye Institute, Henan Eye Hospital, People's Hospital of Zhengzhou UniversityZhengzhouChina
| | - Ye Tao
- Henan Eye Institute, Henan Eye Hospital, People's Hospital of Zhengzhou UniversityZhengzhouChina
- Zhengzhou University School of MedicineZhengzhouChina
| |
Collapse
|
24
|
Li X, Sheng S, Li G, Hu Y, Zhou F, Geng Z, Su J. Research Progress in Hydrogels for Cartilage Organoids. Adv Healthc Mater 2024; 13:e2400431. [PMID: 38768997 DOI: 10.1002/adhm.202400431] [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: 02/04/2024] [Revised: 04/29/2024] [Indexed: 05/22/2024]
Abstract
The repair and regeneration of cartilage has always been a hot topic in medical research. Cartilage organoids (CORGs) are special cartilage tissue created using tissue engineering techniques outside the body. These engineered organoids tissues provide models that simulate the complex biological functions of cartilage, opening new possibilities for cartilage regenerative medicine and treatment strategies. However, it is crucial to establish suitable matrix scaffolds for the cultivation of CORGs. In recent years, utilizing hydrogel to culture stem cells and induce their differentiation into chondrocytes has emerged as a promising method for the in vitro construction of CORGs. In this review, the methods for establishing CORGs are summarized and an overview of the advantages and limitations of using matrigel in the cultivation of such organoids is provided. Furthermore, the importance of cartilage tissue ECM and alternative hydrogel substitutes for Matrigel, such as alginate, peptides, silk fibroin, and DNA derivatives is discussed, and the pros and cons of using these hydrogels for the cultivation of CORGs are outlined. Finally, the challenges and future directions in hydrogel research for CORGs are discussed. It is hoped that this article provides valuable references for the design and development of hydrogels for CORGs.
Collapse
Affiliation(s)
- Xiaolong Li
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Department of Orthopedics and Traumatology, Nanning Hospital of Traditional Chinese Medicine, Guangxi University of Chinese Medicine, Nanning, Guangxi, 530000, China
- Organoid Research Center, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
- School of Medicine, Shanghai University, Shanghai, 200444, China
- School of Life Sciences, Shanghai University, Shanghai, 200444, China
| | - Shihao Sheng
- Department of Orthopedics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Guangfeng Li
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Organoid Research Center, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
- Department of Orthopedics, Shanghai Zhongye Hospital, Shanghai, 200941, China
| | - Yan Hu
- Department of Orthopedics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Fengjin Zhou
- Department of Orthopedics, Honghui Hospital, Xi'an Jiao Tong University, Xi'an, 710000, China
| | - Zhen Geng
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Organoid Research Center, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Jiacan Su
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Organoid Research Center, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
- Department of Orthopedics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| |
Collapse
|
25
|
Blázquez-Carmona P, Ruiz-Mateos R, Barrasa-Fano J, Shapeti A, Martín-Alfonso JE, Domínguez J, Van Oosterwyck H, Reina-Romo E, Sanz-Herrera JA. Quantitative atlas of collagen hydrogels reveals mesenchymal cancer cell traction adaptation to the matrix nanoarchitecture. Acta Biomater 2024; 185:281-295. [PMID: 38992411 DOI: 10.1016/j.actbio.2024.07.002] [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: 02/26/2024] [Revised: 07/01/2024] [Accepted: 07/02/2024] [Indexed: 07/13/2024]
Abstract
Collagen-based hydrogels are commonly used in mechanobiology to mimic the extracellular matrix. A quantitative analysis of the influence of collagen concentration and properties on the structure and mechanics of the hydrogels is essential for tailored design adjustments for specific in vitro conditions. We combined focused ion beam scanning electron microscopy and rheology to provide a detailed quantitative atlas of the mechanical and nanoscale three-dimensional structural alterations that occur when manipulating different hydrogel's physicochemistry. Moreover, we study the effects of such alterations on the phenotype of breast cancer cells and their mechanical interactions with the extracellular matrix. Regardless of the microenvironment's pore size, porosity or mechanical properties, cancer cells are able to reach a stable mesenchymal-like morphology. Additionally, employing 3D traction force microscopy, a positive correlation between cellular tractions and ECM mechanics is observed up to a critical threshold, beyond which tractions plateau. This suggests that cancer cells in a stable mesenchymal state calibrate their mechanical interactions with the ECM to keep their migration and invasiveness capacities unaltered. STATEMENT OF SIGNIFICANCE: The paper presents a thorough study on the mechanical microenvironment in breast cancer cells during their interaction with collagen based hydrogels of different compositions. The hydrogels' microstructure were obtained using state-of-the-art 3D microscopy, namely focused ion beam-scanning electron microscope (FIB-SEM). FIB-SEM was originally applied in this work to reconstruct complex fibered collagen microstructures within the nanometer range, to obtain key microarchitectural parameters. The mechanical microenvironment of cells was recovered using Traction Force Microscopy (TFM). The obtained results suggest that cells calibrate tractions such that they depend on mechanical, microstructural and physicochemical characteristics of the hydrogels, hence revealing a steric hindrance. We hypothesize that cancer cells studied in this paper tune their mechanical state to keep their migration and invasiveness capacities unaltered.
Collapse
Affiliation(s)
- Pablo Blázquez-Carmona
- Escuela Técnica Superior de Ingeniería, Universidad de Sevilla. Avenida Camino de los Descubrimientos s/n, 41092 Sevilla, Spain; Instituto de Biomedicina de Sevilla (IBIS). C. Antonio Maura Montaner, 41013 Sevilla, Spain
| | - Raquel Ruiz-Mateos
- Escuela Técnica Superior de Ingeniería, Universidad de Sevilla. Avenida Camino de los Descubrimientos s/n, 41092 Sevilla, Spain; Instituto de Biomedicina de Sevilla (IBIS). C. Antonio Maura Montaner, 41013 Sevilla, Spain
| | - Jorge Barrasa-Fano
- Department of Mechanical Engineering, Biomechanics Section, KU Leuven, Celestijnenlaan 300. B-3001 Heverlee, Belgium
| | - Apeksha Shapeti
- Department of Mechanical Engineering, Biomechanics Section, KU Leuven, Celestijnenlaan 300. B-3001 Heverlee, Belgium
| | - José Enrique Martín-Alfonso
- Escuela Técnica Superior de Ingeniería, Universidad de Huelva. Avda. de las Fuerzas Armadas s/n, 21007 Huelva, Spain
| | - Jaime Domínguez
- Escuela Técnica Superior de Ingeniería, Universidad de Sevilla. Avenida Camino de los Descubrimientos s/n, 41092 Sevilla, Spain; Instituto de Biomedicina de Sevilla (IBIS). C. Antonio Maura Montaner, 41013 Sevilla, Spain
| | - Hans Van Oosterwyck
- Department of Mechanical Engineering, Biomechanics Section, KU Leuven, Celestijnenlaan 300. B-3001 Heverlee, Belgium
| | - Esther Reina-Romo
- Escuela Técnica Superior de Ingeniería, Universidad de Sevilla. Avenida Camino de los Descubrimientos s/n, 41092 Sevilla, Spain; Instituto de Biomedicina de Sevilla (IBIS). C. Antonio Maura Montaner, 41013 Sevilla, Spain
| | - José Antonio Sanz-Herrera
- Escuela Técnica Superior de Ingeniería, Universidad de Sevilla. Avenida Camino de los Descubrimientos s/n, 41092 Sevilla, Spain; Instituto de Biomedicina de Sevilla (IBIS). C. Antonio Maura Montaner, 41013 Sevilla, Spain.
| |
Collapse
|
26
|
Oliver-Cervelló L, López-Gómez P, Martin-Gómez H, Marion M, Ginebra MP, Mas-Moruno C. Functionalization of Alginate Hydrogels with a Multifunctional Peptide Supports Mesenchymal Stem Cell Adhesion and Reduces Bacterial Colonization. Chemistry 2024; 30:e202400855. [PMID: 39031737 DOI: 10.1002/chem.202400855] [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: 02/29/2024] [Revised: 06/07/2024] [Accepted: 06/19/2024] [Indexed: 07/22/2024]
Abstract
Hydrogels with cell adhesive moieties stand out as promising materials to enhance tissue healing and regeneration. Nonetheless, bacterial infections of the implants represent an unmet major concern. In the present work, we developed an alginate hydrogel modified with a multifunctional peptide containing the RGD cell adhesive motif in combination with an antibacterial peptide derived from the 1-11 region of lactoferrin (LF). The RGD-LF branched peptide was successfully anchored to the alginate backbone by carbodiimide chemistry, as demonstrated by 1H NMR and fluorescence measurements. The functionalized hydrogel presented desirable physicochemical properties (porosity, swelling and rheological behavior) to develop biomaterials for tissue engineering. The viability of mesenchymal stem cells (MSCs) on the peptide-functionalized hydrogels was excellent, with values higher than 85 % at day 1, and higher than 95 % after 14 days in culture. Moreover, the biological characterization demonstrated the ability of the hydrogels to significantly enhance ALP activity of MSCs as well as to decrease bacterial colonization of both Gram-positive and Gram-negative models. Such results prove the potential of the functionalized hydrogels as novel biomaterials for tissue engineering, simultaneously displaying cell adhesive activity and the capacity to prevent bacterial contamination, a dual bioactivity commonly not found for these types of hydrogels.
Collapse
Affiliation(s)
- Lluís Oliver-Cervelló
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Engineering, Universitat Politècnica de Catalunya (UPC), Barcelona, 08019, Spain
- Barcelona Research Center in Multiscale Science and Engineering, UPC, Barcelona, 08019, Spain
| | - Patricia López-Gómez
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Engineering, Universitat Politècnica de Catalunya (UPC), Barcelona, 08019, Spain
- Barcelona Research Center in Multiscale Science and Engineering, UPC, Barcelona, 08019, Spain
| | - Helena Martin-Gómez
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Engineering, Universitat Politècnica de Catalunya (UPC), Barcelona, 08019, Spain
- Barcelona Research Center in Multiscale Science and Engineering, UPC, Barcelona, 08019, Spain
| | - Mahalia Marion
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Engineering, Universitat Politècnica de Catalunya (UPC), Barcelona, 08019, Spain
- Barcelona Research Center in Multiscale Science and Engineering, UPC, Barcelona, 08019, Spain
| | - Maria-Pau Ginebra
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Engineering, Universitat Politècnica de Catalunya (UPC), Barcelona, 08019, Spain
- Barcelona Research Center in Multiscale Science and Engineering, UPC, Barcelona, 08019, Spain
- Centro de Investigación Biomédica en Red, Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Madrid, 28029, Spain
- Institute for Bioengineering of Catalonia (IBEC), Barcelona, 08028, Spain
| | - Carlos Mas-Moruno
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Engineering, Universitat Politècnica de Catalunya (UPC), Barcelona, 08019, Spain
- Barcelona Research Center in Multiscale Science and Engineering, UPC, Barcelona, 08019, Spain
- Centro de Investigación Biomédica en Red, Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Madrid, 28029, Spain
| |
Collapse
|
27
|
Vuorenpää H, Valtonen J, Penttinen K, Koskimäki S, Hovinen E, Ahola A, Gering C, Parraga J, Kelloniemi M, Hyttinen J, Kellomäki M, Aalto-Setälä K, Miettinen S, Pekkanen-Mattila M. Gellan gum-gelatin based cardiac models support formation of cellular networks and functional cardiomyocytes. Cytotechnology 2024; 76:483-502. [PMID: 38933872 PMCID: PMC11196475 DOI: 10.1007/s10616-024-00630-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 04/06/2024] [Indexed: 06/28/2024] Open
Abstract
Cardiovascular diseases remain as the most common cause of death worldwide. To reveal the underlying mechanisms in varying cardiovascular diseases, in vitro models with cells and supportive biomaterial can be designed to recapitulate the essential components of human heart. In this study, we analyzed whether 3D co-culture of cardiomyocytes (CM) with vascular network and with adipose tissue-derived mesenchymal stem/stromal cells (ASC) can support CM functionality. CM were cultured with either endothelial cells (EC) and ASC or with only ASC in hydrazide-modified gelatin and oxidized gellan gum hybrid hydrogel to form cardiovascular multiculture and myocardial co-culture, respectively. We studied functional characteristics of CM in two different cellular set-ups and analyzed vascular network formation, cellular morphology and orientation. The results showed that gellan gum-gelatin hydrogel supports formation of two different cellular networks and functional CM. We detected formation of a modest vascular network in cardiovascular multiculture and extensive ASC-derived alpha smooth muscle actin -positive cellular network in multi- and co-culture. iPSC-CM showed elongated morphology, partly aligned orientation with the formed networks and presented normal calcium transients, beating rates, and contraction and relaxation behavior in both setups. These 3D cardiac models provide promising platforms to study (patho) physiological mechanisms of cardiovascular diseases. Supplementary Information The online version contains supplementary material available at 10.1007/s10616-024-00630-5.
Collapse
Affiliation(s)
- Hanna Vuorenpää
- Adult Stem Cell Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Tays Research Services, Wellbeing Services County of Pirkanmaa, Tampere University Hospital, Tampere, Finland
| | - Joona Valtonen
- Heart Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Kirsi Penttinen
- Heart Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Sanna Koskimäki
- Adult Stem Cell Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Tays Research Services, Wellbeing Services County of Pirkanmaa, Tampere University Hospital, Tampere, Finland
- Heart Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Emma Hovinen
- Adult Stem Cell Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Tays Research Services, Wellbeing Services County of Pirkanmaa, Tampere University Hospital, Tampere, Finland
- Heart Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Antti Ahola
- Computational Biophysics and Imaging Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Christine Gering
- Biomaterials and Tissue Engineering Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Jenny Parraga
- Biomaterials and Tissue Engineering Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Minna Kelloniemi
- Department of Plastic and Reconstructive Surgery, Tampere University Hospital, Tampere, Finland
| | - Jari Hyttinen
- Computational Biophysics and Imaging Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Minna Kellomäki
- Biomaterials and Tissue Engineering Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Katriina Aalto-Setälä
- Heart Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Heart Hospital, Tampere University Hospital, Tampere, Finland
| | - Susanna Miettinen
- Adult Stem Cell Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Tays Research Services, Wellbeing Services County of Pirkanmaa, Tampere University Hospital, Tampere, Finland
| | - Mari Pekkanen-Mattila
- Heart Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| |
Collapse
|
28
|
Pipis N, Stewart KA, Tabatabaei M, Williams LN, Allen JB. Exploring the Fibrous Nature of Single-Stranded DNA-Collagen Complexes: Nanostructural Observations and Physicochemical Insights. ACS OMEGA 2024; 9:32052-32058. [PMID: 39072094 PMCID: PMC11270544 DOI: 10.1021/acsomega.4c04104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 06/26/2024] [Accepted: 07/08/2024] [Indexed: 07/30/2024]
Abstract
Nucleic acid-collagen complexes (NACCs) are a self-assembled biomimetic fibrillary platform arising from the spontaneous complexation of single-stranded DNA (ssDNA) oligonucleotides and collagen. NACCs merge the extracellular matrix functionality of collagen with the tunable bioactivity of ssDNA as aptamers for broad biomedical applications. We hypothesize that NACCs offer a hierarchical architecture across multiple length scales that significantly varies compared to native collagen. We investigate this using atomic force microscopy and electron microscopy (transmission electron microscopy and cryogenic electron microscopy). Results demonstrate key topographical differences induced by adding ssDNA oligonucleotides to collagen type I. NACCs form a dense network of intertwined collagen fiber bundles in the microscale and nanoscale while retaining their characteristic D-band periodicities (∼67 nm). Additionally, our exploration of thermodynamic parameters governing the interaction indicates an entropically favorable NACC formation driven by ssDNA. Thermal analysis demonstrates the preservation of collagen's triple helical domains and a more stabilized polypeptide structure at higher temperatures than native collagen. These findings offer important insights into our understanding of the ssDNA-induced complexation of collagen toward the further establishment of structure-property relationships in NACCs and their future development into practical biomaterials. They also provide pathways for manipulating and enhancing collagenous matrices' properties without requiring complex chemical modifications or fabrication procedures.
Collapse
Affiliation(s)
- Nikolaos Pipis
- J.
Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, Florida 32611, United States
| | - Kevin A. Stewart
- George
& Josephine Butler Polymer Research Laboratory, Department of
Chemistry, Center for Macromolecular Science and Engineering, University of Florida, Gainesville, Florida 32611, United States
| | - Mohammad Tabatabaei
- J.
Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, Florida 32611, United States
| | - Lakiesha N. Williams
- J.
Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, Florida 32611, United States
| | - Josephine B. Allen
- J.
Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, Florida 32611, United States
- Department
of Materials Science & Engineering, University of Florida, Gainesville, Florida 32611, United States
| |
Collapse
|
29
|
Buriti BMADB, Figueiredo PLB, Passos MF, da Silva JKR. Polymer-Based Wound Dressings Loaded with Essential Oil for the Treatment of Wounds: A Review. Pharmaceuticals (Basel) 2024; 17:897. [PMID: 39065747 PMCID: PMC11279661 DOI: 10.3390/ph17070897] [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: 05/27/2024] [Revised: 07/03/2024] [Accepted: 07/03/2024] [Indexed: 07/28/2024] Open
Abstract
Wound healing can result in complex problems, and discovering an effective method to improve the healing process is essential. Polymeric biomaterials have structures similar to those identified in the extracellular matrix of the tissue to be regenerated and also avoid chronic inflammation, and immunological reactions. To obtain smart and effective dressings, bioactive agents, such as essential oils, are also used to promote a wide range of biological properties, which can accelerate the healing process. Therefore, we intend to explore advances in the potential for applying hybrid materials in wound healing. For this, fifty scientific articles dated from 2010 to 2023 were investigated using the Web of Science, Scopus, Science Direct, and PubMed databases. The principles of the healing process, use of polymers, type and properties of essential oils and processing techniques, and characteristics of dressings were identified. Thus, the plants Syzygium romanticum or Eugenia caryophyllata, Origanum vulgare, and Cinnamomum zeylanicum present prospects for application in clinical trials due to their proven effects on wound healing and reducing the incidence of inflammatory cells in the site of injury. The antimicrobial effect of essential oils is mainly due to polyphenols and terpenes such as eugenol, cinnamaldehyde, carvacrol, and thymol.
Collapse
Affiliation(s)
- Bruna Michele A. de B. Buriti
- Instituto de Ciências Exatas e Naturais, Programa de Pós-Graduação em Química, Universidade Federal do Pará, Belém 66075-110, PA, Brazil;
| | - Pablo Luis B. Figueiredo
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal do Pará, Belém 66079-420, PA, Brazil; (P.L.B.F.); (M.F.P.)
| | - Marcele Fonseca Passos
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal do Pará, Belém 66079-420, PA, Brazil; (P.L.B.F.); (M.F.P.)
- Programa de Pós-Graduação em Biotecnologia, Universidade Federal do Pará, Belém 66075-110, PA, Brazil
| | - Joyce Kelly R. da Silva
- Instituto de Ciências Exatas e Naturais, Programa de Pós-Graduação em Química, Universidade Federal do Pará, Belém 66075-110, PA, Brazil;
- Programa de Pós-Graduação em Biotecnologia, Universidade Federal do Pará, Belém 66075-110, PA, Brazil
| |
Collapse
|
30
|
Rybachuk O, Nesterenko Y, Zhovannyk V. Modern advances in spinal cord regeneration: hydrogel combined with neural stem cells. Front Pharmacol 2024; 15:1419797. [PMID: 38994202 PMCID: PMC11236698 DOI: 10.3389/fphar.2024.1419797] [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: 04/18/2024] [Accepted: 06/11/2024] [Indexed: 07/13/2024] Open
Abstract
Severe spinal cord injuries (SCI) lead to loss of functional activity of the body below the injury site, affect a person's ability to self-care and have a direct impact on performance. Due to the structural features and functional role of the spinal cord in the body, the consequences of SCI cannot be completely overcome at the expense of endogenous regenerative potential and, developing over time, lead to severe complications years after injury. Thus, the primary task of this type of injury treatment is to create artificial conditions for the regenerative growth of damaged nerve fibers through the area of the SCI. Solving this problem is possible using tissue neuroengineering involving the technology of replacing the natural tissue environment with synthetic matrices (for example, hydrogels) in combination with stem cells, in particular, neural/progenitor stem cells (NSPCs). This approach can provide maximum stimulation and support for the regenerative growth of axons of damaged neurons and their myelination. In this review, we consider the currently available options for improving the condition after SCI (use of NSC transplantation or/and replacement of the damaged area of the SCI with a matrix, specifically a hydrogel). We emphasise the expediency and effectiveness of the hydrogel matrix + NSCs complex system used for the reconstruction of spinal cord tissue after injury. Since such a complex approach (a combination of tissue engineering and cell therapy), in our opinion, allows not only to creation of conditions for supporting endogenous regeneration or mechanical reconstruction of the spinal cord, but also to strengthen endogenous regeneration, prevent the spread of the inflammatory process, and promote the restoration of lost reflex, motor and sensory functions of the injured area of spinal cord.
Collapse
Affiliation(s)
- Oksana Rybachuk
- Bogomoletz Institute of Physiology NAS of Ukraine, Kyiv, Ukraine
- Institute of Genetic and Regenerative Medicine, M. D. Strazhesko National Scientific Center of Cardiology, Clinical and Regenerative Medicine, National Academy of Medical Sciences of Ukraine, Kyiv, Ukraine
| | | | | |
Collapse
|
31
|
Wachendörfer M, Palkowitz AL, Fischer H. Development of a biofabricated 3D in vitrovessel model for investigating transendothelial migration in stem cell therapy. Biofabrication 2024; 16:035028. [PMID: 38810632 DOI: 10.1088/1758-5090/ad51a5] [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: 01/09/2024] [Accepted: 05/29/2024] [Indexed: 05/31/2024]
Abstract
Systemic stem cell therapies hold promise for treating severe diseases, but their efficiency is hampered by limited migration of injected stem cells across vascular endothelium towards diseased tissues. Understanding transendothelial migration is crucial for improving therapy outcomes. We propose a novel 3Din vitrovessel model that aids to unravel these mechanisms and thereby facilitates stem cell therapy development. Our model simulates inflammation through cytokine diffusion from the tissue site into the vessel. It consists of a biofabricated vessel embedded in a fibrin hydrogel, mimicking arterial wall composition with smooth muscle cells and fibroblasts. The perfusable channel is lined with a functional endothelium which expresses vascular endothelial cadherin, provides an active barrier function, aligns with flow direction and is reconstructed byin situtwo-photon-microscopy. Inflammatory cytokine release (tumor necrosis factorα, stromal-derived factor (1) is demonstrated in both a transwell assay and the 3D model. In proof-of-principle experiments, mesoangioblasts, known as a promising candidate for a stem cell therapy against muscular dystrophies, are injected into the vessel model, showing shear-resistant endothelial adhesion under capillary-like flow conditions. Our 3Din vitromodel offers significant potential to study transendothelial migration mechanisms of stem cells, facilitating the development of improved stem cell therapies.
Collapse
Affiliation(s)
- Mattis Wachendörfer
- Department of Dental Materials and Biomaterials Research, RWTH Aachen University Hospital, Pauwelsstrasse 30, 52074 Aachen, Germany
| | - Alena Lisa Palkowitz
- Department of Dental Materials and Biomaterials Research, RWTH Aachen University Hospital, Pauwelsstrasse 30, 52074 Aachen, Germany
| | - Horst Fischer
- Department of Dental Materials and Biomaterials Research, RWTH Aachen University Hospital, Pauwelsstrasse 30, 52074 Aachen, Germany
| |
Collapse
|
32
|
Gerrits L, Bakker B, Hendriks LD, Engels S, Hammink R, Kouwer PHJ. Tailoring of Physical Properties in Macroporous Poly(isocyanopeptide) Cryogels. Biomacromolecules 2024; 25:3464-3474. [PMID: 38743442 PMCID: PMC11170948 DOI: 10.1021/acs.biomac.4c00086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 05/01/2024] [Accepted: 05/02/2024] [Indexed: 05/16/2024]
Abstract
Over the years, synthetic hydrogels have proven remarkably useful as cell culture matrixes to elucidate the role of the extracellular matrix (ECM) on cell behavior. Yet, their lack of interconnected macropores undermines the widespread use of hydrogels in biomedical applications. To overcome this limitation, cryogels, a class of macroporous hydrogels, are rapidly emerging. Here, we introduce a new, highly elastic, and tunable synthetic cryogel, based on poly(isocyanopeptides) (PIC). Introduction of methacrylate groups on PIC facilitated cryopolymerization through free-radical polymerization and afforded cryogels with an interconnected macroporous structure. We investigated which cryogelation parameters can be used to tune the architectural and mechanical properties of the PIC cryogels by systematically altering cryopolymerization temperature, polymer concentration, and polymer molecular weight. We show that for decreasing cryopolymerization temperatures, there is a correlation between cryogel pore size and stiffness. More importantly, we demonstrate that by simply varying the polymer concentration, we can selectively tune the compressive strength of PIC cryogels without affecting their architecture. This unique feature is highly useful for biomedical applications, as it facilitates decoupling of stiffness from other variables such as pore size. As such, PIC cryogels provide an interesting new biomaterial for scientists to unravel the role of the ECM in cellular functions.
Collapse
Affiliation(s)
- Lotte Gerrits
- Institute
for Molecules and Materials, Radboud University, Heyendaalseweg 135, 6525 AJ Nijmegen, The Netherlands
- Institute
for Chemical Immunology, 6525 GA Nijmegen ,Netherlands
| | - Bram Bakker
- Institute
for Molecules and Materials, Radboud University, Heyendaalseweg 135, 6525 AJ Nijmegen, The Netherlands
- Institute
for Chemical Immunology, 6525 GA Nijmegen ,Netherlands
| | - Lynn D. Hendriks
- Institute
for Molecules and Materials, Radboud University, Heyendaalseweg 135, 6525 AJ Nijmegen, The Netherlands
- Institute
for Chemical Immunology, 6525 GA Nijmegen ,Netherlands
| | - Sjoerd Engels
- Institute
for Molecules and Materials, Radboud University, Heyendaalseweg 135, 6525 AJ Nijmegen, The Netherlands
- Institute
for Chemical Immunology, 6525 GA Nijmegen ,Netherlands
| | - Roel Hammink
- Department
of Medical BioSciences,Radboudumc, Geert Grooteplein 26, 6525 GA Nijmegen, The Netherlands
- Division
of Immunotherapy, Oncode Institute, Radboud
University Medical Center, 6525 GA Nijmegen ,Netherlands
| | - Paul H. J. Kouwer
- Institute
for Molecules and Materials, Radboud University, Heyendaalseweg 135, 6525 AJ Nijmegen, The Netherlands
- Institute
for Chemical Immunology, 6525 GA Nijmegen ,Netherlands
| |
Collapse
|
33
|
Peng Y, Liang S, Meng QF, Liu D, Ma K, Zhou M, Yun K, Rao L, Wang Z. Engineered Bio-Based Hydrogels for Cancer Immunotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2313188. [PMID: 38362813 DOI: 10.1002/adma.202313188] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 02/01/2024] [Indexed: 02/17/2024]
Abstract
Immunotherapy represents a revolutionary paradigm in cancer management, showcasing its potential to impede tumor metastasis and recurrence. Nonetheless, challenges including limited therapeutic efficacy and severe immune-related side effects are frequently encountered, especially in solid tumors. Hydrogels, a class of versatile materials featuring well-hydrated structures widely used in biomedicine, offer a promising platform for encapsulating and releasing small molecule drugs, biomacromolecules, and cells in a controlled manner. Immunomodulatory hydrogels present a unique capability for augmenting immune activation and mitigating systemic toxicity through encapsulation of multiple components and localized administration. Notably, hydrogels based on biopolymers have gained significant interest owing to their biocompatibility, environmental friendliness, and ease of production. This review delves into the recent advances in bio-based hydrogels in cancer immunotherapy and synergistic combinatorial approaches, highlighting their diverse applications. It is anticipated that this review will guide the rational design of hydrogels in the field of cancer immunotherapy, fostering clinical translation and ultimately benefiting patients.
Collapse
Affiliation(s)
- Yuxuan Peng
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Shuang Liang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Qian-Fang Meng
- Institute of Biomedical Health Technology and Engineering, Shenzhen Bay Laboratory, Shenzhen, 518132, China
| | - Dan Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Kongshuo Ma
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Mengli Zhou
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Kaiqing Yun
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Lang Rao
- Institute of Biomedical Health Technology and Engineering, Shenzhen Bay Laboratory, Shenzhen, 518132, China
| | - Zhaohui Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| |
Collapse
|
34
|
De Lauretis A, Øvrebø Ø, Romandini M, Lyngstadaas SP, Rossi F, Haugen HJ. From Basic Science to Clinical Practice: A Review of Current Periodontal/Mucogingival Regenerative Biomaterials. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2308848. [PMID: 38380549 PMCID: PMC11077667 DOI: 10.1002/advs.202308848] [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: 11/17/2023] [Revised: 01/23/2024] [Indexed: 02/22/2024]
Abstract
Periodontitis is a dysbiosis-driven inflammatory disease affecting the tooth-supporting tissues, characterized by their progressive resorption, which can ultimately lead to tooth loss. A step-wise therapeutic approach is employed for periodontitis. After an initial behavioral and non-surgical phase, intra-bony or furcation defects may be amenable to regenerative procedures. This review discusses the regenerative technologies employed for periodontal regeneration, highlighting the current limitations and future research areas. The search, performed on the MEDLINE database, has identified the available biomaterials, including biologicals (autologous platelet concentrates, hydrogels), bone grafts (pure or putty), and membranes. Biologicals and bone grafts have been critically analyzed in terms of composition, mechanism of action, and clinical applications. Although a certain degree of periodontal regeneration is predictable in intra-bony and class II furcation defects, complete defect closure is hardly achieved. Moreover, treating class III furcation defects remains challenging. The key properties required for functional regeneration are discussed, and none of the commercially available biomaterials possess all the ideal characteristics. Therefore, research is needed to promote the advancement of more effective and targeted regenerative therapies for periodontitis. Lastly, improving the design and reporting of clinical studies is suggested by strictly adhering to the Consolidated Standards of Reporting Trials (CONSORT) 2010 statement.
Collapse
Affiliation(s)
- Angela De Lauretis
- Department of Biomaterials, Institute of Clinical Dentistry, Faculty of DentistryUniversity of OsloOslo0455Norway
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta”Politecnico di MilanoMilan20133Italy
| | - Øystein Øvrebø
- Department of Biomaterials, Institute of Clinical Dentistry, Faculty of DentistryUniversity of OsloOslo0455Norway
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta”Politecnico di MilanoMilan20133Italy
| | - Mario Romandini
- Department of Periodontology, Institute of Clinical Dentistry, Faculty of DentistryUniversity of OsloOslo0455Norway
| | - Ståle Petter Lyngstadaas
- Department of Biomaterials, Institute of Clinical Dentistry, Faculty of DentistryUniversity of OsloOslo0455Norway
| | - Filippo Rossi
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta”Politecnico di MilanoMilan20133Italy
| | - Håvard Jostein Haugen
- Department of Biomaterials, Institute of Clinical Dentistry, Faculty of DentistryUniversity of OsloOslo0455Norway
| |
Collapse
|
35
|
Stachowiak M, Mlynarczyk DT, Dlugaszewska J. Wondrous Yellow Molecule: Are Hydrogels a Successful Strategy to Overcome the Limitations of Curcumin? Molecules 2024; 29:1757. [PMID: 38675577 PMCID: PMC11051891 DOI: 10.3390/molecules29081757] [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: 02/29/2024] [Revised: 04/06/2024] [Accepted: 04/07/2024] [Indexed: 04/28/2024] Open
Abstract
Curcumin is a natural compound with a great pharmaceutical potential that involves anticancer, anti-inflammatory, antioxidant, and neuroprotective activity. Unfortunately, its low bioavailability, instability, and poor water solubility significantly deteriorate its clinical use. Many attempts have been made to overcome this issue, and encapsulating curcumin in a hydrogel matrix may improve those properties. Hydrogel formulation is used in many drug delivery forms, including classic types and novel forms such as self-assembly systems or responsive to external factors. Reviewed studies confirmed better properties of hydrogel-stabilized curcumin in comparison to pure compound. The main enhanced characteristics were chemical stability, bioavailability, and water solubility, which enabled these systems to be tested for various diseases. These formulations were evaluated for wound healing properties, effectiveness in treating skin diseases, and anticancer and regenerative activity. Hydrogel formulation significantly improved biopharmaceutical properties, opening the opportunity to finally see curcumin as a clinically approved substance and unravel its therapeutic potential.
Collapse
Affiliation(s)
- Magdalena Stachowiak
- Chair and Department of Chemical Technology of Drugs, Poznan University of Medical Sciences, Rokietnicka 3, 60-806 Poznan, Poland
| | - Dariusz T. Mlynarczyk
- Chair and Department of Chemical Technology of Drugs, Poznan University of Medical Sciences, Rokietnicka 3, 60-806 Poznan, Poland
| | - Jolanta Dlugaszewska
- Chair and Department of Genetics and Pharmaceutical Microbiology, Poznan University of Medical Sciences, Rokietnicka 3, 60-806 Poznan, Poland
| |
Collapse
|
36
|
Martorana A, Lenzuni M, Contardi M, Palumbo FS, Cataldo S, Pettignano A, Catania V, Schillaci D, Summa M, Athanassiou A, Fiorica C, Bertorelli R, Pitarresi G. Schiff Base-Based Hydrogel Embedded with In Situ Generated Silver Nanoparticles Capped by a Hyaluronic Acid-Diethylenetriamine Derivative for Wound Healing Application. ACS APPLIED MATERIALS & INTERFACES 2024. [PMID: 38603548 DOI: 10.1021/acsami.4c00657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/13/2024]
Abstract
In this study, hydrogels were produced using a Schiff base reaction between two hyaluronic acid derivatives: one containing aldehyde groups (HA-Ald) and the other holding a diethylenetriamine with terminal amino groups (HA-DETA). The DETA portion promotes the in situ growth, complexation, and stabilization of silver nanoparticles (AgNPs), eliminating the need for external reducing agents. The reaction between HA-DETA and HA-Ald leads to the formation of imine bonds, which results in dynamically pH-responsive cross-linking. While the DETA capping ability helped in embedding the AgNPs, the on/off pH environmental responsivity of the hydrogel allows for a controlled and on-demand release of the drug, mainly when bacterial infections cause pH variation of the wound bed. The injectable hydrogels resulted in being highly compatible in contact with blood red cells, fibroblasts, and keratinocytes and capable of having a proliferative effect on an in vitro wound scratch model. The pH-responsive hydrogels showed proper antibacterial activity againstPseudomonas aeruginosaandStaphylococcus aureus, common bacterial strains presented in wound infections. Finally, in vivo wound model studies demonstrated an overall speeding up in the wound healing rate and advanced wound conditions in the experimental group treated with the hydrogels compared to control samples.
Collapse
Affiliation(s)
- Annalisa Martorana
- Department of Biological, Chemical, and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Via Archirafi 32, 90123 Palermo, Italy
| | - Martina Lenzuni
- Smart Materials, Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genova, Italy
| | - Marco Contardi
- Smart Materials, Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genova, Italy
- Department of Earth and Environmental Sciences (DISAT), University of Milan-Bicocca, Piazza della Scienza, 20126 Milan, Italy
| | - Fabio S Palumbo
- Department of Biological, Chemical, and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Via Archirafi 32, 90123 Palermo, Italy
| | - Salvatore Cataldo
- Department of Physics and Chemistry - Emilio Segrè, University of Palermo, Viale delle scienze, Ed. 17, 90128 Palermo, Italy
| | - Alberto Pettignano
- Department of Physics and Chemistry - Emilio Segrè, University of Palermo, Viale delle scienze, Ed. 17, 90128 Palermo, Italy
| | - Valentina Catania
- Department of Earth and Marine Sciences (DiSTeM), University of Palermo, Viale delle Scienze Ed. 16, 90128 Palermo, Italy
| | - Domenico Schillaci
- Department of Biological, Chemical, and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Via Archirafi 32, 90123 Palermo, Italy
| | - Maria Summa
- Translational Pharmacology, Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genova, Italy
| | | | - Calogero Fiorica
- Department of Biological, Chemical, and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Via Archirafi 32, 90123 Palermo, Italy
| | - Rosalia Bertorelli
- Translational Pharmacology, Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genova, Italy
| | - Giovanna Pitarresi
- Department of Biological, Chemical, and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Via Archirafi 32, 90123 Palermo, Italy
| |
Collapse
|
37
|
Abbadessa A, Ronca A, Salerno A. Integrating bioprinting, cell therapies and drug delivery towards in vivo regeneration of cartilage, bone and osteochondral tissue. Drug Deliv Transl Res 2024; 14:858-894. [PMID: 37882983 DOI: 10.1007/s13346-023-01437-1] [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] [Accepted: 09/18/2023] [Indexed: 10/27/2023]
Abstract
The biological and biomechanical functions of cartilage, bone and osteochondral tissue are naturally orchestrated by a complex crosstalk between zonally dependent cells and extracellular matrix components. In fact, this crosstalk involves biomechanical signals and the release of biochemical cues that direct cell fate and regulate tissue morphogenesis and remodelling in vivo. Three-dimensional bioprinting introduced a paradigm shift in tissue engineering and regenerative medicine, since it allows to mimic native tissue anisotropy introducing compositional and architectural gradients. Moreover, the growing synergy between bioprinting and drug delivery may enable to replicate cell/extracellular matrix reciprocity and dynamics by the careful control of the spatial and temporal patterning of bioactive cues. Although significant advances have been made in this direction, unmet challenges and open research questions persist. These include, among others, the optimization of scaffold zonality and architectural features; the preservation of the bioactivity of loaded active molecules, as well as their spatio-temporal release; the in vitro scaffold maturation prior to implantation; the pros and cons of each animal model and the graft-defect mismatch; and the in vivo non-invasive monitoring of new tissue formation. This work critically reviews these aspects and reveals the state of the art of using three-dimensional bioprinting, and its synergy with drug delivery technologies, to pattern the distribution of cells and/or active molecules in cartilage, bone and osteochondral engineered tissues. Most notably, this work focuses on approaches, technologies and biomaterials that are currently under in vivo investigations, as these give important insights on scaffold performance at the implantation site and its interaction/integration with surrounding tissues.
Collapse
Affiliation(s)
- Anna Abbadessa
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), IDIS Research Institute, Universidade de Santiago de Compostela, 15782, Santiago de Compostela, Spain.
- Department of Pharmacology, Pharmacy and Pharmaceutical Technology, School of Pharmacy, Universidade de Santiago de Compostela, Campus Vida, Santiago de Compostela, Spain.
| | - Alfredo Ronca
- Institute of Polymers, Composites and Biomaterials, National Research Council, 80125, Naples, Italy.
| | - Aurelio Salerno
- Department of Chemical, Materials and Production Engineering, University of Naples Federico II, 80125, Naples, Italy.
| |
Collapse
|
38
|
Hameed H, Faheem S, Paiva-Santos AC, Sarwar HS, Jamshaid M. A Comprehensive Review of Hydrogel-Based Drug Delivery Systems: Classification, Properties, Recent Trends, and Applications. AAPS PharmSciTech 2024; 25:64. [PMID: 38514495 DOI: 10.1208/s12249-024-02786-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 03/05/2024] [Indexed: 03/23/2024] Open
Abstract
As adaptable biomaterials, hydrogels have shown great promise in several industries, which include the delivery of drugs, engineering of tissues, biosensing, and regenerative medicine. These hydrophilic polymer three-dimensional networks have special qualities like increased content of water, soft, flexible nature, as well as biocompatibility, which makes it excellent candidates for simulating the extracellular matrix and promoting cell development and tissue regeneration. With an emphasis on their design concepts, synthesis processes, and characterization procedures, this review paper offers a thorough overview of hydrogels. It covers the various hydrogel material types, such as natural polymers, synthetic polymers, and hybrid hydrogels, as well as their unique characteristics and uses. The improvements in hydrogel-based platforms for controlled drug delivery are examined. It also looks at recent advances in bioprinting methods that use hydrogels to create intricate tissue constructions with exquisite spatial control. The performance of hydrogels is explored through several variables, including mechanical properties, degradation behaviour, and biological interactions, with a focus on the significance of customizing hydrogel qualities for particular applications. This review paper also offers insights into future directions in hydrogel research, including those that promise to advance the discipline, such as stimuli-responsive hydrogels, self-healing hydrogels, and bioactive hydrogels. Generally, the objective of this review paper is to provide readers with a detailed grasp of hydrogels and all of their potential uses, making it an invaluable tool for scientists and researchers studying biomaterials and tissue engineering.
Collapse
Affiliation(s)
- Huma Hameed
- Faculty of Pharmaceutical Sciences, University of Central Punjab, Lahore, 54000, Pakistan.
| | - Saleha Faheem
- 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
| | - Hafiz Shoaib Sarwar
- Faculty of Pharmaceutical Sciences, University of Central Punjab, Lahore, 54000, Pakistan
| | - Muhammad Jamshaid
- Faculty of Pharmaceutical Sciences, University of Central Punjab, Lahore, 54000, Pakistan
| |
Collapse
|
39
|
Wu Y, Gu X, Chen X, Cui Y, Jiang W, Liu B. Hydrogel: a new material for intravesical drug delivery after bladder cancer surgery. J Mater Chem B 2024; 12:2938-2949. [PMID: 38426380 DOI: 10.1039/d3tb02837b] [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: 03/02/2024]
Abstract
The standard treatment for non-muscle invasive bladder cancer (NMIBC) is transurethral resection of bladder tumor (TURBT). However, this procedure may miss small lesions or incompletely remove them, resulting in cancer recurrence or progression. As a result, intravesical instillation of chemotherapy or immunotherapy drugs is often used as an adjunctive treatment after TURBT to prevent cancer recurrence. In the traditional method, drugs are instilled into the patient's bladder through a urinary catheter under sterile conditions. However, this treatment exposes the bladder mucosa to the drug directly, leading to potential side effects like chemical cystitis. Furthermore, this treatment has several limitations, including a short drug retention period, susceptibility to urine dilution, low drug permeability, lack of targeted effect, and limited long-term clinical efficacy. Hydrogel, a polymer material with a high-water content, possesses solid elasticity and liquid fluidity, making it compatible with tissues and environmentally friendly. It exhibits great potential in various applications. One emerging use of hydrogels is in intravesical instillation. By employing hydrogels, drug dilution is minimized, and drug absorption, retention, and persistence in the bladder are enhanced due to the mucus-adhesive and flotation properties of hydrogel materials. Furthermore, hydrogels can improve drug permeability and offer targeting capabilities. This article critically examines the current applications and future prospects of hydrogels in the treatment of bladder cancer.
Collapse
Affiliation(s)
- Yalong Wu
- China-Japan Union Hospital of Jilin University, Jilin University, Changchun 130033, China.
| | - Xinquan Gu
- China-Japan Union Hospital of Jilin University, Jilin University, Changchun 130033, China.
| | - Xiaoxi Chen
- Nanozyme Medical Center, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China.
| | - Yongliang Cui
- China-Japan Union Hospital of Jilin University, Jilin University, Changchun 130033, China.
| | - Wei Jiang
- Nanozyme Medical Center, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China.
| | - Bin Liu
- China-Japan Union Hospital of Jilin University, Jilin University, Changchun 130033, China.
| |
Collapse
|
40
|
Chandran C, Santra M, Rubin E, Geary ML, Yam GHF. Regenerative Therapy for Corneal Scarring Disorders. Biomedicines 2024; 12:649. [PMID: 38540264 PMCID: PMC10967722 DOI: 10.3390/biomedicines12030649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 03/08/2024] [Accepted: 03/12/2024] [Indexed: 05/09/2024] Open
Abstract
The cornea is a transparent and vitally multifaceted component of the eye, playing a pivotal role in vision and ocular health. It has primary refractive and protective functions. Typical corneal dysfunctions include opacities and deformities that result from injuries, infections, or other medical conditions. These can significantly impair vision. The conventional challenges in managing corneal ailments include the limited regenerative capacity (except corneal epithelium), immune response after donor tissue transplantation, a risk of long-term graft rejection, and the global shortage of transplantable donor materials. This review delves into the intricate composition of the cornea, the landscape of corneal regeneration, and the multifaceted repercussions of scar-related pathologies. It will elucidate the etiology and types of dysfunctions, assess current treatments and their limitations, and explore the potential of regenerative therapy that has emerged in both in vivo and clinical trials. This review will shed light on existing gaps in corneal disorder management and discuss the feasibility and challenges of advancing regenerative therapies for corneal stromal scarring.
Collapse
Affiliation(s)
- Christine Chandran
- Corneal Regeneration Laboratory, Department of Ophthalmology, Mercy Vision Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA; (C.C.); (M.S.); (E.R.); (M.L.G.)
| | - Mithun Santra
- Corneal Regeneration Laboratory, Department of Ophthalmology, Mercy Vision Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA; (C.C.); (M.S.); (E.R.); (M.L.G.)
| | - Elizabeth Rubin
- Corneal Regeneration Laboratory, Department of Ophthalmology, Mercy Vision Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA; (C.C.); (M.S.); (E.R.); (M.L.G.)
| | - Moira L. Geary
- Corneal Regeneration Laboratory, Department of Ophthalmology, Mercy Vision Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA; (C.C.); (M.S.); (E.R.); (M.L.G.)
| | - Gary Hin-Fai Yam
- Corneal Regeneration Laboratory, Department of Ophthalmology, Mercy Vision Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA; (C.C.); (M.S.); (E.R.); (M.L.G.)
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
| |
Collapse
|
41
|
Li YY, Ji SF, Fu XB, Jiang YF, Sun XY. Biomaterial-based mechanical regulation facilitates scarless wound healing with functional skin appendage regeneration. Mil Med Res 2024; 11:13. [PMID: 38369464 PMCID: PMC10874556 DOI: 10.1186/s40779-024-00519-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 01/30/2024] [Indexed: 02/20/2024] Open
Abstract
Scar formation resulting from burns or severe trauma can significantly compromise the structural integrity of skin and lead to permanent loss of skin appendages, ultimately impairing its normal physiological function. Accumulating evidence underscores the potential of targeted modulation of mechanical cues to enhance skin regeneration, promoting scarless repair by influencing the extracellular microenvironment and driving the phenotypic transitions. The field of skin repair and skin appendage regeneration has witnessed remarkable advancements in the utilization of biomaterials with distinct physical properties. However, a comprehensive understanding of the underlying mechanisms remains somewhat elusive, limiting the broader application of these innovations. In this review, we present two promising biomaterial-based mechanical approaches aimed at bolstering the regenerative capacity of compromised skin. The first approach involves leveraging biomaterials with specific biophysical properties to create an optimal scarless environment that supports cellular activities essential for regeneration. The second approach centers on harnessing mechanical forces exerted by biomaterials to enhance cellular plasticity, facilitating efficient cellular reprogramming and, consequently, promoting the regeneration of skin appendages. In summary, the manipulation of mechanical cues using biomaterial-based strategies holds significant promise as a supplementary approach for achieving scarless wound healing, coupled with the restoration of multiple skin appendage functions.
Collapse
Affiliation(s)
- Ying-Ying Li
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Department, Chinese PLA General Hospital and PLA Medical College; PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing, 100048, China
| | - Shuai-Fei Ji
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Department, Chinese PLA General Hospital and PLA Medical College; PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing, 100048, China
| | - Xiao-Bing Fu
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Department, Chinese PLA General Hospital and PLA Medical College; PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing, 100048, China.
| | - Yu-Feng Jiang
- Department of Tissue Regeneration and Wound Repair, Chinese PLA General Hospital, Beijing, 100853, China.
| | - Xiao-Yan Sun
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Department, Chinese PLA General Hospital and PLA Medical College; PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing, 100048, China.
| |
Collapse
|
42
|
Taghizadeh S, Tayebi L, Akbarzadeh M, Lohrasbi P, Savardashtaki A. Magnetic hydrogel applications in articular cartilage tissue engineering. J Biomed Mater Res A 2024; 112:260-275. [PMID: 37750666 DOI: 10.1002/jbm.a.37620] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 09/02/2023] [Accepted: 09/11/2023] [Indexed: 09/27/2023]
Abstract
Articular cartilage defects afflict millions of individuals worldwide, presenting a significant challenge due to the tissue's limited self-repair capability and anisotropic nature. Hydrogel-based biomaterials have emerged as promising candidates for scaffold production in artificial cartilage construction, owing to their water-rich composition, biocompatibility, and tunable properties. Nevertheless, conventional hydrogels typically lack the anisotropic structure inherent to natural cartilage, impeding their clinical and preclinical applications. Recent advancements in tissue engineering (TE) have introduced magnetically responsive hydrogels, a type of intelligent hydrogel that can be remotely controlled using an external magnetic field. These innovative materials offer a means to create the desired anisotropic architecture required for successful cartilage TE. In this review, we first explore conventional techniques employed for cartilage repair and subsequently delve into recent breakthroughs in the application and utilization of magnetic hydrogels across various aspects of articular cartilage TE.
Collapse
Affiliation(s)
- Saeed Taghizadeh
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
- Pharmaceutical Science Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Lobat Tayebi
- Marquette University School of Dentistry, Milwaukee, Wisconsin, USA
| | - Majid Akbarzadeh
- Department of Internal Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Parvin Lohrasbi
- Department of Reproductive Biology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amir Savardashtaki
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
- Infertility Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
43
|
Ozhava D, Bektas C, Lee K, Jackson A, Mao Y. Human Mesenchymal Stem Cells on Size-Sorted Gelatin Hydrogel Microparticles Show Enhanced In Vitro Wound Healing Activities. Gels 2024; 10:97. [PMID: 38391427 PMCID: PMC10887759 DOI: 10.3390/gels10020097] [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: 12/22/2023] [Revised: 01/17/2024] [Accepted: 01/22/2024] [Indexed: 02/24/2024] Open
Abstract
The demand for innovative therapeutic interventions to expedite wound healing, particularly in vulnerable populations such as aging and diabetic patients, has prompted the exploration of novel strategies. Mesenchymal stem cell (MSC)-based therapy emerges as a promising avenue for treating acute and chronic wounds. However, its clinical application faces persistent challenges, notably the low survivability and limited retention time of engraftment in wound environments. Addressing this, a strategy to sustain the viability and functionality of human MSCs (hMSCs) in a graft-able format has been identified as crucial for advanced wound care. Hydrogel microparticles (HMPs) emerge as promising entities in the field of wound healing, showcasing versatile capabilities in delivering both cells and bioactive molecules/drugs. In this study, gelatin HMPs (GelMPs) were synthesized via an optimized mild processing method. GelMPs with distinct diameter sizes were sorted and characterized. The growth of hMSCs on GelMPs with various sizes was evaluated. The release of wound healing promoting factors from hMSCs cultured on different GelMPs were assessed using scratch wound assays and gene expression analysis. GelMPs with a size smaller than 100 microns supported better cell growth and cell migration compared to larger sizes (100 microns or 200 microns). While encapsulation of hMSCs in hydrogels has been a common route for delivering viable hMSCs, we hypothesized that hMSCs cultured on GelMPs are more robust than those encapsulated in hydrogels. To test this hypothesis, hMSCs were cultured on GelMPs or in the cross-linked methacrylated gelatin hydrogel (GelMA). Comparative analysis of growth and wound healing effects revealed that hMSCs cultured on GelMPs exhibited higher viability and released more wound healing activities in vitro. This observation highlights the potential of GelMPs, especially those with a size smaller than 100 microns, as a promising carrier for delivering hMSCs in wound healing applications, providing valuable insights for the optimization of advanced therapeutic strategies.
Collapse
Affiliation(s)
- Derya Ozhava
- Laboratory for Biomaterials Research, Department of Chemistry and Chemical Biology, Rutgers University, 145 Bevier Rd., Piscataway, NJ 08854, USA
- Department of Chemistry and Chemical Processing Technologies, Cumra Vocational School, Selcuk University, 42130 Konya, Turkey
| | - Cemile Bektas
- Laboratory for Biomaterials Research, Department of Chemistry and Chemical Biology, Rutgers University, 145 Bevier Rd., Piscataway, NJ 08854, USA
| | - Kathleen Lee
- Laboratory for Biomaterials Research, Department of Chemistry and Chemical Biology, Rutgers University, 145 Bevier Rd., Piscataway, NJ 08854, USA
| | - Anisha Jackson
- Laboratory for Biomaterials Research, Department of Chemistry and Chemical Biology, Rutgers University, 145 Bevier Rd., Piscataway, NJ 08854, USA
| | - Yong Mao
- Laboratory for Biomaterials Research, Department of Chemistry and Chemical Biology, Rutgers University, 145 Bevier Rd., Piscataway, NJ 08854, USA
| |
Collapse
|
44
|
Teer L, Yaddanapudi K, Chen J. Biophysical Control of the Glioblastoma Immunosuppressive Microenvironment: Opportunities for Immunotherapy. Bioengineering (Basel) 2024; 11:93. [PMID: 38247970 PMCID: PMC10813491 DOI: 10.3390/bioengineering11010093] [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: 11/01/2023] [Revised: 01/14/2024] [Accepted: 01/15/2024] [Indexed: 01/23/2024] Open
Abstract
GBM is the most aggressive and common form of primary brain cancer with a dismal prognosis. Current GBM treatments have not improved patient survival, due to the propensity for tumor cell adaptation and immune evasion, leading to a persistent progression of the disease. In recent years, the tumor microenvironment (TME) has been identified as a critical regulator of these pro-tumorigenic changes, providing a complex array of biomolecular and biophysical signals that facilitate evasion strategies by modulating tumor cells, stromal cells, and immune populations. Efforts to unravel these complex TME interactions are necessary to improve GBM therapy. Immunotherapy is a promising treatment strategy that utilizes a patient's own immune system for tumor eradication and has exhibited exciting results in many cancer types; however, the highly immunosuppressive interactions between the immune cell populations and the GBM TME continue to present challenges. In order to elucidate these interactions, novel bioengineering models are being employed to decipher the mechanisms of immunologically "cold" GBMs. Additionally, these data are being leveraged to develop cell engineering strategies to bolster immunotherapy efficacy. This review presents an in-depth analysis of the biophysical interactions of the GBM TME and immune cell populations as well as the systems used to elucidate the underlying immunosuppressive mechanisms for improving current therapies.
Collapse
Affiliation(s)
- Landon Teer
- Department of Bioengineering, University of Louisville, Louisville, KY 40292, USA;
| | - Kavitha Yaddanapudi
- Department of Microbiology and Immunology, University of Louisville, Louisville, KY 40202, USA
- Immuno-Oncology Program, Brown Cancer Center, Department of Medicine, University of Louisville, Louisville, KY 40202, USA
- Division of Immunotherapy, Department of Surgery, University of Louisville, Louisville, KY 40202, USA
| | - Joseph Chen
- Department of Bioengineering, University of Louisville, Louisville, KY 40292, USA;
| |
Collapse
|
45
|
Brebu M, Dumitriu RP, Pamfil D, Butnaru E, Stoleru E. Riboflavin mediated UV crosslinking of chitosan-gelatin cryogels for loading of hydrophobic bioactive compounds. Carbohydr Polym 2024; 324:121521. [PMID: 37985057 DOI: 10.1016/j.carbpol.2023.121521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 10/18/2023] [Accepted: 10/20/2023] [Indexed: 11/22/2023]
Abstract
Chitosan-gelatin cryogels with good loading capacity of hydrophobic compounds were successfully obtained by UV-induced crosslinking. Using riboflavin as photoinitiator was a suitable alternative to classical carbodiimide crosslinking in obtaining carrier matrices for bioactive hydrophobic compounds. Chitosan had a double role, acting both as a base polymer for the hydrogel network and as co-initiator in riboflavin photo-crosslinking. This co-initiator role of chitosan is due to its electron donor capacity, being well known as a Lewis base type macromolecule. The rheological behaviour of the chitosan-gelatin hydrogel precursor solutions was greatly influenced by riboflavin addition as well as by UV irradiation. As a consequence, the temperature of the sol-gel transition during cooling decreased to 25.5 °C. Compared with classical carbodiimide crosslinking, UV irradiation lead to gels with increased network stability, enhanced elastic behaviour, higher structural strength and almost total stress recovery yield (99 %), the latter indicating self-healing capacity. The cryogels manifested pH responsive swelling, this being highest at close to neutral pH of 7.4. Although hydrophilic in nature, the chitosan-gelatin cryogels crosslinked under the combined effect of riboflavin and UV exposure possess the necessary chemical functionality and morphology that allowed successful embedding of hydrophobic clove essential oil. This was loaded by immersion or fumigation and imparted antioxidant activity to the polymeric matrix.
Collapse
Affiliation(s)
- Mihai Brebu
- "Petru Poni" Institute of Macromolecular Chemistry, Grigore Ghica Voda Alley, 41 A, 700487, Iasi, Romania
| | - Raluca Petronela Dumitriu
- "Petru Poni" Institute of Macromolecular Chemistry, Grigore Ghica Voda Alley, 41 A, 700487, Iasi, Romania
| | - Daniela Pamfil
- "Petru Poni" Institute of Macromolecular Chemistry, Grigore Ghica Voda Alley, 41 A, 700487, Iasi, Romania
| | - Elena Butnaru
- "Petru Poni" Institute of Macromolecular Chemistry, Grigore Ghica Voda Alley, 41 A, 700487, Iasi, Romania
| | - Elena Stoleru
- "Petru Poni" Institute of Macromolecular Chemistry, Grigore Ghica Voda Alley, 41 A, 700487, Iasi, Romania.
| |
Collapse
|
46
|
Maeng SW, Park TY, Park Y, Yoon T, Jung YM, Cha HJ. Self-Healable Adhesive Hydrogel with a Preserved Underwater Adhesive Ability Based on Histidine-Zinc Coordination and a Bioengineered Hybrid Mussel Protein. Biomacromolecules 2024; 25:379-387. [PMID: 38108296 DOI: 10.1021/acs.biomac.3c01025] [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: 12/19/2023]
Abstract
Mussels are marine organisms that are capable of constructing an underwater adhesion between their bodies and rigid structures. It is well known that mussels achieve underwater adhesion through the presence of mussel adhesive proteins (MAPs) that contain high levels of 3,4-dihydroxyphenylalanine (DOPA). Although the extraordinary underwater adhesive properties of mussels are attributed to DOPA, its capacity to play a dual role in surface adhesion and internal cohesion is inherently limited. However, mussels employ a combination of chemical moieties, not just DOPA, along with anatomical components, such as plaque and byssus, in underwater adhesion. This also involves junction proteins that connect the plaque and byssus. In this study, a novel hybrid MAP was bioengineered via the fusion of the plaque protein (foot protein type 1) and the histidine-rich domain of the junction protein (foot protein type 4). To achieve direct adhesion underwater, the adhesive should maintain surface adhesion without disintegrating. Notably, the histidine-Zn-coordinated hybrid MAP hydrogel maintained a high surface adhesion ability even after cross-linking because of the preservation of its unoxidized and non-cross-linked DOPA moieties. The formulated adhesive hydrogel system based on the bioengineered hybrid MAP exhibited self-healing properties, owing to the reversible metal coordination bonds. The developed adhesive hydrogel exhibits outstanding levels of bulk adhesion in underwater environments, highlighting its potential as an effective adhesive biomaterial. Therefore, the introduction of histidine-rich domains into MAPs may be applied in various studies to formulate mussel-inspired adhesives with self-healing properties and to fully utilize the adhesive ability of DOPA.
Collapse
Affiliation(s)
- Seong-Woo Maeng
- Department of Chemical Engineering, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
| | - Tae Yoon Park
- Department of Chemical Engineering, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
| | - Yeonju Park
- Department of Chemistry, Institute for Molecular Science and Fusion Technology, Kangwon Radiation Convergence Research Support Center, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Taehee Yoon
- Department of Chemical Engineering, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
| | - Young Mee Jung
- Department of Chemistry, Institute for Molecular Science and Fusion Technology, Kangwon Radiation Convergence Research Support Center, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Hyung Joon Cha
- Department of Chemical Engineering, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
- Medical Science and Engineering, School of Convergence Science and Technology, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
| |
Collapse
|
47
|
Enoch K, Somasundaram AA. Rheological insights on Carboxymethyl cellulose hydrogels. Int J Biol Macromol 2023; 253:127481. [PMID: 37865366 DOI: 10.1016/j.ijbiomac.2023.127481] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 10/05/2023] [Accepted: 10/15/2023] [Indexed: 10/23/2023]
Abstract
Hydrogels are copiously studied for tissue engineering, drug delivery, and bone regeneration owing to their water content, mechanical strength, and elastic behaviour. The preparation of stable and mechanically strengthened hydrogels without using toxic crosslinkers and expensive approaches is immensely challenging. In this study, we prepared Carboxymethyl cellulose based hydrogels with different polymer concentration via a less expensive physical crosslinking approach without using any toxic crosslinkers and evaluated their mechanical strength. In this hydrogel system, the carbopol concentration was fixed at 1 wt/v% and the Carboxymethyl cellulose concentration was varied between 1 and 5 wt/v%. In this hydrogel system, Carbopol serves as the crosslinker to bridge Carboxymethyl cellulose polymer through hydrogen bonds. Rheological analysis was employed in assessing the mechanical properties of the prepared hydrogel, in particular, the viscoelastic behaviour of the hydrogels. The viscoelastic nature and mechanical strength of the hydrogels increased with an increase in the Carboxymethyl cellulose polymer concentration. Further, our results suggested that gels with Carboxymethyl cellulose concentration between 3 wt/v % and 4 wt/v % with yield stresses of 58.83 Pa and 81.47 Pa, respectively, are potential candidates for use in transdermal drug delivery. The prepared hydrogels possessed high thermal stability and retained their gel network structure even at 50 °C. These findings are beneficial for biomedical applications in transdermal drug delivery and tissue engineering owing to the biocompatibility, stability, and mechanical strength of the prepared hydrogels.
Collapse
Affiliation(s)
- Karolinekersin Enoch
- Soft Matter Laboratory, Department of Physics and Nanotechnology, SRM Institute of Science and Technology, Kattankulathur - 603203, Tamil Nadu, India
| | - Anbumozhi Angayarkanni Somasundaram
- Soft Matter Laboratory, Department of Physics and Nanotechnology, SRM Institute of Science and Technology, Kattankulathur - 603203, Tamil Nadu, India.
| |
Collapse
|
48
|
Ai Y, Dai F, Li W, Xu F, Yang H, Wu J, Yang K, Li L, Ai F, Song L. Photo-crosslinked bioactive BG/BMSCs@GelMA hydrogels for bone-defect repairs. Mater Today Bio 2023; 23:100882. [PMID: 38161508 PMCID: PMC10755535 DOI: 10.1016/j.mtbio.2023.100882] [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: 09/02/2023] [Revised: 11/05/2023] [Accepted: 11/22/2023] [Indexed: 01/03/2024] Open
Abstract
The clinical treatments of bone defects remain a challenge. Hydrogels containing bone marrow mesenchymal stem cells (BMSCs) are extensively used to bone regeneration because of excellent biocompatibility and hydrophilicity. However, the insufficient osteo-induction capacity of the BMSC-loaded hydrogels limits their clinical applications. In this study, bio-active glass (BG) and BMSCs were combined with gelatin methacryloyl (GelMA) to fabricate composite hydrogels via photo-crosslinking, and the regulation of bone regeneration was investigated. In vitro experiments showed that the BG/BMSCs@GelMA hydrogel had excellent cytocompatibility and promoted osteogenic differentiation in BMSCs. Furthermore, the BG/BMSCs@GelMA hydrogel was injected into critical-sized calvarial defects, and the results further confirmed its excellent angiogenetic and bone regeneration capacity. In addition, BG/BMSCs@GelMA promoted the polarization of macrophages towards the M2 phenotype. In summary, this novel composite hydrogel demonstrated remarkable potential for application in bone regeneration due to its immunomodulatory, excellent angiogenetic as well as osteo-induction capacity.
Collapse
Affiliation(s)
- Yufeng Ai
- Center of Stomatology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 33006, China
- The Institute of Periodontal Disease, Nanchang University, Nanchang, Jiangxi, 33006, China
- JXHC Key Laboratory of Periodontology (The Second Affiliated Hospital of Nanchang University), Nanchang, Jiangxi, 33006, China
- The Second Clinical Medical School, Nanchang University, Nanchang, Jiangxi, 33006, China
| | - Fang Dai
- Center of Stomatology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 33006, China
- The Institute of Periodontal Disease, Nanchang University, Nanchang, Jiangxi, 33006, China
- JXHC Key Laboratory of Periodontology (The Second Affiliated Hospital of Nanchang University), Nanchang, Jiangxi, 33006, China
| | - Wenfeng Li
- Center of Stomatology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 33006, China
- The Institute of Periodontal Disease, Nanchang University, Nanchang, Jiangxi, 33006, China
- JXHC Key Laboratory of Periodontology (The Second Affiliated Hospital of Nanchang University), Nanchang, Jiangxi, 33006, China
| | - Fancheng Xu
- Center of Stomatology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 33006, China
- The Institute of Periodontal Disease, Nanchang University, Nanchang, Jiangxi, 33006, China
- JXHC Key Laboratory of Periodontology (The Second Affiliated Hospital of Nanchang University), Nanchang, Jiangxi, 33006, China
- The Second Clinical Medical School, Nanchang University, Nanchang, Jiangxi, 33006, China
| | - Hanwen Yang
- The Second Clinical Medical School, Nanchang University, Nanchang, Jiangxi, 33006, China
| | - Jianxin Wu
- Center of Stomatology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 33006, China
- The Institute of Periodontal Disease, Nanchang University, Nanchang, Jiangxi, 33006, China
- JXHC Key Laboratory of Periodontology (The Second Affiliated Hospital of Nanchang University), Nanchang, Jiangxi, 33006, China
- The Second Clinical Medical School, Nanchang University, Nanchang, Jiangxi, 33006, China
| | - Kaiqiang Yang
- Center of Stomatology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 33006, China
- The Institute of Periodontal Disease, Nanchang University, Nanchang, Jiangxi, 33006, China
- JXHC Key Laboratory of Periodontology (The Second Affiliated Hospital of Nanchang University), Nanchang, Jiangxi, 33006, China
- The Second Clinical Medical School, Nanchang University, Nanchang, Jiangxi, 33006, China
| | - Li Li
- Center of Stomatology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 33006, China
- The Institute of Periodontal Disease, Nanchang University, Nanchang, Jiangxi, 33006, China
- JXHC Key Laboratory of Periodontology (The Second Affiliated Hospital of Nanchang University), Nanchang, Jiangxi, 33006, China
- The Second Clinical Medical School, Nanchang University, Nanchang, Jiangxi, 33006, China
| | - Fanrong Ai
- School of Advanced Manufacturing, Nanchang University, Nanchang, Jiangxi, 33006, China
| | - Li Song
- Center of Stomatology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 33006, China
- The Institute of Periodontal Disease, Nanchang University, Nanchang, Jiangxi, 33006, China
- JXHC Key Laboratory of Periodontology (The Second Affiliated Hospital of Nanchang University), Nanchang, Jiangxi, 33006, China
| |
Collapse
|
49
|
Monteiro LPG, Rodrigues JMM, Mano JF. In situ generated hemostatic adhesives: From mechanisms of action to recent advances and applications. BIOMATERIALS ADVANCES 2023; 155:213670. [PMID: 37952461 DOI: 10.1016/j.bioadv.2023.213670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 10/20/2023] [Accepted: 10/20/2023] [Indexed: 11/14/2023]
Abstract
Conventional surgical closure techniques, such as sutures, clips, or skin closure strips, may not always provide optimal wound closure and may require invasive procedures, which can result in potential post-surgical complications. As result, there is a growing demand for innovative solutions to achieve superior wound closure and improve patient outcomes. To overcome the abovementioned issues, in situ generated hemostatic adhesives/sealants have emerged as a promising alternative, offering a targeted, controllable, and minimally invasive procedure for a wide variety of medical applications. The aim of this review is to provide a comprehensive overview of the mechanisms of action and recent advances of in situ generated hemostatic adhesives, particularly protein-based, thermoresponsive, bioinspired, and photocrosslinkable formulations, as well as the design challenges that must be addressed. Overall, this review aims to enhance a comprehensive understanding of the latest advancements of in situ generated hemostatic adhesives and their mechanisms of action, with the objective of promoting further research in this field.
Collapse
Affiliation(s)
- Luís P G Monteiro
- Department of Chemistry, CICECO-Aveiro Institute of Materials, University of Aveiro, 3810-193 Aveiro, Portugal
| | - João M M Rodrigues
- Department of Chemistry, CICECO-Aveiro Institute of Materials, University of Aveiro, 3810-193 Aveiro, Portugal.
| | - João F Mano
- Department of Chemistry, CICECO-Aveiro Institute of Materials, University of Aveiro, 3810-193 Aveiro, Portugal.
| |
Collapse
|
50
|
Schneider KH, Goldberg BJ, Hasturk O, Mu X, Dötzlhofer M, Eder G, Theodossiou S, Pichelkastner L, Riess P, Rohringer S, Kiss H, Teuschl-Woller AH, Fitzpatrick V, Enayati M, Podesser BK, Bergmeister H, Kaplan DL. Silk fibroin, gelatin, and human placenta extracellular matrix-based composite hydrogels for 3D bioprinting and soft tissue engineering. Biomater Res 2023; 27:117. [PMID: 37978399 PMCID: PMC10656895 DOI: 10.1186/s40824-023-00431-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 09/18/2023] [Indexed: 11/19/2023] Open
Abstract
BACKGROUND There is a great clinical need and it remains a challenge to develop artificial soft tissue constructs that can mimic the biomechanical properties and bioactivity of natural tissue. This is partly due to the lack of suitable biomaterials. Hydrogels made from human placenta offer high bioactivity and represent a potential solution to create animal-free 3D bioprinting systems that are both sustainable and acceptable, as placenta is widely considered medical waste. A combination with silk and gelatin polymers can bridge the biomechanical limitations of human placenta chorion extracellular matrix hydrogels (hpcECM) while maintaining their excellent bioactivity. METHOD In this study, silk fibroin (SF) and tyramine-substituted gelatin (G-TA) were enzymatically crosslinked with human placental extracellular matrix (hpcECM) to produce silk-gelatin-ECM composite hydrogels (SGE) with tunable mechanical properties, preserved elasticity, and bioactive functions. The SGE composite hydrogels were characterized in terms of gelation kinetics, protein folding, and bioactivity. The cyto- and biocompatibility of the SGE composite was determined by in vitro cell culture and subcutaneous implantation in a rat model, respectively. The most cell-supportive SGE formulation was then used for 3-dimensional (3D) bioprinting that induced chemical crosslinking during extrusion. CONCLUSION Addition of G-TA improved the mechanical properties of the SGE composite hydrogels and inhibited crystallization and subsequent stiffening of SF for up to one month. SGE hydrogels exhibit improved and tunable biomechanical properties and high bioactivity for encapsulated cells. In addition, its use as a bioink for 3D bioprinting with free reversible embedding of suspended hydrogels (FRESH) has been validated, opening the possibility to fabricate highly complex scaffolds for artificial soft tissue constructs with natural biomechanics in future.
Collapse
Affiliation(s)
- Karl Heinrich Schneider
- Center for Biomedical Research and Translational Surgery, Medical University of Vienna, 1090, Vienna, Austria
- Ludwig Boltzmann Institute for Cardiovascular Research, Vienna, 1090, Austria
- Department of Biomedical Engineering, Tufts University, Medford, MA, 02155, USA
| | - Benjamin J Goldberg
- Department of Biomedical Engineering, Tufts University, Medford, MA, 02155, USA
| | - Onur Hasturk
- Department of Biomedical Engineering, Tufts University, Medford, MA, 02155, USA
| | - Xuan Mu
- Department of Biomedical Engineering, Tufts University, Medford, MA, 02155, USA
- Roy J Carver Department of Biomedical Engineering, College of Engineering, the University of Iowa, Iowa City, IA, 52242, USA
| | - Marvin Dötzlhofer
- Center for Biomedical Research and Translational Surgery, Medical University of Vienna, 1090, Vienna, Austria
| | - Gabriela Eder
- Center for Biomedical Research and Translational Surgery, Medical University of Vienna, 1090, Vienna, Austria
| | - Sophia Theodossiou
- Department of Biomedical Engineering, Tufts University, Medford, MA, 02155, USA
- Department of Mechanical and Biomedical Engineering, Boise State University, Boise, ID, 83725, USA
| | - Luis Pichelkastner
- Center for Biomedical Research and Translational Surgery, Medical University of Vienna, 1090, Vienna, Austria
| | - Peter Riess
- Center for Biomedical Research and Translational Surgery, Medical University of Vienna, 1090, Vienna, Austria
| | - Sabrina Rohringer
- Center for Biomedical Research and Translational Surgery, Medical University of Vienna, 1090, Vienna, Austria
| | - Herbert Kiss
- Department of Obstetrics and Gynecology, Division of Obstetrics and Feto-Maternal Medicine, Medical University of Vienna, 1090, Vienna, Austria
| | - Andreas H Teuschl-Woller
- Department Life Science Technologies, University of Applied Sciences Technikum Wien, 1200, Vienna, Austria
| | - Vincent Fitzpatrick
- Department of Biomedical Engineering, Tufts University, Medford, MA, 02155, USA
- UMR CNRS 7338 Biomechanics & Bioengineering, Université de Technologie de Compiègne, Sorbonne Universités, 60203, Compiegne, France
| | - Marjan Enayati
- Center for Biomedical Research and Translational Surgery, Medical University of Vienna, 1090, Vienna, Austria
- Ludwig Boltzmann Institute for Cardiovascular Research, Vienna, 1090, Austria
| | - Bruno K Podesser
- Center for Biomedical Research and Translational Surgery, Medical University of Vienna, 1090, Vienna, Austria
- Ludwig Boltzmann Institute for Cardiovascular Research, Vienna, 1090, Austria
| | - Helga Bergmeister
- Center for Biomedical Research and Translational Surgery, Medical University of Vienna, 1090, Vienna, Austria
- Ludwig Boltzmann Institute for Cardiovascular Research, Vienna, 1090, Austria
| | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, MA, 02155, USA.
| |
Collapse
|